1
|
Li Z, Han B, Qi M, Li Y, Duan Y, Yao Y. Modulating macrophage-mediated programmed cell removal: An attractive strategy for cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189172. [PMID: 39151808 DOI: 10.1016/j.bbcan.2024.189172] [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: 01/11/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Macrophage-mediated programmed cell removal (PrCR) is crucial for the identification and elimination of needless cells that maintain tissue homeostasis. The efficacy of PrCR depends on the balance between pro-phagocytic "eat me" signals and anti-phagocytic "don't eat me" signals. Recently, a growing number of studies have shown that tumourigenesis and progression are closely associated with PrCR. In the tumour microenvironment, PrCR activated by the "eat me" signal is counterbalanced by the "don't eat me" signal of CD47/SIRPα, resulting in tumour immune escape. Therefore, targeting exciting "eat me" signalling while simultaneously suppressing "don't eat me" signalling and eventually inducing macrophages to produce effective PrCR will be a very attractive antitumour strategy. Here, we comprehensively review the functions of PrCR-activating signal molecules (CRT, PS, Annexin1, SLAMF7) and PrCR-inhibiting signal molecules (CD47/SIRPα, MHC-I/LILRB1, CD24/Siglec-10, SLAMF3, SLAMF4, PD-1/PD-L1, CD31, GD2, VCAM1), the interactions between these molecules, and Warburg effect. In addition, we highlight the molecular regulatory mechanisms that affect immune system function by exciting or suppressing PrCR. Finally, we review the research advances in tumour therapy by activating PrCR and discuss the challenges and potential solutions to smooth the way for tumour treatment strategies that target PrCR.
Collapse
Affiliation(s)
- Zhenzhen Li
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Bingqian Han
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Menghui Qi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yinchao Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongtao Duan
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China.
| | - Yongfang Yao
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Xing L, Wang Z, Feng Y, Luo H, Dai G, Sang L, Zhang C, Qian J. The biological roles of CD47 in ovarian cancer progression. Cancer Immunol Immunother 2024; 73:145. [PMID: 38832992 PMCID: PMC11150368 DOI: 10.1007/s00262-024-03708-3] [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: 03/18/2024] [Accepted: 04/19/2024] [Indexed: 06/06/2024]
Abstract
Ovarian cancer is one of the most lethal malignant tumors, characterized by high incidence and poor prognosis. Patients relapse occurred in 65-80% after initial treatment. To date, no effective treatment has been established for these patients. Recently, CD47 has been considered as a promising immunotherapy target. In this paper, we reviewed the biological roles of CD47 in ovarian cancer and summarized the related mechanisms. For most types of cancers, the CD47/Sirpα immune checkpoint has attracted the most attention in immunotherapy. Notably, CD47 monoclonal antibodies and related molecules are promising in the immunotherapy of ovarian cancer, and further research is needed. In the future, new immunotherapy regimens targeting CD47 can be applied to the clinical treatment of ovarian cancer patients.
Collapse
Affiliation(s)
- Linan Xing
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Zhao Wang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Yue Feng
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Haixia Luo
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Guijiang Dai
- Department of Comprehensive Office, The Second Affiliated Hospital of MuDanjiang Medical University, Mudanjiang, 157009, People's Republic of China
| | - Lin Sang
- Department of Obstetrics and Gynecology, People's Hospital of Anji, Huzhou, 310022, People's Republic of China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Jianhua Qian
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
3
|
Chen L, Zhao X, Liu X, Ouyang Y, Xu C, Shi Y. Development of small molecule drugs targeting immune checkpoints. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0034. [PMID: 38727005 PMCID: PMC11131045 DOI: 10.20892/j.issn.2095-3941.2024.0034] [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: 01/17/2024] [Accepted: 03/28/2024] [Indexed: 05/29/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are used to relieve and refuel anti-tumor immunity by blocking the interaction, transcription, and translation of co-inhibitory immune checkpoints or degrading co-inhibitory immune checkpoints. Thousands of small molecule drugs or biological materials, especially antibody-based ICIs, are actively being studied and antibodies are currently widely used. Limitations, such as anti-tumor efficacy, poor membrane permeability, and unneglected tolerance issues of antibody-based ICIs, remain evident but are thought to be overcome by small molecule drugs. Recent structural studies have broadened the scope of candidate immune checkpoint molecules, as well as innovative chemical inhibitors. By way of comparison, small molecule drug-based ICIs represent superior oral bioavailability and favorable pharmacokinetic features. Several ongoing clinical trials are exploring the synergetic effect of ICIs and other therapeutic strategies based on multiple ICI functions, including immune regulation, anti-angiogenesis, and cell cycle regulation. In this review we summarized the current progression of small molecule ICIs and the mechanism underlying immune checkpoint proteins, which will lay the foundation for further exploration.
Collapse
Affiliation(s)
- Luoyi Chen
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xinchen Zhao
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xiaowei Liu
- Institute for Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujie Ouyang
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chuan Xu
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Ying Shi
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| |
Collapse
|
4
|
Yang HW, Ju SP, Hsieh YT, Yang YC. Design single-stranded DNA aptamer of cluster of differentiation 47 protein by stochastic tunnelling-basin hopping-discrete molecular dynamics method. J Biomol Struct Dyn 2024; 42:3969-3982. [PMID: 37261868 DOI: 10.1080/07391102.2023.2217511] [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: 01/27/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023]
Abstract
The formation of the Cluster of Differentiation 47 (CD47, PDB code: 2JJT)/signal regulatory protein α (SIRPα) complex is very important as it protects healthy cells from immune clearance while promoting macrophage phagocytosis for tumour elimination. Although several antibodies have been developed for cancer therapy, new function-blocking aptamers are still under development. This study aims to design the aptamer AptCD47, which can block the formation of the CD47/SIRPα complex. This study employs the MARTINI coarse-grained (CG) force field and the stochastic tunnelling-basin hopping-discrete molecular dynamics (STUN-BH-DMD) method to identify the most stable AptCD47/CD47 complexes. Coarse-grained molecular dynamics (CGMD) simulations were used to obtain root-mean-square deviation (RMSD) and root-mean-square fluctuation (RMSF) analyses. The results demonstrate that the formation of AptCD47/CD47 complexes renders the CD47 structure more stable than the single CD47 molecule in a water environment. The minimum energy pathway (MEP) obtained by the nudged elastic band (NEB) method indicates that the binding processes of 5'-ATTCAATTCC-3' and 5'-AGTGCAATCT-3' to CD47 are barrierless, which is much lower than the binding barrier of SIRPα to CD47 of about 14.23 kcal/mol. Therefore, these two AptCD47/CD47 complexes can create a high spatial binding barrier for SIRPα, preventing the formation of a stable CD47/SIRPα complex. The proposed numerical process with the MARTINI CG force field can be used to design CD47 aptamers that efficiently block SIRPα from binding to CD47.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hung-Wei Yang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan City, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan City, Taiwan
| | - Shin-Pon Ju
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yun-Te Hsieh
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Yung-Cheng Yang
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Malinge P, Chauchet X, Bourguignon J, Bosson N, Calloud S, Bautzova T, Borlet M, Laursen M, Kelpsas V, Rose N, Gueneau F, Ravn U, Magistrelli G, Fischer N. Structural analysis of light chain-driven bispecific antibodies targeting CD47 and PD-L1. MAbs 2024; 16:2362432. [PMID: 38849989 PMCID: PMC11164222 DOI: 10.1080/19420862.2024.2362432] [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: 02/16/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
In contrast to natural antibodies that rely mainly on the heavy chain to establish contacts with their cognate antigen, we have developed a bispecific antibody format in which the light chain (LC) drives antigen binding and specificity. To better understand epitope-paratope interactions in this context, we determined the X-ray crystallographic structures of an antigen binding fragment (Fab) in complex with human CD47 and another Fab in complex with human PD-L1. These Fabs contain a κ-LC and a λ-LC, respectively, which are paired with an identical heavy chain (HC). The structural analysis of these complexes revealed the dominant contribution of the LCs to antigen binding, but also that the common HC provides some contacts in both CD47 and PD-L1 Fab complexes. The anti-CD47 Fab was affinity optimized by diversifying complementary-determining regions of the LC followed by phage display selections. Using homology modeling, the contributions of the amino acid modification to the affinity increase were analyzed. Our results demonstrate that, despite a less prominent role in natural antibodies, the LC can mediate high affinity binding to different antigens and neutralize their biological function. Importantly, Fabs containing a common variable heavy (VH) domain enable the generation of bispecific antibodies retaining a truly native structure, maximizing their therapeutic potential.
Collapse
Affiliation(s)
- Pauline Malinge
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | - Xavier Chauchet
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | | | - Nicolas Bosson
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | | | - Tereza Bautzova
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | - Marie Borlet
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | | | | | | | - Franck Gueneau
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | - Ulla Ravn
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | | | - Nicolas Fischer
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| |
Collapse
|
6
|
Dewdney B, Jenkins MR, Best SA, Freytag S, Prasad K, Holst J, Endersby R, Johns TG. From signalling pathways to targeted therapies: unravelling glioblastoma's secrets and harnessing two decades of progress. Signal Transduct Target Ther 2023; 8:400. [PMID: 37857607 PMCID: PMC10587102 DOI: 10.1038/s41392-023-01637-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/29/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023] Open
Abstract
Glioblastoma, a rare, and highly lethal form of brain cancer, poses significant challenges in terms of therapeutic resistance, and poor survival rates for both adult and paediatric patients alike. Despite advancements in brain cancer research driven by a technological revolution, translating our understanding of glioblastoma pathogenesis into improved clinical outcomes remains a critical unmet need. This review emphasises the intricate role of receptor tyrosine kinase signalling pathways, epigenetic mechanisms, and metabolic functions in glioblastoma tumourigenesis and therapeutic resistance. We also discuss the extensive efforts over the past two decades that have explored targeted therapies against these pathways. Emerging therapeutic approaches, such as antibody-toxin conjugates or CAR T cell therapies, offer potential by specifically targeting proteins on the glioblastoma cell surface. Combination strategies incorporating protein-targeted therapy and immune-based therapies demonstrate great promise for future clinical research. Moreover, gaining insights into the role of cell-of-origin in glioblastoma treatment response holds the potential to advance precision medicine approaches. Addressing these challenges is crucial to improving outcomes for glioblastoma patients and moving towards more effective precision therapies.
Collapse
Affiliation(s)
- Brittany Dewdney
- Cancer Centre, Telethon Kids Institute, Nedlands, WA, 6009, Australia.
- Centre For Child Health Research, University of Western Australia, Perth, WA, 6009, Australia.
| | - Misty R Jenkins
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
| | - Sarah A Best
- Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
| | - Saskia Freytag
- Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
| | - Krishneel Prasad
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
| | - Jeff Holst
- School of Biomedical Sciences, University of New South Wales, Sydney, 2052, Australia
| | - Raelene Endersby
- Cancer Centre, Telethon Kids Institute, Nedlands, WA, 6009, Australia
- Centre For Child Health Research, University of Western Australia, Perth, WA, 6009, Australia
| | - Terrance G Johns
- Cancer Centre, Telethon Kids Institute, Nedlands, WA, 6009, Australia
- Centre For Child Health Research, University of Western Australia, Perth, WA, 6009, Australia
| |
Collapse
|
7
|
Laddha K, Sobhia ME. Breaking the 'don't eat me' signal: in silico design of CD47-directed peptides for cancer immunotherapy. Mol Divers 2023:10.1007/s11030-023-10732-5. [PMID: 37759140 DOI: 10.1007/s11030-023-10732-5] [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: 07/07/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
The leading cause of death worldwide is cancer. Although there are various therapies available to treat cancer, finding a successful one can be like searching for a needle in a haystack. Immunotherapy appears to be one of those needles in the haystack of cancer treatment. Immunotherapeutic agents enhance the immune response of the patient's body to tumor cells. One of the immunotherapeutic targets, Cluster of Differentiation 47 (CD47), releases the "don't eat me" signal when it binds to its receptor, Signal Regulatory Protein (SIRPα). Tumor cells use this signal to circumvent the immune system, rendering it ineffective. To stop tumor cells from releasing the "don't eat me" signal, the CD47-SIRPα interaction is specifically targeted in this study. To do so, in silico peptides were designed based on the structural analysis of the interaction between two proteins using point mutations on the interacting residues with the other amino acids. The peptide library was designed and docked on SIRPα using computational tools. Later on, after analyzing the docked complex, the best of them was selected for MD simulation studies of 100 ns. Further analysis after MD studies was carried out to determine the possible potential anti-SIRPα peptides.
Collapse
Affiliation(s)
- Kapil Laddha
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, 160062, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
8
|
Zhang C, Sheng Y, Sun X, Wang Y. New insights for gynecological cancer therapies: from molecular mechanisms and clinical evidence to future directions. Cancer Metastasis Rev 2023; 42:891-925. [PMID: 37368179 PMCID: PMC10584725 DOI: 10.1007/s10555-023-10113-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 05/22/2023] [Indexed: 06/28/2023]
Abstract
Advanced and recurrent gynecological cancers lack effective treatment and have poor prognosis. Besides, there is urgent need for conservative treatment for fertility protection of young patients. Therefore, continued efforts are needed to further define underlying therapeutic targets and explore novel targeted strategies. Considerable advancements have been made with new insights into molecular mechanisms on cancer progression and breakthroughs in novel treatment strategies. Herein, we review the research that holds unique novelty and potential translational power to alter the current landscape of gynecological cancers and improve effective treatments. We outline the advent of promising therapies with their targeted biomolecules, including hormone receptor-targeted agents, inhibitors targeting epigenetic regulators, antiangiogenic agents, inhibitors of abnormal signaling pathways, poly (ADP-ribose) polymerase (PARP) inhibitors, agents targeting immune-suppressive regulators, and repurposed existing drugs. We particularly highlight clinical evidence and trace the ongoing clinical trials to investigate the translational value. Taken together, we conduct a thorough review on emerging agents for gynecological cancer treatment and further discuss their potential challenges and future opportunities.
Collapse
Affiliation(s)
- Chunxue Zhang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 People’s Republic of China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yaru Sheng
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 People’s Republic of China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiao Sun
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 People’s Republic of China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yudong Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030 People’s Republic of China
- Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| |
Collapse
|
9
|
Deng H, Wang G, Zhao S, Tao Y, Zhang Z, Yang J, Lei Y. New hope for tumor immunotherapy: the macrophage-related "do not eat me" signaling pathway. Front Pharmacol 2023; 14:1228962. [PMID: 37484024 PMCID: PMC10358856 DOI: 10.3389/fphar.2023.1228962] [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: 05/25/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
The "do not eat me" signaling pathway is extremely active in tumor cells, providing a means for these cells to elude macrophage phagocytosis and escape immune surveillance. Representative markers of this pathway, such as CD47 and CD24, are highly expressed in numerous tumors. The interaction of SIRPα with CD47 reduces the accumulation of non-myosin ⅡA on the cell membrane. The combination of CD24 and Siglec10 ultimately leads to the recruitment of SHP-1 or SHP-2 to reduce signal transduction. Both of them weaken the ability of macrophages to engulf tumor cells. Blocking the mutual recognition between CD47-SIRPα or CD24-Siglec10 using large molecular proteins or small molecular drugs represents a promising avenue for tumor immunotherapy. Doing so can inhibit signal transduction and enhance macrophage clearance rates of cancer cells. In this paper, we summarize the characteristics of the drugs that affect the "do not eat me" signaling pathway via classical large molecular proteins and small molecule drugs, which target the CD47-SIRPα and CD24-Siglec10 signaling pathways, which target the CD47-SIRPα and CD24-Siglec10 signaling pathways. We expect it will offer insight into the development of new drugs centered on blocking the "do not eat me" signaling pathway.
Collapse
Affiliation(s)
- Han Deng
- General Practice Ward/International Medical Center, General Practice Medical Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Guan Wang
- General Practice Ward/International Medical Center, General Practice Medical Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Shengyan Zhao
- General Practice Ward/International Medical Center, General Practice Medical Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Yiran Tao
- West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhixiong Zhang
- General Practice Ward/International Medical Center, General Practice Medical Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Jinliang Yang
- General Practice Ward/International Medical Center, General Practice Medical Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Yi Lei
- General Practice Ward/International Medical Center, General Practice Medical Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Zhang L, Tian Y, Li M, Wang M, Wu S, Jiang Z, Wang Q, Wang W. Peptide nano 'bead-grafting' for SDT-facilitated immune checkpoints blocking. Chem Sci 2022; 13:14052-14062. [PMID: 36540822 PMCID: PMC9728588 DOI: 10.1039/d2sc02728c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 11/13/2022] [Indexed: 09/21/2023] Open
Abstract
Combination therapies based on immune checkpoint blockade (ICB) are currently the mainstay of cancer treatment, in which the synergetic delivery of multiple drugs is the essential step. Although nanoparticle drugs (NPDs) show satisfactory anticancer effects, the promotion of active co-delivery of NPDs is premature, since the processes are usually difficult to predict and control. Targeting peptide self-assemblies have been widely used as carriers for small-molecular drugs, but remain elusive for NPDs. We describe here peptide-based nano 'bead-grafting' for the active delivery of quantum-dot NPDs through a co-assembly method. Based on a 'de novo' design, we used a 'one-bead-one-compound (OBOC)' combinatorial chemical screening method to select a peptide RT with high affinity for the immune checkpoint CD47, which could also form biocompatible nanofibers and efficiently trap Ag2S quantum dots along the self-assembly path. This system can combine ICB therapy and sonodynamic therapy (SDT) to effectively inhibit tumor growth. Moreover, the tumor antigen produced by SDT can activate the adaptive immune system, which enhances the anti-tumor immune response of the ICB and shows efficient inhibition of both primary and distant tumors. This study provides a new strategy for the active control and delivery of NPDs and a new option for ICB therapy with immune checkpoints that are highly susceptible to systemic side effects.
Collapse
Affiliation(s)
- Limin Zhang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Institute of Engineering Medicine, Beijing Institute of Technology Beijing 100081 PR China
| | - Yuwei Tian
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Institute of Engineering Medicine, Beijing Institute of Technology Beijing 100081 PR China
| | - Mengzhen Li
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Institute of Engineering Medicine, Beijing Institute of Technology Beijing 100081 PR China
| | - Minxuan Wang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Institute of Engineering Medicine, Beijing Institute of Technology Beijing 100081 PR China
| | - Shang Wu
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Institute of Engineering Medicine, Beijing Institute of Technology Beijing 100081 PR China
| | - Zhenqi Jiang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Institute of Engineering Medicine, Beijing Institute of Technology Beijing 100081 PR China
| | - Qiqin Wang
- Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University Guangzhou 510632 China
| | - Weizhi Wang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Institute of Engineering Medicine, Beijing Institute of Technology Beijing 100081 PR China
| |
Collapse
|
11
|
Mishra AK, Ali A, Dutta S, Banday S, Malonia SK. Emerging Trends in Immunotherapy for Cancer. Diseases 2022; 10:60. [PMID: 36135216 PMCID: PMC9498256 DOI: 10.3390/diseases10030060] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Recent advances in cancer immunology have enabled the discovery of promising immunotherapies for various malignancies that have shifted the cancer treatment paradigm. The innovative research and clinical advancements of immunotherapy approaches have prolonged the survival of patients with relapsed or refractory metastatic cancers. Since the U.S. FDA approved the first immune checkpoint inhibitor in 2011, the field of cancer immunotherapy has grown exponentially. Multiple therapeutic approaches or agents to manipulate different aspects of the immune system are currently in development. These include cancer vaccines, adoptive cell therapies (such as CAR-T or NK cell therapy), monoclonal antibodies, cytokine therapies, oncolytic viruses, and inhibitors targeting immune checkpoints that have demonstrated promising clinical efficacy. Multiple immunotherapeutic approaches have been approved for specific cancer treatments, while others are currently in preclinical and clinical trial stages. Given the success of immunotherapy, there has been a tremendous thrust to improve the clinical efficacy of various agents and strategies implemented so far. Here, we present a comprehensive overview of the development and clinical implementation of various immunotherapy approaches currently being used to treat cancer. We also highlight the latest developments, emerging trends, limitations, and future promises of cancer immunotherapy.
Collapse
Affiliation(s)
- Alok K. Mishra
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Amjad Ali
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Shubham Dutta
- MassBiologics, UMass Chan Medical School, Boston, MA 02126, USA
| | - Shahid Banday
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Sunil K. Malonia
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
12
|
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
|