1
|
Hou R, Ren S, Wang R, Różycki B, Hu J. Multiscale Simulations of Membrane Adhesion Mediated by CD47-SIRPα Complexes. J Chem Theory Comput 2025; 21:2030-2042. [PMID: 39960300 PMCID: PMC11866742 DOI: 10.1021/acs.jctc.4c01337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025]
Abstract
Adhesion of biological cells is essential for various processes, including tissue formation, immune responses, and signaling. It involves multiple length scales, ranging from nanometers to micrometers, which are characteristic of (a) the intercellular receptor-ligand binding that mediates the cell adhesion, (b) the spatial distribution of the receptor and ligand proteins in the membranes of adhering cells, (c) adhesion-induced deformations and thermal undulations of the membranes, (d) the overall size of the interface between adhering cells. Therefore, computer simulations of cell membrane adhesion require multiscale modeling and suitable approximations that capture the essential physics of the system under study. Here, we introduce such a multiscale approach to study membrane adhesion mediated by the CD47-SIRPα binding, which is an immunologically relevant process. The synergetic use of coarse-grained molecular dynamics simulations and mesoscale kinetic Monte Carlo simulations allows us to explore both equilibrium properties and dynamical behavior of adhering membranes on the relevant length scales between 1 nm and 1 μm on time scales ranging from 0.1 ns all the way up to about 20 s. The multiscale simulations not only reproduce available experimental data but also give quantitative predictions on binding-induced conformational changes of SIRPα and membrane-mediated cooperativity of the CD47-SIRPα binding as well as fluctuation-induced interactions between the CD47-SIRPα complexes. Our approach is applicable to various membrane proteins and provides invaluable data for comparison with experimental findings.
Collapse
Affiliation(s)
- Ruihan Hou
- Kuang
Yaming Honors School, Nanjing University, Nanjing 210023, China
- Department
of Polymer Science and Engineering, Key Laboratory of High Performance
Polymer Material and Technology of Ministry of Education, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
| | - Shuanglong Ren
- Kuang
Yaming Honors School, Nanjing University, Nanjing 210023, China
| | - Rong Wang
- Department
of Polymer Science and Engineering, Key Laboratory of High Performance
Polymer Material and Technology of Ministry of Education, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
| | - Bartosz Różycki
- Institute
of Physics, Polish Academy of Sciences, Aleja Lotników 32/46, Warsaw 02-668, Poland
| | - Jinglei Hu
- Kuang
Yaming Honors School, Nanjing University, Nanjing 210023, China
- Department
of Polymer Science and Engineering, Key Laboratory of High Performance
Polymer Material and Technology of Ministry of Education, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
| |
Collapse
|
2
|
In H, Park M, Lee H, Han KH. Immune Cell Engagers: Advancing Precision Immunotherapy for Cancer Treatment. Antibodies (Basel) 2025; 14:16. [PMID: 39982231 PMCID: PMC11843982 DOI: 10.3390/antib14010016] [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: 01/17/2025] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Immune cell engagers (ICEs) are an emerging class of immunotherapies designed to harness the immune system's anti-tumor potential through precise targeting and activation of immune effector cells. By engaging T cells, natural killer (NK) cells, and phagocytes, ICEs overcome challenges such as immune evasion and MHC downregulation, addressing critical barriers in cancer treatment. T-cell engagers (TCEs), led by bispecific T-cell engagers (BiTEs), dominate the field, with innovations such as half-life-extended BiTEs, trispecific antibodies, and checkpoint inhibitory T-cell engagers driving their application in hematologic and solid malignancies. NK cell engagers (NKCEs) and phagocyte cell engagers (PCEs) are rapidly progressing, drawing on NK cells' innate cytotoxicity and macrophages' phagocytic abilities to target tumors, particularly in immunosuppressive microenvironments. Since the FDA approval of Blinatumomab in 2014, ICEs have transformed the oncology landscape, with nine FDA-approved products and numerous candidates in clinical trials. Despite challenges such as toxicity, resistance, and limited efficacy in solid tumors, ongoing research into advanced platforms and combination therapies highlights the growing potential of ICEs to provide personalized, scalable, and effective cancer treatments. This review investigates the mechanisms, platforms, research trends, and clinical progress of ICEs, emphasizing their pivotal role in advancing precision immunotherapy and their promise as a cornerstone of next-generation cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Kyung Ho Han
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea
| |
Collapse
|
3
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
4
|
Bond A, Morrissey MA. Biochemical and biophysical mechanisms macrophages use to tune phagocytic appetite. J Cell Sci 2025; 138:JCS263513. [PMID: 39749603 PMCID: PMC11828473 DOI: 10.1242/jcs.263513] [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] [Indexed: 01/04/2025] Open
Abstract
Macrophages phagocytose, or eat, pathogens, dead cells and cancer cells. To activate phagocytosis, macrophages recognize 'eat me' signals like IgG and phosphatidylserine on the target cell surface. Macrophages must carefully adjust their phagocytic appetite to ignore non-specific or transient eat me signal exposure on healthy cells while still rapidly recognizing pathogens and debris. Depending on the context, macrophages can increase their appetite for phagocytosis, to prioritize an effective immune response, or decrease their appetite, to avoid damage to healthy tissue during homeostasis. In this Review, we discuss the biochemical and biophysical mechanisms that macrophages employ to increase or decrease their sensitivity or capacity for phagocytosis. We discuss evidence that macrophages tune their sensitivity via several mechanisms, including altering the balance of activating and inhibitory receptor expression, altering the availability of activating receptors, as well as influencing their clustering and mobility, and modulating inhibitory receptor location. We also highlight how membrane availability limits the capacity of macrophages for phagocytosis and discuss potential mechanisms to promote membrane recycling and increase phagocytic capacity. Overall, this Review highlights recent work detailing the molecular toolkit that macrophages use to alter their appetite.
Collapse
Affiliation(s)
- Annalise Bond
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| | - Meghan A. Morrissey
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
5
|
Dooling LJ, Anlaş AA, Tobin MP, Ontko NM, Marchena T, Wang M, Andrechak JC, Discher DE. Clustered macrophages cooperate to eliminate tumors via coordinated intrudopodia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613918. [PMID: 39345601 PMCID: PMC11430028 DOI: 10.1101/2024.09.19.613918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Macrophages often pervade solid tumors, but their nearest neighbor organization is understudied and potentially enables key functions such as phagocytosis. Here, we observe dynamic macrophage clusters in tumors under conditions that maximize cancer cell phagocytosis and use reductionist approaches to uncover pathways to cluster formation and roles for tumor-intrusive pseudopodia, which we term 'intrudopodia'. Macrophage clusters form over hours on low- adhesion substrates after M1 polarization with interferons, including T cell-derived cytokines, and yet clusters prove fluid on timescales of minutes. Clusters also sort from M2 macrophages that disperse on the same substrates. M1 macrophages upregulate specific cell-cell adhesion receptors but suppress actomyosin contractility, and while both pathways contribute to cluster formation, decreased cortical tension was predicted to unleash pseudopodia. Macrophage neighbors in tumor spheroids indeed extend intrudopodia between adjacent cancer cell junctions - at least when phagocytosis conditions are maximized, and coordinated intrudopodia help detach and individualize cancer cells for rapid engulfment. Macrophage clusters thereby provide a cooperative advantage for phagocytosis to overcome solid tumor cohesion.
Collapse
|
6
|
Jiang C, Sun H, Jiang Z, Tian W, Cang S, Yu J. Targeting the CD47/SIRPα pathway in malignancies: recent progress, difficulties and future perspectives. Front Oncol 2024; 14:1378647. [PMID: 39040441 PMCID: PMC11261161 DOI: 10.3389/fonc.2024.1378647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Since its initial report in 2015, CD47 has garnered significant attention as an innate immune checkpoint, raising expectations to become the next "PD-1." The optimistic early stages of clinical development spurred a flurry of licensing deals for CD47-targeted molecules and company mergers or acquisitions for related assets. However, a series of setbacks unfolded recently, starting with the July 2023 announcement of discontinuing the phase 3 ENHANCE study on Magrolimab plus Azacitidine for higher-risk myelodysplastic syndromes (MDS). Subsequently, in August 2023, the termination of the ASPEN-02 program, assessing Evorpacept in combination with Azacitidine in MDS patients, was disclosed due to insufficient improvement compared to Azacitidine alone. These setbacks have cast doubt on the feasibility of targeting CD47 in the industry. In this review, we delve into the challenges of developing CD47-SIRPα-targeted drugs, analyze factors contributing to the mentioned setbacks, discuss future perspectives, and explore potential solutions for enhancing CD47-SIRPα-targeted drug development.
Collapse
Affiliation(s)
- Chenyang Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Henan Provincial People’s Hospital, Henan University People’s Hospital and Zhengzhou University, Zhengzhou, Henan, China
| | - Hao Sun
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, China
| | - Shundong Cang
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Henan Provincial People’s Hospital, Henan University People’s Hospital and Zhengzhou University, Zhengzhou, Henan, China
| | - Jifeng Yu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Liu T, Zhang M, Zhang J, Kang N, Zheng L, Ding Z. Targeted Delivery of Macrophage Membrane Biomimetic Liposomes Through Intranasal Administration for Treatment of Ischemic Stroke. Int J Nanomedicine 2024; 19:6177-6199. [PMID: 38911498 PMCID: PMC11194020 DOI: 10.2147/ijn.s458656] [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: 01/09/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024] Open
Abstract
Purpose Ginsenoside Rg3 (Rg3) and Panax notoginseng saponins (PNS) can be used for ischemic stroke treatment, however, the lack of targeting to the ischemic region limits the therapeutic effect. To address this, we leveraged the affinity of macrophage membrane proteins for inflamed brain microvascular endothelial cells to develop a macrophage membrane-cloaked liposome loaded with Rg3 and PNS (MM-Lip-Rg3/PNS), which can precisely target brain lesion region through intranasal administration. Methods MM-Lip-Rg3/PNS was prepared by co-extrusion method and was performed by characterization, stability, surface protein, and morphology. The cellular uptake, immune escape ability, and blood-brain barrier crossing ability of MM-Lip-Rg3/PNS were studied in vitro. The in vivo brain targeting, biodistribution and anti-ischemic efficacy of MM-Lip-Rg3/PNS were evaluated in MACO rats, and we determined the diversity of the nasal brain pathway through the olfactory nerve blockade model in rats. Finally, the pharmacokinetics and brain targeting index of MM-Lip-Rg3/PNS were investigated. Results Our results indicated that MM-Lip-Rg3/PNS was spherical with a shell-core structure. MM-Lip-Rg3/PNS can avoid mononuclear phagocytosis, actively bind to inflammatory endothelial cells, and have the ability to cross the blood-brain barrier. Moreover, MM-Lip-Rg3/PNS could specifically target ischemic sites, even microglia, increase the cumulative number of drugs in the brain, improve the inflammatory environment of the brain, and reduce the infarct size. By comparing olfactory nerve-blocking rats with normal rats, it was found that there are direct and indirect pathways for nasal entry into the brain. Pharmacokinetics demonstrated that MM-Lip-Rg3/PNS exhibited stronger brain targeting and prolonged drug half-life. Conclusion MM-Lip-Rg3/PNS might contribute to the accumulation of Rg3 and PNS in the ischemic brain area to improve treatment efficacy. This biomimetic nano-drug delivery system provides a new and promising strategy for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Tianshu Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People’s Republic of China
| | - Mengfan Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jin Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People’s Republic of China
| | - Naijin Kang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People’s Republic of China
| | - Linlin Zheng
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People’s Republic of China
| | - Zhiying Ding
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
8
|
Sue M, Tsubaki T, Ishimoto Y, Hayashi S, Ishida S, Otsuka T, Isumi Y, Kawase Y, Yamaguchi J, Nakada T, Ishiguro J, Nakamura K, Kawaida R, Ohtsuka T, Wada T, Agatsuma T, Kawasaki N. Blockade of SIRPα-CD47 axis by anti-SIRPα antibody enhances anti-tumor activity of DXd antibody-drug conjugates. PLoS One 2024; 19:e0304985. [PMID: 38843278 PMCID: PMC11156334 DOI: 10.1371/journal.pone.0304985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
Signal regulatory protein alpha (SIRPα) is an immune inhibitory receptor on myeloid cells including macrophages and dendritic cells, which binds to CD47, a ubiquitous self-associated molecule. SIRPα-CD47 interaction is exploited by cancer cells to suppress anti-tumor activity of myeloid cells, therefore emerging as a novel immune checkpoint for cancer immunotherapy. In blood cancer, several SIRPα-CD47 blockers have shown encouraging monotherapy activity. However, the anti-tumor activity of SIRPα-CD47 blockers in solid tumors seems limited, suggesting the need for combination therapies to fully exploit the myeloid immune checkpoint in solid tumors. Here we tested whether combination of SIRPα-CD47 blocker with antibody-drug conjugate bearing a topoisomerase I inhibitor DXd (DXd-ADC) would enhance anti-tumor activity in solid tumors. To this end, DS-1103a, a newly developed anti-human SIRPα antibody (Ab), was assessed for the potential combination benefit with datopotamab deruxtecan (Dato-DXd) and trastuzumab deruxtecan (T-DXd), DXd-ADCs targeting human trophoblast cell-surface antigen 2 and human epidermal growth factor receptor 2, respectively. DS-1103a inhibited SIRPα-CD47 interaction and enhanced antibody-dependent cellular phagocytosis of Dato-DXd and T-DXd against human cancer cells. In a whole cancer cell vaccination model, vaccination with DXd-treated cancer cells led to activation of tumor-specific T cells when combined with an anti-mouse SIRPα (anti-mSIRPα) Ab, implying the benefit of combining DXd-ADCs with anti-SIRPα Ab on anti-tumor immunity. Furthermore, in syngeneic mouse models, both Dato-DXd and T-DXd combination with anti-mSIRPα Ab showed stronger anti-tumor activity over the monotherapies. Taken together, this study provides a preclinical rationale of novel therapies for solid tumors combining SIRPα-CD47 blockers with DXd-ADCs.
Collapse
Affiliation(s)
- Mayumi Sue
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Takuya Tsubaki
- Modality Research Laboratories III, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yoko Ishimoto
- Translational Science Department I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Shinko Hayashi
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Saori Ishida
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Takafumi Otsuka
- Research Innovation Planning Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yoshitaka Isumi
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yumi Kawase
- Discovery Research Laboratories V, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Junko Yamaguchi
- Discovery Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Takashi Nakada
- Modality Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Jun Ishiguro
- Discovery Research Laboratories V, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kensuke Nakamura
- Modality Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Reimi Kawaida
- Discovery Research Laboratories V, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Toshiaki Ohtsuka
- Discovery Research Laboratories V, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Teiji Wada
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | - Norihito Kawasaki
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
9
|
Lu J, He R, Liu Y, Zhang J, Xu H, Zhang T, Chen L, Yang G, Zhang J, Liu J, Chi H. Exploiting cell death and tumor immunity in cancer therapy: challenges and future directions. Front Cell Dev Biol 2024; 12:1416115. [PMID: 38887519 PMCID: PMC11180757 DOI: 10.3389/fcell.2024.1416115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Cancer remains a significant global challenge, with escalating incidence rates and a substantial burden on healthcare systems worldwide. Herein, we present an in-depth exploration of the intricate interplay between cancer cell death pathways and tumor immunity within the tumor microenvironment (TME). We begin by elucidating the epidemiological landscape of cancer, highlighting its pervasive impact on premature mortality and the pronounced burden in regions such as Asia and Africa. Our analysis centers on the pivotal concept of immunogenic cell death (ICD), whereby cancer cells succumbing to specific stimuli undergo a transformation that elicits robust anti-tumor immune responses. We scrutinize the mechanisms underpinning ICD induction, emphasizing the release of damage-associated molecular patterns (DAMPs) and tumor-associated antigens (TAAs) as key triggers for dendritic cell (DC) activation and subsequent T cell priming. Moreover, we explore the contributions of non-apoptotic RCD pathways, including necroptosis, ferroptosis, and pyroptosis, to tumor immunity within the TME. Emerging evidence suggests that these alternative cell death modalities possess immunogenic properties and can synergize with conventional treatments to bolster anti-tumor immune responses. Furthermore, we discuss the therapeutic implications of targeting the TME for cancer treatment, highlighting strategies to harness immunogenic cell death and manipulate non-apoptotic cell death pathways for therapeutic benefit. By elucidating the intricate crosstalk between cancer cell death and immune modulation within the TME, this review aims to pave the way for the development of novel cancer therapies that exploit the interplay between cell death mechanisms and tumor immunity and overcome Challenges in the Development and implementation of Novel Therapies.
Collapse
Affiliation(s)
- Jiaan Lu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Ru He
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yang Liu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jinghan Zhang
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Heng Xu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Tianchi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Li Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Jun Zhang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Hayes BH, Wang M, Zhu H, Phan SH, Dooling LJ, Andrechak JC, Chang AH, Tobin MP, Ontko NM, Marchena T, Discher DE. Chromosomal instability induced in cancer can enhance macrophage-initiated immune responses that include anti-tumor IgG. eLife 2024; 12:RP88054. [PMID: 38805560 PMCID: PMC11132682 DOI: 10.7554/elife.88054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
Solid tumors generally exhibit chromosome copy number variation, which is typically caused by chromosomal instability (CIN) in mitosis. The resulting aneuploidy can drive evolution and associates with poor prognosis in various cancer types as well as poor response to T-cell checkpoint blockade in melanoma. Macrophages and the SIRPα-CD47 checkpoint are understudied in such contexts. Here, CIN is induced in poorly immunogenic B16F10 mouse melanoma cells using spindle assembly checkpoint MPS1 inhibitors that generate persistent micronuclei and diverse aneuploidy while skewing macrophages toward a tumoricidal 'M1-like' phenotype based on markers and short-term anti-tumor studies. Mice bearing CIN-afflicted tumors with wild-type CD47 levels succumb similar to controls, but long-term survival is maximized by SIRPα blockade on adoptively transferred myeloid cells plus anti-tumor monoclonal IgG. Such cells are the initiating effector cells, and survivors make de novo anti-cancer IgG that not only promote phagocytosis of CD47-null cells but also suppress tumor growth. CIN does not affect the IgG response, but pairing CIN with maximal macrophage anti-cancer activity increases durable cures that possess a vaccination-like response against recurrence.
Collapse
Affiliation(s)
- Brandon H Hayes
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Mai Wang
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Hui Zhu
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Steven H Phan
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Lawrence J Dooling
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Jason C Andrechak
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Alexander H Chang
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Michael P Tobin
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Nicholas M Ontko
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Tristan Marchena
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Dennis E Discher
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
11
|
von Ofen AJ, Thiel U, Eck J, Gassmann H, Thiede M, Hauer J, Holm PS, Schober SJ. YB-1-based oncolytic virotherapy in combination with CD47 blockade enhances phagocytosis of pediatric sarcoma cells. Front Oncol 2024; 14:1304374. [PMID: 38357194 PMCID: PMC10865101 DOI: 10.3389/fonc.2024.1304374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024] Open
Abstract
Oncolytic viruses (OVs) selectively replicate in tumor cells resulting in lysis, spreading of new infectious units and induction of antitumor immune responses through abrogating an immunosuppressive tumor microenvironment (TME). Due to their mode of action, OVs are ideal combination partners with targeted immunotherapies. One highly attractive combination is the inhibition of the 'don't-eat-me'-signal CD47, which is known to increase the phagocytic potential of tumor-associated macrophages. In this work, we analyzed the combination approach consisting of the YB-1-based oncolytic adenovirus XVir-N-31 (XVir) and the CD47 inhibitor (CD47i) B6.H12.2 concerning its phagocytic potential. We investigate phagocytosis of XVir-, adenovirus wildtype (AdWT)-, and non-infected established pediatric sarcoma cell lines by different monocytic cells. Phagocytes (immature dendritic cells and macrophages) were derived from THP-1 cells and healthy human donors. Phagocytosis of tumor cells was assessed via FACS analysis in the presence and absence of CD47i. Additional characterization of T cell-stimulatory surface receptors as well as chemo-/cytokine analyses were performed. Furthermore, tumor cells were infected and studied for the surface expression of the 'eat-me'-signal calreticulin (CALR) and the 'don't-eat-me'-signal CD47. We herein demonstrate that (1) XVir-infected tumor cells upregulate both CALR and CD47. XVir induces higher upregulation of CD47 than AdWT. (2) XVir-infection enhances phagocytosis in general and (3) the combination of XVir and CD47i compared to controls showed by far superior enhancement of phagocytosis, tumor cell killing and innate immune activation. In conclusion, the combination of CD47i and XVir causes a significant increase in phagocytosis exceeding the monotherapies considerably accompanied by upregulation of T cell-stimulatory receptor expression and inflammatory chemo/-cytokine secretion.
Collapse
Affiliation(s)
- Anna Josefine von Ofen
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Uwe Thiel
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Jennifer Eck
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Hendrik Gassmann
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Melanie Thiede
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Julia Hauer
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Per Sonne Holm
- Department of Urology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Oral and Maxillofacial Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian J. Schober
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
12
|
Hayes BH, Wang M, Zhu H, Phan SH, Dooling LJ, Andrechak JC, Chang AH, Tobin MP, Ontko NM, Marchena T, Discher DE. Chromosomal instability can favor macrophage-mediated immune response and induce a broad, vaccination-like anti-tumor IgG response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.02.535275. [PMID: 37066426 PMCID: PMC10103980 DOI: 10.1101/2023.04.02.535275] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Chromosomal instability (CIN), a state in which cells undergo mitotic aberrations that generate chromosome copy number variations, generates aneuploidy and is thought to drive cancer evolution. Although associated with poor prognosis and reduced immune response, CIN generates aneuploidy-induced stresses that could be exploited for immunotherapies. In such contexts, macrophages and the CD47-SIRPα checkpoint are understudied. Here, CIN is induced pharmacologically induced in poorly immunogenic B16F10 mouse melanoma cells, generating persistent micronuclei and diverse aneuploidy while skewing macrophages towards an anti-cancer M1-like phenotype, based on RNA-sequencing profiling, surface marker expression and short-term antitumor studies. These results further translate to in vivo efficacy: Mice bearing CIN-afflicted tumors with wild-type CD47 levels survive only slightly longer relative to chromosomally stable controls, but long-term survival is maximized when combining macrophage-stimulating anti-tumor IgG opsonization and some form of disruption of the CD47-SIRPα checkpoint. Survivors make multi-epitope, de novo anti-cancer IgG that promote macrophage-mediated phagocytosis of CD47 knockout B16F10 cells and suppress tumoroids in vitro and growth of tumors in vivo . CIN does not greatly affect the level of the IgG response compared to previous studies but does significantly increase survival. These results highlight an unexpected therapeutic benefit from CIN when paired with maximal macrophage anti-cancer activity: an anti-cancer vaccination-like antibody response that can lead to more durable cures and further potentiate cell-mediated acquired immunity.
Collapse
|
13
|
Dooling LJ, Andrechak JC, Hayes BH, Kadu S, Zhang W, Pan R, Vashisth M, Irianto J, Alvey CM, Ma L, Discher DE. Cooperative phagocytosis of solid tumours by macrophages triggers durable anti-tumour responses. Nat Biomed Eng 2023; 7:1081-1096. [PMID: 37095318 PMCID: PMC10791169 DOI: 10.1038/s41551-023-01031-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/27/2023] [Indexed: 04/26/2023]
Abstract
In solid tumours, the abundance of macrophages is typically associated with a poor prognosis. However, macrophage clusters in tumour-cell nests have been associated with survival in some tumour types. Here, by using tumour organoids comprising macrophages and cancer cells opsonized via a monoclonal antibody, we show that highly ordered clusters of macrophages cooperatively phagocytose cancer cells to suppress tumour growth. In mice with poorly immunogenic tumours, the systemic delivery of macrophages with signal-regulatory protein alpha (SIRPα) genetically knocked out or else with blockade of the CD47-SIRPα macrophage checkpoint was combined with the monoclonal antibody and subsequently triggered the production of endogenous tumour-opsonizing immunoglobulin G, substantially increased the survival of the animals and helped confer durable protection from tumour re-challenge and metastasis. Maximizing phagocytic potency by increasing macrophage numbers, by tumour-cell opsonization and by disrupting the phagocytic checkpoint CD47-SIRPα may lead to durable anti-tumour responses in solid cancers.
Collapse
Affiliation(s)
- Lawrence J Dooling
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason C Andrechak
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Brandon H Hayes
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Siddhant Kadu
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - William Zhang
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruby Pan
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - Manasvita Vashisth
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Cory M Alvey
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - Leyuan Ma
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA.
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA.
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Montero E, Isenberg JS. The TSP1-CD47-SIRPα interactome: an immune triangle for the checkpoint era. Cancer Immunol Immunother 2023; 72:2879-2888. [PMID: 37217603 PMCID: PMC10412679 DOI: 10.1007/s00262-023-03465-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
The use of treatments, such as programmed death protein 1 (PD1) or cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies, that loosen the natural checks upon immune cell activity to enhance cancer killing have shifted clinical practice and outcomes for the better. Accordingly, the number of antibodies and engineered proteins that interact with the ligand-receptor components of immune checkpoints continue to increase along with their use. It is tempting to view these molecular pathways simply from an immune inhibitory perspective. But this should be resisted. Checkpoint molecules can have other cardinal functions relevant to the development and use of blocking moieties. Cell receptor CD47 is an example of this. CD47 is found on the surface of all human cells. Within the checkpoint paradigm, non-immune cell CD47 signals through immune cell surface signal regulatory protein alpha (SIRPα) to limit the activity of the latter, the so-called trans signal. Even so, CD47 interacts with other cell surface and soluble molecules to regulate biogas and redox signaling, mitochondria and metabolism, self-renewal factors and multipotency, and blood flow. Further, the pedigree of checkpoint CD47 is more intricate than supposed. High-affinity interaction with soluble thrombospondin-1 (TSP1) and low-affinity interaction with same-cell SIRPα, the so-called cis signal, and non-SIRPα ectodomains on the cell membrane suggests that multiple immune checkpoints converge at and through CD47. Appreciation of this may provide latitude for pathway-specific targeting and intelligent therapeutic effect.
Collapse
Affiliation(s)
- Enrique Montero
- Department of Diabetes Immunology, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, USA
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, USA
| | - Jeffrey S Isenberg
- Department of Diabetes Complications and Metabolism, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, USA.
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
15
|
Hayes BH, Zhu H, Andrechak JC, Dooling LJ, Discher DE. Titrating CD47 by mismatch CRISPR-interference reveals incomplete repression can eliminate IgG-opsonized tumors but limits induction of antitumor IgG. PNAS NEXUS 2023; 2:pgad243. [PMID: 37593202 PMCID: PMC10427748 DOI: 10.1093/pnasnexus/pgad243] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/22/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
Phagocytic elimination of solid tumors by innate immune cells seems attractive for immunotherapy, particularly because of the possibilities for acquired immunity. However, the approach remains challenging, with blockade of the macrophage checkpoint CD47 working in immunodeficient mice and against highly immunogenic tumors but not in the clinic where tumors are poorly immunogenic. Even when mouse tumors of poorly immunogenic B16F10 melanoma are opsonized to drive engulfment with a suitable monoclonal antibody (mAb), anti-CD47 blockade remains insufficient. Using both in vitro immuno-tumoroids and in vivo mouse models, we show with CRISPR interference (CRISPRi) that a relatively uniform minimum repression of CD47 by 80% is needed for phagocytosis to dominate net growth when combined with an otherwise ineffective mAb (anti-Tyrp1). Heterogeneity enriches for CD47-high cells, but mice that eliminate tumors generate prophagocytic IgGs that increase in titer with CD47 repression and with tumor accumulation of macrophages, although deeper repression does not improve survival. Given well-known limitations of antibody permeation into solid tumors, our studies clarify benchmarks for CD47 disruption that should be more clinically feasible and safer but just as effective as complete ablation. Additionally, safe but ineffective opsonization in human melanoma trials suggests that combinations with deep repression of CD47 could prove effective and initiate durable immunity.
Collapse
Affiliation(s)
- Brandon H Hayes
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hui Zhu
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason C Andrechak
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lawrence J Dooling
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
16
|
Xu S, Wang C, Yang L, Wu J, Li M, Xiao P, Xu Z, Xu Y, Wang K. Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy. Front Immunol 2023; 14:1199631. [PMID: 37313405 PMCID: PMC10258331 DOI: 10.3389/fimmu.2023.1199631] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/16/2023] [Indexed: 06/15/2023] Open
Abstract
Unprecedented breakthroughs have been made in cancer immunotherapy in recent years. Particularly immune checkpoint inhibitors have fostered hope for patients with cancer. However, immunotherapy still exhibits certain limitations, such as a low response rate, limited efficacy in certain populations, and adverse events in certain tumors. Therefore, exploring strategies that can improve clinical response rates in patients is crucial. Tumor-associated macrophages (TAMs) are the predominant immune cells that infiltrate the tumor microenvironment and express a variety of immune checkpoints that impact immune functions. Mounting evidence indicates that immune checkpoints in TAMs are closely associated with the prognosis of patients with tumors receiving immunotherapy. This review centers on the regulatory mechanisms governing immune checkpoint expression in macrophages and strategies aimed at improving immune checkpoint therapies. Our review provides insights into potential therapeutic targets to improve the efficacy of immune checkpoint blockade and key clues to developing novel tumor immunotherapies.
Collapse
Affiliation(s)
- Shumin Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Chenyang Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lingge Yang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Jiaji Wu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Mengshu Li
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Peng Xiao
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Yun Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
17
|
Erdem N, Chen KT, Qi M, Zhao Y, Wu X, Garcia I, Ku HT, Montero E, Al-Abdullah IH, Kandeel F, Roep BO, Isenberg JS. Thrombospondin-1, CD47, and SIRPα display cell-specific molecular signatures in human islets and pancreata. Am J Physiol Endocrinol Metab 2023; 324:E347-E357. [PMID: 36791324 PMCID: PMC11967708 DOI: 10.1152/ajpendo.00221.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/10/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
Thrombospondin-1 (TSP1) is a secreted protein minimally expressed in health but increased in disease and age. TSP1 binds to the cell membrane receptor CD47, which itself engages signal regulatory protein α (SIRPα), and the latter creates a checkpoint for immune activation. Individuals with cancer administered checkpoint-blocking molecules developed insulin-dependent diabetes. Relevant to this, CD47 blocking antibodies and SIRPα fusion proteins are in clinical trials. We characterized the molecular signature of TSP1, CD47, and SIRPα in human islets and pancreata. Fresh islets and pancreatic tissue from nondiabetic individuals were obtained. The expression of THBS1, CD47, and SIRPA was determined using single-cell mRNA sequencing, immunofluorescence microscopy, Western blot, and flow cytometry. Islets were exposed to diabetes-affiliated inflammatory cytokines and changes in protein expression were determined. CD47 mRNA was expressed in all islet cell types. THBS1 mRNA was restricted primarily to endothelial and mesenchymal cells, whereas SIRPA mRNA was found mostly in macrophages. Immunofluorescence staining showed CD47 protein expressed by β cells and present in the exocrine pancreas. TSP1 and SIRPα proteins were not seen in islets or the exocrine pancreas. Western blot and flow cytometry confirmed immunofluorescent expression patterns. Importantly, human islets produced substantial quantities of secreted TSP1. Human pancreatic exocrine and endocrine tissue expressed CD47, whereas fresh islets displayed cell surface CD47 and secreted TSP1 at baseline and in inflammation. These findings suggest unexpected effects on islets from agents that intersect TSP1-CD47-SIRPα.NEW & NOTEWORTHY CD47 is a cell surface receptor with two primary ligands, soluble thrombospondin-1 (TSP1) and cell surface signal regulatory protein alpha (SIRPα). Both interactions provide checkpoints for immune cell activity. We determined that fresh human islets display CD47 and secrete TSP1. However, human islet endocrine cells lack SIRPα. These gene signatures are likely important given the increasing use of CD47 and SIRPα blocking molecules in individuals with cancer.
Collapse
Affiliation(s)
- Neslihan Erdem
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
- Department of Molecular & Cellular Endocrinology, City of Hope National Medical Center, Duarte, California, United States
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Kuan-Tsen Chen
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Meirigeng Qi
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Yuqi Zhao
- Integrative Genomics Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Isaac Garcia
- Department of Molecular & Cellular Endocrinology, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Hsun Teresa Ku
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Enrique Montero
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Ismail H Al-Abdullah
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Bart O Roep
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Jeffrey S Isenberg
- Department of Diabetes Complications & Metabolism, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| |
Collapse
|
18
|
Yang H, Xun Y, You H. The landscape overview of CD47-based immunotherapy for hematological malignancies. Biomark Res 2023; 11:15. [PMID: 36726125 PMCID: PMC9893585 DOI: 10.1186/s40364-023-00456-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/22/2023] [Indexed: 02/03/2023] Open
Abstract
Extensive clinical and experimental evidence suggests that macrophages play a crucial role in cancer immunotherapy. Cluster of differentiation (CD) 47, which is found on both healthy and malignant cells, regulates macrophage-mediated phagocytosis by sending a "don't eat me" signal to the signal regulatory protein alpha (SIRPα) receptor. Increasing evidence demonstrates that blocking CD47 interaction with SIRPα can enhance cancer cell clearance by macrophages. Additionally, inhibition of CD47/SIRPα interaction can increase antigen cross-presentation, leading to T-cell priming and an activated adaptive antitumor immune response. Therefore, inhibiting CD47/SIRPα axis has a significant impact on tumor immunotherapy. Studies on CD47 monoclonal antibodies are at the forefront of research, and impressive results have been obtained. Nevertheless, hematotoxicity, especially anemia, has become the most common adverse effect of the CD47 monoclonal antibody. More specific targeted drugs (i.e., bispecific antibodies, SIRPα/Fc fusion protein antibodies, and small-molecule inhibitors) have been developed to reduce hematotoxicity. Here, we review the present usage of CD47 antagonists for the treatment of lymphomas and hematologic neoplasms from the perspectives of structure, function, and clinical trials, including a comprehensive overview of the drugs in development.
Collapse
Affiliation(s)
- Hua Yang
- grid.443369.f0000 0001 2331 8060Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, Guangdong Province 528000 China
| | - Yang Xun
- grid.443369.f0000 0001 2331 8060Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, Guangdong Province 528000 China
| | - Hua You
- grid.488412.3Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Children’s Hospital of Chongqing Medical University, Chongqing, 401122 China ,grid.488412.3Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 401122 China
| |
Collapse
|
19
|
Jiang D, Burger CA, Akhanov V, Liang JH, Mackin RD, Albrecht NE, Andrade P, Schafer DP, Samuel MA. Neuronal signal-regulatory protein alpha drives microglial phagocytosis by limiting microglial interaction with CD47 in the retina. Immunity 2022; 55:2318-2335.e7. [PMID: 36379210 PMCID: PMC9772037 DOI: 10.1016/j.immuni.2022.10.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022]
Abstract
Microglia utilize their phagocytic activity to prune redundant synapses and refine neural circuits during precise developmental periods. However, the neuronal signals that control this phagocytic clockwork remain largely undefined. Here, we show that neuronal signal-regulatory protein alpha (SIRPα) is a permissive cue for microglial phagocytosis in the developing murine retina. Removal of neuronal, but not microglial, SIRPα reduced microglial phagocytosis, increased synpase numbers, and impaired circuit function. Conversely, prolonging neuronal SIRPα expression extended developmental microglial phagocytosis. These outcomes depended on the interaction of presynaptic SIRPα with postsynaptic CD47. Global CD47 deficiency modestly increased microglial phagocytosis, while CD47 overexpression reduced it. This effect was rescued by coexpression of neuronal SIRPα or codeletion of neuronal SIRPα and CD47. These data indicate that neuronal SIRPα regulates microglial phagocytosis by limiting microglial SIRPα access to neuronal CD47. This discovery may aid our understanding of synapse loss in neurological diseases.
Collapse
Affiliation(s)
- Danye Jiang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Courtney A Burger
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Viktor Akhanov
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Justine H Liang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert D Mackin
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nicholas E Albrecht
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pilar Andrade
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Melanie A Samuel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Shi H, Wang X, Li F, Gerlach BD, Yurdagul A, Moore MP, Zeldin S, Zhang H, Cai B, Zheng Z, Valenti L, Tabas I. CD47-SIRPα axis blockade in NASH promotes necroptotic hepatocyte clearance by liver macrophages and decreases hepatic fibrosis. Sci Transl Med 2022; 14:eabp8309. [PMID: 36417485 PMCID: PMC10199725 DOI: 10.1126/scitranslmed.abp8309] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Necroptosis contributes to hepatocyte death in nonalcoholic steatohepatitis (NASH), but the fate and roles of necroptotic hepatocytes (necHCs) in NASH remain unknown. We show here that the accumulation of necHCs in human and mouse NASH liver is associated with an up-regulation of the "don't-eat-me" ligand CD47 on necHCs, but not on apoptotic hepatocytes, and an increase in the CD47 receptor SIRPα on liver macrophages, consistent with impaired macrophage-mediated clearance of necHCs. In vitro, necHC clearance by primary liver macrophages was enhanced by treatment with either anti-CD47 or anti-SIRPα. In a proof-of-concept mouse model of inducible hepatocyte necroptosis, anti-CD47 antibody treatment increased necHC uptake by liver macrophages and inhibited markers of hepatic stellate cell (HSC) activation, which is responsible for liver fibrogenesis. Treatment of two mouse models of diet-induced NASH with anti-CD47, anti-SIRPα, or AAV8-H1-shCD47 to silence CD47 in hepatocytes increased the uptake of necHC by liver macrophages and decreased markers of HSC activation and liver fibrosis. Anti-SIRPα treatment avoided the adverse effect of anemia found in anti-CD47-treated mice. These findings provide evidence that impaired clearance of necHCs by liver macrophages due to CD47-SIRPα up-regulation contributes to fibrotic NASH, and suggest therapeutic blockade of the CD47-SIRPα axis as a strategy to decrease the accumulation of necHCs in NASH liver and dampen the progression of hepatic fibrosis.
Collapse
Affiliation(s)
- Hongxue Shi
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Fang Li
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brennan D. Gerlach
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mary P. Moore
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sharon Zeldin
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Bishuang Cai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ze Zheng
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano and Fondazione Ca’ Granda Ospedale Maggiore Policlinico Milano, Milano 20122, Italy
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
21
|
Jalil AR, Andrechak JC, Hayes BH, Chenoweth DM, Discher DE. Human CD47-Derived Cyclic Peptides Enhance Engulfment of mAb-Targeted Melanoma by Primary Macrophages. Bioconjug Chem 2022; 33:1973-1982. [PMID: 35285229 PMCID: PMC10805119 DOI: 10.1021/acs.bioconjchem.2c00020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD47 on healthy cells, cancer cells, and even engineered particles can inhibit phagocytic clearance by binding SIRPα on macrophages. To mimic and modulate this interaction with peptides that could be used as soluble antagonists or potentially as bioconjugates to surfaces, we made cyclic "nano-Self" peptides based on the key interaction loop of human CD47. Melanoma cells were studied as a standard preclinical cancer model and were antibody-opsonized to adhere to and activate engulfment by primary mouse macrophages. Phagocytosis in the presence of soluble peptides showed cyclic > wildtype > scrambled activity, with the same trend observed with human cells. Opsonized cells that were not engulfed adhered tightly to macrophages, with opposite trends to phagocytosis. Peptide activity is nonetheless higher in human versus mouse assays, consistent with species differences in CD47-SIRPα. Small peptides thus function as soluble antagonists of a major macrophage checkpoint.
Collapse
|
22
|
Jalil AR, Tobin MP, Discher DE. Suppressing or Enhancing Macrophage Engulfment through the Use of CD47 and Related Peptides. Bioconjug Chem 2022; 33:1989-1995. [PMID: 35316023 PMCID: PMC9990087 DOI: 10.1021/acs.bioconjchem.2c00019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Foreign particles and microbes are rapidly cleared by macrophages in vivo, although many key aspects of uptake mechanisms remain unclear. "Self" cells express CD47 which functions as an anti-phagocytic ligand for SIRPα on macrophages, particularly when pro-phagocytic ligands such as antibodies are displayed in parallel. Here, we review CD47 and related "Self" peptides as modulators of macrophage uptake. Nanoparticles conjugated with either CD47 or peptides derived from its SIRPα binding site can suppress phagocytic uptake by macrophages in vitro and in vivo, with similar findings for CD47-displaying viruses. Drugs, dyes, and genes as payloads thus show increased delivery to targeted cells. On the other hand, CD47 expression by cancer cells enables such cells to evade macrophages and immune surveillance. This has motivated development of soluble antagonists to CD47-SIRPα, ranging from blocking antibodies in the clinic to synthetic peptides in preclinical models. CD47 and peptides are thus emerging as dual-use phagocytosis modulators against diseases.
Collapse
|
23
|
Shilts J, Severin Y, Galaway F, Müller-Sienerth N, Chong ZS, Pritchard S, Teichmann S, Vento-Tormo R, Snijder B, Wright GJ. A physical wiring diagram for the human immune system. Nature 2022; 608:397-404. [PMID: 35922511 PMCID: PMC9365698 DOI: 10.1038/s41586-022-05028-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 06/28/2022] [Indexed: 12/14/2022]
Abstract
The human immune system is composed of a distributed network of cells circulating throughout the body, which must dynamically form physical associations and communicate using interactions between their cell-surface proteomes1. Despite their therapeutic potential2, our map of these surface interactions remains incomplete3,4. Here, using a high-throughput surface receptor screening method, we systematically mapped the direct protein interactions across a recombinant library that encompasses most of the surface proteins that are detectable on human leukocytes. We independently validated and determined the biophysical parameters of each novel interaction, resulting in a high-confidence and quantitative view of the receptor wiring that connects human immune cells. By integrating our interactome with expression data, we identified trends in the dynamics of immune interactions and constructed a reductionist mathematical model that predicts cellular connectivity from basic principles. We also developed an interactive multi-tissue single-cell atlas that infers immune interactions throughout the body, revealing potential functional contexts for new interactions and hubs in multicellular networks. Finally, we combined targeted protein stimulation of human leukocytes with multiplex high-content microscopy to link our receptor interactions to functional roles, in terms of both modulating immune responses and maintaining normal patterns of intercellular associations. Together, our work provides a systematic perspective on the intercellular wiring of the human immune system that extends from systems-level principles of immune cell connectivity down to mechanistic characterization of individual receptors, which could offer opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jarrod Shilts
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, UK.
| | - Yannik Severin
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Francis Galaway
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, UK
| | | | - Zheng-Shan Chong
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, UK
| | - Sophie Pritchard
- Cellular Genetics Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Sarah Teichmann
- Cellular Genetics Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Roser Vento-Tormo
- Cellular Genetics Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Berend Snijder
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, UK.
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
24
|
Andrechak JC, Dooling LJ, Tobin MP, Zhang W, Hayes BH, Lee JY, Jin X, Irianto J, Discher DE. CD47-SIRPα Checkpoint Disruption in Metastases Requires Tumor-Targeting Antibody for Molecular and Engineered Macrophage Therapies. Cancers (Basel) 2022; 14:1930. [PMID: 35454837 PMCID: PMC9026896 DOI: 10.3390/cancers14081930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/17/2022] Open
Abstract
The macrophage checkpoint interaction CD47-SIRPα is an emerging target for cancer therapy, but clinical trials of monoclonal anti-CD47 show efficacy only in liquid tumors when combined with tumor-opsonizing IgG. Here, in challenging metastatic solid tumors, CD47 deletion shows no effect on tumor growth unless combined with otherwise ineffective tumor-opsonization, and we likewise show wild-type metastases are suppressed by SIRPα-blocked macrophages plus tumor-opsonization. Lung tumor nodules of syngeneic B16F10 melanoma cells with CD47 deletion show opsonization drives macrophage phagocytosis of B16F10s, consistent with growth versus phagocytosis calculus for exponential suppression of cancer. Wild-type CD47 levels on metastases in lungs of immunocompetent mice and on human metastases in livers of immunodeficient mice show that systemic injection of antibody-engineered macrophages also suppresses growth. Such in vivo functionality can be modulated by particle pre-loading of the macrophages. Thus, even though CD47-SIRPα disruption and tumor-opsonizing IgG are separately ineffective against established metastatic solid tumors, their combination in molecular and cellular therapies prolongs survival.
Collapse
Affiliation(s)
- Jason C. Andrechak
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lawrence J. Dooling
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
| | - Michael P. Tobin
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William Zhang
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brandon H. Hayes
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justine Y. Lee
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
| | - Xiaoling Jin
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
| | - Jerome Irianto
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Dennis E. Discher
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Billerhart M, Schönhofer M, Schueffl H, Polzer W, Pichler J, Decker S, Taschauer A, Maier J, Anton M, Eckmann S, Blaschek M, Heffeter P, Sami H, Ogris M. CD47-targeted cancer immunogene therapy: Secreted SIRPα-Fc fusion protein eradicates tumors by macrophage and NK cell activation. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:192-204. [PMID: 34729396 PMCID: PMC8526499 DOI: 10.1016/j.omto.2021.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022]
Abstract
CD47 protects healthy cells from macrophage attack by binding to signal regulatory protein α (SIRPα), while its upregulation in cancer prevents immune clearance. Systemic treatment with CD47 antibodies requires a weakened Fc-mediated effector function or lower CD47-binding affinity to prevent side effects. Our approach combines “the best of both worlds,” i.e., maximized CD47 binding and full Fc-mediated immune activity, by exploiting gene therapy for paracrine release. We developed a plasmid vector encoding for the secreted fusion protein sCV1-hIgG1, comprising highly efficient CD47-blocking moiety CV1 and Fc domain of human immunoglobulin G1 (IgG1) with maximized immune activation. sCV1-hIgG1 exhibited a potent bystander effect, blocking CD47 on all cells via fusion protein secreted from only a fraction of cells or when transferring transfection supernatant to untransfected cells. The CpG-free plasmid ensured sustained secretion of sCV1-hIgG1. In orthotopic human triple-negative breast cancer in CB17-severe combined immunodeficiency (SCID) mice, ex vivo transfection significantly delayed tumor growth and eradicated one-third of tumors. In intratumoral transfection experiments, CD47 blockage and increased migration of macrophages into the tumor were observed within 17 h of a single injection. Natural killer (NK) cell-mediated lysis of sCV1-hIgG1-expressing cells was demonstrated in vitro. Taken together, this approach also opens the opportunity to block, in principle, any immune checkpoints.
Collapse
Affiliation(s)
- Magdalena Billerhart
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Monika Schönhofer
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Hemma Schueffl
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfram Polzer
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Julia Pichler
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Simon Decker
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Alexander Taschauer
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Julia Maier
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Martina Anton
- Institutes of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81675 Munich, Germany
| | - Sebastian Eckmann
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Manuel Blaschek
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Haider Sami
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Manfred Ogris
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
26
|
Zhao C, Lv H, Tao S, Zhang T, Xu N, Zhu L. Exosomes: Promising nanocarrier for cancer therapy. NANO SELECT 2021. [DOI: 10.1002/nano.202100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Chen‐Chen Zhao
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Hui‐Zhong Lv
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Su‐Wan Tao
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Tong‐Cun Zhang
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Na Xu
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Lian Zhu
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
- School of Chemical and Environmental Engineering Wuhan Polytechnic University Wuhan Hubei China
| |
Collapse
|
27
|
Sharp RC, Brown ME, Shapiro MR, Posgai AL, Brusko TM. The Immunoregulatory Role of the Signal Regulatory Protein Family and CD47 Signaling Pathway in Type 1 Diabetes. Front Immunol 2021; 12:739048. [PMID: 34603322 PMCID: PMC8481641 DOI: 10.3389/fimmu.2021.739048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Background The pathogenesis of type 1 diabetes (T1D) involves complex genetic susceptibility that impacts pathways regulating host immunity and the target of autoimmune attack, insulin-producing pancreatic β-cells. Interactions between risk variants and environmental factors result in significant heterogeneity in clinical presentation among those who develop T1D. Although genetic risk is dominated by the human leukocyte antigen (HLA) class II and insulin (INS) gene loci, nearly 150 additional risk variants are significantly associated with the disease, including polymorphisms in immune checkpoint molecules, such as SIRPG. Scope of Review In this review, we summarize the literature related to the T1D-associated risk variants in SIRPG, which include a protein-coding variant (rs6043409, G>A; A263V) and an intronic polymorphism (rs2281808, C>T), and their potential impacts on the immunoregulatory signal regulatory protein (SIRP) family:CD47 signaling axis. We discuss how dysregulated expression or function of SIRPs and CD47 in antigen-presenting cells (APCs), T cells, natural killer (NK) cells, and pancreatic β-cells could potentially promote T1D development. Major Conclusions We propose a hypothesis, supported by emerging genetic and functional immune studies, which states a loss of proper SIRP:CD47 signaling may result in increased lymphocyte activation and cytotoxicity and enhanced β-cell destruction. Thus, we present several novel therapeutic strategies for modulation of SIRPs and CD47 to intervene in T1D.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- CD47 Antigen/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Genetic Association Studies
- Humans
- Immunotherapy
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Polymorphism, Genetic
- Receptors, Cell Surface/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Robert C. Sharp
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Matthew E. Brown
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Melanie R. Shapiro
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
28
|
Freeman S, Grinstein S. Promoters and Antagonists of Phagocytosis: A Plastic and Tunable Response. Annu Rev Cell Dev Biol 2021; 37:89-114. [PMID: 34152790 DOI: 10.1146/annurev-cellbio-120219-055903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent observations indicate that, rather than being an all-or-none response, phagocytosis is finely tuned by a host of developmental and environmental factors. The expression of key phagocytic determinants is regulated via transcriptional and epigenetic means that confer memory on the process. Membrane traffic, the cytoskeleton, and inside-out signaling control the activation of phagocytic receptors and their ability to access their targets. An exquisite extra layer of complexity is introduced by the coexistence of distinct "eat-me" and "don't-eat-me" signals on targets and of corresponding "eat" and "don't-eat" receptors on the phagocyte surface. Moreover, assorted physical barriers constitute "don't-come-close-to-me" hurdles that obstruct the engagement of ligands by receptors. The expression, mobility, and accessibility of all these determinants can be modulated, conferring extreme plasticity on phagocytosis and providing attractive targets for therapeutic intervention in cancer, atherosclerosis, and dementia. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Spencer Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario M5G0A4, Canada; , .,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario M5G0A4, Canada; , .,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
29
|
Roberts DD, Isenberg JS. CD47 and thrombospondin-1 regulation of mitochondria, metabolism, and diabetes. Am J Physiol Cell Physiol 2021; 321:C201-C213. [PMID: 34106789 DOI: 10.1152/ajpcell.00175.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Thrombospondin-1 (TSP1) is the prototypical member of a family of secreted proteins that modulate cell behavior by engaging with molecules in the extracellular matrix and with receptors on the cell surface. CD47 is widely displayed on many, if not all, cell types and is a high-affinity TSP1 receptor. CD47 is a marker of self that limits innate immune cell activities, a feature recently exploited to enhance cancer immunotherapy. Another major role for CD47 in health and disease is to mediate TSP1 signaling. TSP1 acting through CD47 contributes to mitochondrial, metabolic, and endocrine dysfunction. Studies in animal models found that elevated TSP1 expression, acting in part through CD47, causes mitochondrial and metabolic dysfunction. Clinical studies established that abnormal TSP1 expression positively correlates with obesity, fatty liver disease, and diabetes. The unabated increase in these conditions worldwide and the availability of CD47 targeting drugs justify a closer look into how TSP1 and CD47 disrupt metabolic balance and the potential for therapeutic intervention.
Collapse
Affiliation(s)
- David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
30
|
Kelley SM, Ravichandran KS. Putting the brakes on phagocytosis: "don't-eat-me" signaling in physiology and disease. EMBO Rep 2021; 22:e52564. [PMID: 34041845 DOI: 10.15252/embr.202152564] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Timely removal of dying or pathogenic cells by phagocytes is essential to maintaining host homeostasis. Phagocytes execute the clearance process with high fidelity while sparing healthy neighboring cells, and this process is at least partially regulated by the balance of "eat-me" and "don't-eat-me" signals expressed on the surface of host cells. Upon contact, eat-me signals activate "pro-phagocytic" receptors expressed on the phagocyte membrane and signal to promote phagocytosis. Conversely, don't-eat-me signals engage "anti-phagocytic" receptors to suppress phagocytosis. We review the current knowledge of don't-eat-me signaling in normal physiology and disease contexts where aberrant don't-eat-me signaling contributes to pathology.
Collapse
Affiliation(s)
- Shannon M Kelley
- Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.,VIB-UGent Center for Inflammation Research, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
31
|
Miah M, Goh I, Haniffa M. Prenatal Development and Function of Human Mononuclear Phagocytes. Front Cell Dev Biol 2021; 9:649937. [PMID: 33898444 PMCID: PMC8060508 DOI: 10.3389/fcell.2021.649937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
The human mononuclear phagocyte (MP) system, which includes dendritic cells, monocytes, and macrophages, is a critical regulator of innate and adaptive immune responses. During embryonic development, MPs derive sequentially in yolk sac progenitors, fetal liver, and bone marrow haematopoietic stem cells. MPs maintain tissue homeostasis and confer protective immunity in post-natal life. Recent evidence - primarily in animal models - highlight their critical role in coordinating the remodeling, maturation, and repair of target organs during embryonic and fetal development. However, the molecular regulation governing chemotaxis, homeostasis, and functional diversification of resident MP cells in their respective organ systems during development remains elusive. In this review, we summarize the current understanding of the development and functional contribution of tissue MPs during human organ development and morphogenesis and its relevance to regenerative medicine. We outline how single-cell multi-omic approaches and next-generation ex-vivo organ-on-chip models provide new experimental platforms to study the role of human MPs during development and disease.
Collapse
Affiliation(s)
- Mohi Miah
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Issac Goh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
32
|
Andrejeva G, Capoccia BJ, Hiebsch RR, Donio MJ, Darwech IM, Puro RJ, Pereira DS. Novel SIRPα Antibodies That Induce Single-Agent Phagocytosis of Tumor Cells while Preserving T Cells. THE JOURNAL OF IMMUNOLOGY 2021; 206:712-721. [PMID: 33431660 DOI: 10.4049/jimmunol.2001019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
The signal regulatory protein α (SIRPα)/CD47 axis has emerged as an important innate immune checkpoint that enables cancer cell escape from macrophage phagocytosis. SIRPα expression is limited to macrophages, dendritic cells, and neutrophils-cells enriched in the tumor microenvironment. In this study, we present novel anti-SIRP Abs, SIRP-1 and SIRP-2, as an approach to targeting the SIRPα/CD47 axis. Both SIRP-1 and SIRP-2 bind human macrophage SIRPα variants 1 and 2, the most common variants in the human population. SIRP-1 and SIRP-2 are differentiated among reported anti-SIRP Abs in that they induce phagocytosis of solid and hematologic tumor cell lines by human monocyte-derived macrophages as single agents. We demonstrate that SIRP-1 and SIRP-2 disrupt SIRPα/CD47 interaction by two distinct mechanisms: SIRP-1 directly blocks SIRPα/CD47 and induces internalization of SIRPα/Ab complexes that reduce macrophage SIRPα surface levels and SIRP-2 acts via disruption of higher-order SIRPα structures on macrophages. Both SIRP-1 and SIRP-2 engage FcγRII, which is required for single-agent phagocytic activity. Although SIRP-1 and SIRP-2 bind SIRPγ with varying affinity, they show no adverse effects on T cell proliferation. Finally, both Abs also enhance phagocytosis when combined with tumor-opsonizing Abs, including a highly differentiated anti-CD47 Ab, AO-176, currently being evaluated in phase 1 clinical trials, NCT03834948 and NCT04445701 SIRP-1 and SIRP-2 are novel, differentiated SIRP Abs that induce in vitro single-agent and combination phagocytosis and show no adverse effects on T cell functionality. These data support their future development, both as single agents and in combination with other anticancer drugs.
Collapse
|
33
|
Jalil AR, Hayes BH, Andrechak JC, Xia Y, Chenoweth DM, Discher DE. Multivalent, Soluble Nano-Self Peptides Increase Phagocytosis of Antibody-Opsonized Targets while Suppressing "Self" Signaling. ACS NANO 2020; 14:15083-15093. [PMID: 33186026 PMCID: PMC8489566 DOI: 10.1021/acsnano.0c05091] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Macrophages engulf "foreign" cells and particles, but phagocytosis of healthy cells and cancer cells is inhibited by expression of the ubiquitous membrane protein CD47 which binds SIRPα on macrophages to signal "self". Motivated by some clinical efficacy of anti-CD47 against liquid tumors and based on past studies of CD47-derived polypeptides on particles that inhibited phagocytosis of the particles, here we design soluble, multivalent peptides to bind and block SIRPα. Bivalent and tetravalent nano-Self peptides prove more potent (Keff ∼ 10 nM) than monovalent 8-mers as agonists for phagocytosis of antibody opsonized cells, including cancer cells. Multivalent peptides also outcompete soluble CD47 binding to human macrophages, consistent with SIRPα binding, and the peptides suppress phosphotyrosine in macrophages, consistent with inhibition of SIRPα's "self" signaling. Peptides exhibit minimal folding, but functionality suggests an induced fit into SIRPα's binding pocket. Pre-clinical studies in mice indicate safety, with no anemia that typifies clinical infusions of anti-CD47. Multivalent nano-Self peptides thus constitute an alternative approach to promoting phagocytosis of "self", including cancer cells targeted clinically.
Collapse
|
34
|
Cham LB, Adomati T, Li F, Ali M, Lang KS. CD47 as a Potential Target to Therapy for Infectious Diseases. Antibodies (Basel) 2020; 9:antib9030044. [PMID: 32882841 PMCID: PMC7551396 DOI: 10.3390/antib9030044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/12/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
The integrin associated protein (CD47) is a widely and moderately expressed glycoprotein in all healthy cells. Cancer cells are known to induce increased CD47 expression. Similar to cancer cells, all immune cells can upregulate their CD47 surface expression during infection. The CD47-SIRPa interaction induces an inhibitory effect on macrophages and dendritic cells (dendritic cells) while CD47-thrombospondin-signaling inhibits T cells. Therefore, the disruption of the CD47 interaction can mediate several biologic functions. Upon the blockade and knockout of CD47 reveals an immunosuppressive effect of CD47 during LCMV, influenza virus, HIV-1, mycobacterium tuberculosis, plasmodium and other bacterial pneumonia infections. In our recent study we shows that the blockade of CD47 using the anti-CD47 antibody increases the activation and effector function of macrophages, dendritic cells and T cells during viral infection. By enhancing both innate and adaptive immunity, CD47 blocking antibody promotes antiviral effect. Due to its broad mode of action, the immune-stimulatory effect derived from this antibody could be applicable in nonresolving and (re)emerging infections. The anti-CD47 antibody is currently under clinical trial for the treatment of cancer and could also have amenable therapeutic potential against infectious diseases. This review highlights the immunotherapeutic targeted role of CD47 in the infectious disease realm.
Collapse
|
35
|
Wang Y, Nanda V, Direnzo D, Ye J, Xiao S, Kojima Y, Howe KL, Jarr KU, Flores AM, Tsantilas P, Tsao N, Rao A, Newman AAC, Eberhard AV, Priest JR, Ruusalepp A, Pasterkamp G, Maegdefessel L, Miller CL, Lind L, Koplev S, Björkegren JLM, Owens GK, Ingelsson E, Weissman IL, Leeper NJ. Clonally expanding smooth muscle cells promote atherosclerosis by escaping efferocytosis and activating the complement cascade. Proc Natl Acad Sci U S A 2020; 117:15818-15826. [PMID: 32541024 PMCID: PMC7354942 DOI: 10.1073/pnas.2006348117] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is the process underlying heart attack and stroke. Despite decades of research, its pathogenesis remains unclear. Dogma suggests that atherosclerotic plaques expand primarily via the accumulation of cholesterol and inflammatory cells. However, recent evidence suggests that a substantial portion of the plaque may arise from a subset of "dedifferentiated" vascular smooth muscle cells (SMCs) which proliferate in a clonal fashion. Herein we use multicolor lineage-tracing models to confirm that the mature SMC can give rise to a hyperproliferative cell which appears to promote inflammation via elaboration of complement-dependent anaphylatoxins. Despite being extensively opsonized with prophagocytic complement fragments, we find that this cell also escapes immune surveillance by neighboring macrophages, thereby exacerbating its relative survival advantage. Mechanistic studies indicate this phenomenon results from a generalized opsonin-sensing defect acquired by macrophages during polarization. This defect coincides with the noncanonical up-regulation of so-called don't eat me molecules on inflamed phagocytes, which reduces their capacity for programmed cell removal (PrCR). Knockdown or knockout of the key antiphagocytic molecule CD47 restores the ability of macrophages to sense and clear opsonized targets in vitro, allowing for potent and targeted suppression of clonal SMC expansion in the plaque in vivo. Because integrated clinical and genomic analyses indicate that similar pathways are active in humans with cardiovascular disease, these studies suggest that the clonally expanding SMC may represent a translational target for treating atherosclerosis.
Collapse
Affiliation(s)
- Ying Wang
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305
| | - Vivek Nanda
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305
- Department of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Daniel Direnzo
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Jianqin Ye
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Sophia Xiao
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Yoko Kojima
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Kathryn L Howe
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Kai-Uwe Jarr
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Alyssa M Flores
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Pavlos Tsantilas
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Noah Tsao
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Abhiram Rao
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| | - Alexandra A C Newman
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22904
| | - Anne V Eberhard
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - James R Priest
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Arno Ruusalepp
- Department of Cardiac Surgery, Tartu University Hospital, Tartu, Estonia 50406
| | - Gerard Pasterkamp
- Department of Cardiology, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
- Laboratory of Clinical Chemistry, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, 80333 Munich, Germany
- German Center for Cardiovascular Research (DZHK partner site), 10785 Munich, Germany
| | - Clint L Miller
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA 22904
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22904
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, SE-751 05 Uppsala, Sweden
| | - Simon Koplev
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22904
| | - Erik Ingelsson
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305
- Department of Medical Sciences, Uppsala University, SE-751 05 Uppsala, Sweden
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Nicholas J Leeper
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305;
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305
| |
Collapse
|
36
|
Zhang X, Li W, Sun J, Yang Z, Guan Q, Wang R, Li X, Li Y, Feng Y, Wang Y. How to use macrophages to realise the treatment of tumour. J Drug Target 2020; 28:1034-1045. [PMID: 32603199 DOI: 10.1080/1061186x.2020.1775236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Macrophages (Mø) are immune cells with natural phagocytic ability and play an important role in tumorigenesis, development and metastasis. Mø play a dual role of tumour inhibition and tumour promotion in tumour development due to their two different phenotypes. Mø in the tumour microenvironment have long been referred to as tumour-associated Mø (TAMs). Mø are mainly involved in tumour resistance, cancer metastasis and mediating immunosuppression. Nowadays, Mø and Mø membranes have been widely used in drug delivery systems (DDSs) because of their good biocompatibility, natural phagocytosis and their important role in tumour development. In this review, from the perspective of Mø's role in tumour development, we present strategies and drugs of Mø targeting and focusing on the several types of biomimetic nanoparticles constructed by Mø and Mø membranes in tumour therapy, and discuss the problem of this delivery system in present research and future directions.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Jialin Sun
- Biological Science and Technology Department, Heilongjiang Vocational College for Nationalities, Harbin, P.R. China
| | - Zhixin Yang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Qingxia Guan
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Rui Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Xiuyan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Yongji Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Yufei Feng
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| |
Collapse
|
37
|
Gupta A, Taslim C, Tullius BP, Cripe TP. Therapeutic modulation of the CD47-SIRPα axis in the pediatric tumor microenvironment: working up an appetite. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:550-562. [PMID: 35582455 PMCID: PMC8992496 DOI: 10.20517/cdr.2020.12] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 11/12/2022]
Abstract
Evasion of immune surveillance is one of the hallmarks of cancer. Although the adaptive immune system has been targeted via checkpoint inhibition, many patients do not sustain durable remissions due to the heterogeneity of the tumor microenvironment, so additional strategies are needed. The innate immune system has its own set of checkpoints, and tumors have co-opted this system by expressing surface receptors that inhibit phagocytosis. One of these receptors, CD47, also known as the "don't eat me" signal, has been found to be overexpressed by most cancer histologies and has been successfully targeted by antibodies blocking the receptor or its ligand, signal regulatory protein α (SIRPα). By enabling phagocytosis via antigen-presenting cells, interruption of CD47-SIRPα binding leads to earlier downstream activation of the adaptive immune system. Recent and ongoing clinical trials are demonstrating the safety and efficacy of CD47 blockade in combination with monoclonal antibodies, chemotherapy, or checkpoint inhibitors for adult cancer histologies. The aim of this review is to highlight the current literature and research on CD47, provide an impetus for investigation of its blockade in pediatric cancer histologies, and provide a rationale for new combination therapies in these patients.
Collapse
Affiliation(s)
- Ajay Gupta
- Division of Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Cenny Taslim
- Center for Childhood Cancer and Blood Diseases, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Brian P. Tullius
- Division of Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Timothy P. Cripe
- Division of Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Center for Childhood Cancer and Blood Diseases, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| |
Collapse
|
38
|
Jalil AR, Andrechak JC, Discher DE. Macrophage checkpoint blockade: results from initial clinical trials, binding analyses, and CD47-SIRPα structure-function. Antib Ther 2020; 3:80-94. [PMID: 32421049 PMCID: PMC7206415 DOI: 10.1093/abt/tbaa006] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
The macrophage checkpoint is an anti-phagocytic interaction between signal regulatory protein alpha (SIRPα) on a macrophage and CD47 on all types of cells - ranging from blood cells to cancer cells. This interaction has emerged over the last decade as a potential co-target in cancer when combined with other anti-cancer agents, with antibodies against CD47 and SIRPα currently in preclinical and clinical development for a variety of hematological and solid malignancies. Monotherapy with CD47 blockade is ineffective in human clinical trials against many tumor types tested to date, except for rare cutaneous and peripheral lymphomas. In contrast, pre-clinical results show efficacy in multiple syngeneic mouse models of cancer, suggesting that many of these tumor models are more immunogenic and likely artificial compared to human tumors. However, combination therapies in humans of anti-CD47 with agents such as the anti-tumor antibody rituximab do show efficacy against liquid tumors (lymphoma) and are promising. Here, we review such trials as well as key interaction and structural features of CD47-SIRPα.
Collapse
Affiliation(s)
- AbdelAziz R Jalil
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason C Andrechak
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E Discher
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|