1
|
Yan C, Wang G. Advances in research on flavonoids in tumor immunotherapy (Review). Mol Med Rep 2025; 31:150. [PMID: 40211703 PMCID: PMC11995692 DOI: 10.3892/mmr.2025.13515] [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: 09/03/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer immunotherapy is an approach used in anti‑tumor treatment; however, its efficacy is limited to specific tumor types that are inherently sensitive to immune system modulation. Expanding the scope of indications and enhancing the efficacy of cancer immunotherapy are key goals for continued advancement. Flavonoids modulate the tumor‑immunosuppressive microenvironment. Integrating flavonoids with immunotherapeutic modalities, including cancer vaccines, immune checkpoint inhibitors and adoptive immune‑cell therapy, has potential in terms of augmenting the therapeutic efficacy of immunotherapy. The present review aimed to summarize flavonoids that enhance cancer immunotherapy, focusing on their underlying mechanisms and the application of nanotechnology to overcome inherent limitations such as poor solubility, low bioavailability, rapid metabolism, and instability under physiological conditions, thereby highlighting the potential of flavonoids in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Chaoguang Yan
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| | - Guangchun Wang
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| |
Collapse
|
2
|
Gao Q, Wu H, Li Z, Yang Z, Li L, Sun X, Wu Q, Sui X. Synergistic Strategies for Lung Cancer Immunotherapy: Combining Phytochemicals and Immune-Checkpoint Inhibitors. Phytother Res 2025. [PMID: 40122686 DOI: 10.1002/ptr.8482] [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: 05/21/2024] [Revised: 09/27/2024] [Accepted: 10/14/2024] [Indexed: 03/25/2025]
Abstract
Lung cancer remains one of the most widespread and deadliest malignant tumors globally, with a particularly high mortality rate among all cancers. Recently, immunotherapy, particularly immune checkpoint inhibitors (ICIs), has emerged as a crucial treatment strategy for lung cancer patients, following surgical intervention, radiotherapy, chemotherapy, and targeted drug therapies. However, the therapeutic limitations are caused owing to their low response rate and undesirable side effects such as immune-related pneumonitis. Therefore, developing new strategies to improve the efficacy of ICIs while minimizing immune-related adverse events will be crucial for cancer immunotherapy. The tumor immune microenvironment plays a significant role in the success of lung cancer immunotherapy, and the immunosuppressive characteristics of the immune microenvironment are one of the major obstacles to the poor immunotherapeutic effect. Phytochemicals, naturally occurring compounds in plants, have shown promise in enhancing cancer immunotherapy by remodeling the immunosuppressive microenvironment, offering the potential to increase the efficacy of ICIs. Therefore, this review summarizes the associated mechanisms of phytochemicals remodeling the immunosuppressive microenvironment in lung cancer. Additionally, the review will focus on the synergistic effects of combining phytochemicals with ICIs, aiming to improve anticancer efficacy and reduce side effects, which may hopefully offer novel strategies to overcome current limitations in immunotherapy.
Collapse
Affiliation(s)
- Quan Gao
- Faculty of Medicine and Faculty of Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau SAR, China
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Zhejiang, China
| | - Hao Wu
- Faculty of Medicine and Faculty of Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau SAR, China
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Zhejiang, China
| | - Zhengjun Li
- College of Health Economics Management, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Engineering Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zijing Yang
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Zhejiang, China
| | - Lin Li
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Zhejiang, China
| | - Xueni Sun
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Zhejiang, China
| | - Qibiao Wu
- Faculty of Medicine and Faculty of Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau SAR, China
| | - Xinbing Sui
- Faculty of Medicine and Faculty of Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau SAR, China
- Department of Medical Oncology and School of Pharmacy, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Zhejiang, China
| |
Collapse
|
3
|
Wang M, Yang F, Kong J, Zong Y, Li Q, Shao B, Wang J. Traditional Chinese medicine enhances the effectiveness of immune checkpoint inhibitors in tumor treatment: A mechanism discussion. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:118955. [PMID: 39427737 DOI: 10.1016/j.jep.2024.118955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Immune checkpoint inhibitors (ICIs) have altered the landscape of tumor immunotherapy, offering novel therapeutic approaches alongside surgery, chemotherapy, and radiotherapy and significantly improving survival benefits. However, their clinical efficacy is limited in some patients, and their use may cause immune-related adverse events (irAEs). Integrating traditional Chinese medicine (TCM) with ICIs has demonstrated the potential to boost sensitization and reduce toxicity. Clinical trials and experimental explorations have confirmed that TCM and its active components synergistically enhance the effectiveness of ICIs. AIMS This narrative review summarizes the TCM practices that enhance the clinical efficacy and reduce irAEs of ICIs. This paper also summarizes the mechanism of experimental studies on the synergies of Chinese herbal decoctions, Chinese herbal preparation, and Chinese herbal active ingredients. Most of the studies on TCM combined with ICIs are basic experiments. We discussed the mechanism of TCM enhanced ICIs to provide reference for the research and development of TCM adjuvant immunotherapy. METHODS We conducted a literature search using PubMed and Chinese National Knowledge Infrastructure databases, with a focus on herbal decoction, Chinese medicine preparations, and active ingredients that boost the effectiveness of ICIs and reduce irAEs. The search keywords were "ICIs and traditional Chinese medicine", "PD-1 and traditional Chinese medicine", "PD-L1 and traditional Chinese medicine", "CTLA-4 and traditional Chinese medicine", "IDO1 and traditional Chinese medicine", "Tim-3 and traditional Chinese medicine", "TIGIT and traditional Chinese medicine", "irAEs and traditional Chinese medicine". The search period was from May 2014 to May 2024. Articles involving the use of TCM or its components in combination with ICIs and investigating the underlying mechanisms were screened. Finally, 30 Chinese medicines used in combination with ICIs were obtained to explore the mechanism. In the part of immune checkpoint molecules other than PD-1, there were few studies on the combined application of TCM, so studies involving the regulation of immune checkpoint molecules by TCM were included. RESULTS TCM has been shown to boost the effectiveness of ICIs and reduce irAEs. Researchers indicate that TCM and its active components can work synergistically with ICIs by regulating immune checkpoints PD-1, PD-L1, CTLA-4, and IDO1, regulating intestinal flora, improving tumor microenvironment and more. CONCLUSIONS Combining TCM with ICIs can play a better anti-tumor role, but larger samples and high-quality clinical trials are necessary to confirm this. Many Chinese medicines and their ingredients have been shown to sensitize ICIs in experimental studies, which provides a rich choice for the subsequent development of ICI enhancers.
Collapse
Affiliation(s)
- Manting Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fan Yang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jingwei Kong
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100007, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yuhan Zong
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Bin Shao
- Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Ji Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
4
|
Dong L, Zhou Y, Wang L, Mao X, Wang J, Du Z, Che X, Li Y. Neobavaisoflavone Protects H9c2 Cells Against H 2O 2-Induced Mitochondrial Dysfunction Through ALOX15/PGC1-α Axis. J Biochem Mol Toxicol 2024; 38:e70043. [PMID: 39485322 DOI: 10.1002/jbt.70043] [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: 03/25/2024] [Revised: 09/18/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
Neobavaisoflavone (NBIF) is a natural antioxidant that has a variety of pharmacological activities. To investigate the effects of NBIF on oxidative stress-induced myocardial injury, H9c2 cells were treated with H2O2. Cell counting kit-8 was used to detect cell viability. Intracellular as well as lipid radicals were detected. To measure mitochondrial function, tetramethylrhodamine ethyl ester was used to detect mitochondrial membrane potential. 12- and 15-hydroxyeicosatetraenoic acids (HETE) were measured by LC-MS/MS. ALOX15, which is the upstream protein of 12-, 15-HETE, was also measured by using western blot analysis. The results showed that H2O2 induced lipid peroxidation in cardiomyocytes and caused mitochondrial dysfunction which was relieved by NBIF treatment. Besides, H2O2 significantly increased the production of 12-HETE and 15-HETE and upregulated the expression of ALOX15 while PGC-1α was downregulated and triggered the release of cytochrome c. The treatment of NBIF decreased the expression of ALOX15 and inhibited the activation of caspase-3. NBIF protected mitochondrial membrane integrity through increasing PGC-1α and Nrf1. Our results indicated that NBIF could protect cardiomyocytes against H2O2-induced mitochondrial dysfunction via ALOX15/PGC-1α axis.
Collapse
Affiliation(s)
- Linyue Dong
- Department of TCM Chemistry, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Zhou
- Department of TCM Chemistry, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liyun Wang
- Department of Clinical Laboratory, Yixing People's Hospital, Wuxi, Jiangsu, China
- Department of Endorinology, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - Xuhua Mao
- Department of Clinical Laboratory, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - Junfang Wang
- Department of Clinical Laboratory, Yixing People's Hospital, Wuxi, Jiangsu, China
- Department of Endorinology, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - Zenan Du
- Department of TCM Chemistry, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuyang Che
- Department of TCM Chemistry, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Li
- Department of TCM Chemistry, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Qi Y, Zhang L, Liu Y, Li Y, Liu Y, Zhang Z. Targeted modulation of myeloid-derived suppressor cells in the tumor microenvironment: Implications for cancer therapy. Biomed Pharmacother 2024; 180:117590. [PMID: 39423752 DOI: 10.1016/j.biopha.2024.117590] [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: 08/05/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells originating from the bone marrow, known for their potent immunosuppressive functions that contribute to tumor immune evasion and progression. This paper provides a comprehensive analysis of the multifaceted interactions between MDSCs and tumors, exploring their distinct phenotypes and immunosuppressive mechanisms. Key roles of MDSCs in tumor biology are discussed, including their involvement in the formation of the pre-metastatic niche, facilitation of angiogenesis, enhancement of vascular permeability, suppression of tumor cell apoptosis, and promotion of resistance to cancer therapies. Additionally, the review highlights recent advances in the development of MDSC-targeting therapies, with a focus on their potential to enhance anti-tumor immunity. The therapeutic potential of Traditional Chinese Medicine (TCM) in modulating MDSC quantity and function is also explored, suggesting a novel approach to cancer treatment by integrating traditional and modern therapeutic strategies.
Collapse
Affiliation(s)
- Yafeng Qi
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Liying Zhang
- School of Integrative Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yeyuan Liu
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yangyang Li
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yongqi Liu
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Zhiming Zhang
- Department of Oncology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| |
Collapse
|
6
|
Hao F, Zeng M, Cao B, Liang X, Ye K, Jiao X, Feng W, Zheng X. Neobavaisoflavone Ameliorates Memory Deficits and Brain Damage in Aβ 25-35-Induced Mice by Regulating SIRT1. CNS Neurosci Ther 2024; 30:e70068. [PMID: 39392360 PMCID: PMC11469773 DOI: 10.1111/cns.70068] [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: 08/06/2024] [Revised: 08/29/2024] [Accepted: 09/15/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a common chronic neurodegenerative disease in older people, and there is no specific treatment that can stop or reverse its progression. Neobavaisoflavone (NBIF) is a flavonoid that has been shown to have neuroprotective effects, but its role in AD has not been revealed. The present study investigated the role and mechanism of NBIF on Aβ25-35-induced brain injury. METHODS In this experiment, the AD mouse model was established by injection of Aβ25-35 peptides (200 μM, icv), and Donepezil (Don, 10 mg/kg/days), NBIF-L (15 mg/kg/days), and NBIF-H (30 mg/kg/days) were administered orally for 4 weeks. Learning memory, hippocampal pathological changes, pathological markers, apoptosis, oxidative stress, inflammation, immune cells were measured in mice. Network pharmacology combined with the GEO database led to the identification of SIRT1, a key target for NBIF intervention in AD, and levels of SIRT1, p-STAT3 and FOXO1 were measured. In addition, the antagonistic activity of SIRT1 transfection silencing against NBIF in Aβ25-35-induced in N9 cells and N2a-APP69 cells was investigated to assess whether the effects caused by NBIF were mediated by SIRT1. RESULTS The results showed that NBIF ameliorated learning memory and hippocampal neuronal damage, reduced pathological markers, apoptosis, oxidative stress and neuroinflammation, and modulated immune cells. SIRT1 is a key target for NBIF intervention in AD, and NBIF upregulates SIRT1 and reduces the expression levels of p-STAT3 and FOXO1. Furthermore, silencing SIRT1 effectively reduced the protective effect of NBIF on Aβ25-35-induced N9 cells and N2a-APP69 cells, which indicated that the protective effect of NBIF on AD is related to SIRT1. CONCLUSIONS NBIF ameliorated Aβ25-35-induced brain injury by inhibiting apoptosis, oxidative stress, and neuroinflammation, which may be mediated through SIRT1 signaling. These findings provide a rationale for NBIF in the treatment of AD and help facilitate the development of clinical therapeutic agents for AD.
Collapse
Affiliation(s)
- Fengxiao Hao
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Mengnan Zeng
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Bing Cao
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Xiwen Liang
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Kaili Ye
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Xinmian Jiao
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Weisheng Feng
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Xiaoke Zheng
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| |
Collapse
|
7
|
Ding X, Ma X, Meng P, Yue J, Li L, Xu L. Potential Effects of Traditional Chinese Medicine in Anti-Aging and Aging-Related Diseases: Current Evidence and Perspectives. Clin Interv Aging 2024; 19:681-693. [PMID: 38706635 PMCID: PMC11070163 DOI: 10.2147/cia.s447514] [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: 11/10/2023] [Accepted: 04/17/2024] [Indexed: 05/07/2024] Open
Abstract
Aging and aging-related diseases present a global public health problem. Therefore, the development of efficient anti-aging drugs has become an important area of research. Traditional Chinese medicine is an important complementary and alternative branch of aging-related diseases therapy. Recently, a growing number of studies have revealed that traditional Chinese medicine has a certain delaying effect on the progression of aging and aging-related diseases. Here, we review the progress in research into using traditional Chinese medicine for aging and aging-related diseases (including neurodegenerative diseases, cardiovascular diseases, diabetes, and cancer). Furthermore, we summarize the potential mechanisms of action of traditional Chinese medicine and provide references for further studies on aging and aging-related diseases.
Collapse
Affiliation(s)
- Xue Ding
- Department of Medical, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Xiuxia Ma
- Department of AIDS Clinical Research Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Pengfei Meng
- Department of the First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jingyu Yue
- Department of AIDS Clinical Research Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Liangping Li
- Department of Graduate, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Liran Xu
- Department of the First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| |
Collapse
|
8
|
Hu Y, Liu Y, Zong L, Zhang W, Liu R, Xing Q, Liu Z, Yan Q, Li W, Lei H, Liu X. The multifaceted roles of GSDME-mediated pyroptosis in cancer: therapeutic strategies and persisting obstacles. Cell Death Dis 2023; 14:836. [PMID: 38104141 PMCID: PMC10725489 DOI: 10.1038/s41419-023-06382-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
Pyroptosis is a novel regulated cell death (RCD) mode associated with inflammation and innate immunity. Gasdermin E (GSDME), a crucial component of the gasdermin (GSDM) family proteins, has the ability to convert caspase-3-mediated apoptosis to pyroptosis of cancer cells and activate anti-tumor immunity. Accumulating evidence indicates that GSDME methylation holds tremendous potential as a biomarker for early detection, diagnosis, prognosis, and treatment of tumors. In fact, GSDME-mediated pyroptosis performs a dual role in anti-tumor therapy. On the one side, pyroptotic cell death in tumors caused by GSDME contributes to inflammatory cytokines release, which transform the tumor immune microenvironment (TIME) from a 'cold' to a 'hot' state and significantly improve anti-tumor immunotherapy. However, due to GSDME is expressed in nearly all body tissues and immune cells, it can exacerbate chemotherapy toxicity and partially block immune response. How to achieve a balance between the two sides is a crucial research topic. Meanwhile, the potential functions of GSDME-mediated pyroptosis in anti-programmed cell death protein 1 (PD-1) therapy, antibody-drug conjugates (ADCs) therapy, and chimeric antigen receptor T cells (CAR-T cells) therapy have not yet been fully understood, and how to improve clinical outcomes persists obscure. In this review, we systematically summarize the latest research regarding the molecular mechanisms of pyroptosis and discuss the role of GSDME-mediated pyroptosis in anti-tumor immunity and its potential applications in cancer treatment.
Collapse
Affiliation(s)
- Yixiang Hu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Ya Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Wenyou Zhang
- Department of Pharmacy, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Renzhu Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Qichang Xing
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Zheng Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Qingzi Yan
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Wencan Li
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Haibo Lei
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| | - Xiang Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| |
Collapse
|
9
|
Wang LL, Liu N, Hu S, Xing RR, Wang RQ, Yang L, Chen X. Application of instantaneous nebulization dispersive liquid-phase microextraction combined with HPLC for the determination of chalcone and isoflavone in traditional Chinese medicines. J Sep Sci 2023; 46:e2300326. [PMID: 37485627 DOI: 10.1002/jssc.202300326] [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: 05/08/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/25/2023]
Abstract
A simple and rapid instantaneous nebulization dispersive liquid-phase microextraction method was developed, and combined with high-performance liquid chromatography for determination of the contents of seven analytes in traditional Chinese medicines. In this study, using the sprinkler device to achieve instantaneous synchronous dispersion and extraction, only one spray can rapidly achieve the concentration and enrichment of seven kinds of chalcone and isoflavones. The key factors affecting the extraction efficiency were optimized including the type and volume of extractant, the pH and salt concentration of the sample phase, and the number of dispersion. Under the optimal conditions, the enrichment factor of the target analytes ranged from 103.1 to 180.9, with good linearity and correlation coefficients above 0.9970. The limits of detection ranged from 0.02 to 0.15 ng/mL, with good accuracy (recoveries 91.1 to 108.9%) and precision (relative standard deviations 1.5-7.1%). This method has short extraction time (2 s), low organic solvent consumption and high enrichment effect, so it has a wide application prospects.
Collapse
Affiliation(s)
- Ling-Li Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, P. R. China
| | - Na Liu
- School of Pharmacy, Shanxi Medical University, Taiyuan, P. R. China
| | - Shuang Hu
- School of Pharmacy, Shanxi Medical University, Taiyuan, P. R. China
| | - Rong-Rong Xing
- School of Pharmacy, Shanxi Medical University, Taiyuan, P. R. China
| | - Run-Qin Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, P. R. China
| | - Li Yang
- School of Pharmacy, Shanxi Medical University, Taiyuan, P. R. China
| | - Xuan Chen
- School of Pharmacy, Shanxi Medical University, Taiyuan, P. R. China
| |
Collapse
|
10
|
Zheng H, Wang G, Liu M, Cheng H. Traditional Chinese medicine inhibits PD-1/PD-L1 axis to sensitize cancer immunotherapy: a literature review. Front Oncol 2023; 13:1168226. [PMID: 37397393 PMCID: PMC10312112 DOI: 10.3389/fonc.2023.1168226] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/01/2023] [Indexed: 07/04/2023] Open
Abstract
The Programmed death-1 (PD-1) and its programmed death-ligand 1 (PD-L1) comprise the PD-1/PD-L1 axis and maintain tumor immune evasion. Cancer immunotherapy based on anti-PD-1/PD-L1 antibodies is the most promising anti-tumor treatment available but is currently facing the thorny problem of unsatisfactory outcomes. Traditional Chinese Medicine (TCM), with its rich heritage of Chinese medicine monomers, herbal formulas, and physical therapies like acupuncture, moxibustion, and catgut implantation, is a multi-component and multi-target system of medicine known for enhancing immunity and preventing the spread of disease. TCM is often used as an adjuvant therapy for cancer in clinical practices, and recent studies have demonstrated the synergistic effects of combining TCM with cancer immunotherapy. In this review, we examined the PD-1/PD-L1 axis and its role in tumor immune escape while exploring how TCM therapies can modulate the PD-1/PD-L1 axis to improve the efficacy of cancer immunotherapy. Our findings suggest that TCM therapy can enhance cancer immunotherapy by reducing the expression of PD-1 and PD-L1, regulating T-cell function, improving the tumor immune microenvironment, and regulating intestinal flora. We hope this review may serve as a valuable resource for future studies on the sensitization of immune checkpoint inhibitors (ICIs) therapy.
Collapse
Affiliation(s)
- Huilan Zheng
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Gang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Ming Liu
- Department of Medical Oncology/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongbin Cheng
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Schäfer J, Klösgen VJ, Omer EA, Kadioglu O, Mbaveng AT, Kuete V, Hildebrandt A, Efferth T. In Silico and In Vitro Identification of P-Glycoprotein Inhibitors from a Library of 375 Phytochemicals. Int J Mol Sci 2023; 24:10240. [PMID: 37373385 DOI: 10.3390/ijms241210240] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer therapy with clinically established anticancer drugs is frequently hampered by the development of drug resistance of tumors and severe side effects in normal organs and tissues. The demand for powerful, but less toxic, drugs is high. Phytochemicals represent an important reservoir for drug development and frequently exert less toxicity than synthetic drugs. Bioinformatics can accelerate and simplify the highly complex, time-consuming, and expensive drug development process. Here, we analyzed 375 phytochemicals using virtual screenings, molecular docking, and in silico toxicity predictions. Based on these in silico studies, six candidate compounds were further investigated in vitro. Resazurin assays were performed to determine the growth-inhibitory effects towards wild-type CCRF-CEM leukemia cells and their multidrug-resistant, P-glycoprotein (P-gp)-overexpressing subline, CEM/ADR5000. Flow cytometry was used to measure the potential to measure P-gp-mediated doxorubicin transport. Bidwillon A, neobavaisoflavone, coptisine, and z-guggulsterone all showed growth-inhibitory effects and moderate P-gp inhibition, whereas miltirone and chamazulene strongly inhibited tumor cell growth and strongly increased intracellular doxorubicin uptake. Bidwillon A and miltirone were selected for molecular docking to wildtype and mutated P-gp forms in closed and open conformations. The P-gp homology models harbored clinically relevant mutations, i.e., six single missense mutations (F336Y, A718C, Q725A, F728A, M949C, Y953C), three double mutations (Y310A-F728A; F343C-V982C; Y953A-F978A), or one quadruple mutation (Y307C-F728A-Y953A-F978A). The mutants did not show major differences in binding energies compared to wildtypes. Closed P-gp forms generally showed higher binding affinities than open ones. Closed conformations might stabilize the binding, thereby leading to higher binding affinities, while open conformations may favor the release of compounds into the extracellular space. In conclusion, this study described the capability of selected phytochemicals to overcome multidrug resistance.
Collapse
Affiliation(s)
- Julia Schäfer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Vincent Julius Klösgen
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
- Institute of Bioinformatics, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ejlal A Omer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Onat Kadioglu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Armelle T Mbaveng
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang P.O. Box 67, Cameroon
| | - Victor Kuete
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang P.O. Box 67, Cameroon
| | - Andreas Hildebrandt
- Institute of Bioinformatics, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| |
Collapse
|
12
|
Sun J, Zhao H, Xu W, Jiang GQ. Recent advances in photothermal therapy-based multifunctional nanoplatforms for breast cancer. Front Chem 2022; 10:1024177. [PMID: 36199665 PMCID: PMC9528973 DOI: 10.3389/fchem.2022.1024177] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/02/2022] [Indexed: 11/15/2022] Open
Abstract
Breast cancer (BC) is one of the most common cancers in women worldwide; however, the successful treatment of BC, especially triple-negative breast cancer (TNBC), remains a significant clinical challenge. Recently, photothermal therapy (PTT), which involves the generation of heat under irradiation to achieve photothermal ablation of BC with minimal invasiveness and outstanding spatial–temporal selectivity, has been demonstrated as a novel therapy that can overcome the drawbacks of chemotherapy or surgery. Significantly, when combining PTT with chemotherapy and/or photodynamic therapy, an enhanced synergistic therapeutic effect can be achieved in both primary and metastatic BC tumors. Thus, this review discusses the recent developments in nanotechnology-based photothermal therapy for the treatment of BC and its metastasis to provide potential strategies for future BC treatment.
Collapse
Affiliation(s)
- Jingjun Sun
- Department of Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Breast Surgery, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
- *Correspondence: Jingjun Sun, ; Guo-Qin Jiang,
| | - Haiyan Zhao
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| | - Weixuan Xu
- Department of Breast Surgery, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Guo-Qin Jiang
- Department of Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Jingjun Sun, ; Guo-Qin Jiang,
| |
Collapse
|