51
|
Hao Y, Zhou X, Li Y, Li B, Cheng L. The CD47-SIRPα axis is a promising target for cancer immunotherapies. Int Immunopharmacol 2023; 120:110255. [PMID: 37187126 DOI: 10.1016/j.intimp.2023.110255] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/17/2023]
Abstract
Cluster of differentiation 47(CD47) is a transmembrane protein that is ubiquitously found on the surface of many cells in the body and uniquely overexpressed by both solid and hematologic malignant cells. CD47 interacts with signal-regulatory protein α (SIRPα), to trigger a "don't eat me" signal and thereby achieve cancer immune escape by inhibiting macrophage-mediated phagocytosis. Thus, blocking the CD47-SIRPα phagocytosis checkpoint, for release of the innate immune system, is a current research focus. Indeed, targeting the CD47-SIRPα axis as a cancer immunotherapy has shown promising efficacies in pre-clinical outcomes. Here, we first reviewed the origin, structure, and function of the CD47-SIRPα axis. Then, we reviewed its role as a target for cancer immunotherapies, as well as the factors regulating CD47-SIRPα axis-based immunotherapies. We specifically focused on the mechanism and progress of CD47-SIRPα axis-based immunotherapies and their combination with other treatment strategies. Finally, we discussed the challenges and directions for future research and identified potential CD47-SIRPα axis-based therapies that are suitable for clinical application.
Collapse
Affiliation(s)
- Yu Hao
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China
| | - Yiling Li
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bolei Li
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
52
|
Luo JQ, Liu R, Chen FM, Zhang JY, Zheng SJ, Shao D, Du JZ. Nanoparticle-Mediated CD47-SIRPα Blockade and Calreticulin Exposure for Improved Cancer Chemo-Immunotherapy. ACS NANO 2023; 17:8966-8979. [PMID: 37133900 DOI: 10.1021/acsnano.2c08240] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Enabling macrophages to phagocytose tumor cells holds great potential for cancer therapy but suffers from tremendous challenges because the tumor cells upregulate antiphagocytosis molecules (such as CD47) on their surface. The blockade of CD47 alone is insufficient to stimulate tumor cell phagocytosis in solid tumors due to the lack of "eat me" signals. Herein, a degradable mesoporous silica nanoparticle (MSN) is reported to simultaneously deliver anti-CD47 antibodies (aCD47) and doxorubicin (DOX) for cancer chemo-immunotherapy. The codelivery nanocarrier aCD47-DMSN was constructed by accommodating DOX within the mesoporous cavity, while adsorbing aCD47 on the surface of MSN. aCD47 blocks the CD47-SIRPα axis to disable the "don't eat me" signal, while DOX induces immunogenic tumor cell death (ICD) for calreticulin exposure as an "eat me" signal. This design facilitated the phagocytosis of tumor cells by macrophages, which enhanced antigen cross-presentation and elicited efficient T cell-mediated immune response. In 4T1 and B16F10 murine tumor models, aCD47-DMSN generated a strong antitumor effect after intravenous injection by increasing tumor-infiltration of CD8+ T cells. Taken together, this study offers a nanoplatform to modulate the phagocytosis of macrophages for efficacious cancer chemo-immunotherapy.
Collapse
Affiliation(s)
- Jia-Qi Luo
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Rong Liu
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Fang-Man Chen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China
| | - Jing-Yang Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China
| | - Sui-Juan Zheng
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Dan Shao
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
53
|
Li L, Tian Y. The role of metabolic reprogramming of tumor-associated macrophages in shaping the immunosuppressive tumor microenvironment. Biomed Pharmacother 2023; 161:114504. [PMID: 37002579 DOI: 10.1016/j.biopha.2023.114504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Macrophages are potent immune effector cells in innate immunity and exert dual-effects in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) make up a significant portion of TME immune cells. Similar to M1/M2 macrophages, TAMs are also highly plastic, and their functions are regulated by cytokines, chemokines and other factors in the TME. The metabolic changes in TAMs are significantly associated with polarization towards a protumour or antitumour phenotype. The metabolites generated via TAM metabolic reprogramming in turn promote tumor progression and immune tolerance. In this review, we explore the metabolic reprogramming of TAMs in terms of energy, amino acid and fatty acid metabolism and the potential roles of these changes in immune suppression.
Collapse
Affiliation(s)
- Lunxu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
54
|
Dacek MM, Kurtz KG, Wallisch P, Pierre SA, Khayat S, Bourne CM, Gardner TJ, Vogt KC, Aquino N, Younes A, Scheinberg DA. Potentiating antibody-dependent killing of cancers with CAR T cells secreting CD47-SIRPα checkpoint blocker. Blood 2023; 141:2003-2015. [PMID: 36696633 PMCID: PMC10163312 DOI: 10.1182/blood.2022016101] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 01/03/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown success in the treatment of hematopoietic malignancies; however, relapse remains a significant issue. To overcome this, we engineered "Orexi" CAR T cells to locally secrete a high-affinity CD47 blocker, CV1, at the tumor and treated tumors in combination with an orthogonally targeted monoclonal antibody. Traditional CAR T cells plus the antibody had an additive effect in xenograft models, and this effect was potentiated by CAR T-cell local CV1 secretion. Furthermore, OrexiCAR-secreted CV1 reversed the immunosuppression of myelomonocytoid cells both in vitro and within the tumor microenvironment. Local secretion of the CD47 inhibitor bypasses the CD47 sink found on all cells in the body and may prevent systemic toxicities. This combination of CAR T-cell therapy, local CD47 blockade, and orthogonal antibody may be a combinatorial strategy to overcome the limitations of each monotherapy.
Collapse
Affiliation(s)
- Megan M. Dacek
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY
- Pharmacology Program, Weill Cornell Medicine, New York, NY
| | - Keifer G. Kurtz
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY
- Pharmacology Program, Weill Cornell Medicine, New York, NY
| | - Patrick Wallisch
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY
- Pharmacology Program, Weill Cornell Medicine, New York, NY
| | - Stephanie A. Pierre
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY
- Tri-institutunal MD-PhD Program, Weill Cornell Medicine, New York, NY
| | - Shireen Khayat
- Pharmacology Program, Weill Cornell Medicine, New York, NY
- Immunology Program, Sloan Kettering Institute, New York, NY
| | - Christopher M. Bourne
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY
| | - Thomas J. Gardner
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY
| | - Kristen C. Vogt
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY
- Tri-Institutional PhD Program in Chemical Biology, Weill Cornell Medicine, Memorial Sloan Kettering Cancer Center, The Rockefeller University, New York, NY
| | - Nica Aquino
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anas Younes
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David A. Scheinberg
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY
- Pharmacology Program, Weill Cornell Medicine, New York, NY
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
55
|
van Helden MJ, Zwarthoff SA, Arends RJ, Reinieren-Beeren IMJ, Paradé MCBC, Driessen-Engels L, de Laat-Arts K, Damming D, Santegoeds-Lenssen EWH, van Kuppeveld DWJ, Lodewijks I, Olsman H, Matlung HL, Franke K, Mattaar-Hepp E, Stokman MEM, de Wit B, Glaudemans DHRF, van Wijk DEJW, Joosten-Stoffels L, Schouten J, Boersema PJ, van der Vleuten M, Sanderink JWH, Kappers WA, van den Dobbelsteen D, Timmers M, Ubink R, Rouwendal GJA, Verheijden G, van der Lee MMC, Dokter WHA, van den Berg TK. BYON4228 is a pan-allelic antagonistic SIRPα antibody that potentiates destruction of antibody-opsonized tumor cells and lacks binding to SIRPγ on T cells. J Immunother Cancer 2023; 11:jitc-2022-006567. [PMID: 37068796 DOI: 10.1136/jitc-2022-006567] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Preclinical studies have firmly established the CD47-signal-regulatory protein (SIRP)α axis as a myeloid immune checkpoint in cancer, and this is corroborated by available evidence from the first clinical studies with CD47 blockers. However, CD47 is ubiquitously expressed and mediates functional interactions with other ligands as well, and therefore targeting of the primarily myeloid cell-restricted inhibitory immunoreceptor SIRPα may represent a better strategy. METHOD We generated BYON4228, a novel SIRPα-directed antibody. An extensive preclinical characterization was performed, including direct comparisons to previously reported anti-SIRPα antibodies. RESULTS BYON4228 is an antibody directed against SIRPα that recognizes both allelic variants of SIRPα in the human population, thereby maximizing its potential clinical applicability. Notably, BYON4228 does not recognize the closely related T-cell expressed SIRPγ that mediates interactions with CD47 as well, which are known to be instrumental in T-cell extravasation and activation. BYON4228 binds to the N-terminal Ig-like domain of SIRPα and its epitope largely overlaps with the CD47-binding site. BYON4228 blocks binding of CD47 to SIRPα and inhibits signaling through the CD47-SIRPα axis. Functional studies show that BYON4228 potentiates macrophage-mediated and neutrophil-mediated killing of hematologic and solid cancer cells in vitro in the presence of a variety of tumor-targeting antibodies, including trastuzumab, rituximab, daratumumab and cetuximab. The silenced Fc region of BYON4228 precludes immune cell-mediated elimination of SIRPα-positive myeloid cells, implying anticipated preservation of myeloid immune effector cells in patients. The unique profile of BYON4228 clearly distinguishes it from previously reported antibodies representative of agents in clinical development, which either lack recognition of one of the two SIRPα polymorphic variants (HEFLB), or cross-react with SIRPγ and inhibit CD47-SIRPγ interactions (SIRPAB-11-K322A, 1H9), and/or have functional Fc regions thereby displaying myeloid cell depletion activity (SIRPAB-11-K322A). In vivo, BYON4228 increases the antitumor activity of rituximab in a B-cell Raji xenograft model in human SIRPαBIT transgenic mice. Finally, BYON4228 shows a favorable safety profile in cynomolgus monkeys. CONCLUSIONS Collectively, this defines BYON4228 as a preclinically highly differentiating pan-allelic SIRPα antibody without T-cell SIRPγ recognition that promotes the destruction of antibody-opsonized cancer cells. Clinical studies are planned to start in 2023.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Hugo Olsman
- Sanquin Research, Amsterdam, The Netherlands
| | | | | | | | | | - Benny de Wit
- Byondis BV, Nijmegen, Gelderland, The Netherlands
| | | | | | | | - Jan Schouten
- Byondis BV, Nijmegen, Gelderland, The Netherlands
| | | | | | | | | | | | | | - Ruud Ubink
- Byondis BV, Nijmegen, Gelderland, The Netherlands
| | | | | | | | | | - Timo K van den Berg
- Byondis BV, Nijmegen, Gelderland, The Netherlands
- Sanquin Research, Amsterdam, The Netherlands
| |
Collapse
|
56
|
Ye ZH, Yu WB, Huang MY, Chen J, Lu JJ. Building on the backbone of CD47-based therapy in cancer: Combination strategies, mechanisms, and future perspectives. Acta Pharm Sin B 2023; 13:1467-1487. [PMID: 37139405 PMCID: PMC10149906 DOI: 10.1016/j.apsb.2022.12.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/10/2022] [Accepted: 11/18/2022] [Indexed: 12/27/2022] Open
Abstract
Described as a "don't eat me" signal, CD47 becomes a vital immune checkpoint in cancer. Its interaction with signal regulatory protein alpha (SIRPα) prevents macrophage phagocytosis. In recent years, a growing body of evidences have unveiled that CD47-based combination therapy exhibits a superior anti-cancer effect. Latest clinical trials about CD47 have adopted the regimen of collaborating with other therapies or developing CD47-directed bispecific antibodies, indicating the combination strategy as a general trend of the future. In this review, clinical and preclinical cases about the current combination strategies targeting CD47 are collected, their underlying mechanisms of action are discussed, and ideas from future perspectives are shared.
Collapse
Affiliation(s)
- Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Wei-Bang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jun Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, 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
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, University of Macau, Macao 999078, China
| |
Collapse
|
57
|
Chang R, Chu X, Zhang J, Fu R, Feng C, Jia D, Wang R, Yan H, Li G, Li J. Liposome-Based Co-Immunotherapy with TLR Agonist and CD47-SIRPα Checkpoint Blockade for Efficient Treatment of Colon Cancer. Molecules 2023; 28:molecules28073147. [PMID: 37049910 PMCID: PMC10095745 DOI: 10.3390/molecules28073147] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/22/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Antitumor immunity is an essential component of cancer therapy and is primarily mediated by the innate immune response, which plays a critical role in initiating and shaping the adaptive immune response. Emerging evidence has identified innate immune checkpoints and pattern recognition receptors, such as CD47 and Toll-like receptor 7 (TLR7), as promising therapeutic targets for cancer treatment. Based on the fusion protein Fc-CV1, which comprises a high-affinity SIRPα variant (CV1), and the Fc fragment of the human IgG1 antibody, we exploited a preparation which coupled Fc-CV1 to imiquimod (TLR7 agonist)-loaded liposomes (CILPs) to actively target CT26. WT syngeneic colon tumor models. In vitro studies revealed that CILPs exhibited superior sustained release properties and cell uptake efficiency compared to free imiquimod. In vivo assays proved that CILPs exhibited more efficient accumulation in tumors, and a more significant tumor suppression effect than the control groups. This immunotherapy preparation possessed the advantages of low doses and low toxicity. These results demonstrated that a combination of immune checkpoint blockade (ICB) therapy and innate immunity agonists, such as the Fc-CV1 and imiquimod-loaded liposome preparation utilized in this study, could represent a highly effective strategy for tumor therapy.
Collapse
Affiliation(s)
- Rui Chang
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Xiaohong Chu
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Jibing Zhang
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Rongrong Fu
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Changshun Feng
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Dianlong Jia
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Rui Wang
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Hui Yan
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Guangyong Li
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Jun Li
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| |
Collapse
|
58
|
Kang Y, Yeo M, Choi H, Jun H, Eom S, Park SG, Yoon H, Kim E, Kang S. Lactate oxidase/vSIRPα conjugates efficiently consume tumor-produced lactates and locally produce tumor-necrotic H 2O 2 to suppress tumor growth. Int J Biol Macromol 2023; 231:123577. [PMID: 36758763 DOI: 10.1016/j.ijbiomac.2023.123577] [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: 12/08/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023]
Abstract
Aggressive tumor formation often leads to excessive anaerobic glycolysis and massive production and accumulation of lactate in the tumor microenvironment (TME). To significantly curb lactate accumulation in TME, in this study, lactate oxidase (LOX) was used as a potential therapeutic enzyme and signal regulatory protein α variant (vSIRPα) as a tumor cell targeting ligand. SpyCatcher protein and SpyTag peptide were genetically fused to LOX and vSIRPα, respectively, to form SC-LOX and ST-vSIRPα and tumor-targeting LOX/vSIRPα conjugates were constructed via a SpyCatcher/SpyTag protein ligation system. LOX/vSIRPα conjugates selectively bound to the CD47-overexpressing mouse melanoma B16-F10 cells and effectively consumed lactate produced by the B16-F10 cells, generating adequate amounts of hydrogen peroxide (H2O2), which induces drastic necrotic tumor cell death. Local treatments of B16-F10 tumor-bearing mice with LOX/vSIRPα conjugates significantly suppressed B16-F10 tumor growth in vivo without any severe side effects. Tumor-targeting vSIRPα may allow longer retention of LOX in tumor sites, effectively consuming surrounding lactate in TME and locally generating adequate amounts of cytotoxic H2O2 to suppress tumor growth. The approach restraining the local lactate concentration and H2O2 in TME using LOX and vSIRPα could offer new opportunities for developing enzyme/targeting ligand conjugate-based therapeutic tools for tumor treatment.
Collapse
Affiliation(s)
- Yujin Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Mirae Yeo
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hyukjun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Heejin Jun
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Soomin Eom
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seong Guk Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Haejin Yoon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eunhee Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| |
Collapse
|
59
|
Emerging phagocytosis checkpoints in cancer immunotherapy. Signal Transduct Target Ther 2023; 8:104. [PMID: 36882399 PMCID: PMC9990587 DOI: 10.1038/s41392-023-01365-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 03/09/2023] Open
Abstract
Cancer immunotherapy, mainly including immune checkpoints-targeted therapy and the adoptive transfer of engineered immune cells, has revolutionized the oncology landscape as it utilizes patients' own immune systems in combating the cancer cells. Cancer cells escape immune surveillance by hijacking the corresponding inhibitory pathways via overexpressing checkpoint genes. Phagocytosis checkpoints, such as CD47, CD24, MHC-I, PD-L1, STC-1 and GD2, have emerged as essential checkpoints for cancer immunotherapy by functioning as "don't eat me" signals or interacting with "eat me" signals to suppress immune responses. Phagocytosis checkpoints link innate immunity and adaptive immunity in cancer immunotherapy. Genetic ablation of these phagocytosis checkpoints, as well as blockade of their signaling pathways, robustly augments phagocytosis and reduces tumor size. Among all phagocytosis checkpoints, CD47 is the most thoroughly studied and has emerged as a rising star among targets for cancer treatment. CD47-targeting antibodies and inhibitors have been investigated in various preclinical and clinical trials. However, anemia and thrombocytopenia appear to be formidable challenges since CD47 is ubiquitously expressed on erythrocytes. Here, we review the reported phagocytosis checkpoints by discussing their mechanisms and functions in cancer immunotherapy, highlight clinical progress in targeting these checkpoints and discuss challenges and potential solutions to smooth the way for combination immunotherapeutic strategies that involve both innate and adaptive immune responses.
Collapse
|
60
|
Vaccaro K, Allen J, Whitfield TW, Maoz A, Reeves S, Velarde J, Yang D, Phan N, Bell GW, Hata AN, Weiskopf K. Targeted therapies prime oncogene-driven lung cancers for macrophage-mediated destruction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.03.531059. [PMID: 36945559 PMCID: PMC10028834 DOI: 10.1101/2023.03.03.531059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Macrophage immune checkpoint inhibitors, such as anti-CD47 antibodies, show promise in clinical trials for solid and hematologic malignancies. However, the best strategies to use these therapies remain unknown and ongoing studies suggest they may be most effective when used in combination with other anticancer agents. Here, we developed a novel screening platform to identify drugs that render lung cancer cells more vulnerable to macrophage attack, and we identified therapeutic synergy exists between genotype-directed therapies and anti-CD47 antibodies. In validation studies, we found the combination of genotype-directed therapies and CD47 blockade elicited robust phagocytosis and eliminated persister cells in vitro and maximized anti-tumor responses in vivo. Importantly, these findings broadly applied to lung cancers with various RTK/MAPK pathway alterations-including EGFR mutations, ALK fusions, or KRASG12C mutations. We observed downregulation of β2-microglobulin and CD73 as molecular mechanisms contributing to enhanced sensitivity to macrophage attack. Our findings demonstrate that dual inhibition of the RTK/MAPK pathway and the CD47/SIRPa axis is a promising immunotherapeutic strategy. Our study provides strong rationale for testing this therapeutic combination in patients with lung cancers bearing driver mutations.
Collapse
Affiliation(s)
- Kyle Vaccaro
- Whitehead Institute for Biomedical Research; Cambridge, MA 02142, USA
| | - Juliet Allen
- Whitehead Institute for Biomedical Research; Cambridge, MA 02142, USA
| | - Troy W. Whitfield
- Whitehead Institute for Biomedical Research; Cambridge, MA 02142, USA
| | - Asaf Maoz
- Whitehead Institute for Biomedical Research; Cambridge, MA 02142, USA
- Dana-Farber Cancer Institute; Boston, MA 02115, USA
| | - Sarah Reeves
- Massachusetts General Hospital Cancer Center; Boston, MA 02129, USA
| | - José Velarde
- Whitehead Institute for Biomedical Research; Cambridge, MA 02142, USA
| | - Dian Yang
- Whitehead Institute for Biomedical Research; Cambridge, MA 02142, USA
| | - Nicole Phan
- Massachusetts General Hospital Cancer Center; Boston, MA 02129, USA
| | - George W. Bell
- Whitehead Institute for Biomedical Research; Cambridge, MA 02142, USA
| | - Aaron N. Hata
- Massachusetts General Hospital Cancer Center; Boston, MA 02129, USA
| | - Kipp Weiskopf
- Whitehead Institute for Biomedical Research; Cambridge, MA 02142, USA
- Dana-Farber Cancer Institute; Boston, MA 02115, USA
| |
Collapse
|
61
|
Hu J, Yang Q, Yue Z, Liao B, Cheng H, Li W, Zhang H, Wang S, Tian Q. Emerging advances in engineered macrophages for tumor immunotherapy. Cytotherapy 2023; 25:235-244. [PMID: 36008206 DOI: 10.1016/j.jcyt.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/02/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
Macrophages are versatile antigen-presenting cells. Recent studies suggest that engineered modifications of macrophages may confer better tumor therapy. Genetic engineering of macrophages with specific chimeric antigen receptors offers new possibilities for treatment of solid tumors and has received significant attention. In vitro gene editing of macrophages and infusion into the body can inhibit the immunosuppressive effect of the tumor microenvironment in solid tumors. This strategy is flexible and can be applied to all stages of cancer treatment. In contrast, nongenetic engineering tools are used to block relevant signaling pathways in immunosuppressive responses. In addition, macrophages can be loaded with drugs and engineered into cellular drug delivery systems. Here, we analyze the effect of the chimeric antigen receptor platform on macrophages and other existing engineering modifications of macrophages, highlighting their status, challenges and future perspectives. Indeed, our analyses show that new approaches in the treatment of solid tumors will likely exploit macrophages, an innate immune cell.
Collapse
Affiliation(s)
- Jing Hu
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Qian Yang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zhongyu Yue
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Boting Liao
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Huijuan Cheng
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Wenqi Li
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Honghua Zhang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shuling Wang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Qingchang Tian
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China.
| |
Collapse
|
62
|
Beckett AN, Chockley P, Pruett-Miller SM, Nguyen P, Vogel P, Sheppard H, Krenciute G, Gottschalk S, DeRenzo C. CD47 expression is critical for CAR T-cell survival in vivo. J Immunother Cancer 2023; 11:jitc-2022-005857. [PMID: 36918226 PMCID: PMC10016274 DOI: 10.1136/jitc-2022-005857] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND CD47 is an attractive immunotherapeutic target because it is highly expressed on multiple solid tumors. However, CD47 is also expressed on T cells. Limited studies have evaluated CD47-chimeric antigen receptor (CAR) T cells, and the role of CD47 in CAR T-cell function remains largely unknown. METHODS Here, we describe the development of CD47-CAR T cells derived from a high affinity signal regulatory protein α variant CV1, which binds CD47. CV1-CAR T cells were generated from human peripheral blood mononuclear cells and evaluated in vitro and in vivo. The role of CD47 in CAR T-cell function was examined by knocking out CD47 in T cells followed by downstream functional analyses. RESULTS While CV1-CAR T cells are specific and exhibit potent activity in vitro they lacked antitumor activity in xenograft models. Mechanistic studies revealed CV1-CAR T cells downregulate CD47 to overcome fratricide, but CD47 loss resulted in their failure to expand and persist in vivo. This effect was not limited to CV1-CAR T cells, since CD47 knockout CAR T cells targeting another solid tumor antigen exhibited the same in vivo fate. Further, CD47 knockout T cells were sensitive to macrophage-mediated phagocytosis. CONCLUSIONS These findings highlight that CD47 expression is critical for CAR T-cell survival in vivo and is a 'sine qua non' for successful adoptive T-cell therapy.
Collapse
Affiliation(s)
- Alex N Beckett
- Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, Tennessee, USA.,Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Peter Chockley
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St Jude Children's Research Hospital, Memphis, Tennessee, USA.,Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Phuong Nguyen
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Peter Vogel
- Department of Pathology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Heather Sheppard
- Department of Pathology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Christopher DeRenzo
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
63
|
Li Y, Zhou H, Liu P, Lv D, Shi Y, Tang B, Xu J, Zhong T, Xu W, Zhang J, Zhou J, Ying K, Zhao Y, Sun Y, Jiang Z, Cheng H, Zhang X, Ke Y. SHP2 deneddylation mediates tumor immunosuppression in colon cancer via the CD47/SIRPα axis. J Clin Invest 2023; 133:162870. [PMID: 36626230 PMCID: PMC9927946 DOI: 10.1172/jci162870] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
SIPRα on macrophages binds with CD47 to resist proengulfment signals, but how the downstream signal of SIPRα controls tumor-infiltrating macrophages (TIMs) is still poorly clarified. Here, we report that the CD47/signal regulatory protein α (SIRPα) axis requires the deneddylation of tyrosine phosphatase SHP2. Mechanistically, Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2) was constitutively neddylated on K358 and K364 sites; thus, its autoinhibited conformation was maintained. In response to CD47-liganded SIRPα, SHP2 was deneddylated by sentrin-specific protease 8 (SENP8), which led to the dephosphorylation of relevant substrates at the phagocytic cup and subsequent inhibition of macrophage phagocytosis. Furthermore, neddylation inactivated myeloid-SHP2 and greatly boosted the efficacy of colorectal cancer (CRC) immunotherapy. Importantly, we observed that supplementation with SHP2 allosteric inhibitors sensitized immune treatment-resistant CRC to immunotherapy. Our results emphasize that the CRC subtype that is unresponsive to immunotherapy relies on SIRPαhiSHP2hiNEDD8lo TIMs and highlight the need to further explore the strategy of SHP2 targeting in CRC therapy.
Collapse
Affiliation(s)
- Yiqing Li
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Zhou
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Liu
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Dandan Lv
- Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yichun Shi
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bufu Tang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research at The Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Jiaqi Xu
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Tingting Zhong
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Wangting Xu
- Department of Respiratory Medicine at The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Zhang
- Department of Urology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Medicine at The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kejing Ying
- Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchao Zhao
- Cancer Institute of The Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Sun
- Cancer Institute of The Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhinong Jiang
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, and Department of Cardiology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuehai Ke
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
64
|
Is the new angel better than the old devil? Challenges and opportunities in CD47- SIRPα-based cancer therapy. Crit Rev Oncol Hematol 2023; 184:103939. [PMID: 36774991 DOI: 10.1016/j.critrevonc.2023.103939] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/05/2022] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
The efficacy of immunotherapies is limited due to the impenetrable nature of the tumor microenvironment (TME). The TME of many tumors is immune-privileged, thus allowing them to evade host immunosurveillance. One mechanism through which this occurs is via the overexpression of CD47, a 'don't eat me' protein that can interact with SIRPα on myeloid cells to suppress their phagocytic action. In recent times, many studies are focusing on CD47-SIRPα-dependent immunotherapies to incite a 'seek and eat' interaction between phagocytes and tumors. Thus, in this review, we highlight the basic molecular properties and mechanisms of CD47-SIRPα cascade. In addition, we discuss the major challenges and potential remedies associated with CD47-SIRPα-based immunotherapies.
Collapse
|
65
|
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: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
66
|
Bungert AD, Urbantat RM, Jelgersma C, Bekele BM, Mueller S, Mueller A, Felsenstein M, Dusatko S, Blank A, Ghori A, Boehm-Sturm P, Koch SP, Vajkoczy P, Brandenburg S. Myeloid cell subpopulations compensate each other for Ccr2-deficiency in glioblastoma. Neuropathol Appl Neurobiol 2023; 49:e12863. [PMID: 36346010 DOI: 10.1111/nan.12863] [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: 11/13/2021] [Revised: 08/07/2022] [Accepted: 10/09/2022] [Indexed: 11/11/2022]
Abstract
AIMS Glioblastomas are high-grade brain tumours that are characterised by the accumulation of brain-resident microglia and peripheral macrophages. Recruitment of these myeloid cells can be facilitated by CCR2/CCL2 signalling. Besides the well-known CCR2+ macrophages, we have identified microglia expressing CCR2 in glioma tissues. Thus, we investigated how Ccr2-deficiency of one of the myeloid cell populations affects the other population and tumour biology. METHODS We generated four chimeric groups to analyse single and combined Ccr2-deficiency of microglia and macrophages. On day 21 after tumour cell implantation (GL261), we conducted flow cytometry, immunofluorescence and real-time polymerase chain reaction analyses. Tumour volume and metabolism were determined by magnetic resonance imaging and magnetic resonance spectroscopy. Moreover, in vitro studies were performed with primary microglia and bone marrow-derived macrophages. RESULTS We demonstrated reduced infiltration of macrophages and microglia depending on the lack of Ccr2. However, the total number of myeloid cells remained constant except for the animals with dual Ccr2-knockout. Both microglia and macrophages with Ccr2-deficiency showed impaired expression of proinflammatory molecules and altered phagocytic activity. Despite the altered immunologic phenotype caused by Ccr2-deficiency, glioma progression and metabolism were hardly affected. Alterations were detected solely in apoptosis and proliferation of tumours from animals with specific Ccr2-deficient microglia, whereas vessel stability was increased in mice with Ccr2-knockout in both cell populations. CONCLUSION These results indicate that microglia and macrophages provide a homoeostatic balance within glioma tissue and compensate for the lack of the corresponding counterpart. Moreover, we identified that the CCR2/CCL2 axis is involved in the immunologic function of microglia and macrophages beyond its relevance for migration.
Collapse
Affiliation(s)
- Alexander D Bungert
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ruth M Urbantat
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claudius Jelgersma
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Biniam M Bekele
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Susanne Mueller
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Berlin, Germany
| | - Annett Mueller
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthäus Felsenstein
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Silke Dusatko
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anne Blank
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Adnan Ghori
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Berlin, Germany
| | - Stefan P Koch
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peter Vajkoczy
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Susan Brandenburg
- Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
67
|
Hassan EM, McWhirter S, Walker GC, Martinez-Rubi Y, Zou S. Elimination of Cancer Cells in Co-Culture: Role of Different Nanocarriers in Regulation of CD47 and Calreticulin-Induced Phagocytosis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:3791-3803. [PMID: 36632842 PMCID: PMC9880957 DOI: 10.1021/acsami.2c19311] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
Under healthy conditions, pro- and anti-phagocytic signals are balanced. Cluster of Differentiation 47 (CD47) is believed to act as an anti-phagocytic marker that is highly expressed on multiple types of human cancer cells including acute myeloid leukemia (AML) and lung and liver carcinomas, allowing them to escape phagocytosis by macrophages. Downregulating CD47 on cancer cells discloses calreticulin (CRT) to macrophages and recovers their phagocytic activity. Herein, we postulate that using a modified graphene oxide (GO) carrier to deliver small interfering RNA (siRNA) CD47 (CD47_siRNA) in AML, A549 lung, and HepG2 liver cancer cells in co-culture in vitro will silence CD47 and flag cancer cells for CRT-mediated phagocytosis. Results showed a high knockdown efficiency of CD47 and a significant increase in CRT levels simultaneously by using GO formulation as carriers in all used cancer cell lines. The presence of CRT on cancer cells was significantly higher than levels before knockdown of CD47 and was required to achieve phagocytosis in co-culture with human macrophages. Lipid nanoparticles (LNPs) and modified boron nitride nanotubes (BNPs) were used to carry CD47_siRNA, and the knockdown efficiency values of CD47 were compared in three cancer cells in co-culture, with an achieved knockdown efficiency of >95% using LNPs as carriers. Interestingly, the high efficiency of CD47 knockdown was obtained by using the LNPs and BNP carriers; however, an increase in CRT levels on cancer cells was not required for phagocytosis to happen in co-culture with human macrophages, indicating other pathways' involvement in the phagocytosis process. These findings highlight the roles of 2D (graphene oxide), 1D (boron nitride nanotube), and "0D" (lipid nanoparticle) carriers for the delivery of siRNA to eliminate cancer cells in co-culture, likely through different phagocytosis pathways in multiple types of human cancer cells. Moreover, these results provide an explanation of immune therapies that target CD47 and the potential use of these carriers in screening drugs for such therapies in vitro.
Collapse
Affiliation(s)
- Eman M. Hassan
- Metrology
Research Centre, National Research Council
Canada, 100 Sussex Drive, Ottawa, OntarioK1A0R6, Canada
| | - Samantha McWhirter
- Department
of Chemistry, University of Toronto, 80 St. George St., Toronto, OntarioM5S3H6, Canada
| | - Gilbert C. Walker
- Department
of Chemistry, University of Toronto, 80 St. George St., Toronto, OntarioM5S3H6, Canada
| | - Yadienka Martinez-Rubi
- Security
and Disruptive Technologies, National Research
Council Canada, 100 Sussex Drive, Ottawa, OntarioK1A0R6, Canada
| | - Shan Zou
- Metrology
Research Centre, National Research Council
Canada, 100 Sussex Drive, Ottawa, OntarioK1A0R6, Canada
- Department
of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, OntarioK1S5B6, Canada
| |
Collapse
|
68
|
Li Y, Liu J, Chen W, Wang W, Yang F, Liu X, Sheng Y, Du K, He M, Lyu X, Li H, Zhao L, Wei Z, Wang F, Zheng S, Sui J. A pH-dependent anti-CD47 antibody that selectively targets solid tumors and improves therapeutic efficacy and safety. J Hematol Oncol 2023; 16:2. [PMID: 36650558 PMCID: PMC9844003 DOI: 10.1186/s13045-023-01399-4] [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: 02/27/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND The antiphagocytic molecule CD47 is overexpressed in a wide variety of cancer cells, and antibodies targeting CD47 for cancer therapies are currently under intensive investigation. However, owing to the ubiquitous expression of CD47 on healthy cells, anti-CD47 therapies often achieve only weak therapeutic benefits and can induce severe side effects. Here, we report the generation of a pH-dependent anti-CD47 antibody (BC31M4) which selectively binds to tumors under the acidic solid tumor microenvironment. METHODS BC31M4 was generated using antibody phage display and a pH-dependent selection strategy. The pH-dependent binding and blocking activities of BC31M4 were verified using in vitro assays, and the structural basis of the pH-dependent binding property was characterized. BC31M4's antitumor effect was confirmed by both phagocytosis assays and studies in xenograft models. The tumor selectivity, mechanism of action, PK properties, side effects, and therapeutic efficacy were further evaluated in humanized (hCD47 and its receptor hSIRPα) immunocompetent syngeneic mouse models. RESULTS The crystal structure reveals that two histidines locate within the CDRs of the light chain directly contribute to the pH-dependent binding of BC31M4. BC31M4 promotes macrophage phagocytosis of tumor cells more potently at acidic-pH than at physiological-pH. Our hCD47/hSIRPα humanized syngeneic mouse model results demonstrated that BC31M4 selectively accumulates in tumors but not in normal tissues. BC31M4 causes minimal side effects and exhibits superior PK properties as compared to the other examined anti-CD47 antibodies. When combined with adoptive T cell transfer, BC31M4 efficiently promotes adaptive immune responses against tumors and also induces immune memory. Moreover, we show that BC31M4's antitumor effects rely on an Fc that mediates strong effector functions. CONCLUSIONS Our study illustrates that the development of a tumor-selective, pH-dependent anti-CD47 antibody safely confers strong therapeutic effects against solid tumors, thus providing a promising therapeutic strategy to overcome the challenges of anti-CD47 therapy.
Collapse
Affiliation(s)
- Yulu Li
- Peking University-Tsinghua University-National Institute of Biological Sciences (PTN) Joint Graduate Program, School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences (NIBS), Beijing, China
| | - Juan Liu
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Wei Chen
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Wei Wang
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Fang Yang
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Ximing Liu
- Peking University-Tsinghua University-National Institute of Biological Sciences (PTN) Joint Graduate Program, School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences (NIBS), Beijing, China
| | - Yao Sheng
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Kaixin Du
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Miaomiao He
- National Institute of Biological Sciences (NIBS), Beijing, China.,Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xueyuan Lyu
- National Institute of Biological Sciences (NIBS), Beijing, China.,PTN Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China
| | - Huiyu Li
- National Institute of Biological Sciences (NIBS), Beijing, China.,PTN Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China
| | - Linlin Zhao
- National Institute of Biological Sciences (NIBS), Beijing, China.,PTN Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhizhong Wei
- National Institute of Biological Sciences (NIBS), Beijing, China.,PTN Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China
| | - Fengchao Wang
- National Institute of Biological Sciences (NIBS), Beijing, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Sanduo Zheng
- National Institute of Biological Sciences (NIBS), Beijing, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Jianhua Sui
- National Institute of Biological Sciences (NIBS), Beijing, China. .,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
69
|
Yang J, Deresa I, Ho WH, Long H, Maslyar D, Rosenthal A, Liang SC, Pincetic A. AL008 Enhances Myeloid Antitumor Function by Inhibiting SIRPα Signaling and Activating Fc Receptors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:204-215. [PMID: 36480261 PMCID: PMC9772397 DOI: 10.4049/jimmunol.2200157] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 11/02/2022] [Indexed: 01/03/2023]
Abstract
Antagonizing the CD47-signal regulatory protein (SIRP)α pathway, a critical myeloid checkpoint, promotes antitumor immunity. In this study, we describe the development of AL008, a pan-allelic, SIRPα-specific Ab that triggers the degradation of SIRPα and, concurrently, stimulates FcγR activation of myeloid cells through an engineered Fc domain. AL008 showed superior enhancement of phagocytosis of tumor cells opsonized with antitumor Ag Abs compared with another SIRPα Ab tested. Unlike ligand-blocking SIRPα Abs, AL008 demonstrated single-agent activity by increasing tumor cell engulfment by human monocyte-derived macrophages even in the absence of opsonizing agents. This effect was due to enhanced Fc function, as blocking FcγR2A abrogated AL008-mediated phagocytic activity. AL008 also promoted human monocyte-derived dendritic cell-mediated T cell proliferation. In humanized mouse models, AL008 induced internalization of SIRPα and increased expression of CD86 and HLA-DR on human tumor-associated macrophages, confirming that the mechanism of action is retained in vivo. Monotherapy treatment with AL008 significantly reduced tumor growth in humanized mice implanted with human MDA-MB-231 tumor cells. AL008 also significantly potentiated the effects of T cell checkpoint blockade with anti-programmed death ligand-1 in syngeneic tumor models. This dual and specific mechanism of AL008, to our knowledge, provides a novel therapeutic strategy for targeting myeloid cells for immune activation.
Collapse
Affiliation(s)
| | | | | | - Hua Long
- Alector, Inc., South San Francisco, CA
| | | | | | | | | |
Collapse
|
70
|
Guo Z, Zhang R, Yang AG, Zheng G. Diversity of immune checkpoints in cancer immunotherapy. Front Immunol 2023; 14:1121285. [PMID: 36960057 PMCID: PMC10027905 DOI: 10.3389/fimmu.2023.1121285] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Finding effective treatments for cancer remains a challenge. Recent studies have found that the mechanisms of tumor evasion are becoming increasingly diverse, including abnormal expression of immune checkpoint molecules on different immune cells, in particular T cells, natural killer cells, macrophages and others. In this review, we discuss the checkpoint molecules with enhanced expression on these lymphocytes and their consequences on immune effector functions. Dissecting the diverse roles of immune checkpoints in different immune cells is crucial for a full understanding of immunotherapy using checkpoint inhibitors.
Collapse
Affiliation(s)
- Zhangyan Guo
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - An-Gang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
- *Correspondence: Guoxu Zheng, ; An-Gang Yang,
| | - Guoxu Zheng
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
- *Correspondence: Guoxu Zheng, ; An-Gang Yang,
| |
Collapse
|
71
|
Chen Y, Huang Y, Deng Y, Liu X, Ye J, Li Q, Luo Y, Lin Y, Liang R, Wei J, Zhang J, Li Y. Cancer Therapy Empowered by Extracellular Vesicle-Mediated Targeted Delivery. Biol Pharm Bull 2023; 46:1353-1364. [PMID: 37779037 DOI: 10.1248/bpb.b23-00378] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Extracellular vesicles (EVs) are a class of nanoparticles that mediate signaling molecules delivery between donor and recipient cells. Heterogeneity in the content of EVs and their membrane surface proteins determines their unique targetability. Their low immunogenicity, capability to cross various biological barriers, and superior biocompatibility enable engineering-modified EVs to be ideal drug delivery carriers. In addition, the engineered EVs that emerge in recent years have become a powerful tool for cancer treatment through the selective delivery of bioactive molecules to therapeutic targets, such as tumor cells and stroma. Our review focuses on the various types of EV modifications and their promoting therapeutic capabilities, which provide an innovative means for cancer precision therapy.
Collapse
Affiliation(s)
- Yong Chen
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Yujuan Huang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Yayan Deng
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Xue Liu
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Jiaxiang Ye
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Qiuyun Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Yue Luo
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Yan Lin
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Rong Liang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Jiazhang Wei
- Department of Otolaryngology & Head and Neck, The People's Hospital of Guangxi Zhuang Autonomous Region
- Institute of Oncology, Guangxi Academy of Medical Sciences
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor
| | - Jinyan Zhang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| | - Yongqiang Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital
| |
Collapse
|
72
|
Zhu S, Li Z, Zheng D, Yu Y, Xiang J, Ma X, Xu D, Qiu J, Yang Z, Wang Z, Li J, Sun H, Chen W, Meng X, Lu Y, Ren Q. A cancer cell membrane coated, doxorubicin and microRNA co-encapsulated nanoplatform for colorectal cancer theranostics. Mol Ther Oncolytics 2022; 28:182-196. [PMID: 36820302 PMCID: PMC9937835 DOI: 10.1016/j.omto.2022.12.002] [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: 02/16/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Endogenous microRNAs (miRNA) in tumors are currently under exhaustive investigation as potential therapeutic agents for cancer treatment. Nevertheless, RNase degradation, inefficient and untargeted delivery, limited biological effect, and currently unclear side effects remain unsettled issues that frustrate clinical application. To address this, a versatile targeted delivery system for multiple therapeutic and diagnostic agents should be adapted for miRNA. In this study, we developed membrane-coated PLGA-b-PEG DC-chol nanoparticles (m-PPDCNPs) co-encapsulating doxorubicin (Dox) and miRNA-190-Cy7. Such a system showed low biotoxicity, high loading efficiency, and superior targeting ability. Systematic delivery of m-PPDCNPs in mouse models showed exceptionally specific tumor accumulation. Sustained release of miR-190 inhibited tumor angiogenesis, tumor growth, and migration by regulating a large group of angiogenic effectors. Moreover, m-PPDCNPs also enhanced the sensitivity of Dox by suppressing TGF-β signal in colorectal cancer cell lines and mouse models. Together, our results demonstrate a stimulating and promising m-PPDCNPs nanoplatform for colorectal cancer theranostics.
Collapse
Affiliation(s)
- Sihao Zhu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Ziyuan Li
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Dongye Zheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yue Yu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jing Xiang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xiao Ma
- Research Group Signal Transduction, Department of Psychiatry, Ludwig Maximilian University of Munich, Nussbaumstr.7, 80336 Munich, Germany
| | - Dongqing Xu
- Department of Pediatric Hematology/Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jiajun Qiu
- Department of Otolaryngology Head and Neck Surgery, the Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ziyu Yang
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Zhiyi Wang
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jun Li
- Laboratory Animal Center, Peking University, Beijing 100871, China
| | - Hongfang Sun
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, Gansu Province, China,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou 730000, Gansu Province, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China,NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing 100142, China,Corresponding author.
| | - Yanye Lu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China,Corresponding author.
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China,Corresponding author.
| |
Collapse
|
73
|
Lu Y, Huntoon K, Lee D, Wang Y, Ha J, Qie Y, Li X, Schrank BR, Dong S, Gallup TD, Kang M, Zhao H, An Y, Yang Z, Li J, Kim BYS, Jiang W. Immunological conversion of solid tumours using a bispecific nanobioconjugate for cancer immunotherapy. NATURE NANOTECHNOLOGY 2022; 17:1332-1341. [PMID: 36357792 PMCID: PMC10036139 DOI: 10.1038/s41565-022-01245-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 09/23/2022] [Indexed: 05/06/2023]
Abstract
Solid tumours display a limited response to immunotherapies. By contrast, haematological malignancies exhibit significantly higher response rates to immunotherapies as compared with solid tumours. Among several microenvironmental and biological disparities, the differential expression of unique immune regulatory molecules contributes significantly to the interaction of blood cancer cells with immune cells. The self-ligand receptor of the signalling lymphocytic activation molecule family member 7 (SLAMF7), a molecule that is critical in promoting the body's innate immune cells to detect and engulf cancer cells, is expressed nearly exclusively on the cell surface of haematologic tumours, but not on solid ones. Here we show that a bispecific nanobioconjugate that enables the decoration of SLAMF7 on the surface of solid tumours induces robust phagocytosis and activates the phagocyte cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway, sensitizing the tumours to immune checkpoint blockade. Our findings support an immunological conversion strategy that uses nano-adjuvants to improve the effectiveness of immunotherapies for solid tumours.
Collapse
Affiliation(s)
- Yifei Lu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - DaeYong Lee
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - JongHoon Ha
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yaqing Qie
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuefeng Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benjamin R Schrank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiyan Dong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas D Gallup
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Minjeong Kang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hai Zhao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yi An
- Department of Therapeutic Radiology, Yale New Haven Hospital, New Haven, CT, USA
| | - Zhaogang Yang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
74
|
Liu L, Chen J. Therapeutic antibodies for precise cancer immunotherapy: current and future perspectives. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:555-569. [PMID: 37724258 PMCID: PMC10471122 DOI: 10.1515/mr-2022-0033] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/25/2022] [Indexed: 09/20/2023]
Abstract
Antibodies, as one of the most important components of host adaptive immune system, play an important role in defense of infectious disease, immune surveillance, and autoimmune disease. Due to the development of recombinant antibody technology, antibody therapeutics become the largest and rapidly expanding drug to provide major health benefits to patients, especially for the treatment of cancer patients. Many antibody-based therapeutic strategies have been developed including monoclonal antibodies, antibody-drug conjugates, bispecific and trispecific antibodies and pro-antibodies with promising results from both clinical and pre-clinical trials. However, the response rate and side-effect still vary between patients with undefined mechanisms. Here, we summarized the current and future perspectives of antibody-based cancer immunotherapeutic strategies for designing next-generation drugs.
Collapse
Affiliation(s)
- Longchao Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jiahui Chen
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
75
|
Matthews AH, Pratz KW, Carroll MP. Targeting Menin and CD47 to Address Unmet Needs in Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:5906. [PMID: 36497385 PMCID: PMC9735817 DOI: 10.3390/cancers14235906] [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: 08/30/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 12/02/2022] Open
Abstract
After forty years of essentially unchanged treatment in acute myeloid leukemia (AML), innovation over the past five years has been rapid, with nine drug approvals from 2016 to 2021. Increased understanding of the molecular changes and genetic ontology of disease have led to targeting mutations in isocitrate dehydrogenase, FMS-like tyrosine kinase 3 (FLT3), B-cell lymphoma 2 and hedgehog pathways. Yet outcomes remain variable; especially in defined molecular and genetic subgroups such as NPM1 (Nucleophosmin 1) mutations, 11q23/KMT2A rearranged and TP53 mutations. Emerging therapies seek to address these unmet needs, and all three of these subgroups have promising new therapeutic approaches. Here, we will discuss the normal biological roles of menin in acute leukemia, notably in KMT2A translocations and NPM1 mutation, as well as current drug development. We will also explore how CD47 inhibition may move immunotherapy into front-line settings and unlock new treatment strategies in TP53 mutated disease. We will then consider how these new therapeutic advances may change the management of AML overall.
Collapse
Affiliation(s)
- Andrew H. Matthews
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Keith W. Pratz
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 715 Biomedical Research Building II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
76
|
Gao X, Yang H, Xiao W, Su J, Zhang Y, Wang H, Ni W, Gu Y. Modified exosomal SIRPα variants alleviate white matter injury after intracerebral hemorrhage via microglia/macrophages. Biomater Res 2022; 26:67. [DOI: 10.1186/s40824-022-00311-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/27/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract
Background
Despite limited efficiency, modulation of microglia/macrophages has shown to attenuate neuroinflammation after intracerebral hemorrhage (ICH). In this context, we evaluated the efficacy of modified exosomal signal regulatory protein α (SIRPα) variants (SIRPα-v Exos) in microglia/macrophages and neuroinflammation-associated white matter injury after ICH.
Methods
SIRPα-v Exos were engineered to block CD47-SIRPα interactions. After obtaining SIRPα-v Exos from lentivirus-infected mesenchymal stem cells, C57BL/6 mice suffering from ICH underwent consecutive intravenous injections of SIRPα-v Exos (6 mg/kg) for 14 days. Afterwards, the volume of hematoma and neurological dysfunctions were assessed in mice continuously until 35 days after ICH. In addition, demyelination, electrophysiology and neuroinflammation were evaluated. Furthermore, the mechanisms of microglial regulation by SIRPα-v Exos were investigated in vitro under coculture conditions.
Results
The results demonstrated that the clearance of hematoma in mice suffering from ICH was accelerated after SIRPα-v Exo treatment. SIRPα-v Exos improved long-term neurological dysfunction by ameliorating white matter injury. In addition, SIRPα-v Exos recruited regulatory T cells (Tregs) to promote M2 polarization of microglia/macrophages in the peri-hematoma tissue. In vitro experiments further showed that SIRPα-v Exos regulated primary microglia in a direct and indirect manner in synergy with Tregs.
Conclusion
Our studies revealed that SIRPα-v Exos could accelerate the clearance of hematoma and ameliorate secondary white matter injury after ICH through regulation of microglia/macrophages. SIRPα-v Exos may become a promising treatment for ICH in clinical practice.
Graphical Abstract
Collapse
|
77
|
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: 0.5] [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
|
78
|
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: 4.0] [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
|
79
|
Son J, Hsieh RCE, Lin HY, Krause KJ, Yuan Y, Biter AB, Welsh J, Curran MA, Hong DS. Inhibition of the CD47-SIRPα axis for cancer therapy: A systematic review and meta-analysis of emerging clinical data. Front Immunol 2022; 13:1027235. [PMID: 36439116 PMCID: PMC9691650 DOI: 10.3389/fimmu.2022.1027235] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/30/2022] [Indexed: 08/11/2023] Open
Abstract
CD47-SIRPα interaction acts as a "don't eat me" signal and is exploited by cancer to downregulate innate and adaptive immune surveillance. There has been intense interest to develop a mechanism of blockade, and we aimed to analyze the emerging data from early clinical trials. We performed a systematic review and meta-analysis of relevant databases and conference abstracts including clinical trials using CD47 and/or SIRPα inhibitors in cancer treatment. Nonlinear mixed models were applied for comparison of response and toxicity. We retrieved 317 articles, 24 of which were eligible. These included 771 response-evaluable patients with hematologic (47.1%) and solid tumors (52.9%). Of these, 6.4% experienced complete response, 10.4% partial response, and 26.1% stable disease for a 16.7% objective response rate (ORR), 42.8% disease control rate, and 4.8-month median duration of response. ORR was significantly higher for hematologic cancers (25.3%) than solid cancers (9.1%, p=0.042). Comparing by mechanism, seven CD47 monoclonal antibodies (mAbs) and six selective SIRPα blockers were given alone or combined with checkpoint inhibitors, targeted therapy, and/or chemotherapy. In solid cancers, selective SIRPα blockade showed a higher ORR (16.2%) than anti-CD47 mAbs (2.8%, p=0.079), which was significant for combination therapies (ORR 28.3% vs 3.0%, respectively, p=0.010). Responses were seen in head and neck, colorectal, endometrial, ovarian, hepatocellular, non-small cell lung, and HER2+gastroesophageal cancers. Dose-limiting toxicity (DLT) was seen in 3.3% of patients (5.4% anti-CD47 mAbs, 1.4% selective SIRPα blockers; p=0.01). The frequency of treatment-related adverse events (TRAEs) ≥grade 3 was 18.0%, similar between the two groups (p=0.082), and mostly laboratory abnormalities. For anti-CD47 mAbs, the most common toxicities included grade 1-2 fatigue (27.2%), headache (21.0%), and anemia (20.5%). For selective SIRPα blockers, these included grade 1-2 infusion reaction (23.1%) and fatigue (15.8%). Anti-CD47 mAbs were significantly more likely than selective SIRPα blockers to cause grade 1-2 fever, chills, nausea/vomiting, headache, and anemia. In conclusion, combination therapies using selective SIRPα blockade had higher response rates in solid tumors than anti-CD47 mAb combinations. Hematologic changes were the main TRAEs, and selective SIRPα blockers seemed to have a better grade 1-2 toxicity profile. Treatment was well-tolerated with minimal DLTs.
Collapse
Affiliation(s)
- Ji Son
- Departments of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rodney Cheng-En Hsieh
- Departments of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Heather Y. Lin
- Departments of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kate J. Krause
- Departments of Research Medical Library, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ying Yuan
- Departments of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amadeo B. Biter
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James Welsh
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael A. Curran
- Departments of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David S. Hong
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
80
|
CDKN1A is a target for phagocytosis-mediated cellular immunotherapy in acute leukemia. Nat Commun 2022; 13:6739. [PMID: 36347876 PMCID: PMC9643439 DOI: 10.1038/s41467-022-34548-3] [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: 05/06/2021] [Accepted: 10/28/2022] [Indexed: 11/09/2022] Open
Abstract
Targeting the reprogramming and phagocytic capacities of tumor-associated macrophages (TAMs) has emerged as a therapeutic opportunity for cancer treatment. Here, we demonstrate that tumor cell phagocytosis drives the pro-inflammatory activation of TAMs and identify a key role for the cyclin-dependent kinase inhibitor CDKN1A (p21). Through the transcriptional repression of Signal-Regularity Protein α (SIRPα), p21 promotes leukemia cell phagocytosis and, subsequently, the pro-inflammatory reprogramming of phagocytic macrophages that extends to surrounding macrophages through Interferon γ. In mouse models of human T-cell acute lymphoblastic leukemia (T-ALL), infusion of human monocytes (Mos) engineered to overexpress p21 (p21TD-Mos) leads to Mo differentiation into phagocytosis-proficient TAMs that, after leukemia cell engulfment, undergo pro-inflammatory activation and trigger the reprogramming of bystander TAMs, reducing the leukemic burden and substantially prolonging survival in mice. These results reveal p21 as a trigger of phagocytosis-guided pro-inflammatory TAM reprogramming and highlight the potential for p21TD-Mo-based cellular therapy as a cancer immunotherapy.
Collapse
|
81
|
Feng Q, Ma X, Cheng K, Liu G, Li Y, Yue Y, Liang J, Zhang L, Zhang T, Wang X, Gao X, Nie G, Zhao X. Engineered Bacterial Outer Membrane Vesicles as Controllable Two-Way Adaptors to Activate Macrophage Phagocytosis for Improved Tumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2206200. [PMID: 35985666 DOI: 10.1002/adma.202206200] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/08/2022] [Indexed: 06/15/2023]
Abstract
The most immune cells infiltrating tumor microenvironment (TME), tumor-associated macrophages (TAMs) closely resemble immunosuppressive M2-polarized macrophages. Moreover, tumor cells exhibit high expression of CD47 "don't eat me" signal, which obstructs macrophage phagocytosis. The precise and efficient activation of TAMs is a promising approach to tumor immunotherapy; however, re-education of macrophages remains a challenge. Bacteria-derived outer membrane vesicles (OMVs) are highly immunogenic nanovesicles that can robustly stimulate macrophages. Here, an OMV-based controllable two-way adaptor is reported, in which a CD47 nanobody (CD47nb) is fused onto OMV surface (OMV-CD47nb), with the outer surface coated with a polyethylene glycol (PEG) layer containing diselenide bonds (PEG/Se) to form PEG/Se@OMV-CD47nb. The PEG/Se layer modification not only mitigates the immunogenicity of OMV-CD47nb, thereby remarkedly increasing the dose that can be administered safely through intravenous injection, but also equips the formulation with radiation-triggered controlled release of OMV-CD47nb. Application of radiation to tumors in mice injected with the nanoformulation results in remodeling of TME. As two-way adaptors, OMV-CD47nb activates TAM phagocytosis of tumor cells via multiple pathways, including induction of M1 polarization and blockade of "don't eat me" signal. Moreover, this activation of TAMs results in the stimulation of T cell-mediated antitumor immunity through effective antigen presentation.
Collapse
Affiliation(s)
- Qingqing Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xiaotu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yale Yue
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Lizhuo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Tianjiao Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
82
|
The Impaired Mechanism and Facilitated Therapies of Efferocytosis in Atherosclerosis. J Cardiovasc Pharmacol 2022; 80:407-416. [PMID: 35853202 DOI: 10.1097/fjc.0000000000001311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/21/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Cardiovascular disease is responsible for the largest number of deaths worldwide, and atherosclerosis is the primary cause. Apoptotic cell accumulation in atherosclerotic plaques leads to necrotic core formation and plaque rupture. Emerging findings show that the progression of atherosclerosis appears to suppress the elimination of apoptotic cells. Mechanistically, the reduced edibility of apoptotic cells, insufficient phagocytic capacity of phagocytes, downregulation of bridging molecules, and dysfunction in the polarization of macrophages lead to impaired efferocytosis in atherosclerotic plaques. This review focuses on the characteristics of efferocytosis in plaques and the therapeutic strategies aimed at promoting efferocytosis in atherosclerosis, which would provide novel insights for the development of antiatherosclerotic drugs based on efferocytosis.
Collapse
|
83
|
Zhu J, Cai C, Li J, Xiao J, Duan X. CD47-SIRPα axis in cancer therapy: Precise delivery of CD47-targeted therapeutics and design of anti-phagocytic drug delivery systems. MEDICINE IN DRUG DISCOVERY 2022. [DOI: 10.1016/j.medidd.2022.100139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
84
|
Mucke HA. Patent Highlights April - May 2022. Pharm Pat Anal 2022; 11:139-145. [PMID: 36052651 DOI: 10.4155/ppa-2022-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
85
|
Yu J, Li S, Chen D, Liu D, Guo H, Yang C, Zhang W, Zhang L, Zhao G, Tu X, Peng L, Liu S, Bai X, Song Y, Jiang Z, Zhang R, Tian W. Crystal Structure of Human CD47 in Complex with Engineered SIRPα.D1(N80A). Molecules 2022; 27:molecules27175574. [PMID: 36080360 PMCID: PMC9457805 DOI: 10.3390/molecules27175574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Targeting the CD47/SIRPα signaling pathway represents a novel approach to enhance anti-tumor immunity. However, the crystal structure of the CD47/SIRPα has not been fully studied. This study aims to analyze the structure interface of the complex of CD47 and IMM01, a novel recombinant SIRPα-Fc fusion protein. Methods: IMM01-Fab/CD47 complex was crystalized, and diffraction images were collected. The complex structure was determined by molecular replacement using the program PHASER with the CD47-SIRPαv2 structure (PDB code 2JJT) as a search model. The model was manually built using the COOT program and refined using TLS parameters in REFMAC from the CCP4 program suite. Results: Crystallization and structure determination analysis of the interface of IMM01/CD47 structure demonstrated CD47 surface buried by IMM01. Comparison with the literature structure (PDB ID 2JJT) showed that the interactions of IMM01/CD47 structure are the same. All the hydrogen bonds that appear in the literature structure are also present in the IMM01/CD47 structure. These common hydrogen bonds are stable under different crystal packing styles, suggesting that these hydrogen bonds are important for protein binding. In the structure of human CD47 in complex with human SIRPα, except SER66, the amino acids that form hydrogen bonds are all conserved. Furthermore, comparing with the structure of PDB ID 2JJT, the salt bridge interaction from IMM01/CD47 structure are very similar, except the salt bridge bond between LYS53 in IMM01 and GLU106 in CD47, which only occurs between the B and D chains. However, as the side chain conformation of LYS53 in chain A is slightly different, the salt bridge bond is absent between the A and C chains. At this site between chain A and chain C, there are a salt bridge bond between LYS53 (A) and GLU104 (C) and a salt bridge bond between HIS56 (A) and GLU106 (C) instead. According to the sequence alignment results of SIRPα, SIRPβ and SIRPγ in the literature of PDB ID 2JJT, except ASP100, the amino acids that form common salt bridge bonds are all conserved. Conclusion: Our data demonstrated crystal structure of the IMM01/CD47 complex and provides a structural basis for the structural binding interface and future clinical applications.
Collapse
Affiliation(s)
- Jifeng Yu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Song Li
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Dianze Chen
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Dandan Liu
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Huiqin Guo
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Chunmei Yang
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Wei Zhang
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Li Zhang
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Gui Zhao
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Xiaoping Tu
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Liang Peng
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Sijin Liu
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Xing Bai
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ruliang Zhang
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai 201203, China
- Correspondence:
| |
Collapse
|
86
|
Wang Y, Johnson KCC, Gatti-Mays ME, Li Z. Emerging strategies in targeting tumor-resident myeloid cells for cancer immunotherapy. J Hematol Oncol 2022; 15:118. [PMID: 36031601 PMCID: PMC9420297 DOI: 10.1186/s13045-022-01335-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/09/2022] [Indexed: 12/11/2022] Open
Abstract
Immune checkpoint inhibitors targeting programmed cell death protein 1, programmed death-ligand 1, and cytotoxic T-lymphocyte-associated protein 4 provide deep and durable treatment responses which have revolutionized oncology. However, despite over 40% of cancer patients being eligible to receive immunotherapy, only 12% of patients gain benefit. A key to understanding what differentiates treatment response from non-response is better defining the role of the innate immune system in anti-tumor immunity and immune tolerance. Teleologically, myeloid cells, including macrophages, dendritic cells, monocytes, and neutrophils, initiate a response to invading pathogens and tissue repair after pathogen clearance is successfully accomplished. However, in the tumor microenvironment (TME), these innate cells are hijacked by the tumor cells and are imprinted to furthering tumor propagation and dissemination. Major advancements have been made in the field, especially related to the heterogeneity of myeloid cells and their function in the TME at the single cell level, a topic that has been highlighted by several recent international meetings including the 2021 China Cancer Immunotherapy workshop in Beijing. Here, we provide an up-to-date summary of the mechanisms by which major myeloid cells in the TME facilitate immunosuppression, enable tumor growth, foster tumor plasticity, and confer therapeutic resistance. We discuss ongoing strategies targeting the myeloid compartment in the preclinical and clinical settings which include: (1) altering myeloid cell composition within the TME; (2) functional blockade of immune-suppressive myeloid cells; (3) reprogramming myeloid cells to acquire pro-inflammatory properties; (4) modulating myeloid cells via cytokines; (5) myeloid cell therapies; and (6) emerging targets such as Siglec-15, TREM2, MARCO, LILRB2, and CLEVER-1. There is a significant promise that myeloid cell-based immunotherapy will help advance immuno-oncology in years to come.
Collapse
Affiliation(s)
- Yi Wang
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Margaret E Gatti-Mays
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
- Stefanie Spielman Comprehensive Breast Center, Columbus, OH, USA.
| | - Zihai Li
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
87
|
Flavonoids regulate tumor-associated macrophages - From structure-activity relationship to clinical potential (Review). Pharmacol Res 2022; 184:106419. [PMID: 36041653 DOI: 10.1016/j.phrs.2022.106419] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/13/2022] [Accepted: 08/25/2022] [Indexed: 11/23/2022]
Abstract
In recent years, the strategy for tumor therapy has changed from focusing on the direct killing effect of different types of therapeutic agents on cancer cells to the new mainstream of multi-mode and -pathway combined interventions in the microenvironment of the developing tumor. Flavonoids, with unique tricyclic structures, have diverse and extensive immunomodulatory and anti-cancer activities in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are the most abundant immunosuppressive cells in the TME. The regulation of macrophages to fight cancer is a promising immunotherapeutic strategy. This study covers the most comprehensive cognition of flavonoids in regulating TAMs so far. Far more than a simple list of studies, we try to dig out evidence of crosstalk at the molecular level between flavonoids and TAMs from literature, in order to discuss the most relevant chemical structure and its possible relationship with the multimodal pharmacological activity, as well as systematically build a structure-activity relationship between flavonoids and TAMs. Additionally, we point out the advantages of the macro-control of flavonoids in the TME and discuss the potential clinical implications as well as areas for future research of flavonoids in regulating TAMs. These results will provide hopeful directions for the research of antitumor drugs, while providing new ideas for the pharmaceutical industry to develop more effective forms of flavonoids.
Collapse
|
88
|
Zhang Y, Wang Y, Zhao Z, Peng W, Wang P, Xu X, Zhao C. Glutaminyl cyclases, the potential targets of cancer and neurodegenerative diseases. Eur J Pharmacol 2022; 931:175178. [DOI: 10.1016/j.ejphar.2022.175178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 11/03/2022]
|
89
|
Abstract
The daily removal of billions of apoptotic cells in the human body via the process of efferocytosis is essential for homeostasis. To allow for this continuous efferocytosis, rapid phenotypic changes occur in the phagocytes enabling them to engulf and digest the apoptotic cargo. In addition, efferocytosis is actively anti-inflammatory and promotes resolution. Owing to its ubiquitous nature and the sheer volume of cell turnover, efferocytosis is a point of vulnerability. Aberrations in efferocytosis are associated with numerous inflammatory pathologies, including atherosclerosis, cancer and infections. The recent exciting discoveries defining the molecular machinery involved in efferocytosis have opened many avenues for therapeutic intervention, with several agents now in clinical trials.
Collapse
Affiliation(s)
- Parul Mehrotra
- Unit for Cell Clearance in Health and Disease, VIB Center for Inflammation Research, Ghent, Belgium
| | - Kodi S Ravichandran
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- The Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
90
|
The Generation of Dual-Targeting Fusion Protein PD-L1/CD47 for the Inhibition of Triple-Negative Breast Cancer. Biomedicines 2022; 10:biomedicines10081843. [PMID: 36009390 PMCID: PMC9405206 DOI: 10.3390/biomedicines10081843] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subset of breast cancer with limited therapeutic options. However, its immune evasion mechanisms, characterized by the over-expression of the immune checkpoint molecules PD-L1 and CD47, can be targeted in order to facilitate cancer elimination by cells of innate and adaptive immunity. In this paper, we describe the design, preparation, and evaluation of three novel dual-targeting fusion proteins that were based on the structure frame of prototype IAB (innate and adaptive dependent bispecific fusion protein) and the “Orcutt-type IgG-scFv” molecular model. Three molecules with different spatial conformations were designed to improve antigen–antibody affinity by the addition of Ag–Ab binding sites from the variable region sequences of the anti-PD-L1 monoclonal antibody (mAb) atezolizumab and CV1, a high-affinity receptor of CD47. The results showed that the best-performing among the three proteins designed in this study was protein Pro3; its CV1 N-terminus and Fc domain C-terminus were not sterically hindered. Pro3 was better at boosting T cell proliferation and the engulfment of macrophages than the IAB prototype and, at the same time, retained a level of ADCC activity similar to that of IAB. Through improved design, the novel constructed dual-targeting immunomodulatory protein Pro3 was superior at activating the anti-tumor immune response and has thus shown potential for use in clinical applications.
Collapse
|
91
|
Behrens LM, van den Berg TK, van Egmond M. Targeting the CD47-SIRPα Innate Immune Checkpoint to Potentiate Antibody Therapy in Cancer by Neutrophils. Cancers (Basel) 2022; 14:cancers14143366. [PMID: 35884427 PMCID: PMC9319280 DOI: 10.3390/cancers14143366] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Immunotherapy aims to engage various immune cells in the elimination of cancer cells. Neutrophils are the most abundant leukocytes in the circulation and have unique mechanisms by which they can kill cancer cells opsonized by antibodies. However, neutrophil effector functions are limited by the inhibitory receptor SIRPα, when it interacts with CD47. The CD47 protein is expressed on all cells in the body and acts as a ‘don’t eat me’ signal to prevent tissue damage. Cancer cells can express high levels of CD47 to circumvent tumor elimination. Thus, blocking the interaction between CD47 and SIRPα may enhance anti-tumor effects by neutrophils in the presence of tumor-targeting monoclonal antibodies. In this review, we discuss CD47-SIRPα as an innate immune checkpoint on neutrophils and explore the preliminary results of clinical trials using CD47-SIRPα blocking agents. Abstract In the past 25 years, a considerable number of therapeutic monoclonal antibodies (mAb) against a variety of tumor-associated antigens (TAA) have become available for the targeted treatment of hematologic and solid cancers. Such antibodies opsonize cancer cells and can trigger cytotoxic responses mediated by Fc-receptor expressing immune cells in the tumor microenvironment (TME). Although frequently ignored, neutrophils, which are abundantly present in the circulation and many cancers, have demonstrated to constitute bona fide effector cells for antibody-mediated tumor elimination in vivo. It has now also been established that neutrophils exert a unique mechanism of cytotoxicity towards antibody-opsonized tumor cells, known as trogoptosis, which involves Fc-receptor (FcR)-mediated trogocytosis of cancer cell plasma membrane leading to a lytic/necrotic type of cell death. However, neutrophils prominently express the myeloid inhibitory receptor SIRPα, which upon interaction with the ‘don’t eat me’ signal CD47 on cancer cells, limits cytotoxicity, forming a mechanism of resistance towards anti-cancer antibody therapeutics. In fact, tumor cells often overexpress CD47, thereby even more strongly restricting neutrophil-mediated tumor killing. Blocking the CD47-SIRPα interaction may therefore potentiate neutrophil-mediated antibody-dependent cellular cytotoxicity (ADCC) towards cancer cells, and various inhibitors of the CD47-SIRPα axis are now in clinical studies. Here, we review the role of neutrophils in antibody therapy in cancer and their regulation by the CD47-SIRPα innate immune checkpoint. Moreover, initial results of CD47-SIRPα blockade in clinical trials are discussed.
Collapse
Affiliation(s)
- Leonie M. Behrens
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology Program, 1081 HV Amsterdam, The Netherlands
- Correspondence:
| | - Timo K. van den Berg
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Byondis B.V., 6545 CM Nijmegen, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology Program, 1081 HV Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
92
|
Chen C, Wang R, Chen X, Hou Y, Jiang J. Targeting CD47 as a Novel Immunotherapy for Breast Cancer. Front Oncol 2022; 12:924740. [PMID: 35860564 PMCID: PMC9289165 DOI: 10.3389/fonc.2022.924740] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nowadays, breast cancer has become the most common cancer worldwide with a high mortality rate. Immune checkpoint blockade holds great promise in tumor‐targeted therapy, and CD47 blockade as one immune therapy is undergoing various preclinical studies and clinical trials to demonstrate its safety and efficacy in breast cancer. In this review, we summarized different therapeutic mechanisms targeting CD47 and its prognostic role and therapeutic value in breast cancer.
Collapse
Affiliation(s)
- Can Chen
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Runlu Wang
- Respiratory Division, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xi Chen
- Department of Dermatology, First People’s Hospital, Huzhou, China
| | - Yulong Hou
- Department of Surgery, Huzhou Central Hospital, Huzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- *Correspondence: Jingting Jiang,
| |
Collapse
|
93
|
van Duijn A, Van der Burg SH, Scheeren FA. CD47/SIRPα axis: bridging innate and adaptive immunity. J Immunother Cancer 2022; 10:jitc-2022-004589. [PMID: 35831032 PMCID: PMC9280883 DOI: 10.1136/jitc-2022-004589] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 11/29/2022] Open
Abstract
Myeloid immune cells are frequently present in the tumor environment, and although they can positively contribute to tumor control they often negatively impact anticancer immune responses. One way of inhibiting the positive contributions of myeloid cells is by signaling through the cluster of differentiation 47 (CD47)/signal regulatory protein alpha (SIRPα) axis. The SIRPα receptor is expressed on myeloid cells and is an inhibitory immune receptor that, upon binding to CD47 protein, delivers a ‘don’t eat me’ signal. As CD47 is often overexpressed on cancer cells, treatments targeting CD47/SIRPα have been under active investigation and are currently being tested in clinical settings. Interestingly, the CD47/SIRPα axis is also involved in T cell-mediated antitumor responses. In this perspective we provide an overview of recent studies showing how therapeutic blockade of the CD47/SIRPα axis improves the adaptive immune response. Furthermore, we discuss the interconnection between the myeloid CD47/SIRPα axis and adaptive T cell responses as well as the potential therapeutic role of the CD47/SIRPα axis in tumors with acquired resistance to the classic immunotherapy through major histocompatibility complex downregulation. Altogether this review provides a profound insight for the optimal exploitation of CD47/SIRPα immune checkpoint therapy.
Collapse
Affiliation(s)
- Anneloes van Duijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H Van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
94
|
Zhang B, Shu Y, Hu S, Qi Z, Chen Y, Ma J, Wang Y, Cheng P. In Situ Tumor Vaccine Expressing Anti-CD47 Antibody Enhances Antitumor Immunity. Front Oncol 2022; 12:897561. [PMID: 35837100 PMCID: PMC9273963 DOI: 10.3389/fonc.2022.897561] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
In situ tumor vaccine is a potential cancer therapy due to their advantages in induction of antitumor immune responses. Oncolytic virotherapy utilizes natural or engineered oncolytic viruses to kill tumors selectively, representing a promising in situ tumor vaccine for cancer immunotherapy. In addition to direct oncolysis, oncolytic viruses elicit potent and durable antitumor immune responses by induction of immunogenic cell death of tumors. Membrane protein CD47 overexpressed on tumor cells engages in “don’t eat me” signal that prevents macrophages from engulfing tumor cells. CD47-targeting agents have been tested via preclinical and clinical trials. As potential tumor vaccine vectors, oncolytic viruses can be engineered to express anti-CD47 antibodies to induce potentiated tumor killing. Therefore, we developed an adenovirus-based tumor vaccine loaded with a CD47-targeting nanobody fused with the IgG2a Fc protein. B16-F10 melanoma, A20 lymphoma, and 4T1 breast cancer models in immunocompetent mice were established to evaluated in vivo antitumor efficacy of in situ tumor vaccination. The tumor vaccine armed with a nanobody against CD47 induced durable suppression of the tumor and long-term survival of tumor-bearing mice, and also elevated the number of tumor-infiltrating immune cells with an activated immunophenotype, suggesting that it could remodel the tumor immune microenvironment. Systemic antitumor effects and immune memory were also observed in immunocompetent mice following in situ vaccination with the anti-CD47 tumor vaccines; tumorigenesis was completely inhibited in these mice after tumor re-challenge. The recombinant anti-CD47 tumor vaccine has an effectual antitumor activity and may be a promising antitumor agent.
Collapse
|
95
|
Cao X, Lai SWT, Chen S, Wang S, Feng M. Targeting tumor-associated macrophages for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:61-108. [PMID: 35636930 DOI: 10.1016/bs.ircmb.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Tumor-associated macrophages (TAMs) are one of the most abundant immune components in the tumor microenvironment and play a plethora of roles in regulating tumorigenesis. Therefore, the therapeutic targeting of TAMs has emerged as a new paradigm for immunotherapy of cancer. Herein, the review summarizes the origin, polarization, and function of TAMs in the progression of malignant diseases. The understanding of such knowledge leads to several distinct therapeutic strategies to manipulate TAMs to battle cancer, which include those to reduce TAM abundance, such as depleting TAMs or inhibiting their recruitment and differentiation, and those to harness or boost the anti-tumor activities of TAMs such as blocking phagocytosis checkpoints, inducing antibody-dependent cellular phagocytosis, and reprogramming TAM polarization. In addition, modulation of TAMs may reshape the tumor microenvironment and therefore synergize with other cancer therapeutics. Therefore, the rational combination of TAM-targeting therapeutics with conventional therapies including radiotherapy, chemotherapy, and other immunotherapies is also reviewed. Overall, targeting TAMs presents itself as a promising strategy to add to the growing repertoire of treatment approaches in the fight against cancer, and it is hopeful that these approaches currently being pioneered will serve to vastly improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| | - Seigmund W T Lai
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Siqi Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Sadira Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| |
Collapse
|
96
|
Jarr KU, Kojima Y, Weissman IL, Leeper NJ. 2021 Jeffrey M. Hoeg Award Lecture: Defining the Role of Efferocytosis in Cardiovascular Disease: A Focus on the CD47 (Cluster of Differentiation 47) Axis. Arterioscler Thromb Vasc Biol 2022; 42:e145-e154. [PMID: 35387480 PMCID: PMC9183217 DOI: 10.1161/atvbaha.122.317049] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/21/2022] [Indexed: 01/09/2023]
Abstract
A key feature of atherogenesis is the accumulation of diseased and dying cells within the lesional necrotic core. While the burden of intraplaque apoptotic cells may be driven in part by an increase in programmed cell death, mounting evidence suggests that their presence may primarily be dictated by a defect in programmed cell removal, or efferocytosis. In this brief review, we will summarize the evidence suggesting that inflammation-dependent changes within the plaque render target cells inedible and reduce the appetite of lesional phagocytes. We will present the genetic causation studies, which indicate these phenomena promote lesion expansion and plaque vulnerability, and the interventional data which suggest that these processes can be reversed. Particular emphasis is provided related to the antiphagocytic CD47 (cluster of differentiation 47) do not eat me axis, which has emerged as a novel antiatherosclerotic translational target that is predicted to provide benefit independent of traditional cardiovascular risk factors.
Collapse
Affiliation(s)
- Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yoko Kojima
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Irving L. Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
97
|
Alausa A, Lawal KA, Babatunde OA, Obiwulu ENO, Oladokun OC, Fadahunsi OS, Celestine UO, Moses EU, Rejoice AI, Adegbola PI. Overcoming Immunotherapeutic Resistance in PDAC: SIRPα-CD47 blockade. Pharmacol Res 2022; 181:106264. [PMID: 35597384 DOI: 10.1016/j.phrs.2022.106264] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/15/2022] [Indexed: 11/25/2022]
Abstract
A daily increase in the number of new cases of pancreatic ductal adenocarcinoma remains an issue of contention in cancer research. The data revealed that a global cumulated case of about 500, 000 have been reported. This has made PDAC the fourteenth most occurring tumor case in cancer research. Furthermore, PDAC is responsible for about 466,003 deaths annually, representing the seventh prevalent type of cancer mortality. PDAC has no salient symptoms in its early stages. This has exasperated several attempts to produce a perfect therapeutic agent against PDAC. Recently, immunotherapeutic research has shifted focus to the blockade of checkpoint proteins in the management and of some cancers. Investigations have centrally focused on developing therapeutic agents that could at least to a significant extent block the SIRPα-CD47 signaling cascade (a cascade which prevent phagocytosis of tumors by dendritic cells, via the deactivation of innate immunity and subsequently resulting in tumor regression) with minimal side effects. The concept on the blockade of this interaction as a possible mechanism for inhibiting the progression of PDAC is currently being debated. This review examined the structure--function activity of SIRPα-CD47 interaction while discussing in detail the mechanism of tumor resistance in PDAC. Further, this review details how the blockade of SIRPα-CD47 interaction serve as a therapeutic option in the management of PDAC.
Collapse
Affiliation(s)
- Abdullahi Alausa
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo state.
| | - Khadijat Ayodeji Lawal
- Heamtalogy and Blood Transfusion Unit, Department of Medical Laboratory Science, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - E N O Obiwulu
- Department of Chemical Science, University of Delta, Agbor, Delta State
| | | | | | - Ugwu Obiora Celestine
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Enugu State University of Science and Technology
| | | | | | | |
Collapse
|
98
|
Combining daratumumab with CD47 blockade prolongs survival in preclinical models of pediatric T-ALL. Blood 2022; 140:45-57. [PMID: 35452517 DOI: 10.1182/blood.2021014485] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/18/2022] [Indexed: 11/20/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignant disease affecting children. Although therapy strategies improved, T-cell acute lymphoblastic leukemia (T-ALL) relapse is associated with chemoresistance and a poor prognosis. One strategy to overcome this obstacle is the application of monoclonal antibodies. Here, we show that leukemic cells from T-ALL patients express surface CD38 and CD47, both attractive targets for antibody therapy. We therefore investigated the commercially available CD38 antibody daratumumab (Dara) in combination with a proprietary modified CD47 antibody (Hu5F9-IgG2σ) in vitro and in vivo. Compared to single treatments, this combination significantly increased in vitro antibody-dependent cellular phagocytosis (ADCP) in T-ALL cell lines as well as in random de novo and in relapsed/refractory T-ALL patient derived xenograft (PDX) samples. Similarly, enhanced ADCP was observed when combining Dara with pharmacological inhibition of CD47 interactions using a glutaminyl cyclase inhibitor. Phase II-like preclinical in vivo trials using T-ALL PDX samples in experimental minimal residual disease like (MRD-like) and overt leukemia models revealed a high anti-leukemic efficacy of CD47 blockade alone. However, T-ALL xenograft mice subjected to chemotherapy first (post-chemo MRD) and subsequently co-treated with Dara and Hu5F9-IgG2σ displayed significantly reduced bone marrow infiltration as compared to single treatments. In relapsed and highly refractory T-ALL PDX combined treatment with Dara and Hu5F9-IgG2σ was required to substantially prolong survival as compared to single treatments. These findings suggest that combining CD47 blockade with Dara is a promising therapy for T-ALL, especially for relapsed/refractory disease harbouring a dismal prognosis in patients.
Collapse
|
99
|
Qu T, Li B, Wang Y. Targeting CD47/SIRPα as a therapeutic strategy, where we are and where we are headed. Biomark Res 2022; 10:20. [PMID: 35418166 PMCID: PMC9009010 DOI: 10.1186/s40364-022-00373-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/31/2022] [Indexed: 02/08/2023] Open
Abstract
Immunotherapy using PD-1 and CTLA4 inhibitors to stimulate T cell immunity has achieved significant clinical success. However, only a portion of patients benefit from T cell-based immunotherapy. Macrophages, the most abundant type of innate immune cells in the body, play an important role in eliminating tumor cells and infectious microbes. The phagocytic check point protein CD47 inhibits the phagocytic activity of macrophages through binding to SIRPα expressed on macrophages. Blockade of the interaction between CD47 and SIRPα could restore phagocytic activity and eliminate tumor cells in vitro and in vivo. In this manuscript, we review the mechanism of action and development status of agents (antibodies targeting CD47 and SIRPα, SIRPα-Fc fusion proteins, and bi-specific antibodies) that block CD47/SIRPα interaction in preclinical studies and in the clinical setting. In addition, small molecules, mRNA, and CAR-T/M that target the CD47/SIRPα axis are also reviewed in this article.
Collapse
Affiliation(s)
- Tailong Qu
- College of life Science and Technology, Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong 510632 People’s Republic of China
- Department of Antibody Discovery, Akeso Biopharma, No.6 of Shennong Road, Torch Development District, Zhongshan, 528437 People’s Republic of China
| | - Baiyong Li
- Department of Antibody Discovery, Akeso Biopharma, No.6 of Shennong Road, Torch Development District, Zhongshan, 528437 People’s Republic of China
| | - Yifei Wang
- College of life Science and Technology, Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong 510632 People’s Republic of China
| |
Collapse
|
100
|
Abstract
Nonresolving inflammation contributes to many diseases, including COVID-19 in its fatal and long forms. Our understanding of inflammation is rapidly evolving. Like the immune system of which it is a part, inflammation can now be seen as an interactive component of a homeostatic network with the endocrine and nervous systems. This review samples emerging insights regarding inflammatory memory, inflammatory aging, inflammatory cell death, inflammatory DNA, inflammation-regulating cells and metabolites, approaches to resolving or modulating inflammation, and inflammatory inequity.
Collapse
Affiliation(s)
- Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|