1
|
Reischer A, Leutbecher A, Hiller B, Perini E, White K, Hernández-Cáceres A, Schele A, Tast B, Rohrbacher L, Winter L, Czogalla B, Mahner S, Flaswinkel H, Leonhardt H, Wyder L, Wichmann C, Maenner D, Trillsch F, Kessler M, Hopfner KP, Fenn NC, Subklewe M. Targeted CD47 checkpoint blockade using a mesothelin-directed antibody construct for enhanced solid tumor-specific immunotherapy. Cancer Immunol Immunother 2025; 74:214. [PMID: 40402266 PMCID: PMC12098241 DOI: 10.1007/s00262-025-04032-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/24/2025] [Indexed: 05/23/2025]
Abstract
The immune checkpoint CD47 is highly upregulated in several cancers as an innate immune escape mechanism. CD47 delivers a "don't eat me" signal to its co-receptor signal regulatory protein α (SIRPα), thereby inhibiting phagocytosis. Blocking the CD47-SIRPα axis is a promising immunotherapeutic strategy against cancer. However, early trial data has demonstrated on-target off-leukemia toxicity. In addition, the ubiquitous expression pattern of CD47 might contribute to an antigen sink. In this study, we combined low-affinity CD47 checkpoint blockade and specific tumor targeting in a multivalent and multifunctional antibody construct to prevent CD47-related toxicities. First, we established a local inhibitory checkpoint monoclonal antibody (LicMAb) by fusing two N-terminal extracellular domains of SIRPα to a full-length anti-human mesothelin (MSLN)-IgG1 antibody, a well-described tumor-associated antigen in epithelial ovarian cancer (EOC) and pancreatic ductal adenocarcinoma (PDAC). Next, we evaluated the SIRPα-αMSLN LicMAb for mediating a tumor-restricted immune response as observed by antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). Our data validates CD47 and MSLN as highly upregulated targets expressed on various solid cancer entities, particularly EOC. We show tumor-specific binding and CD47 blocking by the SIRPα-αMSLN LicMAb even in the presence of healthy CD47-expressing cells. Furthermore, the LicMAb induces NK-cell-mediated cytotoxicity and improves phagocytosis of EOC and PDAC tumor cells. Moreover, cell death in EOC-derived organoids was specifically LicMAb-driven. Hence, the SIRPα-αMSLN LicMAb combines a tumor-restricted blockade of the CD47-SIRPα axis with a specific antitumor response while preventing on-target off-tumor toxicities. Our data supports the multifunctional SIRPα-αMSLN LicMAb as a promising approach to treating solid tumors.
Collapse
Affiliation(s)
- Anna Reischer
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Alexandra Leutbecher
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Björn Hiller
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany
| | - Enrico Perini
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany
| | - Kieron White
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | | | - Alexandra Schele
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany
| | - Benjamin Tast
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Lisa Rohrbacher
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Lis Winter
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Bastian Czogalla
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Munich, Germany
| | - Heinrich Flaswinkel
- Faculty of Biology and Center for Molecular Biosystems (BioSysM), Human Biology and BioImaging, LMU Munich, Munich, Germany
| | - Heinrich Leonhardt
- Faculty of Biology and Center for Molecular Biosystems (BioSysM), Human Biology and BioImaging, LMU Munich, Munich, Germany
| | - Lorenza Wyder
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Denis Maenner
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Munich, Germany
| | - Mirjana Kessler
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Munich, Germany
| | | | - Nadja C Fenn
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany.
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany.
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany.
| |
Collapse
|
2
|
Ye YH, Xin HY, Li N, Luo CB, Chen L, Pan JY, Xu Y, Weng F, Tu CY, Ji YY, Fan J, Zhou J, Zhou ZJ, Zhou SL. The intratumoral balance of IgG4 + plasma cells and CD8 + T cells is associated with prognosis of intrahepatic cholangiocarcinoma after curative resection. Dig Liver Dis 2025:S1590-8658(25)00325-1. [PMID: 40307164 DOI: 10.1016/j.dld.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/13/2025] [Accepted: 04/06/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND The tumor microenvironment has shown abilities to influence the progression and prognosis of intrahepatic cholangiocarcinoma (iCCA). However, little is known about the effect of IgG4+ plasma cells in iCCA. METHODS We stained IgG4+plasma cells by immunohistochemistry and performed Kaplan-Meier survival analysis to detect the prognostic value. We also stained CD3, CD4, CD8 and Foxp3 positive T cells to explore its associations with IgG4+plasma cells. RESULTS We found that tumor infiltrated IgG4+plasma cells were associated with poor prognosis of iCCA, rather than IgG4+plasma cells in adjacent liver tissue. The number of IgG4+plasma cells was associated with CD8+ T cells. We divided the iCCA patients into 3 groups by IgG4+plasma cells to CD8+ T cells ratio. The group I (IgG4-/CD8+) had the best prognosis. The group II (IgG4-/CD8- or IgG4+/CD8+) and group III (IgG4+/CD8-) were independent risk factors of recurrence-free survival (HR = 1.69, group II; HR = 2.11, group III) and overall survival (HR = 1.76, group II; HR = 2.38, group III) compared with group I. CONCLUSIONS We examined the distribution of IgG4+ plasma cells in iCCA and discovered its prognostic utility when combined with CD8+ T cell distribution in iCCA.
Collapse
Affiliation(s)
- Yu-Hang Ye
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Hao-Yang Xin
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Ning Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chu-Bin Luo
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Long Chen
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Jing-Yue Pan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ye Xu
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Fan Weng
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Cun-Yang Tu
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Ya-Ya Ji
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jia Fan
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Jian Zhou
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China
| | - Zheng-Jun Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Shao-Lai Zhou
- Liver Surgery Department, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, China.
| |
Collapse
|
3
|
Fan TC, Yeo HL, Hung TH, Chang NC, Tang YH, Yu J, Chen SH, Yu AL. ST3GAL1 regulates cancer cell migration through crosstalk between EGFR and neuropilin-1 signaling. J Biol Chem 2025; 301:108368. [PMID: 40024474 PMCID: PMC11984587 DOI: 10.1016/j.jbc.2025.108368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025] Open
Abstract
Metastasis is a major cause of cancer-related morbidity and mortality. The overexpression of the sialyltransferase ST3GAL1 in breast cancer correlates with metastasis. However, the molecular mechanisms underlying the effect of ST3GAL1 on cell movement are poorly understood. We identified neuropilin-1/NRP1 as a substrate for ST3GAL1. Gene expression analysis revealed that recurrence-free survival (p = 0.0046) and distant metastasis-free survival (p = 0.0003) were significantly shorter in the ST3GAL1HighNRP1High cohort than in the both-low subgroup. We demonstrated that the ST3GAL1-mediated sialylation of NRP1 results in increased binding affinity toward EGFR at the molecular level. At the cellular level, ST3GAL1 silencing impaired cell migration and wound healing ability, which was linked to reduced activities of CAPN2 as a consequence of diminished EGF/EGFR signaling. These data establish a function for the ST3GAL1-mediated sialylation of NRP1, leading to increased EGF/EGFR downstream signaling and enhanced tumor cell motility. Furthermore, ST3GAL1 silencing augmented the sensitivity to cetuximab-mediated cell lysis. Our findings provide novel insight into the mechanisms underlying the function of ST3GAL1 in promoting tumor cell migration through the EGFR/NRP1 pathway. Our results suggest that ST3GAL1 may represent a valuable target for strategies aimed at inhibiting tumor migration.
Collapse
Affiliation(s)
- Tan-Chi Fan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Hui Ling Yeo
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Nai-Chuan Chang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Yun-Hsin Tang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan; Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Shih-Hsiang Chen
- Division of Hematology-Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan.
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan; Genomics Research Center, Academia Sinica, Taipei, Taiwan; Department of Pediatrics/Hematology Oncology, University of California in San Diego, San Diego, California, USA.
| |
Collapse
|
4
|
Hale G. Living in LALA land? Forty years of attenuating Fc effector functions. Immunol Rev 2024; 328:422-437. [PMID: 39158044 PMCID: PMC11659930 DOI: 10.1111/imr.13379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
The Fc region of antibodies is vital for most of their physiological functions, many of which are engaged through binding to a range of Fc receptors. However, these same interactions are not always helpful or wanted when therapeutic antibodies are directed against self-antigens, and can sometimes cause catastrophic adverse reactions. Over the past 40 years, there have been intensive efforts to "silence" unwanted binding to Fc-gamma receptors, resulting in at least 45 different variants which have entered clinical trials. One of the best known is "LALA" (L234A/L235A). However, neither this, nor most of the other variants in clinical use are completely silenced, and in addition, the biophysical properties of many of them are compromised. I review the development of different variants to see what we can learn from their biological properties and use in the clinic. With the rise of powerful new uses of antibody therapy such as bispecific T-cell engagers, antibody-drug conjugates, and checkpoint inhibitors, it is increasingly important to optimize the Fc region as well as the antibody binding site in order to achieve the best combination of safety and efficacy.
Collapse
|
5
|
Izadi A, Godzwon M, Söderlund Strand A, Schmidt T, Kumlien Georén S, Drosten C, Ohlin M, Nordenfelt P. Protective Non-neutralizing anti-N-terminal Domain mAb Maintains Fc-mediated Function against SARS-COV-2 Variants up to BA.2.86-JN.1 with Superfluous In Vivo Protection against JN.1 Due to Attenuated Virulence. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:678-689. [PMID: 39018495 PMCID: PMC11335326 DOI: 10.4049/jimmunol.2300675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/25/2024] [Indexed: 07/19/2024]
Abstract
Substantial evidence supports that Fc-mediated effector functions of anti-spike Abs contribute to anti-SARS-Cov-2 protection. We have previously shown that two non-neutralizing but opsonic mAbs targeting the receptor-binding domain and N-terminal domain (NTD), Ab81 and Ab94, respectively, are protective against lethal Wuhan SARS-CoV-2 infection in K18-hACE2 mice. In this article, we investigated whether these protective non-neutralizing Abs maintain Fc-mediated function and Ag binding against mutated SARS-CoV-2 variants. Ab81 and Ab94 retained their nanomolar affinity and Fc-mediated function toward Omicron and its subvariants, such as BA.2, BA.4, BA.5, XBB, XBB1.5, and BQ1.1. However, when encountering the more heavily mutated BA.2.86, Ab81 lost its function, whereas the 10 new mutations in the NTD did not affect Ab94. In vivo experiments with Ab94 in K18-hACE2 mice inoculated with a stringent dose of 100,000 PFU of the JN.1 variant revealed unexpected results. Surprisingly, this variant exhibited low disease manifestation in this animal model with no weight loss or death in the control group. Still, assessment of mice using a clinical scoring system showed better protection for Ab94-treated mice, indicating that Fc-mediated functions are still beneficial. Our work shows that a protective anti-receptor-binding domain non-neutralizing mAb lost reactivity when BA.2.86 emerged, whereas the anti-NTD mAb was still functional. Finally, this work adds new insight into the evolution of the SARS-CoV-2 virus by reporting that JN.1 is substantially less virulent in vivo than previous strains.
Collapse
Affiliation(s)
- Arman Izadi
- Department of Clinical Sciences Lund, Division of Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Anna Söderlund Strand
- Department of Laboratory Medicine, Clinical Microbiology, Skåne University Hospital Lund, Lund University, Lund, Sweden
| | - Tobias Schmidt
- Department of Clinical Sciences Lund, Division of Pediatrics, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Christian Drosten
- German Center for Infection Research, Berlin, Germany
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mats Ohlin
- Department of Immunotechnology, Lund University, Lund, Sweden
- SciLifeLab Drug Discovery and Development, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Department of Clinical Sciences Lund, Division of Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Laboratory Medicine, Clinical Microbiology, Skåne University Hospital Lund, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Zeng J, Fang Y, Zhang Z, Lv Z, Wang X, Huang Q, Tian Z, Li J, Xu W, Zhu W, Yu J, Liu T, Qian Q. Antitumor activity of Z15-0-2, a bispecific nanobody targeting PD-1 and CTLA-4. Oncogene 2024; 43:2244-2252. [PMID: 38806619 PMCID: PMC11245388 DOI: 10.1038/s41388-024-03066-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
The combination of programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) antibodies has potential for enhancing clinical efficacy. We described the development and antitumor activity of Z15-0, a bispecific nanobody targeting both the PD-1 and CTLA-4 pathways simultaneously. We designed and optimized the mRNA sequence encoding Z15-0, referred to as Z15-0-2 and through a series of in vitro and in vivo experiments, we established that the optimized Z15-0-2 mRNA sequence significantly increased the expression of the bispecific nanobody. Administration of Z15-0-2 mRNA to tumor-bearing mice led to greater inhibition of tumor growth compared to controls. In aggregate, we introduced a novel bispecific nanobody and have re-engineered it to boost expression of mRNA, representing a new drug development paradigm.
Collapse
Affiliation(s)
- Jianyao Zeng
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yuan Fang
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Zixuan Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhenzhen Lv
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Xiaodie Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Qian Huang
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Zhidan Tian
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Jiaguo Li
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Wenfeng Xu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Weimin Zhu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Jing Yu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Tao Liu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China.
| | - Qijun Qian
- School of Medicine, Shanghai University, Shanghai, 200444, China.
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China.
- Shanghai Mengchao Cancer Hospital, Shanghai University, Shanghai, 201805, China.
| |
Collapse
|
7
|
Izadi A, Karami Y, Bratanis E, Wrighton S, Khakzad H, Nyblom M, Olofsson B, Happonen L, Tang D, Sundwall M, Godzwon M, Chao Y, Toledo AG, Schmidt T, Ohlin M, Nilges M, Malmström J, Bahnan W, Shannon O, Malmström L, Nordenfelt P. The hinge-engineered IgG1-IgG3 hybrid subclass IgGh 47 potently enhances Fc-mediated function of anti-streptococcal and SARS-CoV-2 antibodies. Nat Commun 2024; 15:3600. [PMID: 38678029 PMCID: PMC11055898 DOI: 10.1038/s41467-024-47928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
Streptococcus pyogenes can cause invasive disease with high mortality despite adequate antibiotic treatments. To address this unmet need, we have previously generated an opsonic IgG1 monoclonal antibody, Ab25, targeting the bacterial M protein. Here, we engineer the IgG2-4 subclasses of Ab25. Despite having reduced binding, the IgG3 version promotes stronger phagocytosis of bacteria. Using atomic simulations, we show that IgG3's Fc tail has extensive movement in 3D space due to its extended hinge region, possibly facilitating interactions with immune cells. We replaced the hinge of IgG1 with four different IgG3-hinge segment subclasses, IgGhxx. Hinge-engineering does not diminish binding as with IgG3 but enhances opsonic function, where a 47 amino acid hinge is comparable to IgG3 in function. IgGh47 shows improved protection against S. pyogenes in a systemic infection mouse model, suggesting that IgGh47 has promise as a preclinical therapeutic candidate. Importantly, the enhanced opsonic function of IgGh47 is generalizable to diverse S. pyogenes strains from clinical isolates. We generated IgGh47 versions of anti-SARS-CoV-2 mAbs to broaden the biological applicability, and these also exhibit strongly enhanced opsonic function compared to the IgG1 subclass. The improved function of the IgGh47 subclass in two distant biological systems provides new insights into antibody function.
Collapse
Affiliation(s)
- Arman Izadi
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Yasaman Karami
- Université de Lorraine, CNRS, Inria, LORIA, F-54000, Nancy, France
- Institut Pasteur, Université Paris cite, CNRS UMR3528, Structural Bioinformatics Unit, Department of Structural Biology and Chemistry, F-75015, Paris, France
| | - Eleni Bratanis
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sebastian Wrighton
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Hamed Khakzad
- Université de Lorraine, CNRS, Inria, LORIA, F-54000, Nancy, France
| | - Maria Nyblom
- Department of Biology & Lund Protein Production Platform (LP3), Lund University, Lund, Sweden
| | - Berit Olofsson
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lotta Happonen
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Di Tang
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Martin Sundwall
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Magdalena Godzwon
- Department of Immunotechnology and SciLifeLab Drug Discovery and Development Platform, Lund University, Lund, Sweden
| | - Yashuan Chao
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Alejandro Gomez Toledo
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Tobias Schmidt
- Department of Clinical Sciences Lund, Division of Pediatrics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Mats Ohlin
- Department of Immunotechnology and SciLifeLab Drug Discovery and Development Platform, Lund University, Lund, Sweden
| | - Michael Nilges
- Institut Pasteur, Université Paris cite, CNRS UMR3528, Structural Bioinformatics Unit, Department of Structural Biology and Chemistry, F-75015, Paris, France
| | - Johan Malmström
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Wael Bahnan
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Oonagh Shannon
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Lars Malmström
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden.
- Department of Laboratory Medicine, Clinical Microbiology, Skåne University Hospital Lund, Lund University, Lund, Sweden.
| |
Collapse
|
8
|
Lu Q, Yang D, Li H, Zhu Z, Zhang Z, Chen Y, Yang N, Li J, Wang Z, Niu T, Tong A. Delivery of CD47-SIRPα checkpoint blocker by BCMA-directed UCAR-T cells enhances antitumor efficacy in multiple myeloma. Cancer Lett 2024; 585:216660. [PMID: 38266806 DOI: 10.1016/j.canlet.2024.216660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/02/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
In the treatment of relapsed or refractory multiple myeloma patients, BCMA-directed autologous CAR-T cells have showed excellent anti-tumor activity. However, their widespread application is limited due to the arguably cost and time-consuming. Multiple myeloma cells highly expressed CD47 molecule and interact with the SIRPα ligand on the surface of macrophages, in which evade the clearance of macrophages through the activation of "don't eat me" signal. In this study, a BCMA-directed universal CAR-T cells, BC404-UCART, secreting a CD47-SIRPα blocker was developed using CRISPR/Cas9 gene-editing system. BC404-UCART cells significantly inhibited tumor growth and prolonged the survival of mice in the xenograft model. The anti-tumor activity of BC404-UCART cells was achieved via two mechanisms, on the one hand, the UCAR-T cells directly killed tumor cells, on the other hand, the BC404-UCART cells enhanced the phagocytosis of macrophages by secreting anti-CD47 nanobody hu404-hfc fusion that blocked the "don't eat me" signal between macrophages and tumor cells, which provides a potential strategy for the development of novel "off-the-shelf" cellular immunotherapies for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Qizhong Lu
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Donghui Yang
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, 712100, China
| | - Hexian Li
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhixiong Zhu
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yongdong Chen
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nian Yang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jia Li
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zeng Wang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Thaller AL, Jönsson F, Fiquet O, Marie S, Doisne JM, Girelli-Zubani G, Eri T, Fernandes P, Tatirovsky E, Langa-Vives F, Bruhns P, Strick-Marchand H, Di Santo JP. A human immune system (HIS) mouse model that dissociates roles for mouse and human FcR + cells during antibody-mediated immune responses. Eur J Immunol 2023; 53:e2350454. [PMID: 37621208 DOI: 10.1002/eji.202350454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Human immune system (HIS) mice provide a model to study human immune responses in vivo. Currently available HIS mouse models may harbor mouse Fc Receptor (FcR)-expressing cells that exert potent effector functions following administration of human Ig. Previous studies showed that the ablation of the murine FcR gamma chain (FcR-γ) results in loss of antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis in vivo. We created a new FcR-γ-deficient HIS mouse model to compare host (mouse) versus graft (human) effects underlying antibody-mediated immune responses in vivo. FcR-γ-deficient HIS recipients lack expression and function of mouse activating FcRs and can be stably and robustly reconstituted with human immune cells. By screening blood B-cell depletion by rituximab Ig variants, we found that human FcγRs-mediated IgG1 effects, whereas mouse activating FcγRs were dominant in IgG4 effects. Complement played a role as an IgG1 variant (IgG1 K322A) lacking complement binding activity was largely ineffective. Finally, we provide evidence that FcγRIIIA on human NK cells could mediate complement-independent B-cell depletion by IgG1 K322A. We anticipate that our FcR-γ-deficient HIS model will help clarify mechanisms of action of exogenous administered human antibodies in vivo.
Collapse
Affiliation(s)
- Anna Louisa Thaller
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Antibodies in Therapy and Pathology Unit, Université Paris Cité, Inserm U1222, Paris, France
| | - Oriane Fiquet
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Solenne Marie
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Jean-Marc Doisne
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Giulia Girelli-Zubani
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Toshiki Eri
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Priyanka Fernandes
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Evgeny Tatirovsky
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Francina Langa-Vives
- Institut Pasteur, Mouse Genetics Engineering Platform, Université Paris Cité, Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Antibodies in Therapy and Pathology Unit, Université Paris Cité, Inserm U1222, Paris, France
| | - Hélène Strick-Marchand
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| |
Collapse
|
10
|
Rispens T, Huijbers MG. The unique properties of IgG4 and its roles in health and disease. Nat Rev Immunol 2023; 23:763-778. [PMID: 37095254 PMCID: PMC10123589 DOI: 10.1038/s41577-023-00871-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
IgG4 is the least abundant subclass of IgG in human serum and has unique functional features. IgG4 is largely unable to activate antibody-dependent immune effector responses and, furthermore, undergoes Fab (fragment antigen binding)-arm exchange, rendering it bispecific for antigen binding and functionally monovalent. These properties of IgG4 have a blocking effect, either on the immune response or on the target protein of IgG4. In this Review, we discuss the unique structural characteristics of IgG4 and how these contribute to its roles in health and disease. We highlight how, depending on the setting, IgG4 responses can be beneficial (for example, in responses to allergens or parasites) or detrimental (for example, in autoimmune diseases, in antitumour responses and in anti-biologic responses). The development of novel models for studying IgG4 (patho)physiology and understanding how IgG4 responses are regulated could offer insights into novel treatment strategies for these IgG4-associated disease settings.
Collapse
Affiliation(s)
- Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Maartje G Huijbers
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
11
|
Heterogeneity and Functions of Tumor-Infiltrating Antibody Secreting Cells: Lessons from Breast, Ovarian, and Other Solid Cancers. Cancers (Basel) 2022; 14:cancers14194800. [PMID: 36230721 PMCID: PMC9563085 DOI: 10.3390/cancers14194800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary B cells are gaining increasing recognition as important contributors to the tumor microenvironment, influencing, positively or negatively, tumor growth, patient survival, and response to therapies. Antibody secreting cells (ASCs) constitute a variable fraction of tumor-infiltrating B cells in most solid tumors, and they produce tumor-specific antibodies that can drive distinct immune responses depending on their isotypes and specificities. In this review, we discuss the current knowledge of the heterogeneity of ASCs infiltrating solid tumors and how both their canonical and noncanonical functions shape antitumor immunity, with a special emphasis on breast and ovarian cancers. Abstract Neglected for a long time in cancer, B cells and ASCs have recently emerged as critical actors in the tumor microenvironment, with important roles in shaping the antitumor immune response. ASCs indeed exert a major influence on tumor growth, patient survival, and response to therapies. The mechanisms underlying their pro- vs. anti-tumor roles are beginning to be elucidated, revealing the contributions of their secreted antibodies as well as of their emerging noncanonical functions. Here, concentrating mostly on ovarian and breast cancers, we summarize the current knowledge on the heterogeneity of tumor-infiltrating ASCs, we discuss their possible local or systemic origin in relation to their immunoglobulin repertoire, and we review the different mechanisms by which antibody (Ab) subclasses and isoforms differentially impact tumor cells and anti-tumor immunity. We also discuss the emerging roles of cytokines and other immune modulators produced by ASCs in cancer. Finally, we propose strategies to manipulate the tumor ASC compartment to improve cancer therapies.
Collapse
|
12
|
Minici C, Testoni S, Della-Torre E. B-Lymphocytes in the Pathophysiology of Pancreatic Adenocarcinoma. Front Immunol 2022; 13:867902. [PMID: 35359944 PMCID: PMC8963963 DOI: 10.3389/fimmu.2022.867902] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic adenocarcinoma is highly infiltrated by B lymphocytes but the relevance of these immune cells in tumor development has been surprisingly overlooked until recently. Based on available evidence from other solid tumors, interaction between B lymphocytes and neoplastic cells is probably not uniformly stimulatory or inhibitory. Although presentation of tumor antigens to T cells and production of antitumor immunoglobulins might intuitively suggest a prominent tumor suppressive activity, specific subsets of B lymphocytes can secrete growth factors for neoplastic cells and immunosuppressive cytokines thus promoting escape from immunosurveillance and cancer progression. Because many of these mechanisms might also be implicated in the development of PDAC, and immune-modulation of B-cell activity is nowadays possible at different levels, determining the role of B-lymphocytes in this lethal cancer becomes of utmost importance to design novel therapeutic strategies. This review aims to discuss the emerging role of B cells in PDAC tumorigenesis, progression, and associated stromal reaction.
Collapse
Affiliation(s)
- Claudia Minici
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sabrina Testoni
- Pancreato-Biliary Endoscopy and Endosonography Division, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuel Della-Torre
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
13
|
Jin Y, Schladetsch MA, Huang X, Balunas MJ, Wiemer AJ. Stepping forward in antibody-drug conjugate development. Pharmacol Ther 2022; 229:107917. [PMID: 34171334 PMCID: PMC8702582 DOI: 10.1016/j.pharmthera.2021.107917] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 01/03/2023]
Abstract
Antibody-drug conjugates (ADCs) are cancer therapeutic agents comprised of an antibody, a linker and a small-molecule payload. ADCs use the specificity of the antibody to target the toxic payload to tumor cells. After intravenous administration, ADCs enter circulation, distribute to tumor tissues and bind to the tumor surface antigen. The antigen then undergoes endocytosis to internalize the ADC into tumor cells, where it is transported to lysosomes to release the payload. The released toxic payloads can induce apoptosis through DNA damage or microtubule inhibition and can kill surrounding cancer cells through the bystander effect. The first ADC drug was approved by the United States Food and Drug Administration (FDA) in 2000, but the following decade saw no new approved ADC drugs. From 2011 to 2018, four ADC drugs were approved, while in 2019 and 2020 five more ADCs entered the market. This demonstrates an increasing trend for the clinical development of ADCs. This review summarizes the recent clinical research, with a specific focus on how the in vivo processing of ADCs influences their design. We aim to provide comprehensive information about current ADCs to facilitate future development.
Collapse
Affiliation(s)
- Yiming Jin
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Megan A Schladetsch
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Xueting Huang
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Marcy J Balunas
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Andrew J Wiemer
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
14
|
Dash R, Singh SK, Chirmule N, Rathore AS. Assessment of Functional Characterization and Comparability of Biotherapeutics: a Review. AAPS J 2021; 24:15. [PMID: 34931298 DOI: 10.1208/s12248-021-00671-0] [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: 09/15/2021] [Accepted: 11/30/2021] [Indexed: 11/30/2022] Open
Abstract
The development of monoclonal antibody (mAb) biosimilars is a complex process. The key to their successful development and commercialization is an in-depth understanding of the key product attributes that impact safety and efficacy and the strategies to control them. Functional assessment of mAb is a crucial part of the comparability of biopharmaceutical drugs. The development of a relevant and robust functional assay requires an interdisciplinary approach and sufficient flexibility to balance regulatory concerns as well as dynamics and variability during the manufacturing process. Although many advanced tools are available to study and compare the potency and bioactivity of the protein, most of these techniques suffer from major shortcomings that limit their routine use. These include the complexity of the task, establishment of the relevance of the chosen method with the mechanism of action (MOA) of the biosimilar, cost and extended time of analysis, and often the ambiguity in interpretation of the resulting data. To overcome or to address these challenges, the use of multiple orthogonal state-of-the-art techniques is a necessary prerequisite.
Collapse
Affiliation(s)
- Rozaleen Dash
- Department of Chemical Engineering, DBT Center of Excellence for Biopharmaceutical Technology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Sumit Kumar Singh
- Department of Chemical Engineering, DBT Center of Excellence for Biopharmaceutical Technology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.,School of Biochemical Engineering, IIT-BHU, Varanasi, India
| | | | - Anurag S Rathore
- Department of Chemical Engineering, DBT Center of Excellence for Biopharmaceutical Technology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
15
|
Development of a novel humanized mouse model for improved evaluation of in vivo anti-cancer effects of anti-PD-1 antibody. Sci Rep 2021; 11:21087. [PMID: 34702924 PMCID: PMC8548333 DOI: 10.1038/s41598-021-00641-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer in the clinic. Further discovery of novel drugs or therapeutic protocols that enhance efficacy requires reliable animal models that recapitulate human immune responses to ICI treatment in vivo. In this study, we utilized an immunodeficient NOG mouse substrain deficient for mouse FcγR genes, NOG-FcγR−/− mice, to evaluate the anti-cancer effects of nivolumab, an anti-programmed cell death-1 (PD-1) antibody. After reconstitution of human immune systems by human hematopoietic stem cell transplantation (huNOG-FcγR−/− mice), four different programmed death-ligand 1 (PD-L1)-positive human cancer cell lines were tested. Among them, the growth of three cell lines was strongly suppressed by nivolumab in huNOG-FcγR−/− mice, but not in conventional huNOG mice. Accordingly, immunohistochemistry demonstrated the enhanced infiltration of human T cells into tumor parenchyma in only nivolumab-treated huNOG-FcγR−/− mice. Consistently, the number of human T cells was increased in the spleen in huNOG-FcγR−/− mice by nivolumab but not in huNOG mice. Furthermore, human PD-L1 expression was strongly induced in the spleen of huNOG-FcγR−/− mice. Collectively, our results suggest that the anti-cancer effects of anti-PD-1 antibodies can be detected more clearly in NOG-FcγR−/− mice than in NOG mice.
Collapse
|
16
|
Zirngibl F, Ivasko SM, Grunewald L, Klaus A, Schwiebert S, Ruf P, Lindhofer H, Astrahantseff K, Andersch L, Schulte JH, Lode HN, Eggert A, Anders K, Hundsdoerfer P, Künkele A. GD2-directed bispecific trifunctional antibody outperforms dinutuximab beta in a murine model for aggressive metastasized neuroblastoma. J Immunother Cancer 2021; 9:jitc-2021-002923. [PMID: 34285106 PMCID: PMC8292814 DOI: 10.1136/jitc-2021-002923] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background Neuroblastoma is the most common extracranial solid tumor of childhood. Patients with high-risk disease undergo extremely aggressive therapy and nonetheless have cure rates below 50%. Treatment with the ch14.18 monoclonal antibody (dinutuximab beta), directed against the GD2 disialoganglioside, improved 5-year event-free survival in high-risk patients when administered in postconsolidation therapy and was recently implemented in standard therapy. Relapse still occurred in 57% of these patients, necessitating new therapeutic options. Bispecific trifunctional antibodies (trAbs) are IgG-like molecules directed against T cells and cancer surface antigens, redirecting T cells (via their CD3 specificity) and accessory immune cells (via their functioning Fc-fragment) toward tumor cells. We sought proof-of-concept for GD2/CD3-directed trAb efficacy against neuroblastoma. Methods We used two GD2-specific trAbs differing only in their CD3-binding specificity: EKTOMUN (GD2/human CD3) and SUREK (GD2/mouse Cd3). This allowed trAb evaluation in human and murine experimental settings. Tumor-blind trAb and the ch14.18 antibody were used as controls. A coculture model of human peripheral blood mononuclear cells (PBMCs) and neuroblastoma cell lines was established to evaluate trAb antitumor efficacy by assessing expression of T-cell surface markers for activation, proinflammatory cytokine release and cytotoxicity assays. Characteristics of tumor-infiltrating T cells and response of neuroblastoma metastases to SUREK treatment were investigated in a syngeneic immunocompetent neuroblastoma mouse model mimicking minimal residual disease. Results We show that EKTOMUN treatment caused effector cell activation and release of proinflammatory cytokines in coculture with neuroblastoma cell lines. Furthermore, EKTOMUN mediated GD2-dependent cytotoxic effects in human neuroblastoma cell lines in coculture with PBMCs, irrespective of the level of target antigen expression. This effect was dependent on the presence of accessory immune cells. Treatment with SUREK reduced the intratumor Cd4/Cd8 ratio and activated tumor infiltrating T cells in vivo. In a minimal residual disease model for neuroblastoma, we demonstrated that single-agent treatment with SUREK strongly reduced or eliminated neuroblastoma metastases in vivo. SUREK as well as EKTOMUN demonstrated superior tumor control compared with the anti-GD2 antibody, ch14.18. Conclusions Here we provide proof-of-concept for EKTOMUN preclinical efficacy against neuroblastoma, presenting this bispecific trAb as a promising new agent to fight neuroblastoma.
Collapse
Affiliation(s)
- Felix Zirngibl
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany .,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sara M Ivasko
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Laura Grunewald
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anika Klaus
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Silke Schwiebert
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peter Ruf
- Trion Research, Martinsried, Germany
| | | | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lena Andersch
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Partner Site Berlin CCCC, German Cancer Consortium, Berlin, Berlin, Germany
| | - Holger N Lode
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Partner Site Berlin CCCC, German Cancer Consortium, Berlin, Berlin, Germany
| | - Kathleen Anders
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Partner Site Berlin CCCC, German Cancer Consortium, Berlin, Berlin, Germany
| | - Patrick Hundsdoerfer
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Pediatrics, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Annette Künkele
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Partner Site Berlin CCCC, German Cancer Consortium, Berlin, Berlin, Germany
| |
Collapse
|
17
|
Okada R, Furusawa A, Vermeer DW, Inagaki F, Wakiyama H, Kato T, Nagaya T, Choyke PL, Spanos WC, Allen CT, Kobayashi H. Near-infrared photoimmunotherapy targeting human-EGFR in a mouse tumor model simulating current and future clinical trials. EBioMedicine 2021; 67:103345. [PMID: 33933782 PMCID: PMC8102756 DOI: 10.1016/j.ebiom.2021.103345] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 11/25/2022] Open
Abstract
Background near-infrared photoimmunotherapy (NIR-PIT) is a cancer treatment that uses antibody-photoabsorber (IRDye700DX, IR700) conjugates (APCs) which bind to target cells and are photoactivated by NIR light inducing rapid necrotic cell death. NIR-PIT targeting human epidermal growth factor receptor (hEGFR) has been shown to destroy hEGFR expressing human tumor cells and to be effective in immunodeficient mouse models. NIR-PIT can also be targeted to cells in the tumor microenvironment, for instance, CD25-targeted NIR-PIT can be used to selectively deplete regulatory T cells (Tregs) within a tumor. The aim of this study was to evaluate the combined therapeutic efficacy of hEGFR and CD25-targeted NIR-PIT in a newly established hEGFR expressing murine oropharyngeal cell line (mEERL-hEGFR). Methods panitumumab conjugated with IR700 (pan-IR700) was used as the cancer cell-directed component of NIR-PIT and anti-CD25-F(ab′)2-IR700 was used as the tumor microenvironment-directed component of NIR-PIT. Efficacy was evaluated using tumor-bearing mice in four groups: (1) non-treatment group (control), (2) pan-IR700 based NIR-PIT (pan-PIT), (3) anti-CD25-F(ab′)2-IR700 based NIR-PIT (CD25-PIT), (4) combined NIR-PIT with pan-IR700 and anti-CD25- F(ab′)2-IR700 (combined PIT). Findings the combined PIT group showed the greatest inhibition of tumor growth. Destruction of cancer cells likely leads to an immune response which is amplified by the loss of Tregs in the tumor microenvironment. Interpretation combined hEGFR and CD25-targeted NIR-PIT is a promising treatment for hEGFR expressing cancers in which Treg cells play an immunosuppressive role.
Collapse
Affiliation(s)
- Ryuhei Okada
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Aki Furusawa
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Daniel W Vermeer
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD 57104, United States
| | - Fuyuki Inagaki
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Hiroaki Wakiyama
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Takuya Kato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Tadanobu Nagaya
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - William C Spanos
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD 57104, United States; Department of Surgery, University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, United States
| | - Clint T Allen
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, United States
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
18
|
Chen X, Liang R, Zhu X. Anti-EGFR therapies in nasopharyngeal carcinoma. Biomed Pharmacother 2020; 131:110649. [PMID: 32836074 DOI: 10.1016/j.biopha.2020.110649] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/12/2020] [Accepted: 08/16/2020] [Indexed: 01/18/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common malignant tumor in Southern China and South-East Asia. Regardless of initiative high response to radiotherapy, parts of patients still have relapses and metastases. It is reported that epidermal growth factor receptor (EGFR) is highly expressed in most of NPC and is a poor prognostic factor. Targeting EGFR therapies including monoclonal antibodies and EGFR tyrosine kinase inhibitors (EGFR-TKIs), offer different benefits and toxicities for patients with NPC. Herein, we summarize the clinical evidence of anti-EGFR therapies in the management of NPC and provide a direction for the treatment and research of NPC in the future.
Collapse
Affiliation(s)
- Xishan Chen
- Department of Oncology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, 545000, China; Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, PR China
| | - Renba Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, PR China
| | - Xiaodong Zhu
- Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, PR China; Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, PR China.
| |
Collapse
|
19
|
Zhao C, Gao Y, Yu N, Li T, Zhang Y, Zhang H, Lu G, Gao Y, Guo X. Unidirectional transport of IgG by neonatal Fc receptor in human thyrocytes varies across different IgG subclasses. Mol Cell Endocrinol 2018; 477:103-111. [PMID: 29908223 DOI: 10.1016/j.mce.2018.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/22/2018] [Accepted: 06/11/2018] [Indexed: 10/14/2022]
Abstract
Neonatal Fc receptor (FcRn) is down-regulated in Hashimoto's thyroiditis (HT) thyrocytes and mediates IgG endocytosis in thyrocytes. The serum distribution of IgG subclasses (of TgAb and TPOAb) differs between HT patients and normal individuals. We aimed to explore the direction and regulation of FcRn-mediated IgG transport in thyrocyte monolayers and the difference between various IgG subclass transport. IgG was transported by FcRn from the basolateral to apical side in the thyrocyte monolayers grown on Transwell filters and the transport was inhibited by IFN-γ and TNF-α. Stimulation by T3 and TSH down-regulated FcRn expression in thyrocytes. IgG1 was transported preferentially over IgG2 and IgG4, which might be related to the differences in FcRn-binding affinities as shown by SPR. FcRn mediates unidirectional IgG transport in thyrocytes in a tissue-specific manner. Down-regulation of FcRn is speculated to play a protective role in HT pathogenesis by mainly reducing IgG1 transport in thyrocytes.
Collapse
Affiliation(s)
- Chenxu Zhao
- Department of Endocrinology, Peking University First Hospital, Beijing, 100034, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, 100034, China
| | - Nan Yu
- Department of Endocrinology, Peking University First Hospital, Beijing, 100034, China
| | - Tiancheng Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, 100034, China; Centre for Cancer Research, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Yang Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, 100034, China.
| | - Hong Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, 100034, China
| | - Guizhi Lu
- Department of Endocrinology, Peking University First Hospital, Beijing, 100034, China
| | - Yanming Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, 100034, China
| | - Xiaohui Guo
- Department of Endocrinology, Peking University First Hospital, Beijing, 100034, China
| |
Collapse
|
20
|
Liu J, Li Y, Lu Z, Gu J, Liang Y, Huang E, Wang Z, Zhang H, Wang L, Zhang D, Yu H, Liu R, Chu Y. Deceleration of glycometabolism impedes IgG-producing B-cell-mediated tumor elimination by targeting SATB1. Immunology 2018; 156:56-68. [PMID: 30171602 DOI: 10.1111/imm.12998] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022] Open
Abstract
B lymphocytes, known as antibody producers, mediate tumor cell destruction in the manner of antibody-dependent cell-mediated cytotoxicity; however, their anti-tumor function seems to be weakened during tumorigenesis, while the underlying mechanisms remain unclear. In this study, we found that IgG mediated anti-tumor effects, but IgG-producing B cells decreased in various tumors. Considering the underlying mechanism, glycometabolism was noteworthy. We found that tumor-infiltrating B cells were glucose-starved and accompanied by a deceleration of glycometabolism. Both inhibition of glycometabolism and deprivation of glucose through tumor cells, or glucose-free treatment, reduced the differentiation of B cells into IgG-producing cells. In this process, special AT-rich sequence-binding protein-1 (SATB1) was significantly silenced in B cells. Down-regulating SATB1 by inhibiting glycometabolism or RNA interference reduced the binding of signal transducer and activator of transcription 6 (STAT6) to the promoter of germline Cγ gene, subsequently resulting in fewer B cells producing IgG. Our findings provide the first evidence that glycometabolic inhibition by tumorigenesis suppresses differentiation of B cells into IgG-producing cells, and altering glycometabolism may be promising in improving the anti-tumor effect of B cells.
Collapse
Affiliation(s)
- Jiajing Liu
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yifan Li
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhou Lu
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jie Gu
- Department of Thoracic Surgery, The Affiliated Zhongshan Hospital of Fudan University, Shanghai, China
| | - Yun Liang
- Department of Orthopedics, The Affiliated Zhongshan Hospital of Fudan University, Shanghai, China
| | - Enyu Huang
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhiming Wang
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hushan Zhang
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Dan Zhang
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hongxiu Yu
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ronghua Liu
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Expression and characterization of a recombinant porcinized antibody against the E2 protein of classical swine fever virus. Appl Microbiol Biotechnol 2017; 102:961-970. [DOI: 10.1007/s00253-017-8647-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 10/18/2022]
|
22
|
Joh J, Chilton PM, Wilcher SA, Zahin M, Park J, Proctor ML, Ghim SJ, Jenson AB. T cell-mediated antitumor immune response eliminates skin tumors induced by mouse papillomavirus, MmuPV1. Exp Mol Pathol 2017; 103:181-190. [PMID: 28939161 DOI: 10.1016/j.yexmp.2017.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/28/2022]
Abstract
Previous studies of naturally occurring mouse papillomavirus (PV) MmuPV1-induced tumors in B6.Cg-Foxn1nu/nu mice suggest that T cell deficiency is necessary and sufficient for the development of such tumors. To confirm this, MmuPV1-induced tumors were transplanted from T cell-deficient mice into immunocompetent congenic mice. Consequently, the tumors regressed and eventually disappeared. The elimination of MmuPV1-infected skin/tumors in immunocompetent mice was consistent with the induction of antitumor T cell immunity. This was confirmed by adoptive cell experiments using hyperimmune splenocytes collected from graft-recipient mice. In the present study, such splenocytes were injected into T cell-deficient mice infected with MmuPV1, and they eliminated both early-stage and fully formed tumors. We clearly show that anti-tumor T cell immunity activated during tumor regression in immunocompetent mice effectively eliminates tumors developing in T cell-deficient congenic mice. The results corroborate the notion that PV-induced tumors are strongly linked to the immune status of the host, and that PV antigens are major anti-tumor antigens. Successful anti-PV T cell responses should, therefore, lead to effective anti-tumor immune therapy in human PV-infected patients.
Collapse
Affiliation(s)
- Joongho Joh
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Medicine, University of Louisville, Louisville, KY, USA.
| | - Paula M Chilton
- Christine M. Kleinert Institute for Hand & Microsurgery, 225 Abraham Flexner Way, Suite 850, Louisville, KY, USA
| | - Sarah A Wilcher
- Research Resources Center, 530 South Jackson Street, Louisville, KY, USA
| | - Maryam Zahin
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Jino Park
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Mary L Proctor
- Research Resources Center, 530 South Jackson Street, Louisville, KY, USA
| | - Shin-Je Ghim
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Alfred B Jenson
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
23
|
Saito H, Miyatani K, Kono Y, Murakami Y, Kuroda H, Matsunaga T, Fukumoto Y, Takano S, Osaki T, Fujiwara Y. Decreased Serum Concentration of Total IgG Is Related to Tumor Progression in Gastric Cancer Patients. Yonago Acta Med 2017; 60:119-125. [PMID: 28701895 PMCID: PMC5502224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/01/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND There is accumulating evidence that shows cell-mediated immunity regulated by T cells is impaired in cancer patients. Unfortunately, the mechanisms by which B cells participate in tumor immunity are only partially understood. METHODS The serum concentration of Immunoglobulin G (IgG) was measured by Enzyme-Linked ImmunoSorbent Assay (ELISA) in patients with gastric cancer. Immunohistochemistry was also performed using the anti- cluster of differentiation (CD)134 antibody to evaluate the number of plasma cells in the tumor tissue. RESULTS The total serum IgG concentration was significantly lower in patients with lymph node metastasis compared with patients without metastasis. The serum concentration of total IgG at stage III/IV was significantly lower compared with tumors classified as stage I/II. A decreased serum concentration of total IgG and IgG1 was significantly related to a poor prognosis for gastric cancer patients. Furthermore, multivariate analysis indicated that the serum concentration of IgG and lymph node metastasis were independent prognostic indicators for poorer survival. The number of plasma cells was significantly lower in gastric cancer tissue compared with non-cancerous gastric mucosa. CONCLUSION A decreased serum concentration of IgG was closely related to poor prognosis, indicating the possibility that impaired antibody-mediated immunity is associated with tumor progression in patients with gastric cancer.
Collapse
Affiliation(s)
- Hiroaki Saito
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Kozo Miyatani
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Yusuke Kono
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Yuki Murakami
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Hirohiko Kuroda
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Tomoyuki Matsunaga
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Yoji Fukumoto
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Shuichi Takano
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Tomohiro Osaki
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Yoshiyuki Fujiwara
- Division of Surgical Oncology, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| |
Collapse
|
24
|
Wu J, Ma XL, Tian L, Zhang CY, Wang BL, Hu YY, Gao XH, Zhou Y, Shen MN, Peng YF, Pan BS, Zhou J, Fan J, Yang XR, Guo W. Serum IgG4:IgG Ratio Predicts Recurrence of Patients with Hepatocellular Carcinoma after Curative Resection. J Cancer 2017; 8:1338-1346. [PMID: 28638447 PMCID: PMC5479238 DOI: 10.7150/jca.18030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 02/14/2017] [Indexed: 12/20/2022] Open
Abstract
Aim: IgG4 is associated with a Th1-to-Th2 switch, which plays a vital role in metastasis, in patients with malignances; thus, we aimed to investigate its clinical significance in predicting hepatocellular carcinoma (HCC) recurrence in the present study. Methods: The correlation between serum IgG4:IgG ratio and recurrence was analyzed in a cohort of 195 patients undergoing curative resection in 2012. Another 100 patients were analyzed in a prospective independent cohort during 2012-2013 to validate the value of serum IgG4. Serum IgG4 and total IgG concentrations were measured with an automatic immune analyzer and the optimal cutoff value for serum IgG4 levels was determined by X-tile software. Results: Our data revealed that serum IgG4:IgG were significantly elevated in patients with tumor recurrence (P<0.05). A cutoff IgG:IgG4 ratio of 0.08 was set to stratify HCC patients into high (>0.08) and low (≤0.08) groups. High serum IgG4:IgG ratio correlated with significantly shorter time-to-recurrence (median 11.85 months vs. 39.20, P=0.005). Univariate and multivariate analyses demonstrated that serum IgG4:IgG ratio is an independent indicator of tumor recurrence and this retained its clinical significance even in conventional low-recurrence-risk subgroups, including patients with low α-fetoprotein and early-stage diseases. Conclusion: Our results demonstrated that elevated serum IgG4:IgG ratio is associated with poor clinical outcomes in HCC patients and therefore, and can serve as a novel prognostic predictor for HCC patients undergoing resection. Analyzing serum IgG4 would be useful to tailor individualized therapies for patients.
Collapse
Affiliation(s)
- Jiong Wu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Xiao-Lu Ma
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Lu Tian
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Chun-Yan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Bei-Li Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yu-Yi Hu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Xing-Hui Gao
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yan Zhou
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Min-Na Shen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yin-Fei Peng
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Bai-Shen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jian Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jia Fan
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan hospital, Fudan University, Shanghai 200032, P. R. China
| | - Xin-Rong Yang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan hospital, Fudan University, Shanghai 200032, P. R. China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
25
|
Dekkers G, Bentlage AEH, Stegmann TC, Howie HL, Lissenberg-Thunnissen S, Zimring J, Rispens T, Vidarsson G. Affinity of human IgG subclasses to mouse Fc gamma receptors. MAbs 2017; 9:767-773. [PMID: 28463043 DOI: 10.1080/19420862.2017.1323159] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human IgG is the main antibody class used in antibody therapies because of its efficacy and longer half-life, which are completely or partly due to FcγR-mediated functions of the molecules. Preclinical testing in mouse models are frequently performed using human IgG, but no detailed information on binding of human IgG to mouse FcγRs is available. The orthologous mouse and human FcγRs share roughly 60-70% identity, suggesting some incompatibility. Here, we report binding affinities of all mouse and human IgG subclasses to mouse FcγR. Human IgGs bound to mouse FcγR with remarkably similar binding strengths as we know from binding to human ortholog receptors, with relative affinities IgG3>IgG1>IgG4>IgG2 and FcγRI>>FcγRIV>FcγRIII>FcγRIIb. This suggests human IgG subclasses to have similar relative FcγR-mediated biological activities in mice.
Collapse
Affiliation(s)
- Gillian Dekkers
- a Department of Experimental Immunohematology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , The Netherlands
| | - Arthur E H Bentlage
- a Department of Experimental Immunohematology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , The Netherlands
| | - Tamara C Stegmann
- a Department of Experimental Immunohematology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , The Netherlands
| | - Heather L Howie
- b Department of Transfusion Medicine , Bloodworks Northwest Research Institute , Seattle , Washington , USA
| | - Suzanne Lissenberg-Thunnissen
- a Department of Experimental Immunohematology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , The Netherlands
| | - James Zimring
- b Department of Transfusion Medicine , Bloodworks Northwest Research Institute , Seattle , Washington , USA
| | - Theo Rispens
- c Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center , University of Amsterdam , The Netherlands
| | - Gestur Vidarsson
- a Department of Experimental Immunohematology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , The Netherlands
| |
Collapse
|
26
|
Kinder M, Greenplate AR, Strohl WR, Jordan RE, Brezski RJ. An Fc engineering approach that modulates antibody-dependent cytokine release without altering cell-killing functions. MAbs 2016; 7:494-504. [PMID: 25933349 PMCID: PMC4622058 DOI: 10.1080/19420862.2015.1022692] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cytotoxic therapeutic monoclonal antibodies (mAbs) often mediate target cell-killing by eliciting immune effector functions via Fc region interactions with cellular and humoral components of the immune system. Key functions include antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC). However, there has been increased appreciation that along with cell-killing functions, the induction of antibody-dependent cytokine release (ADCR) can also influence disease microenvironments and therapeutic outcomes. Historically, most Fc engineering approaches have been aimed toward modulating ADCC, ADCP, or CDC. In the present study, we describe an Fc engineering approach that, while not resulting in impaired ADCC or ADCP, profoundly affects ADCR. As such, when peripheral blood mononuclear cells are used as effector cells against mAb-opsonized tumor cells, the described mAb variants elicit a similar profile and quantity of cytokines as IgG1. In contrast, although the variants elicit similar levels of tumor cell-killing as IgG1 with macrophage effector cells, the variants do not elicit macrophage-mediated ADCR against mAb-opsonized tumor cells. This study demonstrates that Fc engineering approaches can be employed to uncouple macrophage-mediated phagocytic and subsequent cell-killing functions from cytokine release.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- ADCP, antibody-dependent cellular phagocytosis
- ADCR, antibody-dependent cytokine release
- APCs, antigen-presenting cells
- CDC, complement-dependent cytotoxicity
- DC, dendritic cell
- Fc gamma receptors
- FcγR, Fc gamma receptor
- IFN, interferon
- IL, interleukin
- NK, natural killer
- PBMC, peripheral blood mononuclear cell
- TNF, tumor necrosis factor
- antibody-dependent cellular phagocytosis
- cytokine release
- interferon gamma
- interleukin 10
- mAbs, monoclonal antibodies
- monocyte-derived macrophages
- natural killer cells
Collapse
Affiliation(s)
- Michelle Kinder
- a Biologics Research; Janssen Research & Development, LLC; Spring House , PA , USA
| | | | | | | | | |
Collapse
|
27
|
Miyatani K, Saito H, Murakami Y, Watanabe J, Kuroda H, Matsunaga T, Fukumoto Y, Osaki T, Nakayama Y, Umekita Y, Ikeguchi M. A high number of IgG4-positive cells in gastric cancer tissue is associated with tumor progression and poor prognosis. Virchows Arch 2016; 468:549-57. [PMID: 26951261 DOI: 10.1007/s00428-016-1914-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/12/2016] [Accepted: 02/11/2016] [Indexed: 12/24/2022]
Abstract
IgG4-related disease is a newly defined disease characterized by elevated serum IgG4 levels and infiltration of affected organs by IgG4-positive plasma cells. Recently, increased IgG4 levels were reported to be closely related with malignancy. To assess the relationship between IgG4 and the progression of gastric cancer, we immunohistochemically stained in this study gastric cancer tissue samples for IgG4-positive cells using an anti-IgG4 antibody. In addition, pre- and postoperative serum concentrations of IgG4 were measured, using an enzyme-linked immunosorbent assay. In gastric cancer samples, the number of CD138-positive plasma cells was significantly lower and the number of IgG4-positive cells significantly higher than in non-cancerous gastric mucosa. The number of IgG4-positive cells was significantly correlated with gross tumor appearance, tumor depth, lymph node metastasis, venous invasion, and lymphatic invasion. Prognosis was significantly poorer in patients with a high number of IgG4-positive cells than in those with a low number. Multivariate analysis indicated that both the number of IgG4-positive cells and the depth of tumor invasion were independently prognostic of survival. In conclusion, in gastric cancer, the number of IgG4-positive cells is increased and this is closely associated with gastric cancer progression.
Collapse
Affiliation(s)
- Kozo Miyatani
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan
| | - Hiroaki Saito
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan.
| | - Yuki Murakami
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan
| | - Joji Watanabe
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan
| | - Hirohiko Kuroda
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan
| | - Tomoyuki Matsunaga
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan
| | - Yoji Fukumoto
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan
| | - Tomohiro Osaki
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan
| | - Yuji Nakayama
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University, Yonago, 683-8503, Japan
| | - Yoshihisa Umekita
- Department of Pathology, Division of Organ Pathology, Tottori University School of Medicine, Yonago, 683-8503, Japan
| | - Masahide Ikeguchi
- Department of Surgery, Division of Surgical Oncology, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan
| |
Collapse
|
28
|
Gao P, Pinkston KL, Wilganowski N, Robinson H, Azhdarinia A, Zhu B, Sevick-Muraca EM, Harvey BR. Deglycosylation of mAb by EndoS for improved molecular imaging. Mol Imaging Biol 2015; 17:195-203. [PMID: 25135058 DOI: 10.1007/s11307-014-0781-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Monoclonal antibodies (mAbs) have been shown preclinically as reliable targeting moieties for antigen imaging using near-infrared fluorescence (NIRF) molecular imaging. However, crystallizable fragment-gamma receptor (FcγRs) expressed on immune cells also bind mAbs through defined epitopes on the constant fragment (Fc) of IgG. Herein, we evaluate the potential impact Fc interactions have on mAb agent imaging specificity. PROCEDURE Through the removal of conserved glycans within the Fc domain, shown to have Fc/FcγR interactions, we evaluate their impact on non-specific binding/accumulation of a NIRF-labeled mAb-based imaging agent in lymph nodes (LNs) in inflamed animals and in an orthotopic prostate cancer animal model of LN metastasis. RESULTS Deglycosylation of a murine mAb against the human epithelial cell adhesion marker using endoglycosidase EndoS significantly reduced non-specific binding in the LNs of inflamed animals and in cancer-negative LNs of tumor-bearing animals. Sensitivity remained unchanged while improvement in imaging specificity increased imaging accuracy. CONCLUSION The reduction of non-specific binding through deglycosylation of a mAb-based imaging agent shows that reducing Fc/FcγR interactions can improve imaging accuracy.
Collapse
Affiliation(s)
- Peng Gao
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, 1825 Pressler Street, Houston, TX, 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Influence of IgG Subclass on Human Antimannan Antibody-Mediated Resistance to Hematogenously Disseminated Candidiasis in Mice. Infect Immun 2015; 84:386-94. [PMID: 26573736 DOI: 10.1128/iai.00890-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/06/2015] [Indexed: 01/23/2023] Open
Abstract
Candida albicans is a yeast-like pathogen and can cause life-threatening systemic candidiasis. Its cell surface is enriched with mannan that is resistant to complement activation. Previously, we developed the recombinant human IgG1 antimannan antibody M1g1. M1g1 was found to promote complement activation and phagocytosis and protect mice from systemic candidiasis. Here, we evaluate the influence of IgG subclass on antimannan antibody-mediated protection. Three IgG subclass variants of M1g1 were constructed: M1g2, M1g3, and M1g4. The IgG subclass identity for each variant was confirmed with DNA sequence and subclass-specific antibodies. These variants contain identical M1 Fabs and exhibited similar binding affinities for C. albicans yeast and purified mannan. Yeast cells and hyphae recovered from the kidney of antibody-treated mice with systemic candidiasis showed uniform binding of each variant, indicating constitutive expression of the M1 epitope and antibody opsonization in the kidney. All variants promoted deposition of both murine and human C3 onto the yeast cell surface, with M1g4 showing delayed activation, as determined by flow cytometry and immunofluorescence microscopy. M1g4-mediated complement activation was found to be associated with its M1 Fab that activates the alternative pathway in an Fc-independent manner. Treatment with each subclass variant extended the survival of mice with systemic candidiasis (P < 0.001). However, treatment with M1g1, M1g3, or M1g4, but not with M1g2, also reduced the kidney fungal burden (P < 0.001). Thus, the role of human antimannan antibody in host resistance to systemic candidiasis is influenced by its IgG subclass.
Collapse
|
30
|
Production of monoclonal antibodies in plants for cancer immunotherapy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:306164. [PMID: 26550566 PMCID: PMC4624878 DOI: 10.1155/2015/306164] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 09/02/2015] [Indexed: 12/17/2022]
Abstract
Plants are considered as an alternative platform for recombinant monoclonal antibody (mAb) production due to the improvement and diversification of transgenic techniques. The diversity of plant species offers a multitude of possibilities for the valorization of genetic resources. Moreover, plants can be propagated indefinitely, providing cheap biomass production on a large scale in controlled conditions. Thus, recent studies have shown the successful development of plant systems for the production of mAbs for cancer immunotherapy. However, their several limitations have to be resolved for efficient antibody production in plants.
Collapse
|
31
|
Shao Y, Fu R, Liu H, Wang Y, Ding S, Wang H, Li L, Shao Z. IgG autoantibody subclasses altered in immuno-related hemocytopenia. Cell Immunol 2015; 294:13-20. [PMID: 25666506 DOI: 10.1016/j.cellimm.2015.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/07/2015] [Accepted: 01/20/2015] [Indexed: 11/30/2022]
Abstract
Recently, we have detected autoantibodies to bone marrow (BM) hemopoietic cells in some idiopathic cytopenia of undetermined significance (ICUS), terming as immunorelated hemocytopenia. Immunoglobulin G consists of 4 subclasses which have different biological functions in many diseases. In this study, we analyzed the alterations of IgG subclasses in 27 IRH patients compared with 20 ICUS patients and 20 normal controls. The results showed that IgG1 and IgG3 levels were increased in the bone marrow supernatant in IRH patients, and had inverse correlations with hematopoietic function. Meanwhile IgG1 level had a positive correlation with the proportion of CD5+ B lymphocytes. Using immunohistochemical staining, IgG1 were also detected on bone marrow nucleated cells in IRH patients. All these results indicated that IgG1 on bone marrow cells in some IRH patients might be involved in the destruction of hematopoietic cells leading to cytopenia and might be a novel therapeutic target in future.
Collapse
Affiliation(s)
- Yuanyuan Shao
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan St, Heping District, Tianjin 300052, PR China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan St, Heping District, Tianjin 300052, PR China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan St, Heping District, Tianjin 300052, PR China
| | - Yihao Wang
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan St, Heping District, Tianjin 300052, PR China
| | - Shaoxue Ding
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan St, Heping District, Tianjin 300052, PR China
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan St, Heping District, Tianjin 300052, PR China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan St, Heping District, Tianjin 300052, PR China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan St, Heping District, Tianjin 300052, PR China.
| |
Collapse
|
32
|
Jonnalagadda M, Mardiros A, Urak R, Wang X, Hoffman LJ, Bernanke A, Chang WC, Bretzlaff W, Starr R, Priceman S, Ostberg JR, Forman SJ, Brown CE. Chimeric antigen receptors with mutated IgG4 Fc spacer avoid fc receptor binding and improve T cell persistence and antitumor efficacy. Mol Ther 2014; 23:757-68. [PMID: 25366031 DOI: 10.1038/mt.2014.208] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 10/17/2014] [Indexed: 12/22/2022] Open
Abstract
The success of adoptive therapy using chimeric antigen receptor (CAR)-expressing T cells partly depends on optimal CAR design. CARs frequently incorporate a spacer/linker region based on the constant region of either IgG1 or IgG4 to connect extracellular ligand-binding with intracellular signaling domains. Here, we evaluated the potential for the IgG4-Fc linker to result in off-target interactions with Fc gamma receptors (FcγRs). As proof-of-principle, we focused on a CD19-specific scFv-IgG4-CD28-zeta CAR and found that, in contrast to CAR-negative cells, CAR+ T cells bound soluble FcγRs in vitro and did not engraft in NSG mice. We hypothesized that mutations to avoid FcγR binding would improve CAR+ T cell engraftment and antitumor efficacy. Thus, we generated CD19-specific CARs with IgG4-Fc spacers that had either been mutated at two sites (L235E; N297Q) within the CH2 region (CD19R(EQ)) or incorporated a CH2 deletion (CD19Rch2Δ). These mutations reduced binding to soluble FcγRs without altering the ability of the CAR to mediate antigen-specific lysis. Importantly, CD19R(EQ) and CD19Rch2Δ T cells exhibited improved persistence and more potent CD19-specific antilymphoma efficacy in NSG mice. Together, these studies suggest that optimal CAR function may require the elimination of cellular FcγR interactions to improve T cell persistence and antitumor responses.
Collapse
Affiliation(s)
- Mahesh Jonnalagadda
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Armen Mardiros
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Ryan Urak
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Xiuli Wang
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Lauren J Hoffman
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Alyssa Bernanke
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Wen-Chung Chang
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - William Bretzlaff
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Renate Starr
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Saul Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Julie R Ostberg
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Christine E Brown
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
33
|
Josephs DH, Spicer JF, Karagiannis P, Gould HJ, Karagiannis SN. IgE immunotherapy: a novel concept with promise for the treatment of cancer. MAbs 2014; 6:54-72. [PMID: 24423620 DOI: 10.4161/mabs.27029] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The importance of antibodies in activating immune responses against tumors is now better appreciated with the emergence of checkpoint blockade antibodies and with engineered antibody Fc domains featuring enhanced capacity to focus potent effector cells against cancer cells. Antibodies designed with Fc regions of the IgE class can confer natural, potent, long-lived immune surveillance in tissues through tenacious engagement of high-affinity cognate Fc receptors on distinct, often tumor-resident immune effector cells, and through ability to activate these cells under tumor-induced Th2-biased conditions. Here, we review the properties that make IgE a contributor to the allergic response and a critical player in the protection against parasites, which also support IgE as a novel anti-cancer modality. We discuss IgE-based active and passive immunotherapeutic approaches in disparate in vitro and in vivo model systems, collectively suggesting the potential of IgE immunotherapies in oncology. Translation toward clinical application is now in progress.
Collapse
Affiliation(s)
- Debra H Josephs
- Cutaneous Medicine and Immunotherapy Unit; St. John's Institute of Dermatology; Division of Genetics and Molecular Medicine & NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London; London, UK; Division of Cancer Studies; King's College London; Guy's Hospital; London, UK
| | - James F Spicer
- Division of Cancer Studies; King's College London; Guy's Hospital; London, UK
| | - Panagiotis Karagiannis
- Cutaneous Medicine and Immunotherapy Unit; St. John's Institute of Dermatology; Division of Genetics and Molecular Medicine & NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London; London, UK
| | - Hannah J Gould
- Randall Division of Cell and Molecular Biophysics & Division of Asthma; Allergy and Lung Biology; MRC and Asthma UK Centre for Allergic Mechanisms of Asthma; King's College London; Guy's Campus; London, UK
| | - Sophia N Karagiannis
- Cutaneous Medicine and Immunotherapy Unit; St. John's Institute of Dermatology; Division of Genetics and Molecular Medicine & NIHR Biomedical Research Centre at Guy's and St. Thomas's Hospitals and King's College London; London, UK
| |
Collapse
|
34
|
Annels NE, Simpson GR, Denyer M, McGrath SE, Falgari G, Killick E, Eeles R, Stebbing J, Pchejetski D, Cutress R, Murray N, Michael A, Pandha H. Spontaneous antibodies against Engrailed-2 (EN2) protein in patients with prostate cancer. Clin Exp Immunol 2014; 177:428-38. [PMID: 24654775 PMCID: PMC4226594 DOI: 10.1111/cei.12332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2014] [Indexed: 11/27/2022] Open
Abstract
We reported the expression of the homeodomain-containing transcription factor Engrailed-2 (EN2) in prostate cancer and showed that the presence of EN2 protein in the urine was highly predictive of prostate cancer. This study aimed to determine whether patients with prostate cancer have EN2 autoantibodies, what the prevalence of these antibodies is and whether they are associated with disease stage. The spontaneous immunoglobulin (Ig)G immune response against EN2 and for comparison the tumour antigen New York Esophageal Squamous Cell Carcinoma 1 (NY-ESO-1), were tested by enzyme-linked immunosorbent assay (ELISA) in three different cohorts of prostate cancer patients as well as a group of men genetically predisposed to prostate cancer. Thirty-two of 353 (9·1%) of the SUN cohort representing all stages of prostate cancer demonstrated EN2 IgG responses, 12 of 107 patients (11·2%) in the advanced prostate cancer patients showed responses, while only four of 121 patients (3·3%) with castrate-resistant prostate cancer showed EN2 autoantibodies. No significant responses were found in the predisposed group. Anti-EN2 IgG responses were significantly higher in patients with prostate cancer compared to healthy control males and similarly prevalent to anti-NY-ESO-1 responses. While EN2 autoantibodies are not a useful diagnostic or monitoring tool, EN2 immunogenicity provides the rationale to pursue studies using EN2 as an immunotherapeutic target.
Collapse
Affiliation(s)
- N E Annels
- Oncology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Hilary Koprowski, MD: A Lifetime of Work. Monoclon Antib Immunodiagn Immunother 2014; 33:1-43. [DOI: 10.1089/mab.2014.kop.biblio] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
36
|
Seo Y, Ishii Y, Ochiai H, Fukuda K, Akimoto S, Hayashida T, Okabayashi K, Tsuruta M, Hasegawa H, Kitagawa Y. Cetuximab-mediated ADCC activity is correlated with the cell surface expression level of EGFR but not with the KRAS/BRAF mutational status in colorectal cancer. Oncol Rep 2014; 31:2115-22. [PMID: 24626880 DOI: 10.3892/or.2014.3077] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/14/2014] [Indexed: 01/26/2023] Open
Abstract
Cetuximab, an IgG1 monoclonal antibody against the epidermal growth factor receptor (EGFR), is widely used for the treatment of metastatic colorectal cancer (mCRC). One of the mechanisms of action is considered to be antibody-dependent cell-mediated cytotoxicity (ADCC) triggered by Fcγ-R on natural killer cells. However, whether ADCC is associated with EGFR expression and/or the mutational status of EGF downstream effectors (KRAS and BRAF) in colorectal cancer (CRC) remains unclear. The aim of the present study was to verify whether ADCC activities are associated with the cell surface expression levels of EGFR and/or the mutational status of KRAS and BRAF. Five human CRC cell lines with different cell surface expression levels of EGFR and different KRAS and BRAF mutational statuses were selected to evaluate ADCC activity using peripheral blood mononuclear cells (PBMCs) from healthy human donors. Furthermore, tumor cells from resected specimens of CRC patients were used to evaluate the cell surface expression level of EGFR using immunohistochemistry and the KRAS and BRAF mutational statuses using direct sequencing, while the ADCC activity was examined using PBMCs from the same CRC patients. A strong correlation was observed between the expression levels of EGFR and the ADCC activities in the cell lines (correlation coefficient: 0.949; P=0.003). Of the 13 resected specimens, a high ADCC activity level was significantly observed in tumor cells with high expression levels of cell surface EGFR, when compared with that in the tumor cells with low expression levels (P=0.027). In both CRC cell lines and tumor cells from CRC patients, the ADCC activities were significantly associated with the cell surface expression levels of EGFR [standard partial regression coefficients: 0.911 (P=0.017) and 0.660 (P=0.018), respectively], but not with the mutational status of KRAS and BRAF [standard partial regression coefficient: -0.101 (P=0.631) and 0.160 (P=0.510), respectively]. Cetuximab-mediated ADCC activity may be correlated with the cell surface expression level of EGFR, regardless of the mutational statuses of KRAS and BRAF, in CRC.
Collapse
Affiliation(s)
- Yuki Seo
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yoshiyuki Ishii
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroki Ochiai
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazumasa Fukuda
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shingo Akimoto
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masashi Tsuruta
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hirotoshi Hasegawa
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
37
|
Li C, Chacko AM, Hu J, Hasegawa K, Swails J, Grasso L, El-Deiry WS, Nicolaides N, Muzykantov VR, Divgi CR, Coukos G. Antibody-based tumor vascular theranostics targeting endosialin/TEM1 in a new mouse tumor vascular model. Cancer Biol Ther 2014; 15:443-51. [PMID: 24553243 DOI: 10.4161/cbt.27825] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Tumor endothelial marker 1 (TEM1, endosialin) is a tumor vascular marker with significant diagnostic and therapeutic potential. However, in vivo small animal models to test affinity reagents specifically targeted to human (h)TEM1 are limited. We describe a new mouse tumor model where tumor vascular endothelial cells express hTEM1 protein. METHODS Immortalized murine endothelial cells MS1 were engineered to express hTEM1 and firefly luciferase and were inoculated in nude mice either alone, to form hemangioma-like endothelial grafts, or admixed with ID8 ovarian tumor cells, to form chimeric endothelial-tumor cell grafts. MORAb-004, a monoclonal humanized IgG 1 antibody specifically recognizing human TEM1 was evaluated for targeted theranostic applications, i.e., for its ability to affect vascular grafts expressing hTEM1 as well as being a tool for molecular positron emission tomography (PET) imaging. RESULTS Naked MORAb-004 treatment of mice bearing angioma grafts or chimeric endothelial-tumor grafts significantly suppressed the ability of hTEM1-positive endothelial cells, but not control endothelial cells, to form grafts and dramatically suppressed local angiogenesis. In addition, highly efficient radioiodination of MORAb-004 did not impair its affinity for hTEM1, and [ (124)I]-MORAb-004-PET enabled non-invasive visualization of tumors enriched with hTEM1-positive, but not hTEM1 negative vasculature with high degree of specificity and sensitivity. CONCLUSION The development of a new robust endothelial graft model expressing human tumor vascular proteins will help accelerate the development of novel theranostics targeting the tumor vasculature, which exhibit affinity specifically to human targets but not their murine counterparts. Our results also demonstrate the theranostic potential of MORAb-004 as PET imaging tracer and naked antibody therapy for TEM1-positive tumor.
Collapse
Affiliation(s)
- Chunsheng Li
- Ovarian Cancer Research Center; University of Pennsylvania; Philadelphia, PA USA
| | - Ann-Marie Chacko
- Institute for Translational Medicine and Therapeutics; University of Pennsylvania; Philadelphia, PA USA; Division of Nuclear Medicine and Clinical Molecular Imaging; Department of Radiology; University of Pennsylvania; Philadelphia, PA USA
| | - Jia Hu
- Ovarian Cancer Research Center; University of Pennsylvania; Philadelphia, PA USA
| | - Kosei Hasegawa
- Ovarian Cancer Research Center; University of Pennsylvania; Philadelphia, PA USA
| | - Jennifer Swails
- Ovarian Cancer Research Center; University of Pennsylvania; Philadelphia, PA USA
| | | | - Wafik S El-Deiry
- Department of Medicine, Hematology/Oncology; University of Pennsylvania; Philadelphia, PA USA
| | | | - Vladimir R Muzykantov
- Institute for Translational Medicine and Therapeutics; University of Pennsylvania; Philadelphia, PA USA
| | - Chaitanya R Divgi
- Division of Nuclear Medicine and Clinical Molecular Imaging; Department of Radiology; University of Pennsylvania; Philadelphia, PA USA
| | - George Coukos
- Ovarian Cancer Research Center; University of Pennsylvania; Philadelphia, PA USA
| |
Collapse
|
38
|
Terme M, Dorvillius M, Cochonneau D, Chaumette T, Xiao W, Diccianni MB, Barbet J, Yu AL, Paris F, Sorkin LS, Birklé S. Chimeric antibody c.8B6 to O-acetyl-GD2 mediates the same efficient anti-neuroblastoma effects as therapeutic ch14.18 antibody to GD2 without antibody induced allodynia. PLoS One 2014; 9:e87210. [PMID: 24520328 PMCID: PMC3919714 DOI: 10.1371/journal.pone.0087210] [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: 11/01/2013] [Accepted: 12/19/2013] [Indexed: 11/21/2022] Open
Abstract
Background Anti-GD2 antibody is a proven therapy for GD2-postive neuroblastoma. Monoclonal antibodies against GD2, such as chimeric mAb ch14.18, have become benchmarks for neuroblastoma therapies. Pain, however, can limit immunotherapy with anti-GD2 therapeutic antibodies like ch14.18. This adverse effect is attributed to acute inflammation via complement activation on GD2-expressing nerves. Thus, new strategies are needed for the development of treatment intensification strategies to improve the outcome of these patients. Methodology/Principal Findings We established the mouse-human chimeric antibody c.8B6 specific to OAcGD2 in order to reduce potential immunogenicity in patients and to fill the need for a selective agent that can kill neuroblastoma cells without inducing adverse neurological side effects caused by anti-GD2 antibody immunotherapy. We further analyzed some of its functional properties compared with anti-GD2 ch14.18 therapeutic antibody. With the exception of allodynic activity, we found that antibody c.8B6 shares the same anti-neuroblastoma attributes as therapeutic ch14.18 anti-GD2 mAb when tested in cell-based assay and in vivo in an animal model. Conclusion/Significance The absence of OAcGD2 expression on nerve fibers and the lack of allodynic properties of c.8B6–which are believed to play a major role in mediating anti-GD2 mAb dose-limiting side effects–provide an important rationale for the clinical application of c.8B6 in patients with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Mickaël Terme
- ATLAB Pharma, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- INSERM U.892, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- CNRS 6299, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
| | - Mylène Dorvillius
- ATLAB Pharma, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
| | - Denis Cochonneau
- INSERM U.892, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- CNRS 6299, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
| | - Tanguy Chaumette
- INSERM U.892, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- CNRS 6299, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- Université de Nantes, UFR des Sciences Pharmaceutiques et Biologiques, Nantes, France
| | - Wenhua Xiao
- Department of Anesthesia, Mc Gill University, Montreal, Quebec, Canada
| | - Mitchell B. Diccianni
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Jacques Barbet
- INSERM U.892, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- CNRS 6299, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
| | - Alice L. Yu
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Center of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - François Paris
- INSERM U.892, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- CNRS 6299, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
| | - Linda S. Sorkin
- Department of Anesthesiology, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Stéphane Birklé
- INSERM U.892, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- CNRS 6299, Centre de Recherche en Cancérologie de Nantes-Angers, Institut de Recherche en Santé de l’Université de Nantes, Nantes, France
- Université de Nantes, UFR des Sciences Pharmaceutiques et Biologiques, Nantes, France
- * E-mail:
| |
Collapse
|
39
|
Wanderley JLM, Thorpe PE, Barcinski MA, Soong L. Phosphatidylserine exposure on the surface of Leishmania amazonensis amastigotes modulates in vivo infection and dendritic cell function. Parasite Immunol 2013; 35:109-119. [PMID: 23163958 DOI: 10.1111/pim.12019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Accepted: 10/23/2012] [Indexed: 12/14/2022]
Abstract
Leishmania amazonensis parasites can cause diverse forms of leishmaniasis in humans and persistent lesions in most inbred strains of mice. In both cases, the infection is characterized by a marked immunosuppression of the host. We previously showed that amastigote forms of the parasite make use of surface-exposed phosphatidylserine (PS) molecules to infect host cells and promote alternative macrophage activation, leading to uncontrolled intracellular proliferation of the parasites. In this study, we demonstrated that treatment of infected mice with a PS-targeting monoclonal antibody ameliorated parasite loads and lesion development, which correlated with increased proliferative responses by lymphocytes. In addition, we observed an enhanced dendritic cell (DC) activation and antigen presentation in vitro. Our data imply that the recognition of PS exposed on the surface of amastigotes plays a role in down-modulating DC functions, in a matter similar to that of apoptotic cell clearance. This study provides new information regarding the mechanism of immune suppression in Leishmania infection.
Collapse
Affiliation(s)
- J L M Wanderley
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Campus UFRJ Macaé, Pólo Universitário, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - P E Thorpe
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - M A Barcinski
- Parasitology Department, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, Brazil.,Laboratory of Cellular Biology, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - L Soong
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.,Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
40
|
Oshima T, Sato S, Kato J, Ito Y, Watanabe T, Tsuji I, Hori A, Kurokawa T, Kokubo T. Nectin-2 is a potential target for antibody therapy of breast and ovarian cancers. Mol Cancer 2013; 12:60. [PMID: 23758976 PMCID: PMC3698035 DOI: 10.1186/1476-4598-12-60] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/31/2013] [Indexed: 01/22/2023] Open
Abstract
Background Nectin-2 is a Ca2+-independent cell-cell adhesion molecule that is one of the plasma membrane components of adherens junctions. However, little has been reported about the involvement of Nectin-2 in cancer. Methods To determine the expression of Nectin-2 in cancer tissues and cancer cell lines, we performed gene expression profile analysis, immunohistochemistry studies, and flow cytometry analysis. We also investigated the potential of this molecule as a target for antibody therapeutics to treat cancers by generating and characterizing an anti-Nectin-2 rabbit polyclonal antibody (poAb) and 256 fully human anti-Nectin-2 monoclonal antibodies (mAbs). In addition, we tested anti-Nectin-2 mAbs in several in vivo tumor growth inhibition models to investigate the primary mechanisms of action of the mAbs. Results In the present study, we found that Nectin-2 was over-expressed in clinical breast and ovarian cancer tissues by using gene expression profile analysis and immunohistochemistry studies. Nectin-2 was over-expressed in various cancer cell lines as well. Furthermore, the polyclonal antibody specific to Nectin-2 suppressed the in vitro proliferation of OV-90 ovarian cancer cells, which express endogenous Nectin-2 on the cell surface. The anti-Nectin-2 mAbs we generated were classified into 7 epitope bins. The anti-Nectin-2 mAbs demonstrated antibody-dependent cellular cytotoxicity (ADCC) and epitope bin-dependent features such as the inhibition of Nectin-2-Nectin-2 interaction, Nectin-2-Nectin-3 interaction, and in vitro cancer cell proliferation. A representative anti-Nectin-2 mAb in epitope bin VII, Y-443, showed anti-tumor effects against OV-90 cells and MDA-MB-231 breast cancer cells in mouse therapeutic models, and its main mechanism of action appeared to be ADCC. Conclusions We observed the over-expression of Nectin-2 in breast and ovarian cancers and anti-tumor activity of anti-Nectin-2 mAbs via strong ADCC. These findings suggest that Nectin-2 is a potential target for antibody therapy against breast and ovarian cancers.
Collapse
Affiliation(s)
- Tsutomu Oshima
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraokahigashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Karagiannis P, Gilbert AE, Josephs DH, Ali N, Dodev T, Saul L, Correa I, Roberts L, Beddowes E, Koers A, Hobbs C, Ferreira S, Geh JL, Healy C, Harries M, Acland KM, Blower PJ, Mitchell T, Fear DJ, Spicer JF, Lacy KE, Nestle FO, Karagiannis SN. IgG4 subclass antibodies impair antitumor immunity in melanoma. J Clin Invest 2013; 123:1457-74. [PMID: 23454746 PMCID: PMC3613918 DOI: 10.1172/jci65579] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 01/03/2013] [Indexed: 12/15/2022] Open
Abstract
Host-induced antibodies and their contributions to cancer inflammation are largely unexplored. IgG4 subclass antibodies are present in IL-10-driven Th2 immune responses in some inflammatory conditions. Since Th2-biased inflammation is a hallmark of tumor microenvironments, we investigated the presence and functional implications of IgG4 in malignant melanoma. Consistent with Th2 inflammation, CD22+ B cells and IgG4(+)-infiltrating cells accumulated in tumors, and IL-10, IL-4, and tumor-reactive IgG4 were expressed in situ. When compared with B cells from patient lymph nodes and blood, tumor-associated B cells were polarized to produce IgG4. Secreted B cells increased VEGF and IgG4, and tumor cells enhanced IL-10 secretion in cocultures. Unlike IgG1, an engineered tumor antigen-specific IgG4 was ineffective in triggering effector cell-mediated tumor killing in vitro. Antigen-specific and nonspecific IgG4 inhibited IgG1-mediated tumoricidal functions. IgG4 blockade was mediated through reduction of FcγRI activation. Additionally, IgG4 significantly impaired the potency of tumoricidal IgG1 in a human melanoma xenograft mouse model. Furthermore, serum IgG4 was inversely correlated with patient survival. These findings suggest that IgG4 promoted by tumor-induced Th2-biased inflammation may restrict effector cell functions against tumors, providing a previously unexplored aspect of tumor-induced immune escape and a basis for biomarker development and patient-specific therapeutic approaches.
Collapse
Affiliation(s)
- Panagiotis Karagiannis
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Amy E. Gilbert
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Debra H. Josephs
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Niwa Ali
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Tihomir Dodev
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Louise Saul
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Isabel Correa
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Luke Roberts
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Emma Beddowes
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Alexander Koers
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Carl Hobbs
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Silvia Ferreira
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Jenny L.C. Geh
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Ciaran Healy
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Mark Harries
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Katharine M. Acland
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Philip J. Blower
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Tracey Mitchell
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - David J. Fear
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - James F. Spicer
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Katie E. Lacy
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Frank O. Nestle
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Sophia N. Karagiannis
- National Institute for Health Research (NIHR) Biomedical Research Centre at Guy’s and St. Thomas’ Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy Unit, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, United Kingdom.
Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, Guy’s Campus, London, United Kingdom.
Skin Tumour Unit, St. John’s Institute of Dermatology, Guy’s Hospital, King’s College London, and Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Division of Imaging Sciences, Rayne Institute, King’s College London School of Medicine, St. Thomas’ Hospital, and King’s College London, London, United Kingdom.
Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom.
Department of Plastic Surgery at Guy’s, King’s, and St. Thomas’ Hospitals, London, United Kingdom.
Clinical Oncology, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.
Department of Academic Oncology, Division of Cancer Studies, King’s College London, Guy’s Hospital, London, United Kingdom
| |
Collapse
|
42
|
Gerdes CA, Nicolini VG, Herter S, van Puijenbroek E, Lang S, Roemmele M, Moessner E, Freytag O, Friess T, Ries CH, Bossenmaier B, Mueller HJ, Umaña P. GA201 (RG7160): A Novel, Humanized, Glycoengineered Anti-EGFR Antibody with Enhanced ADCC and Superior In Vivo Efficacy Compared with Cetuximab. Clin Cancer Res 2012. [DOI: 10.1158/1078-0432.ccr-12-0989] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
|
44
|
Overdijk MB, Verploegen S, Ortiz Buijsse A, Vink T, Leusen JHW, Bleeker WK, Parren PWHI. Crosstalk between human IgG isotypes and murine effector cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:3430-8. [PMID: 22956577 DOI: 10.4049/jimmunol.1200356] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Development of human therapeutic Abs has led to reduced immunogenicity and optimal interactions with the human immune system in patients. Humanization had as a consequence that efficacy studies performed in mouse models, which represent a crucial step in preclinical development, are more difficult to interpret because of gaps in our knowledge of the activation of murine effector cells by human IgG (hIgG) remain. We therefore developed full sets of human and mouse isotype variants of human Abs targeting epidermal growth factor receptor and CD20 to explore the crosstalk with mouse FcγRs (mFcγRs) and murine effector cells. Analysis of mFcγR binding demonstrated that hIgG1 and hIgG3 bound to all four mFcγRs, with hIgG3 having the highest affinity. hIgG1 nevertheless was more potent than hIgG3 in inducing Ab-dependent cellular cytotoxicity (ADCC) and Ab-dependent cellular phagocytosis with mouse NK cells, mouse polymorphonuclear leukocytes, and mouse macrophages. hIgG4 bound to all mFcγRs except mFcγRIV and showed comparable interactions with murine effector cells to hIgG3. hIgG4 is thus active in the murine immune system, in contrast with its inert phenotype in the human system. hIgG2 bound to mFcγRIIb and mFcγRIII, and induced potent ADCC with mouse NK cells and mouse polymorphonuclear leukocytes. hIgG2 induced weak ADCC and, remarkably, was unable to induce Ab-dependent cellular phagocytosis with mouse macrophages. Finally, the isotypes were studied in s.c. and i.v. tumor xenograft models, which confirmed hIgG1 to be the most potent human isotype in mouse models. These data enhance our understanding of the crosstalk between hIgGs and murine effector cells, permitting a better interpretation of human Ab efficacy studies in mouse models.
Collapse
|
45
|
Parekh BS, Berger E, Sibley S, Cahya S, Xiao L, LaCerte MA, Vaillancourt P, Wooden S, Gately D. Development and validation of an antibody-dependent cell-mediated cytotoxicity-reporter gene assay. MAbs 2012; 4:310-8. [PMID: 22531445 DOI: 10.4161/mabs.19873] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Humanized monoclonal antibodies (mAbs) are the fastest growing class of biological therapeutics that are being developed for various medical indications, and more than 30 mAbs are already approved and in the market place. Antibody-dependent cell-mediated cytotoxicity (ADCC) is an important biological function attributed to the mechanism of action of several therapeutic antibodies, particularly oncology targeting mAbs. The ADCC assay is a complicated and highly variable assay. Thus, the use of an ADCC assay as a lot release test or a stability test for clinical trial batches of mAbs has been a substantial challenge to install in quality control laboratories. We describe here the development and validation of an alternate approach, an ADCC-reporter gene assay that is based on the key attributes of the PBMC-based ADCC assay. We tested the biological relevance of this assay using an anti-CD20 based model and demonstrated that this ADCC-reporter assay correlated well with standard ADCC assays when induced with the drugable human isotypes [IgG1, IgG2, IgG4, IgG4S > P (S228P) and IgG4PAA (S228P, F234A, L235A)] and with IgG1 isotype variants with varying amounts of fucosylation. This data demonstrates that the ADCC-reporter gene assay has performance characteristics (accuracy, precision and robustness) to be used not only as a potency assay for lot release and stability testing for antibody therapeutics, but also as a key assay for the characterization and process development of therapeutic molecules.
Collapse
Affiliation(s)
- Bhavin S Parekh
- BioProduct Research and Development, Eli Lilly and Company, Indianapolis, IN, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Castel V, Segura V, Cañete A. Treatment of high-risk neuroblastoma with anti-GD2 antibodies. Clin Transl Oncol 2012; 12:788-93. [DOI: 10.1007/s12094-010-0600-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
47
|
Zeitlin L, Pettitt J, Scully C, Bohorova N, Kim D, Pauly M, Hiatt A, Ngo L, Steinkellner H, Whaley KJ, Olinger GG. Enhanced potency of a fucose-free monoclonal antibody being developed as an Ebola virus immunoprotectant. Proc Natl Acad Sci U S A 2011; 108:20690-4. [PMID: 22143789 PMCID: PMC3251097 DOI: 10.1073/pnas.1108360108] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
No countermeasures currently exist for the prevention or treatment of the severe sequelae of Filovirus (such as Ebola virus; EBOV) infection. To overcome this limitation in our biodefense preparedness, we have designed monoclonal antibodies (mAbs) which could be used in humans as immunoprotectants for EBOV, starting with a murine mAb (13F6) that recognizes the heavily glycosylated mucin-like domain of the virion-attached glycoprotein (GP). Point mutations were introduced into the variable region of the murine mAb to remove predicted human T-cell epitopes, and the variable regions joined to human constant regions to generate a mAb (h-13F6) appropriate for development for human use. We have evaluated the efficacy of three variants of h-13F6 carrying different glycosylation patterns in a lethal mouse EBOV challenge model. The pattern of glycosylation of the various mAbs was found to correlate to level of protection, with aglycosylated h-13F6 providing the least potent efficacy (ED(50) = 33 μg). A version with typical heterogenous mammalian glycoforms (ED(50) = 11 μg) had similar potency to the original murine mAb. However, h-13F6 carrying complex N-glycosylation lacking core fucose exhibited superior potency (ED(50) = 3 μg). Binding studies using Fcγ receptors revealed enhanced binding of nonfucosylated h-13F6 to mouse and human FcγRIII. Together the results indicate the presence of Fc N-glycans enhances the protective efficacy of h-13F6, and that mAbs manufactured with uniform glycosylation and a higher potency glycoform offer promise as biodefense therapeutics.
Collapse
|
48
|
Souto JC, Vila L, Brú A. Polymorphonuclear neutrophils and cancer: intense and sustained neutrophilia as a treatment against solid tumors. Med Res Rev 2011; 31:311-63. [PMID: 19967776 DOI: 10.1002/med.20185] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Polymorphonuclear neutrophils (PMN) are the most abundant circulating immune cells and represent the first line of immune defense against infection. This review of the biomedical literature of the last 40 years shows that they also have a powerful antitumoral effect under certain circumstances. Typically, the microenvironment surrounding a solid tumor possesses many of the characteristics of chronic inflammation, a condition considered very favorable for tumor growth and spread. However, there are many circumstances that shift the chronic inflammatory state toward an acute inflammatory response around a tumor. This shift seems to convert PMN into very efficient anticancer effector cells. Clinical reports of unexpected antitumoral effects linked to the prolonged use of granulocyte colony-stimulating factor, which stimulates an intense and sustained neutrophilia, suggest that an easy way to fight solid tumors would be to encourage the development of intense peritumoral PMN infiltrates. Specifically designed clinical trials are urgently needed to evaluate the safety and efficacy of such drug-induced neutrophilia in patients with solid tumors. This antitumoral role of neutrophils may provide new avenues for the clinical treatment of cancer.
Collapse
Affiliation(s)
- Juan Carlos Souto
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | | | | |
Collapse
|
49
|
|
50
|
Surrogate approaches in development of monoclonal antibodies. Drug Discov Today 2009; 14:1159-65. [DOI: 10.1016/j.drudis.2009.09.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 12/31/2022]
|