1
|
Chen LJ, Liu HY, Xiao ZY, Qiu T, Zhang D, Zhang LJ, Han FY, Chen GJ, Xu XM, Zhu JH, Ding YQ, Wang SY, Ye YP, Jiao HL. IGF2BP3 promotes the progression of colorectal cancer and mediates cetuximab resistance by stabilizing EGFR mRNA in an m 6A-dependent manner. Cell Death Dis 2023; 14:581. [PMID: 37658049 PMCID: PMC10474290 DOI: 10.1038/s41419-023-06099-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 09/03/2023]
Abstract
Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), an RNA-binding protein, is associated with tumorigenesis and progression. However, the exact molecular mechanisms of IGF2BP3 in colorectal cancer (CRC) oncogenesis, progression, and drug resistance remain unclear. This study found that IGF2BP3 was upregulated in CRC tissues. Clinically, the elevated IGF2BP3 level is predictive of a poor prognosis. Functionally, IGF2BP3 enhances CRC tumorigenesis and progression both in vitro and in vivo. Mechanistically, IGF2BP3 promotes epidermal growth factor receptor (EGFR) mRNA stability and translation and further activates the EGFR pathway by serving as a reader in an N6-methyladenosine (m6A)-dependent manner by cooperating with METTL14. Furthermore, IGF2BP3 increases the drug resistance of CRC cells to the EGFR-targeted antibody cetuximab. Taken together, our results demonstrated that IGF2BP3 was a functional and clinical oncogene of CRC. Targeting IGF2BP3 and m6A modification may therefore offer rational therapeutic targets for patients with CRC.
Collapse
Affiliation(s)
- Li-Jie Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Hui-Ye Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Zhi-Yuan Xiao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
- Department of Pathology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, China
| | - Ting Qiu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Dan Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Ling-Jie Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Fang-Yi Han
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Guo-Jun Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Xue-Mei Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Jiong-Hua Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yan-Qing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Shu-Yang Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Ya-Ping Ye
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.
| | - Hong-Li Jiao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.
| |
Collapse
|
2
|
Xu W, Zhang L, Chen Z, Wang H, Yan Z. Pingyangmycin Activates Oral Carcinoma Cell Autophagy via the Phosphorylation of the PI3K/AKT/mTOR Axis to Achieve the Purpose of Treating Oral Carcinoma. Emerg Med Int 2022; 2022:4522873. [PMID: 36072614 PMCID: PMC9441391 DOI: 10.1155/2022/4522873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/29/2022] [Indexed: 11/21/2022] Open
Abstract
Objective The aim of the study is to investigate the role of pingyangmycin (PYM) in oral carcinoma (OC) cell autophagy via the PI3K/AKT/mTOR axis. Methods 200 μL PYM culture solution with a concentration of 100 μg/ml (low PYM (L-PYM) group), 300 μg/ml (middle PYM (M-PYM) group), 500 μg/ml (high PYM (H-PYM) group), and the same amount of conventional medium (normal control (NC)) were added to the purchased OC cell line SCC-25, respectively, and the PI3K/AKT/mTOR pathway expression, autophagy protein levels, cell activity, and apoptosis rate were determined. Subsequently, we selected OC cells co-cultured with PYM with the concentration of the most significant intervention effect and 740Y-P, a specific activator of the PI3K/AKT/mTOR axis, and those treated with 740Y-P alone for the aforementioned detection. Results L-PYM, M-PYM, and H-PYM groups all showed decreased PI3K, AKT, mTOR, and phosphorylated protein levels (P < 0.05). Beclin1 and LC3-II protein levels and apoptosis rate of PYM-intervened OC cells increased, but the activity decreased (P < 0.05). Under 740Y-P intervention, the PI3K/AKT/mTOR pathway was activated, cell activity was increased, and the apoptosis rate and autophagy were decreased (P < 0.05). Simultaneous use of PYM and 740Y-P led to no difference in cell condition compared with NC (P > 0.05P>0.05). Conclusion PYM can activate OC cell autophagy by inhibiting the phosphorylation of the PI3K/AKT/mTOR axis, and thus, achieving the goal of killing tumor cells.
Collapse
Affiliation(s)
- Wei Xu
- Department of Oral and Maxillofacial Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China
| | - Laijian Zhang
- Department of Oral and Maxillofacial Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China
| | - Zhi Chen
- Department of Oral and Maxillofacial Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China
| | - Hao Wang
- Department of Oral and Maxillofacial Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China
| | - Zhongyi Yan
- Department of Oral and Maxillofacial Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China
| |
Collapse
|
3
|
He S, Zhao C, Tao H, Sheng W, Gao R, Liu X, Zhen Y. A recombinant scFv antibody-based fusion protein that targets EGFR associated with IMPDH2 downregulation and its drug conjugate show therapeutic efficacy against esophageal cancer. Drug Deliv 2022; 29:1243-1256. [PMID: 35416106 PMCID: PMC9048960 DOI: 10.1080/10717544.2022.2063454] [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] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to evaluate the anti-tumor efficacy of the epidermal growth factor receptor (EGFR)-targeting recombinant fusion protein Fv-LDP-D3 and its antibody-drug conjugate Fv-LDP-D3-AE against esophageal cancer. Fv-LDP-D3, consisting of the fragment variable (Fv) of an anti-EGFR antibody, the apoprotein of lidamycin (LDP), and the third domain of human serum albumin (D3), exhibited a high binding affinity for EGFR-overexpressing esophageal cancer cells, inhibited EGFR phosphorylation and down-regulated inosine monophosphate dehydrogenase type II (IMPDH2) expression. Fv-LDP-D3 was taken up by cancer cells through intensive macropinocytosis; it inhibited the proliferation and induced the apoptosis of esophageal cancer cells. In vivo imaging revealed that Fv-LDP-D3 displayed specific tumor-site accumulation and a long-lasting retention over a 26-day period. Furthermore, Fv-LDP-D3-AE, a pertinent antibody-drug conjugate prepared by integrating the enediyne chromophore of lidamycin into the Fv-LDP-D3 molecule, displayed highly potent cytotoxicity, inhibited migration and invasion, induced apoptosis and DNA damage, arrested cells at G2/M phase, and caused mitochondrial damage in esophageal cancer cells. More importantly, both of Fv-LDP-D3 and Fv-LDP-D3-AE markedly inhibited the growth of esophageal cancer xenografts in athymic mice at well tolerated doses. The present results indicate that Fv-LDP-D3, and Fv-LDP-D3-AE exert prominent antitumor efficacy associated with targeting EGFR, suggesting their potential as promising candidates for targeted therapy against esophageal cancer.
Collapse
Affiliation(s)
- Shiming He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chunyan Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hongyu Tao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Weijin Sheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ruijuan Gao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yongsu Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
He S, Xu J, Liu X, Zhen Y. Advances and challenges in the treatment of esophageal cancer. Acta Pharm Sin B 2021; 11:3379-3392. [PMID: 34900524 PMCID: PMC8642427 DOI: 10.1016/j.apsb.2021.03.008] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/24/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Esophageal cancer (EC) is one of the most common cancers with high morbidity and mortality rates. EC includes two histological subtypes, namely esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). ESCC primarily occurs in East Asia, whereas EAC occurs in Western countries. The currently available treatment strategies for EC include surgery, chemotherapy, radiation therapy, molecular targeted therapy, and combinations thereof. However, the prognosis remains poor, and the overall five-year survival rate is very low. Therefore, achieving the goal of effective treatment remains challenging. In this review, we discuss the latest developments in chemotherapy and molecular targeted therapy for EC, and comprehensively analyze the application prospects and existing problems of immunotherapy. Collectively, this review aims to provide a better understanding of the currently available drugs through in-depth analysis, promote the development of new therapeutic agents, and eventually improve the treatment outcomes of patients with EC.
Collapse
Affiliation(s)
- Shiming He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Jian Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Yongsu Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
5
|
Time series expression pattern of key genes reveals the molecular process of esophageal cancer. Biosci Rep 2021; 40:222161. [PMID: 32068233 PMCID: PMC7048673 DOI: 10.1042/bsr20191985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/24/2019] [Accepted: 12/16/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Esophageal cancer is one of the most poorly diagnosed and fatal cancers in the world. Although a series of studies on esophageal cancer have been reported, the molecular pathogenesis of the disease is still elusive. Aim: To investigate the molecular process of esophageal cancer comprehensively and deeply. Methods: Differential expression analysis was performed to identify differentially expressed genes (DEGs) in different stages of esophageal cancer. Then exacting gene interaction modules and hub genes were identified in module interaction network. Further, though survival analysis, methylation analysis, pivot analysis, and enrichment analysis, some important molecules and related function or pathway were identified to elucidate potential mechanism in esophageal cancer. Results: A total of 7457 DEGs and 14 gene interaction modules were identified. These module genes were significantly involved in the positive regulation of protein transport, gastric acid secretion, insulin-like growth factor receptor binding and other biological processes (BPs), as well as p53 signaling pathway, ERBB signaling pathway and epidermal growth factor receptor (EGFR) signaling pathway. Then, transcription factors (TFs) (including HIF1A) and ncRNAs (including CRNDE and hsa-mir-330-3p) significantly regulate dysfunction modules were identified. Further, survival analysis showed that GNGT2 was closely related to survival of esophageal cancer. And DEGs with strong methylation regulation ability were identified, including SST and SH3GL2. Conclusion: These works not only help us to reveal the potential regulatory factors in the development of disease, but also deepen our understanding of its deterioration mechanism.
Collapse
|
6
|
Shan CK, Du YB, Zhai XT, Wang YX, Li Y, Gong JH, Ge ZJ, Liu XJ, Zhen YS. Pingyangmycin enhances the antitumor efficacy of anti-PD-1 therapy associated with tumor-infiltrating CD8 + T cell augmentation. Cancer Chemother Pharmacol 2021; 87:425-436. [PMID: 33388950 DOI: 10.1007/s00280-020-04209-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/20/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE To investigate the antitumor efficacy of pingyangmycin (PYM) in combination with anti-PD-1 antibody and determine the capability of PYM to induce immunogenic cell death (ICD) in cancer cells. METHODS The murine 4T1 breast cancer and B16 melanoma models were used for evaluation of therapeutic efficacy of the combination of PYM with anti-PD-1 antibody. The ELISA kits were used to quantify the ICD related ATP and HMGB1 levels. The Transwell assay was conducted to determine the chemotaxis ability of THP-1 cell in vitro. The flow cytometry was used to measure reactive oxygen species level and analyze the ratio of immune cell subsets. RESULTS PYM induced ICD in murine 4T1 breast cancer and B16 melanoma cells and increased the release of nucleic acid fragments that may further promote the monocytic chemotaxis. In the 4T1 murine breast cancer model, PYM alone, anti-PD-1 antibody alone, and their combination suppressed tumor growth by 66.3%, 16.1% and 77.6%, respectively. PYM markedly enhanced the therapeutic efficacy of anti-PD-1 antibody against 4T1 breast cancer. The calculated CDI (coefficient of drug interaction) indicated synergistic effect. Evaluated by graphic analysis, the nucleated cells intensity in the femur bone marrow remained unchanged. Histopathological observations revealed no noticeable toxico-pathological changes in the lung and various organs, indicating that the PYM and anti-PD-1 antibody combination exerted enhanced efficacy at well-tolerated dosage level. By the combination treatment, a panel of immunological changes emerged. The ratio of CD3+ cells, NK cells and NKT cells increased and Tregs decreased in peripheral blood. The DCs increased in the spleen. Prominent changes occurred in tumor infiltrating lymphocytes. The ratio of CD8+ cells increased, while that of CD4+ cells decreased; however, the ratio of CD3+ cells remained unchanged, implying that certain immunological responses emerged in the tumor microenvironment. PYM alone could also increase CD8+ cells and reduce CD4+ cells in tumor infiltrating lymphocytes. CONCLUSIONS The studies indicate that PYM, as an ICD inducer with mild myelosuppression effect, may enhance the therapeutic efficacy of anti-PD-1 antibody in association with tumor infiltrating CD8+ T cell augmentation.
Collapse
Affiliation(s)
- Chuan-Kun Shan
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Bo Du
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Tian Zhai
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue-Xuan Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Hua Gong
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Juan Ge
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiu-Jun Liu
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yong-Su Zhen
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
7
|
Gong JH, Zheng YB, Zhang MR, Wang YX, Yang SQ, Wang RH, Miao QF, Liu XJ, Zhen YS. Dexamethasone enhances the antitumor efficacy of Gemcitabine by glucocorticoid receptor signaling. Cancer Biol Ther 2020; 21:332-343. [PMID: 31906826 DOI: 10.1080/15384047.2019.1702399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Gemcitabine (Gem) is currently used as the first-line therapy for liver and pancreatic cancer but has limited efficacy in most cases. Dexamethasone (Dex) have been applied as a chemoprotectant and chemosensitizer in cancer chemotherapy. This study further explored the potential of combination of Gem and Dex and tested the hypothesis that glucocorticoid receptor signaling is essential for the synergistic antitumor activity. In the HepG2 and AsPC-1 xenograft models, the combination treatment showed a significantly synergistic antitumor activity. Immunohistochemistry of post-treatment tumors showed a significant decrease in proliferation and angiogenesis as compared to either of the treatments alone. Dex alone and the combination with Gem inhibited the expression of glucocorticoid receptor. The combination of Dex and Gem showed synergistic cytotoxicity in cell lines in vitro. The antiproliferative synergism is prevented by used glucocorticoid receptor (GR) small interfering RNA, demonstrating that the glucocorticoid receptor is required for the antiproliferative synergism of Gem and Dex. The inhibition of glucocorticoid receptor signaling pathway and induction of apoptosis via activation of caspases 3, 8 and 9, PARP, contributed to the synergistic effect of this combination therapy. These results demonstrate that Dex could potentiate the antitumor efficacy of Gem. The synergistic antitumor activity of the combination of Dex and Gem was through glucocorticoid receptor signaling. Taken together, a combination of Dex and Gem shows a significant synergistic antitumor activity and lesser toxicity both in vitro and in vivo and could be a combination chemotherapy for the treatment of highly expression of glucocorticoid receptor patients.
Collapse
Affiliation(s)
- Jian-Hua Gong
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yan-Bo Zheng
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Meng-Ran Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue-Xuan Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Si-Qi Yang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rui-Hai Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qing-Fang Miao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiu-Jun Liu
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yong-Su Zhen
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Liu Q, Li P, Yang Z, Qu B, Qin C, Meng S, Fang H, Wu R, Cheng T, Yang D. Multi-stage surgery combined with radiotherapy for treatment of giant anterior chest wall keloid: A case report. Medicine (Baltimore) 2020; 99:e18886. [PMID: 31977896 PMCID: PMC7004727 DOI: 10.1097/md.0000000000018886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
RATIONALE Giant keloids often have indications for surgical resection, but postoperative reconstruction of the skin and high recurrence of keloids are a challenge for clinical treatment. This article reports a rare successful treatment of a giant keloid in the anterior chest wall by multistage surgery combined with radiotherapy, which is why this case is meaningful. PATIENT CONCERNS A 66-year-old woman presented a giant keloid with ulcerations and severe itching on the anterior chest wall. She had a history of keloid disease for more than 10 years, and had been treated by multiple operations, with no success. DIAGNOSES The patient was diagnosed as keloid based on her history and symptoms. Histopathology findings supported our diagnosis. INTERVENTIONS We successfully excised the keloid after 5 operations and 2 rounds of electron-beam radiotherapy, which was applied at 24 hours after the 4th and 5th operation. OUTCOMES There was no sign of recurrence over the follow-up period of 24 months. LESSONS The combination of multistage surgery and radiotherapy presents as a good choice for the treatment of giant keloids.
Collapse
Affiliation(s)
- Qingwu Liu
- School of Clinical Medicine, Beijing University of Chinese Medicine
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University
| | - Ping Li
- School of Clinical Medicine, Beijing University of Chinese Medicine
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University
| | - Zhishan Yang
- School of Clinical Medicine, Beijing University of Chinese Medicine
- China-Japan Friendship Hospital, Beijing, China
| | - Baoquan Qu
- School of Clinical Medicine, Beijing University of Chinese Medicine
- China-Japan Friendship Hospital, Beijing, China
| | - Chunfang Qin
- School of Clinical Medicine, Beijing University of Chinese Medicine
- China-Japan Friendship Hospital, Beijing, China
| | - Shengnan Meng
- School of Clinical Medicine, Beijing University of Chinese Medicine
- China-Japan Friendship Hospital, Beijing, China
| | - Huijuan Fang
- School of Clinical Medicine, Beijing University of Chinese Medicine
- China-Japan Friendship Hospital, Beijing, China
| | - Ruiying Wu
- School of Clinical Medicine, Beijing University of Chinese Medicine
- China-Japan Friendship Hospital, Beijing, China
| | - Tiantian Cheng
- School of Clinical Medicine, Beijing University of Chinese Medicine
- China-Japan Friendship Hospital, Beijing, China
| | - Dingquan Yang
- School of Clinical Medicine, Beijing University of Chinese Medicine
- China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
9
|
DpdtbA-Induced Growth Inhibition in Human Esophageal Cancer Cells Involved Inactivation of the p53/EGFR/AKT Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5414670. [PMID: 31354907 PMCID: PMC6636558 DOI: 10.1155/2019/5414670] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022]
Abstract
Esophageal cancer (ESC) is one of the most deadly diseases for human. p53 in most cancers, including ESC cell, is mutated, and the mutated p53 losses its original function and acquires “gain of function” that allows for promoting the hallmarks of cancer, such as antiapoptosis, metastasis, invasion, angiogenesis, and resistance to chemotherapy. Targeting p53 through either introducing wild-type or degrading mutated p53 is an important strategy in cancer therapy. Di-2,2′-pyridine ketone dithiocarbamate s-butyric acid (DpdtbA) has significant growth inhibition against gastric cancer lines in previous study. Similar action in ESC cell lines but a novel molecular mechanism was observed in the present study. The results showed that DpdtbA exhibited an excellent antiproliferative effect for ESC cell lines (IC50 ≤ 4.5 ± 0.4 μM for Kyse 450, 3.2 ± 0.6 μM for Kyse 510 cell, and 10.0 ± 0.6 μM for Kyse 150) and led to cell cycle arrest at the S phase which correlated to CDK2 downregulation. The mechanistic study suggested that growth inhibition was related to ROS-mediated apoptosis, and ROS production was due to SOD inhibition initiated by DpdtbA rather than occurrence of ferritinophagy. In addition, DpdtbA also induced a downregulation of EGFR, p53, and AKT, which hinted that mutant p53 still played a role in the regulation of its downstream targets. Further study revealed that the downregulation of p53 was through stub1- (chip-) mediated autophagic degradation rather than MDM2-mediated ubiquitination. Taken together, the DpdtbA-induced growth inhibition in a mechanism was through inactivating the p53/EGFR/AKT signal pathway.
Collapse
|
10
|
Yang Y, Tian Z, Ding Y, Li X, Zhang Z, Yang L, Zhao F, Ren F, Guo R. EGFR-Targeted Immunotoxin Exerts Antitumor Effects on Esophageal Cancers by Increasing ROS Accumulation and Inducing Apoptosis via Inhibition of the Nrf2-Keap1 Pathway. J Immunol Res 2018; 2018:1090287. [PMID: 30596104 PMCID: PMC6286775 DOI: 10.1155/2018/1090287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 09/20/2018] [Accepted: 10/30/2018] [Indexed: 12/11/2022] Open
Abstract
Previously, we developed a novel EGFR-targeted antibody (denoted as Pan), which has superior antitumor activity against EGFR-overexpressed tumors. However, it shows marginal effect on the growth of esophageal cancers. Therefore, the variable region of Pan was fused to a fragment of Pseudomonas exotoxin A (PE38) to create the immunotoxin, denoted as Ptoxin (PT). Results indicated that PT shows more effective antitumor activity as compared with Pan both on EGFR-overexpressed KYSE-450 and KYSE-150 esophageal cancer cells, especially on KYSE-450 cells. Moreover, treatment of PT induces regression of KYSE-450 tumor xenografts in nude mice. Furthermore, we investigated the potential mechanism involved in the enhanced antitumor effects of PT. Data showed that PT was more potent in reducing the phosphorylation of EGFR and ERK1/2. More importantly, we for the first time found that PT was more effective than Pan in inducing ROS accumulation by suppression of the Nrf2-Keap1 antioxidant pathway, and then induced apoptosis in KYSE-450 esophageal cancer cells, which may partly explain the more sensitive response of KYSE-450 to PT treatment. To conclude, our study provides a promising therapeutic approach for immunotoxin-based esophageal cancer treatment.
Collapse
Affiliation(s)
- Yun Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai, China
| | - Ziyin Tian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yanke Ding
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiaojing Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Ziheng Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Liu Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Fangyu Zhao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Feng Ren
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Rui Guo
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
11
|
Zhu H, Wang C, Wang J, Chen D, Deng J, Deng J, Fan J, Badakhshi H, Huang X, Zhang L, Cai J, Guo S, Qian W, Nie Y, Li Q, Zhao K. A subset of esophageal squamous cell carcinoma patient-derived xenografts respond to cetuximab, which is predicted by high EGFR expression and amplification. J Thorac Dis 2018; 10:5328-5338. [PMID: 30416780 DOI: 10.21037/jtd.2018.09.18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Epidermal growth factor receptor (EGFR) is reportedly overexpressed in most esophageal tumors, but most targeted therapies showed no efficacy in non-selected patients. This study aims at investigating the adaptive cetuximab subset in a cohort of esophageal squamous cell carcinoma (ESCC) patient-derived xenografts (PDXs). Methods A large panel of ESCC PDXs has been established. The copy number, mRNA expression and immunohistochemistry (IHC) of key EGFR pathways have been examined along with cetuximab response. A preclinical trial on a randomly selected cohort of 16 ESCC PDXs was conducted, and the genomic annotations of these models were compared against the efficacy readout of the mouse trial. Results The trial identified that 7 of 16 (43.8%) responded to cetuximab (ΔT/ΔC <0 as responders). The gene amplification and expression analysis indicated that EGFR copy number ≥5 (P=0.035), high EGFR mRNA expression (P=0.001) and IHC score of 2-3 (P=0.034) are associated with tumor growth inhibition by cetuximab, suggesting EGFR may function as a single predictive biomarker for cetuximab response in ESCC. Conclusions Overall, our results suggest that an ESCC subtype with EGFR amplification and overexpression benefits from cetuximab treatment, which warrants further clinical confirmation.
Collapse
Affiliation(s)
- Hanting Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chunyu Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | - Dawei Chen
- Crown Bioscience, Inc., San Diego, CA, USA
| | - Jiaying Deng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | - Jianhong Fan
- Department of Gynaecology, Renhe Hospital, Shanghai 200431, China
| | - Harun Badakhshi
- Department of Radiation Oncology, Charité School of Medicine and Centre for Cancer Medicine, Berlin, Germany
| | | | | | - Jie Cai
- Crown Bioscience, Inc., San Diego, CA, USA
| | - Sheng Guo
- Crown Bioscience, Inc., San Diego, CA, USA
| | - Wubin Qian
- Crown Bioscience, Inc., San Diego, CA, USA
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digest Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Qixiang Li
- Crown Bioscience, Inc., San Diego, CA, USA.,State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Cai Y, Sun R, He KF, Zhao YF, Zhao JH. Sclerotherapy for the recurrent granulomatous epulis with pingyangmycin. Med Oral Patol Oral Cir Bucal 2017; 22:e214-e218. [PMID: 28160580 PMCID: PMC5359704 DOI: 10.4317/medoral.21422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 12/15/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Relapse of granulomatous epulis is common after surgery because of local irritations, hormonal level in vivo, or incomplete resection. Currently, if recurrence occurs, then extraction of the teeth adjacent to the lesion is commonly performed, which may influence the aesthetics or masticatory function. Thus, a more effective and less aggressive treatment method is urgently demanded, particularly for the recurring lesion. This study investigated the effects of the intralesional pingyangmycin (PYM) injections for the recurrent granulomatous epulis and assessed the complications. MATERIAL AND METHODS A total of 16 patients with recurrent granulomatous epulis underwent intralesional PYM injections, between July 2010 and June 2014. The effects and complications of the treatment were retrospectively reviewed. RESULTS The total number of injections performed was 48 (for all patients). The median number of injections per patient was three (range, two to four). All cases completely recovered with no recurrence and resorption of the alveolar bone after a follow-up of more than 12 months. The complications included slight bleeding, local swelling and pain following injection. All these symptoms resolved 7 to 10 days after the injection. CONCLUSIONS In summary, intralesional PYM injections may be a preferred option for recurring granulomatous epulis.
Collapse
Affiliation(s)
- Y Cai
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, 237, LuoYu Road, Wuhan, 430079, P. R. China,
| | | | | | | | | |
Collapse
|
13
|
He Y, Lan Y, Liu Y, Yu H, Han Z, Li X, Zhang L. Pingyangmycin and Bleomycin Share the Same Cytotoxicity Pathway. Molecules 2016; 21:molecules21070862. [PMID: 27376254 PMCID: PMC6274306 DOI: 10.3390/molecules21070862] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/13/2016] [Accepted: 06/15/2016] [Indexed: 01/22/2023] Open
Abstract
Pingyangmycin is an anticancer drug known as bleomycin A5 (A5), discovered in the Pingyang County of Zhejiang Province of China. Bleomycin (BLM) is a mixture of mainly two compounds (A2 and B2), which is on the World Health Organization’s list of essential medicines. Both BLM and A5 are hydrophilic molecules that depend on transporters or endocytosis receptors to get inside of cells. Once inside, the anticancer activities rely on their abilities to produce DNA breaks, thus leading to cell death. Interestingly, the half maximal inhibitory concentration (IC50) of BLMs in different cancer cell lines varies from nM to μM ranges. Different cellular uptake, DNA repair rate, and/or increased drug detoxification might be some of the reasons; however, the molecules and signaling pathways responsible for these processes are largely unknown. In the current study, we purified the A2 and B2 from the BLM and tested the cytotoxicities and the molecular mechanisms of each individual compound or in combination with six different cell lines, including a Chinese hamster ovary (CHO) cell line defective in glycosaminoglycan biosynthesis. Our data suggested that glycosaminoglycans might be involved in the cellular uptake of BLMs. Moreover, both BLM and A5 shared similar signaling pathways and are involved in cell cycle and apoptosis in different cancer cell lines.
Collapse
Affiliation(s)
- Yanli He
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Ying Lan
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Yong Liu
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Haibo Yu
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China.
| | - Zhangrun Han
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Xiulian Li
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Lijuan Zhang
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
14
|
Cort A, Ozben T, Melchiorre M, Chatgilialoglu C, Ferreri C, Sansone A. Effects of bleomycin and antioxidants on the fatty acid profile of testicular cancer cell membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:434-41. [PMID: 26656160 DOI: 10.1016/j.bbamem.2015.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/01/2015] [Accepted: 12/03/2015] [Indexed: 12/21/2022]
Abstract
Bleomycin is used in chemotherapy regimens for the treatment of patients having testicular germ-cell tumor (TGCT). There is no study in the literature investigating the effects of bleomycin on membrane lipid profile in testicular cancer cells. We investigated membrane fatty acid (FA) profiles isolated, derivatized and analyzed by gas chromatography of NTera-2 testicular cancer cells incubated with bleomycin (Bleo) for 24 h in the absence and presence of N-Acetyl-L-Cysteine (NAC) and curcumin (Cur) as commonly used antioxidant adjuvants. At the same time the MAPK pathway and EGFR levels were followed up. Bleomycin treatment increased significantly saturated fatty acids (SFA) of phospholipids at the expense of monounsaturated (MUFA) and polyunsaturated fatty acids (PUFA). Bleomycin also led to a significant increase in the trans lipid isomers of oleic and arachidonic acids due to its free radical producing effect. Incubation with bleomycin increased the p38 MAPK and JNK levels and downregulated EGFR pathway. Coincubation of bleomycin with NAC reversed effects caused by bleomycin. Our results highlight the important role of membrane fatty acid remodeling occurring during the use of bleomycin and its concurrent use with antioxidants which can adjuvate the cytotoxic effects of the chemotherapeutic agents.
Collapse
Affiliation(s)
- A Cort
- Department of Medical Biochemistry, School of Medicine, Faculty of Health Sciences SANKO University, Gaziantep, Turkey; Department of Nutrition and Dietetics, Faculty of Health Sciences SANKO University, Gaziantep, Turkey; Institute for the Organic Synthesis and Photoreactivity, Consiglio Nazionale delle Ricerche, Bologna, Italy; Akdeniz University, Medical Faculty, Department of Biochemistry, Antalya, Turkey
| | - T Ozben
- Akdeniz University, Medical Faculty, Department of Biochemistry, Antalya, Turkey.
| | - M Melchiorre
- Institute for the Organic Synthesis and Photoreactivity, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - C Chatgilialoglu
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", Aghia Paraskevi, Athens, Greece
| | - C Ferreri
- Institute for the Organic Synthesis and Photoreactivity, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - A Sansone
- Institute for the Organic Synthesis and Photoreactivity, Consiglio Nazionale delle Ricerche, Bologna, Italy
| |
Collapse
|
15
|
Gao R, Li L, Shang B, Zhao C, Sheng W, Li D. A Gelatinases-targeting scFv-based Fusion Protein Shows Enhanced Antitumour Activity with Endostar against Hepatoma. Basic Clin Pharmacol Toxicol 2015; 117:105-16. [PMID: 25615234 DOI: 10.1111/bcpt.12379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 01/09/2015] [Indexed: 12/26/2022]
Abstract
Gelatinases play important roles in tumour invasion and metastasis and are thus considered promising targets for cancer therapy. In this study, a new single-chain variable fragment (scFv)-based fusion protein Fv-LDP, composed of the anti-gelatinases scFv and lidamycin apoprotein (LDP), was prepared, and its combination with angiogenesis inhibitor Endostar was then investigated. The fusion protein Fv-LDP specifically bound to various tumour cells, and its binding capability to human pulmonary giant cell carcinoma (PG) cells was higher than that of LDP. Fv-LDP inhibited the expression and secretion of gelatinases and could be internalized into tumour cells via endocytosis. Fv-LDP also suppressed the growth of human hepatoma cells and murine hepatoma 22 transplanted in Kunming mice in various degrees. In addition, Endostar could enhance the synergistic or additive inhibition of Fv-LDP on the growth, migration or invasion of human hepatoma cells shown by a colony formation assay and a transwell-based migration or invasion assay, respectively. In vivo, Fv-LDP/Endostar combination showed a significantly synergistic effect on the growth of a human hepatoma xenograft, with an inhibition rate of 80.8% compared with the Fv-LDP (44.1%) or Endostar (8.9%)-treated group. The above-mentioned results indicate that the fusion protein Fv-LDP is effective against transplantable hepatoma in mice and human hepatoma xenografts in athymic mice. Moreover, Endostar can potentiate the inhibition effect of Fv-LDP on the growth of human hepatoma cells and xenografts. These data will provide a new combined strategy for improving the therapeutic efficacy of treatments for hepatoma or other gelatinase-overexpressing tumours.
Collapse
Affiliation(s)
- Ruijuan Gao
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Liang Li
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Boyang Shang
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chunyan Zhao
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Weijin Sheng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Diandong Li
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Li XQ, Ouyang ZG, Zhang SH, Liu H, Shang Y, Li Y, Zhen YS. Synergy of enediyne antibiotic lidamycin and temozolomide in suppressing glioma growth with potentiated apoptosis induction. J Neurooncol 2014; 119:91-100. [PMID: 24842385 DOI: 10.1007/s11060-014-1477-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 04/30/2014] [Indexed: 12/15/2022]
Abstract
The present work evaluated the synergistic efficacy of an enediyne antibiotic lidamycin (LDM) plus temozolomide (TMZ) against glioma in vitro and in vivo. LDM plus TMZ inhibited the proliferations of rat glioma C6 cells and human glioma U87 cells more efficiently than the single usage of LDM or TMZ. In addition, LDM also potentiated the apoptosis inductions by TMZ in rat C6 cells and human U87 cells. Meanwhile, the results of TdT-mediated dUTP Nick End Labeling assay for subcutaneous U87 tumor sections indicated an enhanced apoptosis induction in vivo by LDM plus TMZ, which confirmed the high potency of the combination for glioma therapy. As determined by Western blot, apoptosis signal pathways in C6 cells and U87 cells were markedly affected by the synergistic alteration of P53, bax, procaspase 3, and bcd-2 expression. In both subcutaneous U87 xenograft and C6 intracerebral orthotopic implant model, TMZ-induced glioma growth suppression was dramatically potentiated by LDM. As shown, the combination therapy efficiently reduced the tumor volumes and tumor weights of the human glioma U87 xenograft. Kaplan-Meier assay revealed that LDM plus TMZ dramatically prolonged the life span of C6 intracerebral tumor-bearing rats with decreased tumor size. This study indicates that the combination of LDM with TMZ might be a promising strategy for glioma therapy.
Collapse
Affiliation(s)
- Xing-Qi Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Tu JB, Li QY, Jiang F, Hu XY, Ma RZ, Dong Q, Zhang H, Pattar P, Li SX. Pingyangmycin stimulates apoptosis in human hemangioma-derived endothelial cells through activation of the p53 pathway. Mol Med Rep 2014; 10:301-5. [PMID: 24789513 DOI: 10.3892/mmr.2014.2174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 03/17/2014] [Indexed: 11/05/2022] Open
Abstract
Pingyangmycin (also known as Bleomycin A5) is produced by Streptomyces verticillus var. pingyangensis n.sp., and has anti‑tumor activities against a variety of tumor cells. The aim of the present study was to determine the molecular mechanism(s) underlying the therapeutic effects of pingyangmycin against infantile hemangiomas. Human hemangioma‑derived endothelial cells (HemECs) were treated with pingyangmycin at varying concentrations (100, 200 or 300 µg/ml), and the morphological changes and apoptosis levels were assessed. The gene expression changes were determined by cDNA microarray technology. Transmission electron microscopy examination revealed that the pingyangmycin‑treated HemECs exhibited typical apoptotic characteristics, including chromatin condensation and the formation of apoptotic bodies. Annexin‑V staining demonstrated that pingyangmycin caused a significant and dose‑dependent induction of apoptosis in the HemECs. In the pingyangmycin‑treated HemECs, 4,752 genes demonstrated at least 2‑fold expression changes at the mRNA level. Quantitative polymerase chain reaction confirmed that pingyangmycin significantly upregulated the expression of p53, p53‑induced protein with death domain, Bax, p53 upregulated modulator of apoptosis and p53 inducible gene 3, and downregulated the expression of murine double minute 2. The data demonstrated that the pro‑apoptotic activity of pingyangmycin against infantile hemangiomas involves p53 pathway activation.
Collapse
Affiliation(s)
- Jun-Bo Tu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Quan-Yan Li
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Fei Jiang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiao-Yi Hu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Rui-Zhao Ma
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Qiang Dong
- Department of Oral and Maxillofacial Surgery, Weihai Stomatological Hospital, Weihai, Shandong 264200, P.R. China
| | - Hao Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Parukjan Pattar
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shi-Xian Li
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
18
|
Qi W, Guo J, Wu S, Su B, Zhang L, Pan J, Zhang J. Synergistic effect of nanosecond pulsed electric field combined with low-dose of pingyangmycin on salivary adenoid cystic carcinoma. Oncol Rep 2014; 31:2220-8. [PMID: 24604118 DOI: 10.3892/or.2014.3063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/29/2014] [Indexed: 11/05/2022] Open
Abstract
Adenoid cystic carcinoma (ACC) is one of the most common malignant neoplasms in salivary glands. To evaluate the therapeutic effects of nanosecond pulsed electric field (nsPEF) combined with pingyangmycin (PYM) on salivary gland adenoid cystic carcinoma (SACC), ACC high metastatic cell line (SACC-LM) and low metastatic cell line (SACC‑83) were tested by CCK-8 assay, cell clonogenic assay, flow cytometry and Transwell assay. Extracellular matrix metalloproteinase inducer (EMMPRIN) expression was tested by western blotting to verify the synergistic mechanism of nsPEF and PYM. The results showed that nsPEF inhibited the cell proliferation of both cell lines, and the inhibitory effect was strongly associated with time and electrical field strength. Moreover, PYM combined with nsPEF may enhance the suppression effect significantly, even at a very low dose (0.01 µg/ml). The synergistic effects may contribute to the downregulation of EMMPRIN expression resulting from the application of nsPEF. For SACC, nsPEF combined with chemotherapy agents may be a valuable strategy not only to improve the treatment effect and prognosis, but also to reduce the side-effects of chemotherapy.
Collapse
Affiliation(s)
- Wei Qi
- Department of General Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Jinsong Guo
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Shan Wu
- College of Engineering, Peking University, Beijing 100871, P.R. China
| | - Bo Su
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Lei Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Jie Pan
- Department of General Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Jue Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| |
Collapse
|
19
|
Li B, Zheng YB, Li DD, Zhen YS. Preparation and evaluation of a CD13/APN-targeting and hydrolase-resistant conjugate that comprises pingyangmycin and NGR motif-integrated apoprotein. J Pharm Sci 2014; 103:1204-13. [PMID: 24504597 DOI: 10.1002/jps.23893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/31/2013] [Accepted: 01/17/2014] [Indexed: 11/10/2022]
Abstract
We have chemically synthesized NGR-LDP-PYM, a novel CD13/aminopeptidase (APN)-targeting and hydrolase-resistant conjugate by cross-linking of the antitumor antibiotic pingyangmycin (bleomycin A5 , PYM) to an engineered NGR motif-integrated apoprotein (NGR-LDP) with a noncleavable linker. This protein-drug conjugate not only basically retains the original properties of PYM but also can specifically deliver PYM to the CD13/APN-expressing tumor cells. Furthermore, the resulting conjugate exhibits more resistance to hydrolysis of recombinant human bleomycin hydrolase than parental PYM. These results may be useful for improving the therapeutic efficacy of PYM and have implications in the treatment of PYM-refractory and CD13/APN-overexpressing tumors.
Collapse
Affiliation(s)
- Bin Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | | | | | | |
Collapse
|
20
|
Li XQ, Ouyang ZG, Zhang SH, Liu H, Shang Y, Li Y, Zhen YS. Synergistic inhibition of angiogenesis and glioma cell-induced angiogenesis by the combination of temozolomide and enediyne antibiotic lidamycin. Cancer Biol Ther 2014; 15:398-408. [PMID: 24424202 DOI: 10.4161/cbt.27626] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Present work mainly evaluated the inhibitory effects of lidamycin (LDM), an enediyne antibiotic, on angiogenesis or glioma-induced angiogenesis in vitro and in vivo, especially its synergistic anti-angiogenesis with temozolomide (TMZ). LDM alone efficiently inhibited proliferations and induced apoptosis of rat brain microvessel endothelial cells (rBMEC). LDM also interrupted the tube formation of rat brain microvessel endothelial cells (rBMEC) and rat aortic ring spreading. The blockade of rBMEC invasion and C6 cell-induced rBMEC migration by LDM was associated with decrease of VEGF secretion in a co-culture system. TMZ dramatically potentiated the effects of LDM on anti-proliferation, apoptosis induction, and synergistically inhibited angiogenesis events. As determined by western blot and ELISA, the interaction of tumor cells and the rBMEC was markedly interrupted by LDM plus TMZ with synergistic regulations of VEGF induced angiogenesis signal pathway, tumor cell invasion/migration, and apoptosis signal pathway. Immunofluorohistochemistry of CD31 and VEGF showed that LDM plus TMZ resulted in synergistic decrease of microvessel density (MVD) and VEGF expression in human glioma U87 cell subcutaneous xenograft. This study indicates that the high efficacy of LDM and the synergistic effects of LDM plus TMZ against glioma are mediated, at least in part, by the potentiated anti-angiogenesis.
Collapse
Affiliation(s)
- Xing-Qi Li
- College of Life Science & Technology; Heilongjiang Bayi Agricultural University; Daqing, PR China; Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Zhi-Gang Ouyang
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Sheng-Hua Zhang
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Hong Liu
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Yue Shang
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Yi Li
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| | - Yong-Su Zhen
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing, PR China
| |
Collapse
|
21
|
Measurement of downstream kinase activity modulation as indicator of epidermal growth factor receptor inhibitor efficacy. Anal Biochem 2013; 448:65-7. [PMID: 24316115 DOI: 10.1016/j.ab.2013.11.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 11/19/2013] [Accepted: 11/28/2013] [Indexed: 12/28/2022]
Abstract
The acoustic membrane micro particle (AMMP) technology has been used to quantify single analytes out of multiple sample types. In this study the technology is used to reveal molecular interactions of components of kinase pathways. Specifically, the downstream kinase activity of the EGFR receptor in the presence or absence of EGFR inhibitors is investigated. These experiments substantiate that EGFR stimulation predominantly activates the MEK/ERK pathway. The EGFR inhibitors tested had varying effectiveness at preventing phosphorylation at the EGFR or downstream kinase activity. These experiments reveal the use of the AMMP technology for observing multiple signaling pathways in a single experiment.
Collapse
|
22
|
Huang Y, Li P, Xia S, Zhuo Y, Wu L. Proapoptotic effect and the mechanism of action of pingyangmycin on cavernous hemangiomas. Exp Ther Med 2013; 7:473-477. [PMID: 24396428 PMCID: PMC3881047 DOI: 10.3892/etm.2013.1428] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 11/07/2013] [Indexed: 12/12/2022] Open
Abstract
This study aimed to investigate the proapoptotic effects and the mechanism of action of pingyangmycin (PY) on cavernous hemangioma. The rat spleen was used as a model of cavernous hemangioma. PY was injected into the spleen and the pathological changes were observed at different time-points. Apoptosis was detected using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and transmission electron microscopy (TEM). The expression levels of the apoptosis-related protein, caspase-3, were determined using immunohistochemistry and image analysis. Rats injected with normal saline were the control group. Injection of normal saline did not damage rat spleens. On days 2 and 5 following PY injection, the spleens exhibited slight swelling. On days 8 and 14, atrophic changes were observed and the splenic sinus endothelial cells were damaged. At various time-points following PY injection, the apoptotic cells were observed by TEM. The TUNEL assay showed that apoptosis occurred widely among the splenic sinus endothelial cells and other splenic cells. The apoptotic rate and caspase-3 expression levels increased with prolonged PY exposure. PY induced apoptosis of splenic sinus endothelial cells through the caspase-3 activation pathway, and resulted in endothelial cell necrosis and fibroblast hyperplasia.
Collapse
Affiliation(s)
- Yideng Huang
- Department of Otorhinolaryngology, The 118th Hospital of Chinese PLA, Wenzhou, Zhejiang 325000, P.R. China
| | - Ping Li
- Department of Otorhinolaryngology, General Hospital of Chengdu Military Region of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Siwen Xia
- Department of Otorhinolaryngology, The 118th Hospital of Chinese PLA, Wenzhou, Zhejiang 325000, P.R. China
| | - Yang Zhuo
- Department of Otorhinolaryngology, The 118th Hospital of Chinese PLA, Wenzhou, Zhejiang 325000, P.R. China
| | - Longjun Wu
- Department of Otorhinolaryngology, The 118th Hospital of Chinese PLA, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
23
|
Zhang SH, Zhang H, He HW, Li L, Li XQ, Zhang YP, Shao RG. Lidamycin up-regulates the expression of thymidine phosphorylase and enhances the effects of capecitabine on the growth and pulmonary metastases of murine breast carcinoma. Cancer Chemother Pharmacol 2013; 72:777-88. [DOI: 10.1007/s00280-013-2253-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/26/2013] [Indexed: 12/27/2022]
|
24
|
Hong L, Han Y, Brain L. Epidermal growth factor receptor: an important target in esophageal cancer. Expert Opin Ther Targets 2013; 17:1179-85. [PMID: 23855932 DOI: 10.1517/14728222.2013.820709] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Even after complete tumor removal by surgery, the clinical outcomes remain poor in patients with advanced esophageal cancer, justifying the need for new treatment options. Epidermal growth factor receptor (EGFR) is a molecular target for antibody-based therapy in various cancer types, and it may play important roles in the development of esophageal cancer. AREAS COVERED This review evaluates the expression, function, and mechanism of EGFR in esophageal cancer and analyzes its value for the prognosis and therapy of esophageal cancer. Future developments toward the clinical applications of EGFR to cancer treatment are also envisaged. EXPERT OPINION EGFR may function as an ideal therapeutic target for esophageal cancer. Further investigation of epidermal growth-factor-receptor-mediated pathways will push insight into the novel strategies of target therapy for esophageal cancer. More clinical trials should be performed to promote the success of therapeutic-clinical use of EGFR and its targets in esophageal cancer.
Collapse
Affiliation(s)
- Liu Hong
- Fourth Military Medical University, Xijing Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Cancer Biology , Xi'an, 710032, Shaanxi Province , China +86 29 84773974 ; +86 29 82539041 ;
| | | | | |
Collapse
|
25
|
Cai Y, Wang R, Yang S, Zhao Y, Zhao J. Sclerotherapy for the mucoceles of the anterior lingual salivary glands with pingyangmycin. Oral Dis 2013; 20:473-6. [PMID: 23848975 DOI: 10.1111/odi.12155] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 06/11/2013] [Accepted: 06/19/2013] [Indexed: 01/21/2023]
Affiliation(s)
- Y Cai
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedical Engineering of Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Wuhan University Wuhan China
| | - R Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedical Engineering of Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
| | - S‐F Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedical Engineering of Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Wuhan University Wuhan China
| | - Y‐F Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedical Engineering of Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Wuhan University Wuhan China
| | - J‐H Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory of Oral Biomedical Engineering of Ministry of Education School & Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Wuhan University Wuhan China
| |
Collapse
|