1
|
Gong R, Tan JL, Liu G, Liu XF, Ma L, Shi S. Mechanism of disturbed endothelial cell function on angiogenesis following ischemic brain stroke (Review). Exp Ther Med 2025; 29:61. [PMID: 39991719 PMCID: PMC11843205 DOI: 10.3892/etm.2025.12811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/10/2025] [Indexed: 02/25/2025] Open
Abstract
The present study focused on the mechanisms of post-ischemic stroke (IS) angiogenesis from the perspective of endothelial cells (ECs) dysfunction. First, it emphasized the importance of hypoxia-inducible factor-1α in the function of ECs under hypoxic conditions, particularly in promoting angiogenesis and improving cerebral blood supply. Secondly, inflammatory cytokines and adhesion factors (for example, selectins, the immunoglobulin superfamily and integrins) influence the function and angiogenesis of ECs through various mechanisms and signaling pathways following IS. In addition, the effects of oxidative stress on ECs function and angiogenesis were explored, along with the potential of antioxidant strategies to improve EC function and promote angiogenesis. Based on these insights, the present study proposed new therapeutic strategies to ameliorate endothelial dysfunction and promote angiogenesis following IS, including small-molecule drugs targeting specific molecules, gene therapy and traditional Chinese medicine treatments. Finally, the importance of translating these laboratory findings into clinical applications was emphasized, alongside the need for advanced imaging techniques to monitor the dynamic processes of post-IS angiogenesis and evaluate the efficacy of novel therapeutic interventions. These explorations aimed at providing a more comprehensive understanding of EC function and the regulatory mechanisms of a deeper understanding of angiogenesis following IS, offering new intervention strategies for IS treatment.
Collapse
Affiliation(s)
- Rui Gong
- Department of Rehabilitation of Chinese Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Jin-Lang Tan
- Department of Rehabilitation of Chinese Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Gang Liu
- Department of Acupuncture, Moxibustion and Tuina, The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Xiao-Fang Liu
- Department of Acupuncture, Moxibustion and Tuina, The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Le Ma
- Department of Rehabilitation of Chinese Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Shuai Shi
- Department of Acupuncture, Moxibustion and Tuina, The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
2
|
You J, Ha S, Kim D, Kim HG, Kim SH, Jeong JH, Oh C, Baek NI, Jung JH, Kim JA, Lee YM. The inhibition of endothelial DLL4-NOTCH1 signaling by 2'-hydroxyflavanone enhances anti-PD-1 therapy in melanoma. Arch Pharm Res 2025; 48:351-364. [PMID: 40172769 DOI: 10.1007/s12272-025-01539-z] [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: 11/04/2024] [Accepted: 03/14/2025] [Indexed: 04/04/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapeutics; however limited patient responses necessitate combination strategies to improve therapeutic efficacy. Among potential combination partners, drugs targeting DLL4-NOTCH1 signaling pathway-a critical regulator of vascular function-show promise as angiogenesis modulators, but their clinical development have been hindered by safety concerns. To address this challenge, we adopted a novel approach by screening natural compounds with a long history of human consumption. Building upon our earlier findings, we identified three inhibitors of DLL4-NOTCH1 signaling: steppogenin, sanggenon F, and dehydrovomifoliol. Steppogenin inhibited both DLL4 and NOTCH1 activities, while sanggenon F and dehydrovomifoliol selectively suppressed DLL4 and NOTCH1 activity, respectively. We assessed their impact on key angiogenic processes, including endothelial cell migration, sprouting, and proliferation, and elucidated the relative contributions of selective DLL4 or NOTCH1 inhibition to the anti-angiogenic effect. By comparing structurally similar compounds, we identified the 2'-hydroxyflavanone moiety as a key element for DLL4 inhibition. Notably, combining steppogenin with an ICI demonstrated that a nature-derived angiogenesis inhibitor can boost the anti-cancer effect of ICI in a mouse melanoma allograft model. This comprehensive analysis of structure-activity relationships and in vivo therapeutic evaluation provides valuable insights into the development of novel anti-angiogenic compounds for combination therapy with ICIs in cancer treatment.
Collapse
Affiliation(s)
- Jihye You
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Seunghwan Ha
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Doyoung Kim
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hyoung-Geun Kim
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung Hee University, Yongin, 17104, Korea
| | - Se Ha Kim
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ji-Hak Jeong
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea
| | - Changmin Oh
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea
| | - Nam-In Baek
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung Hee University, Yongin, 17104, Korea
| | - Jong Hwa Jung
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea
| | - Jeong Ah Kim
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - You Mie Lee
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, 80 Daehak-Ro, Buk-Gu, Daegu, 41566, Republic of Korea.
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
3
|
Liu J, Zhou F, Tang Y, Li L, Li L. Progress in Lactate Metabolism and Its Regulation via Small Molecule Drugs. Molecules 2024; 29:5656. [PMID: 39683818 DOI: 10.3390/molecules29235656] [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: 10/17/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Lactate, once viewed as a byproduct of glycolysis and a metabolic "waste", is now recognized as an energy-providing substrate and a signaling molecule that modulates cellular functions under pathological conditions. The discovery of histone lactylation in 2019 marked a paradigm shift, with subsequent studies revealing that lactate can undergo lactylation with both histone and non-histone proteins, implicating it in the pathogenesis of various diseases, including cancer, liver fibrosis, sepsis, ischemic stroke, and acute kidney injury. Aberrant lactate metabolism is associated with disease onset, and its levels can predict disease outcomes. Targeting lactate production, transport, and lactylation may offer therapeutic potential for multiple diseases, yet a systematic summary of the small molecules modulating lactate and its metabolism in various diseases is lacking. This review outlines the sources and clearance of lactate, as well as its roles in cancer, liver fibrosis, sepsis, ischemic stroke, myocardial infarction, and acute kidney injury, and summarizes the effects of small molecules on lactate regulation. It aims to provide a reference and direction for future research.
Collapse
Affiliation(s)
- Jin Liu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Feng Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yang Tang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Linghui Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ling Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
4
|
Hefny SM, El-Moselhy TF, El-Din N, Giovannuzzi S, Bin Traiki T, Vaali-Mohammed MA, El-Dessouki AM, Yamaguchi K, Sugiura M, Shaldam MA, Supuran CT, Abdulla MH, Eldehna WM, Tawfik HO. Discovery and Mechanistic Studies of Dual-Target Hits for Carbonic Anhydrase IX and VEGFR-2 as Potential Agents for Solid Tumors: X-ray, In Vitro, In Vivo, and In Silico Investigations of Coumarin-Based Thiazoles. J Med Chem 2024; 67:7406-7430. [PMID: 38642371 DOI: 10.1021/acs.jmedchem.4c00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2024]
Abstract
A dual-targeting approach is predicted to yield better cancer therapy outcomes. Consequently, a series of coumarin-based thiazoles (5a-h, 6, and 7a-e) were designed and constructed as potential carbonic anhydrase (CA) and VEGFR-2 suppressors. The inhibitory actions of the target compounds were assessed against CA isoforms IX and VEGFR-2. The assay results showed that coumarin-based thiazoles 5a, 5d, and 5e can effectively inhibit both targets. 5a, 5d, and 5e cytotoxic effects were tested on pancreatic, breast, and prostate cancer cells (PANC1, MCF7, and PC3). Further mechanistic investigation disclosed the ability of 5e to interrupt the PANC1 cell progression in the S stage by triggering the apoptotic cascade, as seen by increased levels of caspases 3, 9, and BAX, alongside the Bcl-2 decline. Moreover, the in vivo efficacy of compound 5e as an antitumor agent was evaluated. Also, molecular docking and dynamics displayed distinctive interactions between 5e and CA IX and VEGFR-2 binding pockets.
Collapse
Affiliation(s)
- Salma M Hefny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Tarek F El-Moselhy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Nabaweya El-Din
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Simone Giovannuzzi
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Sesto Fiorentino, Firenze Italy
| | - Thamer Bin Traiki
- Department of Surgery, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | | | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, sixth of October City, Giza 12566, Egypt
| | - Koki Yamaguchi
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Masaharu Sugiura
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Sesto Fiorentino, Firenze Italy
| | - Maha-Hamadien Abdulla
- Department of Surgery, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
5
|
Wang Y, Ai Q, Gu M, Guan H, Yang W, Zhang M, Mao J, Lin Z, Liu Q, Liu J. Comprehensive overview of different medicinal parts from Morus alba L.: chemical compositions and pharmacological activities. Front Pharmacol 2024; 15:1364948. [PMID: 38694910 PMCID: PMC11061381 DOI: 10.3389/fphar.2024.1364948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/25/2024] [Indexed: 05/04/2024] Open
Abstract
Morus alba L., a common traditional Chinese medicine (TCM) with a centuries-old medicinal history, owned various medicinal parts like Mori folium, Mori ramulus, Mori cortex and Mori fructus. Different medical parts exhibit distinct modern pharmacological effects. Mori folium exhibited analgesic, anti-inflammatory, hypoglycemic action and lipid-regulation effects. Mori ramulus owned anti-bacterial, anti-asthmatic and diuretic activities. Mori cortex showed counteraction action of pain, inflammatory, bacterial, and platelet aggregation. Mori fructus could decompose fat, lower blood lipids and prevent vascular sclerosis. The main chemical components in Morus alba L. covered flavonoids, phenolic compounds, alkaloids, and amino acids. This article comprehensively analyzed the recent literature related to chemical components and pharmacological actions of M. alba L., summarizing 198 of ingredients and described the modern activities of different extracts and the bioactive constituents in the four parts from M. alba L. These results fully demonstrated the medicinal value of M. alba L., provided valuable references for further comprehensive development, and layed the foundation for the utilization of M. alba L.
Collapse
Affiliation(s)
- Yumei Wang
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Qing Ai
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
- School of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Meiling Gu
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
- School of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Hong Guan
- Office of Academic Research, Qiqihar Medical University, Qiqihar, China
| | - Wenqin Yang
- Office of Academic Research, Qiqihar Medical University, Qiqihar, China
| | - Meng Zhang
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
- School of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Jialin Mao
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Zhao Lin
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Qi Liu
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Jicheng Liu
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
6
|
Andriolo LG, Cammisotto V, Spagnoli A, Alunni Fegatelli D, Chicone M, Di Rienzo G, Dell’Anna V, Lobreglio G, Serio G, Pignatelli P. Overview of angiogenesis and oxidative stress in cancer. World J Meta-Anal 2023; 11:253-265. [DOI: 10.13105/wjma.v11.i6.253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/07/2023] [Accepted: 07/10/2023] [Indexed: 09/13/2023] Open
Abstract
Neoplasms can be considered as a group of aberrant cells that need more vascular supply to fulfill all their functions. Therefore, they promote angiogenesis through the same neovascularization pathway used physiologically. Angiogenesis is a process characterized by a heterogeneous distribution of oxygen caused by the tumor and oxidative stress; the latter being one of the most powerful stimuli of angiogenesis. As a result of altered tumor metabolism due to hypoxia, acidosis occurs. The angiogenic process and oxidative stress can be detected by measuring serum and tissue biomarkers. The study of the mechanisms underlying angiogenesis and oxidative stress could lead to the identification of new biomarkers, ameliorating the selection of patients with neoplasms and the prediction of their response to possible anti-tumor therapies. In particular, in the treatment of patients with similar clinical tumor phenotypes but different prognoses, the new biomarkers could be useful. Moreover, they may lead to a better understanding of the mechanisms underlying drug resistance. Experimental studies show that blocking the vascular supply results in antiproliferative activity in vivo in neuroendocrine tumor cells, which require a high vascular supply.
Collapse
Affiliation(s)
- Luigi Gaetano Andriolo
- Department of General and Specialistic Surgery Paride Stefanini, Policlinico Umberto I, University of Rome Sapienza, Rome 06100, Italy
- Unità Operativa Complessa Chirurgia Toracica, Ospedale Vito Fazzi, Lecce 73100, Italy
| | - Vittoria Cammisotto
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences, University of Rome Sapienza, Rome 06100, Italy
| | - Alessandra Spagnoli
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome 06100, Italy
| | - Danilo Alunni Fegatelli
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome 06100, Italy
| | - Michele Chicone
- Department of Clinical Pathology and Microbiology, Ospedale Vito Fazzi, Lecce 73100, Italy
| | - Gaetano Di Rienzo
- Unità Operativa Complessa Chirurgia Toracica, Ospedale Vito Fazzi, Lecce 73100, Italy
| | | | - Giambattista Lobreglio
- Department of Clinical Pathology and Microbiology, Ospedale Vito Fazzi, Lecce 73100, Italy
| | - Giovanni Serio
- Pathological Anatomy Unit, Ospedale Vito Fazzi, Lecce 73100, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences, University of Rome Sapienza, Rome 06100, Italy
| |
Collapse
|
7
|
Zhang H, Huang H, Wu S, He X, Chen J, Zheng X, Chen L, Wang Z. Single-cell RNA sequencing reveals the effects of anti-PD-L1 therapy on 3LL lung cancer model and its tumor microenvironment. Med Oncol 2023; 40:285. [PMID: 37653265 DOI: 10.1007/s12032-023-02156-w] [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: 07/07/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023]
Abstract
PD-L1 is expressed on antigen-presenting cells and tumor cells, thus allows tumor cells to escape immune surveillance. Moreover, targeting PD-L1 was also recommended and selected as important immune checkpoint inhibitors (ICIs) strategy in the treatment of advanced cancers due to the safety and activity. However, the detailed alteration of tumor microenvironment (TME) upon anti-PD-L1 therapy in lung cancer tumor model still needs to be resolved. In our present study, first, we characterized PD-L1 expression in human lung adenocarcinoma tissues by using public data, then we established the subcutaneous tumor-bearing model by using murine lung cancer cell line 3LL to perform the anti-PD-L1 therapy and the single-cell RNA sequencing (scRNA-seq) to reveal the remodeling of TME. We confirmed that PD-L1 blockade significantly inhibited tumor progression in 3LL mouse lung cancer model. The scRNA-seq depicted the detailed TME landscape of 3LL tumor model upon anti-PD-L1 treatment. Five major populations according to the marker genes were identified, including tumor cells, stromal cells, myeloid cells, T cells, and NK cells. In addition, we found that anti-PD-L1 treatment enhanced tumor immunogenicity and promoted inflammation in TME and promoted cancer-associated fibroblasts (CAFs)-mediated T-cell migration and infiltration. We also found that anti-PD-L1 treatment can increase dendritic cells (DCs) population and enhance the antigen-presenting ability to CD8+T cells and promote the transition of monocytes to macrophages and tumor-associated macrophages 2 (TAM2) to TAM1. We also revealed that Nfatc1 was up-regulated in the anti-PD-L1 treatment group, the frequencies of effector CD8+T cells, exhausted CD8+T cells, cycling T cells, and NKT were increased, and the frequencies of conventional CD4+T cells, Treg, IFN-induced T cells, and γδT cells were decreased. Therefore, our scRNA-seq data of the lung cancer tumor model upon anti-PD-L1 treatment made a comprehensive presentation and description about the remodeling of TME and will benefit us to understand the underlying mechanisms and to design combinational therapeutic strategies based on anti-PD-L1 therapy against lung cancer.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Geriatrics, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
| | - Hao Huang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
| | - Xian He
- Department of Geriatrics, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
| | - Junjun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, China.
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China.
| | - Zhigang Wang
- Department of Geriatrics, The Third Affiliated Hospital of Suzhou University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|