1.AAZ2 induces mitochondrial-dependent apoptosis by targeting PDK1 in gastric cancer.
Yi LI ; Wenyan SHE ; Xiaoran XU ; Yixin LIU ; Xinyu WANG ; Sheng TIAN ; Shiyi LI ; Miao WANG ; Chaochao YU ; Pan LIU ; Tianhe HUANG ; Yongchang WEI
Journal of Zhejiang University. Science. B 2023;24(3):232-247
		                        		
		                        			
		                        			Drastic surges in intracellular reactive oxygen species (ROS) induce cell apoptosis, while most chemotherapy drugs lead to the accumulation of ROS. Here, we constructed an organic compound, arsenical N-(4-(1,3,2-dithiarsinan-2-yl)phenyl)acrylamide (AAZ2), which could prompt the ROS to trigger mitochondrial-dependent apoptosis in gastric cancer (GC). Mechanistically, by targeting pyruvate dehydrogenase kinase 1 (PDK1), AAZ2 caused metabolism alteration and the imbalance of redox homeostasis, followed by the inhibition of phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway and leading to the activation of B-cell lymphoma 2 (Bcl2)/Bcl2-associated X (Bax)/caspase-9 (Cas9)/Cas3 cascades. Importantly, our in vivo data demonstrated that AAZ2 could inhibit the growth of GC xenograft. Overall, our data suggested that AAZ2 could contribute to metabolic abnormalities, leading to mitochondrial-dependent apoptosis by targeting PDK1 in GC.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			Stomach Neoplasms/drug therapy*
		                        			;
		                        		
		                        			Reactive Oxygen Species/metabolism*
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-akt/metabolism*
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-bcl-2
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			
		                        		
		                        	
2.GDF15 negatively regulates chemosensitivity via TGFBR2-AKT pathway-dependent metabolism in esophageal squamous cell carcinoma.
Yingxi DU ; Yarui MA ; Qing ZHU ; Yong FU ; Yutong LI ; Ying ZHANG ; Mo LI ; Feiyue FENG ; Peng YUAN ; Xiaobing WANG
Frontiers of Medicine 2023;17(1):119-131
		                        		
		                        			
		                        			Treating patients with esophageal squamous cell carcinoma (ESCC) is challenging due to the high chemoresistance. Growth differentiation factor 15 (GDF15) is crucial in the development of various types of tumors and negatively related to the prognosis of ESCC patients according to our previous research. In this study, the link between GDF15 and chemotherapy resistance in ESCC was further explored. The relationship between GDF15 and the chemotherapy response was investigated through in vitro and in vivo studies. ESCC patients with high levels of GDF15 expression showed an inferior chemotherapeutic response. GDF15 improved the tolerance of ESCC cell lines to low-dose cisplatin by regulating AKT phosphorylation via TGFBR2. Through an in vivo study, we further validated that the anti-GDF15 antibody improved the tumor inhibition effect of cisplatin. Metabolomics showed that GDF15 could alter cellular metabolism and enhance the expression of UGT1A. AKT and TGFBR2 inhibition resulted in the reversal of the GDF15-induced expression of UGT1A, indicating that TGFBR2-AKT pathway-dependent metabolic pathways were involved in the resistance of ESCC cells to cisplatin. The present investigation suggests that a high level of GDF15 expression leads to ESCC chemoresistance and that GDF15 can be targeted during chemotherapy, resulting in beneficial therapeutic outcomes.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Esophageal Squamous Cell Carcinoma/drug therapy*
		                        			;
		                        		
		                        			Cisplatin/metabolism*
		                        			;
		                        		
		                        			Esophageal Neoplasms/metabolism*
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-akt/metabolism*
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/genetics*
		                        			;
		                        		
		                        			Growth Differentiation Factor 15/therapeutic use*
		                        			;
		                        		
		                        			Receptor, Transforming Growth Factor-beta Type II/therapeutic use*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			
		                        		
		                        	
3.Physical exercise suppresses hepatocellular carcinoma progression by alleviating hypoxia and attenuating cancer stemness through the Akt/GSK-3β/β-catenin pathway.
Chu-Lan XIAO ; Zhi-Peng ZHONG ; Can LÜ ; Bing-Jie GUO ; Jiao-Jiao CHEN ; Tong ZHAO ; Zi-Fei YIN ; Bai LI
Journal of Integrative Medicine 2023;21(2):184-193
		                        		
		                        			OBJECTIVE:
		                        			Physical exercise, a common non-drug intervention, is an important strategy in cancer treatment, including hepatocellular carcinoma (HCC). However, the mechanism remains largely unknown. Due to the importance of hypoxia and cancer stemness in the development of HCC, the present study investigated whether the anti-HCC effect of physical exercise is related to its suppression on hypoxia and cancer stemness.
		                        		
		                        			METHODS:
		                        			A physical exercise intervention of swimming (30 min/d, 5 d/week, for 4 weeks) was administered to BALB/c nude mice bearing subcutaneous human HCC tumor. The anti-HCC effect of swimming was assessed in vivo by tumor weight monitoring, hematoxylin and eosin (HE) staining, and immunohistochemistry (IHC) detection of proliferating cell nuclear antigen (PCNA) and Ki67. The expression of stemness transcription factors, including Nanog homeobox (NANOG), octamer-binding transcription factor 4 (OCT-4), v-Myc avian myelocytomatosis viral oncogene homolog (C-MYC) and hypoxia-inducible factor-1α (HIF-1α), was detected using real-time reverse transcription polymerase chain reaction. A hypoxia probe was used to explore the intratumoral hypoxia status. Western blot was used to detect the expression of HIF-1α and proteins related to protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β)/β-catenin signaling pathway. The IHC analysis of platelet endothelial cell adhesion molecule-1 (CD31), and the immunofluorescence co-location of CD31 and desmin were used to analyze tumor blood perfusion. SMMC-7721 cells were treated with nude mice serum. The inhibition effect on cancer stemness in vitro was detected using suspension sphere experiments and the expression of stemness transcription factors. The hypoxia status was inferred by measuring the protein and mRNA levels of HIF-1α. Further, the expression of proteins related to Akt/GSK-3β/β-catenin signaling pathway was detected.
		                        		
		                        			RESULTS:
		                        			Swimming significantly reduced the body weight and tumor weight in nude mice bearing HCC tumor. HE staining and IHC results showed a lower necrotic area ratio as well as fewer PCNA or Ki67 positive cells in mice receiving the swimming intervention. Swimming potently alleviated the intratumoral hypoxia, attenuated the cancer stemness, and inhibited the Akt/GSK-3β/β-catenin signaling pathway. Additionally, the desmin+/CD31+ ratio, rather than the number of CD31+ vessels, was significantly increased in swimming-treated mice. In vitro experiments showed that treating cells with the serum from the swimming intervention mice significantly reduced the formation of SMMC-7721 cell suspension sphere, as well as the mRNA expression level of stemness transcription factors. Consistent with the in vivo results, HIF-1α and Akt/GSK-3β/β-catenin signaling pathway were also inhibited in cells treated with serum from swimming group.
		                        		
		                        			CONCLUSION
		                        			Swimming alleviated hypoxia and attenuated cancer stemness in HCC, through suppression of the Akt/GSK-3β/β-catenin signaling pathway. The alleviation of intratumoral hypoxia was related to the increase in blood perfusion in the tumor. Please cite this article as: Xiao CL, Zhong ZP, Lü C, Guo BJ, Chen JJ, Zhao T, Yin ZF, Li B. Physical exercise suppresses hepatocellular carcinoma progression by alleviating hypoxia and attenuating cancer stemness through the Akt/GSK-3β/β-catenin pathway. J Integr Med. 2023; 21(2): 184-193.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Carcinoma, Hepatocellular/drug therapy*
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-akt/metabolism*
		                        			;
		                        		
		                        			Proliferating Cell Nuclear Antigen/therapeutic use*
		                        			;
		                        		
		                        			Mice, Nude
		                        			;
		                        		
		                        			Glycogen Synthase Kinase 3 beta/genetics*
		                        			;
		                        		
		                        			beta Catenin/therapeutic use*
		                        			;
		                        		
		                        			Liver Neoplasms/drug therapy*
		                        			;
		                        		
		                        			Desmin/therapeutic use*
		                        			;
		                        		
		                        			Ki-67 Antigen
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Hypoxia
		                        			;
		                        		
		                        			RNA, Messenger/therapeutic use*
		                        			;
		                        		
		                        			Cell Proliferation
		                        			
		                        		
		                        	
4.FOXA1 in prostate cancer.
Hui-Yu DONG ; Lei DING ; Tian-Ren ZHOU ; Tao YAN ; Jie LI ; Chao LIANG
Asian Journal of Andrology 2023;25(3):287-295
		                        		
		                        			
		                        			Most prostate cancers initially respond to androgen deprivation therapy (ADT). With the long-term application of ADT, localized prostate cancer will progress to castration-resistant prostate cancer (CRPC), metastatic CRPC (mCRPC), and neuroendocrine prostate cancer (NEPC), and the transcriptional network shifted. Forkhead box protein A1 (FOXA1) may play a key role in this process through multiple mechanisms. To better understand the role of FOXA1 in prostate cancer, we review the interplay among FOXA1-targeted genes, modulators of FOXA1, and FOXA1 with a particular emphasis on androgen receptor (AR) function. Furthermore, we discuss the distinct role of FOXA1 mutations in prostate cancer and clinical significance of FOXA1. We summarize possible regulation pathways of FOXA1 in different stages of prostate cancer. We focus on links between FOXA1 and AR, which may play different roles in various types of prostate cancer. Finally, we discuss FOXA1 mutation and its clinical significance in prostate cancer. FOXA1 regulates the development of prostate cancer through various pathways, and it could be a biomarker for mCRPC and NEPC. Future efforts need to focus on mechanisms underlying mutation of FOXA1 in advanced prostate cancer. We believe that FOXA1 would be a prognostic marker and therapeutic target in prostate cancer.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Androgen Antagonists/therapeutic use*
		                        			;
		                        		
		                        			Androgens/metabolism*
		                        			;
		                        		
		                        			Hepatocyte Nuclear Factor 3-alpha/metabolism*
		                        			;
		                        		
		                        			Mutation
		                        			;
		                        		
		                        			Prostatic Neoplasms, Castration-Resistant/drug therapy*
		                        			;
		                        		
		                        			Receptors, Androgen/metabolism*
		                        			
		                        		
		                        	
5.Expert consensus on PD-L1 expression testing in esophageal carcinoma in China.
Li Yan XUE ; Yin LI ; Jing HUANG
Chinese Journal of Oncology 2023;45(4):291-297
		                        		
		                        			
		                        			In recent years, immunotherapy represented by immune checkpoint inhibitors programmed death 1 (PD-1) has made great progress in the treatment of esophageal cancer and is rewriting the global paradigm for the treatment of esophageal cancer. According to current data, only a small number of patients with esophageal cancer could benefit from immunotherapy. Therefore, it is a challenge to screen the potential beneficiaries of PD-1 inhibitors. Studies have shown that the expression level of programmed death-ligand 1 (PD-L1) in esophageal cancer is closely associated with the efficacy of PD-1 inhibitors, and PD-L1 is the most important predictive biomarker of the efficacy of PD-1 inhibitors. With the clinical application of different PD-1 inhibitors and PD-L1 protein expression detection platforms, clarifying the clinical significance and timing of detection of PD-L1 protein expression in esophageal cancer, and establishing a standardized PD-L1 testing procedure, are of great significance to improve the accuracy of detection and reduce the difference between laboratories, so as to maximize the therapeutic benefits for patients. This consensus was finally reached, based on the combination of literature, expert experience, and internal discussion and voting of committee members, to provide an accurate and reliable evidence for clinicians to make decisions.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			B7-H1 Antigen/metabolism*
		                        			;
		                        		
		                        			Immune Checkpoint Inhibitors/therapeutic use*
		                        			;
		                        		
		                        			Consensus
		                        			;
		                        		
		                        			Esophageal Neoplasms/drug therapy*
		                        			;
		                        		
		                        			Immunotherapy/methods*
		                        			;
		                        		
		                        			Lung Neoplasms/pathology*
		                        			
		                        		
		                        	
6.Hydroxysafflor yellow A inhibits proliferation, migration, and chemoresistance of colorectal cancer cells through Akt/mTOR-autophagy pathway.
Li WANG ; Fang LI ; Ni-Ni GU ; Hui SHEN ; Cai-Li HAN ; Kai-Yang LI ; Rui-Yang YAN ; Jue WANG ; Zhi-Kuan MI
China Journal of Chinese Materia Medica 2023;48(2):517-524
		                        		
		                        			
		                        			In recent years, the clinical treatment of colorectal cancer(CRC) has made great progress, but chemoresistance is still one of the main reasons for reducing the survival rate of patients with colorectal cancer. Therefore, ameliorating chemotherapy resis-tance is an urgent problem to be solved. The purpose of this study was to investigate the regulatory role and related molecular mechanisms of hydroxysafflor yellow A(HSYA) in colorectal cancer cell proliferation, migration, and 5-fluorouracil(5-FU) chemoresistance. In this study, HCT116 and HT-29 cells were used as research subjects. Firstly, methyl thiazolyl tetrazolium(MTT) assay and colony formation assay were used to detect and analyze the effect of HSYA on the proliferation of CRC cells. Secondly, the effect of HSYA on the cell cycle in CRC cells was analyzed by cell cycle assay. Furthermore, the effect of HSYA on the migration of CRC cells was analyzed by wound-healing assay and Transwell assay. Based on the above, the influences of HSYA on 5-FU chemoresistance of CRC cells and related molecular mechanisms were explored and analyzed. The results showed that HSYA significantly inhibited the proliferation and migration of CRC cells, and arrested the cell cycle in G_0/G_1 phase. In addition, HSYA significantly ameliorated the chemoresistance of CRC cells to 5-FU. The results of acridine orange staining and Western blot showed that the autophagy activity of CRC cells in the HSYA and 5-FU combined treatment group was significantly higher than that in the 5-FU single drug treatment group. As compared with the 5-FU single drug treatment group, the phosphorylation levels of protein kinase B(Akt) and mammalian target of rapamycin(mTOR) in the HSYA and 5-FU combined treatment group were significantly reduced, indicating that the Akt/mTOR signaling pathway in the combined treatment group was down-regulated in CRC cells. In conclusion, HSYA may upregulate autophagy activity through the Akt/mTOR signaling pathway, thereby inhibiting the proliferation and migration of CRC cells and ameliorating the chemoresistance to 5-FU.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-akt/metabolism*
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			TOR Serine-Threonine Kinases/metabolism*
		                        			;
		                        		
		                        			Fluorouracil/pharmacology*
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Autophagy
		                        			;
		                        		
		                        			Colorectal Neoplasms/drug therapy*
		                        			
		                        		
		                        	
7.Lung Squamous Cell Carcinoma with EML4-ALK Fusion and TP53 Co-mutation Treated with Ensartinib: A Case Report and Literature Review.
Donglai LV ; Chunwei XU ; Chong WANG ; Qiuju SANG
Chinese Journal of Lung Cancer 2023;26(1):78-82
		                        		
		                        			
		                        			Lung squamous cell carcinoma (LSCC) accounts for approximately 30% of non-small cell lung cancer (NSCLC) cases and is the second most common histological type of lung cancer. Anaplastic lymphoma kinase (ALK)-positive NSCLC accounts for only 2%-5% of all NSCLC cases, and is almost exclusively detected in patients with lung adenocarcinoma. Thus, ALK testing is not routinely performed in the LSCC population, and the efficacy of such treatment for ALK-rearranged LSCC remains unknown. Echinoderm microtubule associated protein like 4 (EML4)-ALK (V1) and TP53 co-mutations were identified by next generation sequencing (NGS) in this patient with advanced LSCC. On December 3, 2020, Ensatinib was taken orally and the efficacy was evaluated as partial response (PR). The progression-free survival (PFS) was 19 months. When the disease progressed, the medication was changed to Loratinib. To our knowledge, Enshatinib created the longest PFS of ALK-mutant LSCC patients treated with targeted therapy since literature review. Herein, we described one case treated by Enshatinib involving a patient with both EML4-ALK and TP53 positive LSCC, and the relevant literatures were reviewed for discussing the treatment of this rare disease.
.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Carcinoma, Non-Small-Cell Lung/drug therapy*
		                        			;
		                        		
		                        			Lung Neoplasms/pathology*
		                        			;
		                        		
		                        			Anaplastic Lymphoma Kinase/metabolism*
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/genetics*
		                        			;
		                        		
		                        			Mutation
		                        			;
		                        		
		                        			Cytoskeletal Proteins/genetics*
		                        			;
		                        		
		                        			Lung/pathology*
		                        			;
		                        		
		                        			Oncogene Proteins, Fusion/genetics*
		                        			;
		                        		
		                        			Protein Kinase Inhibitors/therapeutic use*
		                        			;
		                        		
		                        			Tumor Suppressor Protein p53/genetics*
		                        			
		                        		
		                        	
8.Application of PROTACs in Hematological Malignancies--Review.
Journal of Experimental Hematology 2023;31(6):1921-1924
		                        		
		                        			
		                        			Proteolysis-targeting chimeras (PROTACs) are heterobifunctional small molecules by utilizing the ubiquitin proteasome system (UPS) to degrade proteins of interest. PROTACs have exhibited unprecedented efficacy and specificity in degrading various oncogenic proteins because of their unique mechanism of action, ability to target "undruggable" and mutant proteins. A series of PROTACs have been developed to degrade multiple key protein targets for the treatment of hematologic malignancy. Notably, PROTACs that target BCL-XL, IRAK4, STAT3 and BTK have entered clinical trials. The known PROTACs that have the potential to be used to treat various hematological malignancies are systematically summarized in this review.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Hematologic Neoplasms/drug therapy*
		                        			;
		                        		
		                        			Proteasome Endopeptidase Complex/metabolism*
		                        			;
		                        		
		                        			Ubiquitin-Protein Ligases/metabolism*
		                        			;
		                        		
		                        			Proteolysis Targeting Chimera
		                        			
		                        		
		                        	
9.Construction of A Nomogram Prediction Model for PD-L1 Expression in Non-small Cell Lung Cancer Based on 18F-FDG PET/CT Metabolic Parameters.
Luoluo HAO ; Lifeng WANG ; Mengyao ZHANG ; Jiaming YAN ; Feifei ZHANG
Chinese Journal of Lung Cancer 2023;26(11):833-842
		                        		
		                        			BACKGROUND:
		                        			In recent years, immunotherapy represented by programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) immunosuppressants has greatly changed the status of non-small cell lung cancer (NSCLC) treatment. PD-L1 has become an important biomarker for screening NSCLC immunotherapy beneficiaries, but how to easily and accurately detect whether PD-L1 is expressed in NSCLC patients is a difficult problem for clinicians. The aim of this study was to construct a Nomogram prediction model of PD-L1 expression in NSCLC patients based on 18F-fluorodeoxy glucose (18F-FDG) positron emission tomography/conputed tomography (PET/CT) metabolic parameters and to evaluate its predictive value.
		                        		
		                        			METHODS:
		                        			Retrospective collection of 18F-FDG PET/CT metabolic parameters, clinicopathological information and PD-L1 test results of 155 NSCLC patients from Inner Mongolia People's Hospital between September 2016 and July 2021. The patients were divided into the training group (n=117) and the internal validation group (n=38), and another 51 cases of NSCLC patients in our hospital between August 2021 and July 2022 were collected as the external validation group according to the same criteria. Then all of them were categorized according to the results of PD-L1 assay into PD-L1+ group and PD-L1- group. The metabolic parameters and clinicopathological information of patients in the training group were analyzed by univariate and binary Logistic regression, and a Nomogram prediction model was constructed based on the screened independent influencing factors. The effect of the model was evaluated by receiver operating characteristic (ROC) curve, calibration curve and decision curve analysis (DCA) in both the training group and the internal and external validation groups.
		                        		
		                        			RESULTS:
		                        			Binary Logistic regression analysis showed that metabolic tumor volume (MTV), gender and tumor diameter were independent influences on PD-L1 expression. Then a Nomogram prediction model was constructed based on the above independent influences. The ROC curve for the model in the training group shows an area under the curve (AUC) of 0.769 (95%CI: 0.683-0.856) with an optimal cutoff value of 0.538. The AUC was 0.775 (95%CI: 0.614-0.936) in the internal validation group and 0.752 (95%CI: 0.612-0.893) in the external validation group. The calibration curves were tested by the Hosmer-Lemeshow test and showed that the training group (χ2=0.040, P=0.979), the internal validation group (χ2=2.605, P=0.271), and the external validation group (χ2=0.396, P=0.820) were well calibrated. The DCA curves show that the model provides clinical benefit to patients over a wide range of thresholds (training group: 0.00-0.72, internal validation group: 0.00-0.87, external validation group: 0.00-0.66).
		                        		
		                        			CONCLUSIONS
		                        			The Nomogram prediction model constructed on the basis of 18F-FDG PET/CT metabolic parameters has greater application value in predicting PD-L1 expression in NSCLC patients.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Carcinoma, Non-Small-Cell Lung/drug therapy*
		                        			;
		                        		
		                        			Positron Emission Tomography Computed Tomography
		                        			;
		                        		
		                        			Lung Neoplasms/drug therapy*
		                        			;
		                        		
		                        			Fluorodeoxyglucose F18/therapeutic use*
		                        			;
		                        		
		                        			Nomograms
		                        			;
		                        		
		                        			Retrospective Studies
		                        			;
		                        		
		                        			B7-H1 Antigen/metabolism*
		                        			;
		                        		
		                        			Glucose/therapeutic use*
		                        			;
		                        		
		                        			Positron-Emission Tomography/methods*
		                        			
		                        		
		                        	
10.Anticancer Activity of Diosgenin and Its Molecular Mechanism.
Qun-Li REN ; Qian WANG ; Xin-Qun ZHANG ; Miao WANG ; Huan HU ; Jun-Jie TANG ; Xiong-Tong YANG ; Ying-Hui RAN ; Huan-Huan LIU ; Zhi-Xing SONG ; Jian-Guo LIU ; Xiao-Lan LI
Chinese journal of integrative medicine 2023;29(8):738-749
		                        		
		                        			
		                        			Diosgenin, a steroidal sapogenin, obtained from Trigonella foenum-graecum, Dioscorea, and Rhizoma polgonati, has shown high potential and interest in the treatment of various cancers such as oral squamous cell carcinoma, laryngeal cancer, esophageal cancer, liver cancer, gastric cancer, lung cancer, cervical cancer, prostate cancer, glioma, and leukemia. This article aims to provide an overview of the in vivo, in vitro, and clinical studies reporting the diosgenin's anticancer effects. Preclinical studies have shown promising effects of diosgenin on inhibiting tumor cell proliferation and growth, promoting apoptosis, inducing differentiation and autophagy, inhibiting tumor cell metastasis and invasion, blocking cell cycle, regulating immunity and improving gut microbiome. Clinical investigations have revealed clinical dosage and safety property of diosgenin. Furthermore, in order to improve the biological activity and bioavailability of diosgenin, this review focuses on the development of diosgenin nano drug carriers, combined drugs and the diosgenin derivatives. However, further designed trials are needed to unravel the diosgenin's deficiencies in clinical application.
		                        		
		                        		
		                        		
		                        			Male
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/drug therapy*
		                        			;
		                        		
		                        			Diosgenin/metabolism*
		                        			;
		                        		
		                        			Mouth Neoplasms/drug therapy*
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Prostatic Neoplasms/drug therapy*
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail