1
|
Bess SN, Igoe MJ, Muldoon TJ. The Physiological and Therapeutic Role of CD47 in Macrophage Function and Cancer. Immunol Invest 2024:1-35. [PMID: 39415597 DOI: 10.1080/08820139.2024.2415409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
BACKGROUND Immunotherapy is an emerging strategy in cancer therapeutics aimed at modulating the immune system to inhibit pro-tumor pathways and increase a tumor's sensitivity to chemotherapy. Several clinically approved immunotherapy treatments, such as monoclonal antibody treatments, have been successful in solid tumors such as breast, colorectal, and pancreatic. However, an outstanding challenge of these strategies is tumor cell resistance. One target of interest for immune cell modulation is targeting macrophages that enter the tumor microenvironment. More specifically, an immune checkpoint of interest is CD47. CD47 is a transmembrane protein that inhibits phagocytic activity by acting as a "don't eat me" signal. In both mice and humans, healthy cells can express CD47, while solid malignancies like colorectal and breast cancer express it most strongly. METHODS Analysis of literature data on the physiological and functional roles of tissue-resident macrophages, along with the structure and mechanisms of action of the CD47 pathway was explored. We also explored how CD47 can influence different aspects of the tumor microenvironment (i.e. cellular metabolism and hypoxia) in addition to current clinical strategies and challenges associated with targeting CD47. RESULTS Overall, it was discovered that CD47 is overexpressed in a variety of cancer types in addition to normal tissue, making it a promising treatment regimen to enhance the capability of macrophages to phagocytose tumor cells. However, treatment efficacy is varied in pre-clinical and clinical models due to various challenges such as off-target effects. CONCLUSION This review emphasizes the diverse functionality of macrophages in normal and cancerous tissue, while also emphasizing the importance of macrophage targeting and their clinical significance.
Collapse
Affiliation(s)
- Shelby N Bess
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Matthew J Igoe
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Timothy J Muldoon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
2
|
Patel Y, Manturthi S, Tiwari S, Gahunia E, Courtemanche A, Gandelman M, Côté M, Gadde S. Development of Pro-resolving and Pro-efferocytic Nanoparticles for Atherosclerosis Therapy. ACS Pharmacol Transl Sci 2024; 7:3086-3095. [PMID: 39416959 PMCID: PMC11475319 DOI: 10.1021/acsptsci.4c00292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 10/19/2024]
Abstract
Atherosclerosis is a major contributor to cardiovascular diseases with a high global prevalence. It is characterized by the formation of lipid-laden plaques in the arteries, which eventually lead to plaque rupture and thrombosis. While the current lipid-lowering therapies are generally effective in lowering the risk of cardiovascular events, they do not address the underlying causes of disease. Defective resolution of inflammation and impaired efferocytosis are the main driving forces of atherosclerosis. Macrophages recognize cells for clearance by the expression of "eat me" and "do not eat me" signals, including the CD47-SIRPα axis. However, the "do not eat me" signal CD47 is overexpressed in atherosclerotic plaques, leading to compromised efferocytosis and secondary necrosis. In this context, prophagocytic antibodies have been explored to stimulate the clearance of apoptotic cells, but they are nonspecific and impact healthy tissues. In macrophages, downstream of signal regulatory protein α, lie protein tyrosine phosphatases, SHP 1/2, which can serve as effective targets for selectively phagocytosing apoptotic cells. While increasing the efferocytosis targets the end stages of lesion development, the underlying issue of inflammation still persists. Simultaneously increasing efferocytosis and reducing inflammation can be effective therapeutic strategies for managing atherosclerosis. For instance, IL-10 is a key anti-inflammatory mediator that enhances efferocytosis via phosphoSTAT3 (pSTAT3) activation. In this study, we developed a combination nanotherapy by encapsulating an SHP-1 inhibitor (NSC 87877) and IL-10 in a single nanoparticle platform [(S + IL)-NPs] to enhance efferocytosis and inflammation resolution. Our studies suggest that (S + IL)-NPs successfully encapsulated both agents, entered the macrophages, and delivered the agents into intracellular compartments. Additionally, (S + IL)-NPs decreased inflammation by suppressing pro-inflammatory markers and enhancing anti-inflammatory mediators. They also exhibited the potential for improved phagocytic activity via pSTAT3 activation. Our nanomedicine-mediated upregulation of the anti-inflammatory and efferocytic responses in macrophages shows promise for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yukta Patel
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Shireesha Manturthi
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Saras Tiwari
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Esha Gahunia
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Amandine Courtemanche
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Michelle Gandelman
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Marceline Côté
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Suresh Gadde
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa−Carleton
Institute for Biomedical Engineering (OCIBME), Ottawa K1N 6N5, Canada
| |
Collapse
|
3
|
Tan Z, Zhang Q, Wang Y, Wang Y, Zhang S, Xing X, Liu H, Shen Z, Sang W. Clinicopathological analysis of immunohistochemical CD47 and signal-regulatory protein-α expression in Extranodal Natural killer/T-cell lymphoma. Ann Hematol 2024; 103:3033-3042. [PMID: 38886192 DOI: 10.1007/s00277-024-05852-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The interaction between CD47 and signal-regulatory protein-alpha (SIRPα) inhibits phagocytosis, and their clinicopathological characteristics have been evaluated in various diseases. However, the significance of CD47 and SIRPα expression, as well as the combined effect, in Extranodal Natural killer/T-cell Lymphoma (ENKTL) remains uncertain. METHODS In total, 76 newly diagnosed ENKTL patients (mean age 49.9 years, 73.7% male) were included in this study. CD47 and SIRPα expression were examined by immunohistochemistry. Survival analyses were conducted through Kaplan-Meier curves and the Cox regression model. RESULTS Seventy-one (93.4%) cases were categorized as the CD47 positive group and 59 (77.6%) cases were categorized as the SIRPα positive group. CD47-negative cases had more advanced-stage illness (P = 0.001), while SIRPα-positive cases showed significantly lower levels of high-density lipoprotein (P < 0.001). In univariable analysis, CD47, SIRPα expression, and their combination were significantly associated with prognosis (P < 0.05). In multivariable analysis, only positive SIRPα expression remained significantly associated with superior overall survival (Hazard ratio [HR] 0.446; 95% confidence interval [CI] 0.207-0.963; P = 0.004). Furthermore, SIRPα expression could re-stratify the survival of patients in ECOG (< 2), advanced CA stage, PINK (HR), CD38-positive, PD1-positive, and CD30-positive groups. CONCLUSIONS SIRPα status was a potential independent prognostic factor for ENKTL. The prognostic significance of CD47 expression and the interaction between CD47 and SIRPα in ENKTL need further investigation.
Collapse
Affiliation(s)
- Zaixiang Tan
- Research Center of Health Policy and Health Management, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Qing Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Ying Wang
- Research Center of Health Policy and Health Management, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Department of Personnel, Suqian First Hospital, Suqian, Jiangsu, 223800, China
| | - Yubo Wang
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Shuo Zhang
- Department of Hematology, Linyi People's Hospital, Linyi, Shandong, 276002, China
| | - Xing Xing
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Hui Liu
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Ziyuan Shen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Wei Sang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, 221006, China.
| |
Collapse
|
4
|
Gong Y, Gao W, Zhang J, Dong X, Zhu D, Ma G. Engineering nanoparticles-enabled tumor-associated macrophages repolarization and phagocytosis restoration for enhanced cancer immunotherapy. J Nanobiotechnology 2024; 22:341. [PMID: 38890636 PMCID: PMC11184870 DOI: 10.1186/s12951-024-02622-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are pivotal within the immunosuppressive tumor microenvironment (TME), and recently, have attracted intensive attention for cancer treatment. However, concurrently to promote TAMs repolarization and phagocytosis of cancer cells remains challenging. Here, a TAMs-targeted albumin nanoparticles-based delivery system (M@SINPs) was constructed for the co-delivery of photosensitizer IR820 and SHP2 inhibitor SHP099 to potentiate macrophage-mediated cancer immunotherapy. M@SINPs under laser irradiation can generate the intracellular reactive oxygen species (ROS) and facilitate M2-TAMs to an M1 phenotype. Meanwhile, inhibition of SHP2 could block the CD47-SIRPa pathway to restore M1 macrophage phagocytic activity. M@SINPs-mediated TAMs remodeling resulted in the immunostimulatory TME by repolarizing TAMs to an M1 phenotype, restoring its phagocytic function and facilitating intratumoral CTLs infiltration, which significantly inhibited tumor growth. Furthermore, M@SINPs in combination with anti-PD-1 antibody could also improve the treatment outcomes of PD-1 blockade and exert the synergistic anticancer effects. Thus, the macrophage repolarization/phagocytosis restoration combination through M@SINPs holds promise as a strategy to concurrently remodel TAMs in TME for improving the antitumor efficiency of immune checkpoint block and conventional therapy.
Collapse
Affiliation(s)
- Yonghua Gong
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Wenyue Gao
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Jinyang Zhang
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Xia Dong
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China.
| | - Dunwan Zhu
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China.
| | - Guilei Ma
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China.
| |
Collapse
|
5
|
Wang J, Zhu N, Su X, Gao Y, Yang R. Novel tumor-associated macrophage populations and subpopulations by single cell RNA sequencing. Front Immunol 2024; 14:1264774. [PMID: 38347955 PMCID: PMC10859433 DOI: 10.3389/fimmu.2023.1264774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/30/2023] [Indexed: 02/15/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are present in almost all solid tumor tissues. 16They play critical roles in immune regulation, tumor angiogenesis, tumor stem cell activation, tumor invasion and metastasis, and resistance to therapy. However, it is unclear how TAMs perform these functions. With the application of single-cell RNA sequencing (scRNA-seq), it has become possible to identify TAM subpopulations associated with distinct functions. In this review, we discuss four novel TAM subpopulations in distinct solid tumors based on core gene signatures by scRNA-seq, including FCN1 +, SPP1 +, C1Q + and CCL18 + TAMs. Functional enrichment and gene expression in scRNA-seq data from different solid tumor tissues found that FCN1 + TAMs may induce inflammation; SPP1 + TAMs are potentially involved in metastasis, angiogenesis, and cancer cell stem cell activation, whereas C1Q + TAMs participate in immune regulation and suppression; And CCL18 + cells are terminal immunosuppressive macrophages that not only have a stronger immunosuppressive function but also enhance tumor metastasis. SPP1 + and C1Q + TAM subpopulations can be further divided into distinct populations with different functions. Meanwhile, we will also present emerging evidence highlighting the separating macrophage subpopulations associated with distinct functions. However, there exist the potential disconnects between cell types and subpopulations identified by scRNA-seq and their actual function.
Collapse
Affiliation(s)
- Juanjuan Wang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Ningning Zhu
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Xiaomin Su
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yunhuan Gao
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Rongcun Yang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
6
|
Moon SY, Han M, Ryu G, Shin SA, Lee JH, Lee CS. Emerging Immune Checkpoint Molecules on Cancer Cells: CD24 and CD200. Int J Mol Sci 2023; 24:15072. [PMID: 37894750 PMCID: PMC10606340 DOI: 10.3390/ijms242015072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer immunotherapy strategies are based on the utilization of immune checkpoint inhibitors to instigate an antitumor immune response. The efficacy of immune checkpoint blockade, directed at adaptive immune checkpoints, has been demonstrated in select cancer types. However, only a limited subset of patients has exhibited definitive outcomes characterized by a sustained response after discontinuation of therapy. Recent investigations have highlighted the significance of immune checkpoint molecules that are overexpressed in cancer cells and inhibit myeloid lineage immune cells within a tumor microenvironment. These checkpoints are identified as potential targets for anticancer immune responses. Notably, the immune checkpoint molecules CD24 and CD200 have garnered attention owing to their involvement in tumor immune evasion. CD24 and CD200 are overexpressed across diverse cancer types and serve as signaling checkpoints by engaging their respective receptors, Siglec-10 and CD200 receptor, which are expressed on tumor-associated myeloid cells. In this review, we summarized and discussed the latest advancements and insights into CD24 and CD200 as emergent immune checkpoint moieties, further delving into their therapeutic potentials for cancer treatment.
Collapse
Affiliation(s)
- Sun Young Moon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Minjoo Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Gyoungah Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Seong-Ah Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Jun Hyuck Lee
- Research Unit of Cryogenic Novel Material, Korea Polar Research Institute, Incheon 21990, Republic of Korea;
- Department of Polar Sciences, University of Science and Technology, Incheon 21990, Republic of Korea
| | - Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| |
Collapse
|
7
|
Wang Y, Barrett A, Hu Q. Targeting Macrophages for Tumor Therapy. AAPS J 2023; 25:80. [PMID: 37589825 DOI: 10.1208/s12248-023-00845-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
Macrophages, as one of the most abundant tumor-infiltrating cells, play an important role in tumor development and metastasis. The frequency and polarization of tumor-associated macrophages (TAMs) correlate with disease progression, tumor metastasis, and resistance to various treatments. Pro-inflammatory M1 macrophages hold the potential to engulf tumor cells. In contrast, anti-inflammatory M2 macrophages, which are predominantly present in tumors, potentiate tumor progression and immune escape. Targeting macrophages to modulate the tumor immune microenvironment can ameliorate the tumor-associated immunosuppression and elicit an anti-tumor immune response. Strategies to repolarize TAMs, deplete TAMs, and block inhibitory signaling hold great potential in tumor therapy. Besides, biomimetic carriers based on macrophages have been extensively explored to prolong circulation, enhance tumor-targeted delivery, and reduce the immunogenicity of therapeutics to augment therapeutic efficacy. Moreover, the genetic engineering of macrophages with chimeric antigen receptor (CAR) allows them to recognize tumor antigens and perform tumor cell-specific phagocytosis. These strategies will expand the toolkit for treating tumors, especially for solid tumors, drug-resistant tumors, and metastatic tumors. Herein, we introduce the role of macrophages in tumor progression, summarize the recent advances in macrophage-centered anticancer therapy, and discuss their challenges as well as future applications. Graphical abstract.
Collapse
Affiliation(s)
- Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A
| | - Allie Barrett
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A..
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A..
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A..
| |
Collapse
|
8
|
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
|
9
|
Tang Z, Wang X, Tang M, Wu J, Zhang J, Liu X, Gao F, Fu Y, Tang P, Li C. Overcoming the On-Target Toxicity in Antibody-Mediated Therapies via an Indirect Active Targeting Strategy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206912. [PMID: 36683161 PMCID: PMC10037698 DOI: 10.1002/advs.202206912] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Antibody-based therapies could be led astray when target receptors are expressed on nontarget sites, and the on-target toxicity poses critical challenges to clinical applications. Here, a biomimetic indirect active targeting (INTACT) strategy is proposed based on receptor expression disparities between nontarget sites and the targets. By prebinding the antibodies using cell membrane vesicles with appropriate receptor expressions, the INTACT strategy could filter out the interactions on nontarget sites due to their inferior receptor expression, whereas ensure on-demand release at the targets by competitive binding. The strategy is verified on CD47 antibody, realizing drastic alleviation of its clinically concerned hematotoxicity on a series of animal models including humanized patient-derived xenograft platforms, accompanied by preferable therapeutic effects. Furthermore, the INTACT strategy proves extensive applicability for various systems including antibody, antibody-drug conjugate, and targeted delivery systems, providing a potential platform refining the specificity for frontier antibody-related therapies.
Collapse
Affiliation(s)
- Zhongjie Tang
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Xiaoyou Wang
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Mei Tang
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Jin Wu
- Department of Breast and Thyroid SurgerySouthwest HospitalChongqing400038P. R. China
| | - Jiexuan Zhang
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Xinlong Liu
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Feiyan Gao
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Yu Fu
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Peng Tang
- Department of Breast and Thyroid SurgerySouthwest HospitalChongqing400038P. R. China
| | - Chong Li
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| |
Collapse
|
10
|
Wang L, Jia Q, Chu Q, Zhu B. Targeting tumor microenvironment for non-small cell lung cancer immunotherapy. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:18-29. [PMID: 39170874 PMCID: PMC11332857 DOI: 10.1016/j.pccm.2022.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 08/23/2024]
Abstract
The tumor microenvironment (TME) is composed of different cellular and non-cellular elements. Constant interactions between tumor cells and the TME are responsible for tumor initiation, tumor progression, and responses to therapies. Immune cells in the TME can be classified into two broad categories, namely adaptive and innate immunity. Targeting these immune cells has attracted substantial research and clinical interest. Current research focuses on identifying key molecular players and developing targeted therapies. These approaches may offer more efficient ways of treating different cancers. In this review, we explore the heterogeneity of the TME in non-small cell lung cancer, summarize progress made in targeting the TME in preclinical and clinical studies, discuss the potential predictive value of the TME in immunotherapy, and highlight the promising effects of bispecific antibodies in the era of immunotherapy.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
11
|
Karizak AZ, Salmasi Z, Gheibihayat SM, Asadi M, Ghasemi Y, Tajbakhsh A, Savardashtaki A. Understanding the regulation of "Don't Eat-Me" signals by inflammatory signaling pathways in the tumor microenvironment for more effective therapy. J Cancer Res Clin Oncol 2023; 149:511-529. [PMID: 36342520 DOI: 10.1007/s00432-022-04452-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/22/2022] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Receptor/ligand pair immune checkpoints are inhibitors that regulate immunity as vital "Don't Find-Me" signals to the adaptive immune system, additionally, the essential goals of anti-cancer therapy. Moreover, the immune checkpoints are involved in treatment resistance in cancer therapy. The immune checkpoints as a signal from "self" and their expression on healthy cells prevent phagocytosis. Cells (e.g., senescent and/or apoptotic cells) with low immune checkpoints, such as low CD47 and/or PD-L1, are phagocytosed, which is necessary for tissue integrity and homeostasis maintenance. In other words, cancer cells induce increased CD47 expression in the tumor microenvironment (TME), avoiding their clearance by immune cells. PD-L1 and/or CD47 expression tumors have also been employed as biomarkers to guide cure prospects. Thus, targeting innate and adaptive immune checkpoints might improve the influence of the treatments on tumor cells. However, the CD47 regulation in the TME stands intricate, so much of this process has stayed a riddle. In this line, less attention has been paid to cytokines in TME. Cytokines are significant regulators of tumor immune surveillance, and they do this by controlling the actions of the immune cell. Recently, it has been suggested that different types of cytokines at TME might cooperate with others that contribute to the regulation of CD47 and/or PD-L1. MATERIALS AND METHODS The data were searched in available databases and a Web Search engine (PubMed, Scopus, and Google Scholar) using related keywords in the title, abstract, and keywords. CONCLUSION Given the significant role of pro/anti-inflammatory signaling in the TME, we discuss the present understanding of pro/anti-inflammatory signaling implications in "Don't Eat-Me" regulation signals, particularly CD47, in the pathophysiology of cancers and come up with innovative opinions for the clinical transformation and personalized medicine.
Collapse
Affiliation(s)
- Ashkan Zare Karizak
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Marzieh Asadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 71362 81407, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran. .,Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 71362 81407, Iran. .,Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
12
|
Liu M, Liu L, Song Y, Li W, Xu L. Targeting macrophages: a novel treatment strategy in solid tumors. J Transl Med 2022; 20:586. [PMID: 36510315 PMCID: PMC9743606 DOI: 10.1186/s12967-022-03813-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
In the tumor microenvironment (TME), tumor-associated macrophages (TAMs) are the most abundant immune cells, which act as a key regulator in tumorigenesis and progression. Increasing evidence have demonstrated that the TME alters the nature of macrophages to maintain dynamic tissue homeostasis, allowing TAMs to acquire the ability to stimulate angiogenesis, promote tumor metastasis and recurrence, and suppress anti-tumor immune responses. Furthermore, tumors with high TAM infiltration have poor prognoses and are resistant to treatment. In the field of solid tumor, the exploration of tumor-promoting mechanisms of TAMs has attracted much attention and targeting TAMs has emerged as a promising immunotherapeutic strategy. Currently, the most common therapeutic options for targeting TAMs are as follows: the deletion of TAMs, the inhibition of TAMs recruitment, the release of phagocytosis by TAMs, and the reprogramming of macrophages to remodel their anti-tumor capacity. Promisingly, the study of chimeric antigen receptor macrophages (CAR-Ms) may provide even greater benefit for patients with solid tumors. In this review, we discuss how TAMs promote the progression of solid tumors as well as summarize emerging immunotherapeutic strategies that targeting macrophages.
Collapse
Affiliation(s)
- Mengmeng Liu
- grid.414008.90000 0004 1799 4638Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008 China ,grid.207374.50000 0001 2189 3846Academy of Medical Sciences of Zhengzhou University, Zhengzhou, 450052 China
| | - Lina Liu
- grid.414008.90000 0004 1799 4638Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Yongping Song
- grid.412633.10000 0004 1799 0733Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Wei Li
- grid.412633.10000 0004 1799 0733Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Linping Xu
- grid.414008.90000 0004 1799 4638Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008 China
| |
Collapse
|
13
|
Dang W, Tao Y, Xu X, Zhao H, Zou L, Li Y. The role of lung macrophages in acute respiratory distress syndrome. Inflamm Res 2022; 71:1417-1432. [PMID: 36264361 PMCID: PMC9582389 DOI: 10.1007/s00011-022-01645-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/22/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute and diffuse inflammatory lung injury in a short time, one of the common severe manifestations of the respiratory system that endangers human life and health. As an innate immune cell, macrophages play a key role in the inflammatory response. For a long time, the role of pulmonary macrophages in ARDS has tended to revolve around the polarization of M1/M2. However, with the development of single-cell RNA sequencing, fate mapping, metabolomics, and other new technologies, a deeper understanding of the development process, classification, and function of macrophages in the lung are acquired. Here, we discuss the function of pulmonary macrophages in ARDS from the two dimensions of anatomical location and cell origin and describe the effects of cell metabolism and intercellular interaction on the function of macrophages. Besides, we explore the treatments for targeting macrophages, such as enhancing macrophage phagocytosis, regulating macrophage recruitment, and macrophage death. Considering the differences in responsiveness of different research groups to these treatments and the tremendous dynamic changes in the gene expression of monocyte/macrophage, we discussed the possibility of characterizing the gene expression of monocyte/macrophage as the biomarkers. We hope that this review will provide new insight into pulmonary macrophage function and therapeutic targets of ARDS.
Collapse
Affiliation(s)
- Wenpei Dang
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yiming Tao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Xinxin Xu
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Hui Zhao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Lijuan Zou
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yongsheng Li
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
14
|
Oh HH, Park YL, Park SY, Myung E, Im CM, Yu HJ, Han B, Seo YJ, Kim KH, Myung DS, Cho SB, Lee WS, Park D, Joo YE. CD47 mediates the progression of colorectal cancer by inducing tumor cell apoptosis and angiogenesis. Pathol Res Pract 2022; 240:154220. [DOI: 10.1016/j.prp.2022.154220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/07/2022] [Accepted: 11/12/2022] [Indexed: 11/16/2022]
|
15
|
Landeira-Viñuela A, Arias-Hidalgo C, Juanes-Velasco P, Alcoceba M, Navarro-Bailón A, Pedreira CE, Lecrevisse Q, Díaz-Muñoz L, Sánchez-Santos JM, Hernández ÁP, García-Vaquero ML, Góngora R, De Las Rivas J, González M, Orfao A, Fuentes M. Unravelling soluble immune checkpoints in chronic lymphocytic leukemia: Physiological immunomodulators or immune dysfunction. Front Immunol 2022; 13:965905. [PMID: 36248816 PMCID: PMC9554405 DOI: 10.3389/fimmu.2022.965905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a lymphoid neoplasm characterized by the accumulation of mature B cells. The diagnosis is established by the detection of monoclonal B lymphocytes in peripheral blood, even in early stages [monoclonal B-cell lymphocytosis (MBLhi)], and its clinical course is highly heterogeneous. In fact, there are well-characterized multiple prognostic factors that are also related to the observed genetic heterogenicity, such as immunoglobulin heavy chain variable region (IGHV) mutational status, del17p, and TP53 mutations, among others. Moreover, a dysregulation of the immune system (innate and adaptive immunity) has been observed in CLL patients, with strong impact on immune surveillance and consequently on the onset, evolution, and therapy response. In addition, the tumor microenvironment is highly complex and heterogeneous (i.e., matrix, fibroblast, endothelial cells, and immune cells), playing a critical role in the evolution of CLL. In this study, a quantitative profile of 103 proteins (cytokines, chemokines, growth/regulatory factors, immune checkpoints, and soluble receptors) in 67 serum samples (57 CLL and 10 MBLhi) has been systematically evaluated. Also, differential profiles of soluble immune factors that discriminate between MBLhi and CLL (sCD47, sCD27, sTIMD-4, sIL-2R, and sULBP-1), disease progression (sCD48, sCD27, sArginase-1, sLAG-3, IL-4, and sIL-2R), or among profiles correlated with other prognostic factors, such as IGHV mutational status (CXCL11/I-TAC, CXCL10/IP-10, sHEVM, and sLAG-3), were deciphered. These results pave the way to explore the role of soluble immune checkpoints as a promising source of biomarkers in CLL, to provide novel insights into the immune suppression process and/or dysfunction, mostly on T cells, in combination with cellular balance disruption and microenvironment polarization leading to tumor escape.
Collapse
Affiliation(s)
- Alicia Landeira-Viñuela
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Carlota Arias-Hidalgo
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Pablo Juanes-Velasco
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Alcoceba
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Almudena Navarro-Bailón
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Carlos Eduardo Pedreira
- Systems and Computing Department Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia-Programa de Engenharia de Sistemas e Computação (COPPE-PESC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Quentin Lecrevisse
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Laura Díaz-Muñoz
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | | - Ángela-Patricia Hernández
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Department of Pharmaceutical Sciences, Organic Chemistry Section, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| | - Marina L. García-Vaquero
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Rafael Góngora
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center Instituto Universitario de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca (CIC-IBMCC, CSIC/USAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Salamanca, Spain
| | - Marcos González
- Department of Hematology, University Hospital of Salamanca, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00233, Center Research-Centre Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC) Consejo Superior de Investigaciones Científicas - Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (CSIC-USAL, IBSAL), Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Manuel Fuentes
- Department of Medicine and General Service of Cytometry, Centro de Investigación Biomédica en Red Cáncer (CIBERONC)- CB16/12/00400, Cancer Research Centre-Instituto Universitario de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas - Universidad de Salamanca (CSIC-USAL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Proteomics Unit, Cancer Research Centre-IBMCC, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca-Consejo Superior de Investigaciones Científicas (CSIC), Salamanca, Spain
- *Correspondence: Manuel Fuentes,
| |
Collapse
|
16
|
Feng Z, Wang Y, Xu H, Guo Y, Xia W, Zhao C, Zhao X, Wu J. Recent advances in bacterial therapeutics based on sense and response. Acta Pharm Sin B 2022; 13:1014-1027. [PMID: 36970195 PMCID: PMC10031265 DOI: 10.1016/j.apsb.2022.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/26/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022] Open
Abstract
Intelligent drug delivery is a promising strategy for cancer therapies. In recent years, with the rapid development of synthetic biology, some properties of bacteria, such as gene operability, excellent tumor colonization ability, and host-independent structure, make them ideal intelligent drug carriers and have attracted extensive attention. By implanting condition-responsive elements or gene circuits into bacteria, they can synthesize or release drugs by sensing stimuli. Therefore, compared with traditional drug delivery, the usage of bacteria for drug loading has better targeting ability and controllability, and can cope with the complex delivery environment of the body to achieve the intelligent delivery of drugs. This review mainly introduces the development of bacterial-based drug delivery carriers, including mechanisms of bacterial targeting to tumor colonization, gene deletions or mutations, environment-responsive elements, and gene circuits. Meanwhile, we summarize the challenges and prospects faced by bacteria in clinical research, and hope to provide ideas for clinical translation.
Collapse
Affiliation(s)
- Zhuo Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Yuchen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Haiheng Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Yunfei Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Wen Xia
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Chenxuan Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Xiaozhi Zhao
- Department of Andrology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210093, China
- Corresponding authors. Tel.: +025 83592629.
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
- Corresponding authors. Tel.: +025 83592629.
| |
Collapse
|
17
|
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
|
18
|
Zhao H, Song S, Ma J, Yan Z, Xie H, Feng Y, Che S. CD47 as a promising therapeutic target in oncology. Front Immunol 2022; 13:757480. [PMID: 36081498 PMCID: PMC9446754 DOI: 10.3389/fimmu.2022.757480] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 07/28/2022] [Indexed: 11/30/2022] Open
Abstract
CD47 is ubiquitously expressed on the surface of cells and plays a critical role in self-recognition. By interacting with SIRPα, TSP-1 and integrins, CD47 modulates cellular phagocytosis by macrophages, determines life span of individual erythrocytes, regulates activation of immune cells, and manipulates synaptic pruning during neuronal development. As such, CD47 has recently be regarded as one of novel innate checkpoint receptor targets for cancer immunotherapy. In this review, we will discuss increasing awareness about the diverse functions of CD47 and its role in immune system homeostasis. Then, we will discuss its potential therapeutic roles against cancer and outlines, the possible future research directions of CD47- based therapeutics against cancer.
Collapse
Affiliation(s)
- Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shuangshuang Song
- Department of Nuclear Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junwei Ma
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhiyong Yan
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongwei Xie
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Feng
- Department of Emergency, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shusheng Che
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Shusheng Che,
| |
Collapse
|