201
|
Xu J, Bai S, Cao Y, Liu L, Fang Y, Du J, Luo L, Chen M, Shen B, Zhang Q. miRNA-221-3p in Endothelial Progenitor Cell-Derived Exosomes Accelerates Skin Wound Healing in Diabetic Mice. Diabetes Metab Syndr Obes 2020; 13:1259-1270. [PMID: 32368119 PMCID: PMC7183783 DOI: 10.2147/dmso.s243549] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Patients with diabetic cutaneous ulcers experience financial burden and a lower quality of life and life expectancy. Endothelial progenitor cell (EPC)-derived exosomes facilitate skin wound healing by positively modulating vascular endothelial cell function. Exosomes play their important regulatory role through microRNA (miRNA). We explored the potential role and molecular mechanisms of miRNA in EPC-derived exosome healing of diabetic skin wounds. METHODS Exosomes were isolated from the media of EPCs derived from mice bone marrow. High-throughput sequencing was used to detect the expression of exosome miRNA, and miRNA target genes were predicted using online databases. A diabetic mouse skin wound model was established, and wounds were treated with exosomes, miRNA-221-3p, or phosphate-buffered saline. RESULTS Exosomes from EPCs accelerated skin wound healing in both control and diabetic mice. High-throughput sequencing showed that miRNA-221-3p was highly expressed in EPC-derived exosomes. Skin wound healing in control and diabetic mice was significantly enhanced by EPC-derived exosomes and miRNA-221-3p administration. Immunohistochemical analyses showed that EPC-derived exosomes and miRNA-221-3p increased protein expression levels of the angiogenesis-related factors VEGF, CD31 and cell proliferation marker Ki67. Bioinformatics analyses indicated that miRNA-221-3p may be involved in the AGE-RAGE signaling pathway in diabetic complications, cell cycle, and the p53 signaling pathway. CONCLUSION We concluded that miRNA-221-3p is one of the high-expressed miRNAs in EPC-derived exosomes and promoted skin wound healing in diabetic mice. The finding uncovers the molecular mechanism of EPC-derived exosomes and provides a potential novel approach to the clinical treatment of diabetic skin wounds.
Collapse
Affiliation(s)
- Juan Xu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui230022, People’s Republic of China
| | - Suwen Bai
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui230032, People’s Republic of China
| | - Yadi Cao
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui230022, People’s Republic of China
| | - Lei Liu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui230022, People’s Republic of China
| | - Yang Fang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui230032, People’s Republic of China
| | - Juan Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui230032, People’s Republic of China
| | - Li Luo
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui230022, People’s Republic of China
| | - Mingwei Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui230022, People’s Republic of China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui230032, People’s Republic of China
- Bing Shen Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui230022, People’s Republic of ChinaTel +86-551-65161132Fax +86-551-65161126 Email
| | - Qiu Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui230022, People’s Republic of China
- Correspondence: Qiu Zhang Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui230022, People’s Republic of China Tel/Fax +86-551-62923631 Email
| |
Collapse
|
202
|
Schoninger SF, Blain SW. The Ongoing Search for Biomarkers of CDK4/6 Inhibitor Responsiveness in Breast Cancer. Mol Cancer Ther 2020; 19:3-12. [PMID: 31909732 PMCID: PMC6951437 DOI: 10.1158/1535-7163.mct-19-0253] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/02/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022]
Abstract
CDK4 inhibitors (CDK4/6i), such as palbociclib, ribociclib, and abemaciclib, are approved in combination with hormonal therapy as a front-line treatment for metastatic HR+, HER2- breast cancer. Their targets, CDK4 and CDK6, are cell-cycle regulatory proteins governing the G1-S phase transition across many tissue types. A key challenge remains to uncover biomarkers to identify those patients that may benefit from this class of drugs. Although CDK4/6i addition to estrogen modulation therapy essentially doubles the median progression-free survival, overall survival is not significantly increased. However, in reality only a subset of treated patients respond. Many patients exhibit primary resistance to CDK4/6 inhibition and do not derive any benefit from these agents, often switching to chemotherapy within 6 months. Some patients initially benefit from treatment, but later develop secondary resistance. This highlights the need for complementary or companion diagnostics to pinpoint patients who would respond. In addition, because CDK4 is a bona fide target in other tumor types where CDK4/6i therapy is currently in clinical trials, the lack of target identification may obscure benefit to a subset of patients there as well. This review summarizes the current status of CDK4/6i biomarker test development, both in clinical trials and at the bench, with particular attention paid to those which have a strong biological basis as well as supportive clinical data.
Collapse
Affiliation(s)
| | - Stacy W Blain
- Departments of Pediatrics and Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York.
| |
Collapse
|
203
|
Downregulation of class II phosphoinositide 3-kinase PI3K-C2β delays cell division and potentiates the effect of docetaxel on cancer cell growth. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:472. [PMID: 31752944 PMCID: PMC6873561 DOI: 10.1186/s13046-019-1472-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023]
Abstract
Background Alteration of signalling pathways regulating cell cycle progression is a common feature of cancer cells. Several drugs targeting distinct phases of the cell cycle have been developed but the inability of many of them to discriminate between normal and cancer cells has strongly limited their clinical potential because of their reduced efficacy at the concentrations used to limit adverse side effects. Mechanisms of resistance have also been described, further affecting their efficacy. Identification of novel targets that can potentiate the effect of these drugs or overcome drug resistance can provide a useful strategy to exploit the anti-cancer properties of these agents to their fullest. Methods The class II PI3K isoform PI3K-C2β was downregulated in prostate cancer PC3 cells and cervical cancer HeLa cells using selective siRNAs and the effect on cell growth was determined in the absence or presence of the microtubule-stabilizing agent/anti-cancer drug docetaxel. Mitosis progression was monitored by time-lapse microscopy. Clonogenic assays were performed to determine the ability of PC3 and HeLa cells to form colonies upon PI3K-C2β downregulation in the absence or presence of docetaxel. Cell multi-nucleation was assessed by immunofluorescence. Tumour growth in vivo was assessed using a xenograft model of PC3 cells upon PI3K-C2β downregulation and in combination with docetaxel. Results Downregulation of PI3K-C2β delays mitosis progression in PC3 and HeLa cells, resulting in reduced ability to form colonies in clonogenic assays in vitro. Compared to control cells, PC3 cells lacking PI3K-C2β form smaller and more compact colonies in vitro and they form tumours more slowly in vivo in the first weeks after cells implant. Stable and transient PI3K-C2β downregulation potentiates the effect of low concentrations of docetaxel on cancer cell growth. Combination of PI3K-C2β downregulation and docetaxel almost completely prevents colonies formation in clonogenic assays in vitro and strongly inhibits tumour growth in vivo. Conclusions These data reveal a novel role for the class II PI3K PI3K-C2β during mitosis progression. Furthermore, data indicate that blockade of PI3K-C2β might represent a novel strategy to potentiate the effect of docetaxel on cancer cell growth.
Collapse
|
204
|
Pharmacological basis and new insights of quercetin action in respect to its anti-cancer effects. Biomed Pharmacother 2019; 121:109604. [PMID: 31733570 DOI: 10.1016/j.biopha.2019.109604] [Citation(s) in RCA: 346] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/14/2019] [Accepted: 10/26/2019] [Indexed: 12/23/2022] Open
Abstract
Quercetin is a kind of flavonoid compounds that comes from nature and is widely existed in the daily diet. Previous studies have found that quercetin has many effects such as anti-inflammatory, anti-oxidation and anti-cancer. Both in vivo and in vitro experiments have demonstrated that quercetin can exert anti-tumor effects by altering cell cycle progression, inhibiting cell proliferation, promoting apoptosis, inhibiting angiogenesis and metastasis progression, and affecting autophagy. This review summarizes the evidence for the pharmacological potential and inhibition of quercetin on cancers, supporting the viewpoint that quercetin should be adequately considered as a therapeutic agent against various cancers.
Collapse
|
205
|
Zhong Y, Jiang L, Long X, Zhou Y, Deng S, Lin H, Li X. Clinical Significance And Integrative Analysis Of Kinesin Family Member 18B In Lung Adenocarcinoma. Onco Targets Ther 2019; 12:9249-9264. [PMID: 31807007 PMCID: PMC6842302 DOI: 10.2147/ott.s227438] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/03/2019] [Indexed: 12/14/2022] Open
Abstract
Background Kinesin family member 18B (KIF18B) is a member of the kinesin-8 superfamily, and functions as an oncogene in human cancers. However, its expression profile and role in lung adenocarcinoma (LUAD) remain unclear. Materials and methods We examined the expression profile of KIF18B using quantitative real-time reverse transcription polymerase chain reaction and immunohistochemistry in fresh clinical samples. Using data downloaded from the Cancer Genome Atlas database and Gene Expression Omnibus, we explored the clinical significance of KIF18B, potential mechanisms of its dysregulation and its underlying biological function in LUAD. Results KIF18B was significantly over-expressed in LUAD tissues relative to normal tissues. High KIF18B expression was associated with smoking history, positive nodal invasion, advanced clinical stage, death status and poorer prognosis. Cox regression analyses revealed that KIF18B overexpression was an independent prognostic biomarker for poor overall survival (OS) and recurrence-free survival in LUAD. In addition, KIF18B mutation was observed in 2.2% of LUAD cases. DNA copy number variation was correlated with upregulated expression of KIF18B in LUAD tissues and cell lines. The methylation level of some KIF18B DNA CpG sites was negatively associated with its mRNA expression. KIF18B was predictively targeted by miR-125a-5p, which was downregulated in LUAD tissues, inversely correlated with KIF18B mRNA expression and significantly associated with poor OS. Furthermore, gene set enrichment analysis revealed that genes positively co-expressed with KIF18B were mainly enriched in cell cycle signaling pathways. Conclusion Our results indicate that KIF18B is a promising prognostic biomarker for LUAD. DNA amplification, hypomethylation as well as miR-125a-5p downregulation may be involved in the mechanism of KIF18B dysregulation in LUAD. KIF18B might function as a novel oncogene through cell cycle regulation pathways in LUAD.
Collapse
Affiliation(s)
- Yonglong Zhong
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Lingyu Jiang
- Intensive Care Unit, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Xiaomao Long
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Yifan Zhou
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Shen Deng
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Hui Lin
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Xiangwei Li
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| |
Collapse
|
206
|
Li P, Zhang X, Gu L, Zhou J, Deng D. P16 methylation increases the sensitivity of cancer cells to the CDK4/6 inhibitor palbociclib. PLoS One 2019; 14:e0223084. [PMID: 31652270 PMCID: PMC6814222 DOI: 10.1371/journal.pone.0223084] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Abstract
The P16 (CDKN2Aink4a) gene is an endogenous CDK4/6 inhibitor. Palbociclib (PD0332991) is an anti-CDK4/6 chemical for cancer treatment. P16 is most frequently inactivated by copy number deletion and DNA methylation in cancers. It is well known that cancer cells with P16 deletion are more sensitive to palbociclib than those without. However, whether P16 methylation is related to palbociclib sensitivity is not known. By analyzing public pharmacogenomic datasets, we found that the IC50 of palbociclib in cancer cell lines (n = 522) was positively correlated with both the P16 expression level and P16 gene copy number. Our experimental results further showed that cancer cell lines with P16 methylation were more sensitive to palbociclib than those without. To determine whether P16 methylation directly increased the sensitivity of cancer cells to palbociclib, we induced P16 methylation in the lung cancer cell lines H661 and HCC827 and the gastric cancer cell line BGC823 via an engineered P16-specific DNA methyltransferase (P16-Dnmt) and found that the sensitivity of these cells to palbociclib was significantly increased. The survival rate of P16-Dnmt cells was significantly lower than that of vector control cells 48 hrs post treatment with palbociclib (10 μM). Notably, palbociclib treatment also selectively inhibited the proliferation of the P16-methylated subpopulation of P16-Dnmt cells, further indicating that P16 methylation can increase the sensitivity of cells to this CDK4/6 inhibitor. These results were confirmed in an animal experiment. In conclusion, inactivation of the P16 gene by DNA methylation can increase the sensitivity of cancer cells to palbociclib.
Collapse
Affiliation(s)
- Paiyun Li
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xuehong Zhang
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Liankun Gu
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jing Zhou
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing, China
- * E-mail:
| |
Collapse
|
207
|
Gilad Y, Eliaz Y, Yu Y, Han SJ, O'Malley BW, Lonard DM. Drug-induced PD-L1 expression and cell stress response in breast cancer cells can be balanced by drug combination. Sci Rep 2019; 9:15099. [PMID: 31641154 PMCID: PMC6805932 DOI: 10.1038/s41598-019-51537-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/28/2019] [Indexed: 12/21/2022] Open
Abstract
The impact of chemotherapy on tumor-immune system interaction can be either beneficial or harmful, which is represented by the immunogenic cell death (ICD) paradigm or overexpression of the immunosuppressive protein – programmed death ligand 1 (PD-L1). In this study we explore the impact of steroid receptor coactivator inhibitor, other targeted anti-cancer compounds and traditional chemotherapeutic agents on the expression of PD-L1 in four breast cancer (BC) cell lines. Our results show that these agents induce PD-L1 expression, yet the magnitude of this induction varies substantially across the different compounds. In addition, we utilized the E0771 ER + BC cells as a model to examine in greater detail the relationship between pharmacological pressure, cell stress and the induction of PD-L1. Our results imply that drug induced PD-L1 expression occurs in the broader context of cell-stress, without conferring acquired drug-resistance. Furthermore, a balance between BC cytotoxicity, induction of cell-stress and the overexpression of PD-L1 can be achieved through the selection of appropriate combinations of anti-cancer compounds. Therefore, we propose that drug combination can be employed not only for increasing the direct kill of cancer cells, but also as a strategy to minimize the activation of immunosuppressive and cancer cell pro-survival program responses during drug treatment.
Collapse
Affiliation(s)
- Yosi Gilad
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Tx, USA
| | - Yossi Eliaz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Tx, USA
| | - Yang Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Tx, USA
| | - Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Tx, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Tx, USA.
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Tx, USA.
| |
Collapse
|
208
|
Wang S, Hu H, Zhong B, Shi D, Qing X, Cheng C, Deng X, Zhang Z, Shao Z. Bruceine D inhibits tumor growth and stem cell-like traits of osteosarcoma through inhibition of STAT3 signaling pathway. Cancer Med 2019; 8:7345-7358. [PMID: 31631559 PMCID: PMC6885873 DOI: 10.1002/cam4.2612] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 09/19/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Patients with osteosarcoma exhibiting resistance to chemotherapy or presenting with metastasis usually have a poor prognosis. Osteosarcoma stem cells (OSCs) are a potential cause of tumor metastasis, relapse, and chemotherapy resistance. Therefore, it is necessary to develop novel therapeutic drugs, which not only kill osteosarcoma cells but also target OSCs. This study aims to explore the anti‐osteosarcoma effects of Bruceine D (BD), a natural compound derived from Brucea javanica, and investigate its underlying mechanisms. Results demonstrated that BD could significantly inhibit cell proliferation and migration, induce cell cycle arrest, and promote apoptosis in osteosarcoma cells. Besides, BD could also suppress the sphere‐forming and self‐renewal ability of OSCs. Mechanistically, the inhibitory role of BD on osteosarcoma cell growth and migration including OSC stemness was partially executed through the inhibition of STAT3 signaling pathway. More importantly, BD showed significant anti‐osteosarcoma activity without obvious side effects in vivo. Collectively, the results of this study demonstrated that BD exerts a strong inhibitory effect on tumor growth and stem cell like traits of osteosarcoma which may be partially due to STAT3 inhibition, suggesting that BD maybe a promising therapeutic candidate against osteosarcoma.
Collapse
Affiliation(s)
- Shangyu Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongzhi Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binlong Zhong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Deyao Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangcheng Qing
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyu Deng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
209
|
Balasubramanian PK, Lee Y, Kim Y. Identification of Ligand‐binding Hotspot Residues of CDK4 Using Molecular Docking and Molecular Dynamics Simulation. B KOREAN CHEM SOC 2019. [DOI: 10.1002/bkcs.11873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | - Yeongjoon Lee
- Department of Bioscience and BiotechnologyKonkuk University Seoul 05029 South Korea
| | - Yangmee Kim
- Department of Bioscience and BiotechnologyKonkuk University Seoul 05029 South Korea
| |
Collapse
|
210
|
Peng Y, Wang L, Wu L, Zhang L, Nie G, Guo M. Methylation of SLFN11 promotes gastric cancer growth and increases gastric cancer cell resistance to cisplatin. J Cancer 2019; 10:6124-6134. [PMID: 31762822 PMCID: PMC6856579 DOI: 10.7150/jca.32511] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
Abstract
Background and Aim: Human SLFN11 gene encodes a protein with structural similarity to RNA helicases, which was reported to sensitize cancer cells to DNA-damaging agents. This study explored the epigenetic regulation and mechanism of SLFN11 in human gastric cancer. Methods: Eight human gastric cancer cell lines and 201 cases of primary gastric cancer were analyzed. Methylation specific PCR, flow cytometry, xenograft mouse model and siRNA technique were employed. Results: SLFN11 was methylated in 29.9% (60/201) of primary gastric cancer. The expression of SLFN11 was regulated by promoter region methylation. Methylation of SLFN11 was significantly associated with tumor size (p < 0.05). SLFN11 suppressed gastric cancer growth both in vitro and in vivo and enhanced the ability of cisplatin to induce S-phrase arrest and apoptosis in gastric cancer cells. Conclusions: SLFN11 is frequently methylated in human gastric cancer, and its expression is regulated by promoter region methylation. Our results demonstrate that SLFN11 is a tumor suppressor in human gastric cancer, and methylation of SLFN11 may serve as a cisplatin resistant marker in human gastric cancer.
Collapse
Affiliation(s)
- Yaojun Peng
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Li Wang
- Department of Surgery, The Affiliated Cancer Hospital of Zhengzhou University, #127 Dongming Road, Zhengzhou, Henan Province 450008, China
| | - Liangliang Wu
- Department of Oncology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Ling Zhang
- Department of Surgery, The Affiliated Cancer Hospital of Zhengzhou University, #127 Dongming Road, Zhengzhou, Henan Province 450008, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, North Road No. 1, Zhongguancun, Beijing, 100190, China
| | - Mingzhou Guo
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| |
Collapse
|
211
|
Yan Y, Xu Z, Chen X, Wang X, Zeng S, Zhao Z, Qian L, Li Z, Wei J, Huo L, Li X, Gong Z, Sun L. Novel Function of lncRNA ADAMTS9-AS2 in Promoting Temozolomide Resistance in Glioblastoma via Upregulating the FUS/MDM2 Ubiquitination Axis. Front Cell Dev Biol 2019; 7:217. [PMID: 31632968 PMCID: PMC6783494 DOI: 10.3389/fcell.2019.00217] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/18/2019] [Indexed: 02/05/2023] Open
Abstract
Background LncRNAs have been shown to play essential roles in cancer therapeutic response. However, the detailed mechanism of lncRNAs in temozolomide (TMZ) resistance in glioblastoma (GBM) remain to be elucidated. Methods To elucidate the mechanism maintaining TMZ resistance, we constructed two TMZ-resistant GBM cell lines (T98G-R/U118-R). LncRNAs from four public datasets were reanalyzed, and the candidate lncRNA ADAMTS9-AS2 was evaluated in TMZ-treated GBM patients and in vitro cell lines. Results Reanalysis of lncRNA expression profiles identified ADAMTS9-AS2 as significantly overexpressed in TMZ-resistant GBM cells and as positively associated with the IC50 of TMZ in GBM cells. Overexpression of ADAMTS9-AS2 was also significantly associated with poor TMZ response and shorter progression-free survival (PFS) in TMZ-treated GBM patients. Knockdown of ADAMTS9-AS2 inhibited proliferation and attenuated the IC50 of TMZ, as well as mitigating invasion and migration in TMZ-resistant GBM cells. Subsequent investigations indicated that reduced expression of ADAMTS9-AS2 significantly suppressed expression of the FUS protein, which was predicted as a direct substrate of ADAMTS9-AS2. Expression trends of FUS were directly correlated with those of ADAMTS9-AS2, as shown by increasing concentrations and prolonged treatment with TMZ. RNA pull-down and RIP assays indicated that both endogenous and exogenous ADAMTS9-AS2 directly binds to the RRM and Znf_RanBP2 domains of FUS, consequently increasing FUS protein expression. Knockdown of ADAMTS9-AS2 reduced the half-life of FUS and decreased FUS protein stability via K48 ubiquitin degradation. Moreover, the E3 ubiquitin-protein ligase MDM2 interacts with and down regulates FUS, while the RRM and Znf_RanBP2 domains of FUS facilitate its binding with MDM2. ADAMTS9-AS2 decreased the interaction between MDM2 and FUS, which mediates FUS K48 ubiquitination. Additionally, knockdown of the ADAMTS9-AS2/FUS signaling axis significantly alleviated progression and metastasis in TMZ-resistant cells. Conclusion ADAMTS9-AS2 possessed a novel function that promotes TMZ resistance via upregulating the FUS/MDM2 axis in GBM cells. The RRM or Znf_RanBP2 domains of FUS facilitate the combination of ADAMTS9-AS2 and FUS, competitively inhibiting MDM2-dependent FUS K48 ubiquitination and resulting in enhanced FUS stability and TMZ resistance. Our results suggest that the ADAMTS9-AS2/FUS/MDM2 axis may represent a suitable prognostic biomarker and a potential target in TMZ-resistant GBM therapy.
Collapse
Affiliation(s)
- Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zijin Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Long Qian
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Li
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wei
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Huo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lunquan Sun
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
212
|
Loretan L, Moskovszky LE, Kurrer M, Exner GU, Trojan A. Efficacy of a CDK4/6 Inhibitor in a Patient with Breast Cancer and Liposarcoma: A Case Report and Review of the Literature. Breast Care (Basel) 2019; 14:325-328. [PMID: 31798393 PMCID: PMC6883449 DOI: 10.1159/000493370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The cyclin D/cyclin-dependent kinase (CDK)4/6 inhibitor of the CDK4 (INK4)/retinoblastoma (Rb) pathway plays a crucial role in cell cycle progression. Selective CDK4/6 inhibitors specifically target a variety of tumors, with the main focus on hormone receptor(HR)-positive and human epidermal growth factor receptor 2(HER2)-negative breast cancer (BC). CASE REPORT We report on the efficacy of neoadjuvant palbociclib and letrozole application in a patient suffering from invasive estrogen receptor (ER)+/HER2- BC and concurrent well-differentiated and dedifferentiated liposarcoma (WD-DDLPS) of the thigh. Clinical and histological workup upon surgery revealed significant regressive changes in both the liposarcoma and the BC. The 24-month follow-up shows no signs of disease. CONCLUSION CDK4/6 inhibitors exhibit a high therapeutic potential, although reliable prognostic markers need to be identified.
Collapse
Affiliation(s)
| | | | - Michael Kurrer
- Gemeinschaftspraxis Pathologie Zürich, Zürich, Switzerland
| | | | | |
Collapse
|
213
|
Recurrent CCND3 mutations in MLL-rearranged acute myeloid leukemia. Blood Adv 2019; 2:2879-2889. [PMID: 30381403 DOI: 10.1182/bloodadvances.2018019398] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 09/13/2018] [Indexed: 12/12/2022] Open
Abstract
In acute myeloid leukemia (AML), MLL (KMT2A) rearrangements are among the most frequent chromosomal abnormalities; however, knowledge of the genetic landscape of MLL-rearranged AML is limited. In this study, we performed whole-exome sequencing (n = 9) and targeted sequencing (n = 56) of samples from pediatric MLL-rearranged AML patients enrolled in the Japanese Pediatric Leukemia/Lymphoma Study Group AML-05 study. Additionally, we analyzed 105 pediatric t(8;21) AML samples and 30 adult MLL-rearranged AML samples. RNA-sequencing data from 31 patients published in a previous study were also reanalyzed. As a result, we identified 115 mutations in pediatric MLL-rearranged AML patients (2.1 mutations/patient), with mutations in signaling pathway genes being the most frequently detected (60.7%). Mutations in genes associated with epigenetic regulation (21.4%), transcription factors (16.1%), and the cohesin complex (8.9%) were also commonly detected. Novel CCND3 mutations were identified in 5 pediatric MLL-rearranged AML patients (8.9%) and 2 adult MLL-rearranged AML patients (3.3%). Recurrent mutations of CCND1 (n = 3, 2.9%) and CCND2 (n = 8, 7.6%) were found in pediatric t(8;21) AML patients, whereas no CCND3 mutations were found, suggesting that D-type cyclins exhibit a subtype-specific mutation pattern in AML. Treatment of MLL-rearranged AML cell lines with CDK4/6 inhibitors (abemaciclib and palbociclib) blocked G1 to S phase cell-cycle progression and impaired proliferation. Pediatric MLL-MLLT3-rearranged AML patients with coexisting mutations (n = 16) had significantly reduced relapse-free survival and overall survival compared with those without coexisting mutations (n = 9) (P = .048 and .046, respectively). These data provide insights into the genetics of MLL-rearranged AML and suggest therapeutic strategies.
Collapse
|
214
|
Niu Y, Xu J, Sun T. Cyclin-Dependent Kinases 4/6 Inhibitors in Breast Cancer: Current Status, Resistance, and Combination Strategies. J Cancer 2019; 10:5504-5517. [PMID: 31632494 PMCID: PMC6775706 DOI: 10.7150/jca.32628] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 08/06/2019] [Indexed: 12/26/2022] Open
Abstract
Dysregulated activation of the cyclin-dependent kinases (CDKs) 4/6, leading to uncontrolled cell division, is hallmark of cancers. Further study of the cell cycle will advance the cancer treatment. As powerful and effective drugs, inhibitors of CDK 4/6 have been widely used in clinical practice for several malignancies, particularly against breast cancers driven by the estrogen receptor (ER). Three CDK4/6 inhibitors, including palbociclib (PD0332991), ribociclib (LEE011) and abemaciclib (LY2835219), have been approved by the US Food and Drug Administration (FDA) for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced or metastatic breast cancer. However, CDK4/6 inhibitors act downstream of many mitogenic signaling pathways, and this has implications for resistance. It is worth to note that the mechanisms of resistance are not very clear. Up to now, a small number of preclinical and clinical studies have explored potential mechanisms of CDK4/6 inhibitors resistance in breast cancer. On this basis, rational and effective combination therapy is under development. Here we review the current knowledge about the mechanisms and efficacy of CDK4/6 inhibitors, and summarize data on resistance mechanisms to make future combination therapies more accurate and reasonable.
Collapse
Affiliation(s)
- Ying Niu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Junnan Xu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China.,Key Laboratory of Liaoning Breast Cancer Research, Shenyang, Liaoning 110042, P.R. China
| | - Tao Sun
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
215
|
Song X, Gan Q, Zhang X, Zhang J. Synthesis and Biological Evaluation of Novel 99mTc-Labeled Palbociclib Derivatives Targeting Cyclin-Dependent Kinase 4/6 (CDK4/6) as Potential Cancer Imaging Agents. Mol Pharm 2019; 16:4213-4222. [PMID: 31424939 DOI: 10.1021/acs.molpharmaceut.9b00540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer results from cell proliferation that exceeds normal growth control. There are various specific proteins that control and regulate the cell cycle, such as cyclin-dependent kinases (CDKs), cyclins, and retinoblastoma protein (pRb). The aberration of the cyclin D-CDK4/6-INK4-pRb pathway occurs frequently in cancers; thus, CDK4/6 is an attractive target for the development of radiopharmaceuticals for tumor imaging. In this study, we chose palbociclib, which was approved by the FDA for treating ER+/HER2- advanced breast cancer as the target vector and the isonitrile group, which can coordinate strongly with the [99mTc(CO)3]+ core as the bifunctional chelator, to develop four novel 99mTc-labeled radiotracers for tumor imaging. The ligands (L2, L3, L4, and L5) were synthesized by reacting palbociclib with isocyanide-containing active esters and then radiolabeling with a [99mTc(CO)3]+ core to produce radiotracers (99mTc-L2, 99mTc-L3, 99mTc-L4, and 99mTc-L5) with high radiochemical purity (>95%) and good stability in vitro. The structures of the 99mTc complexes were identified by preparation and characterization of the corresponding stable rhenium complexes. Partition coefficient results indicated that these complexes were lipophilic. A kinase inhibition assay demonstrated the high affinity of the stable Re complexes for CDK4. A cell study showed that all four complexes had substantial uptake by MCF-7 cells and could be significantly inhibited by palbociclib and nonradiolabeled ligand, indicating a CDK4/6-specific uptake mechanism. Biodistribution studies in nude mice bearing MCF-7 tumors showed that the complexes had obvious accumulation in tumors at 2 h postinjection. 99mTc-L2 exhibited the highest tumor uptake and tumor/blood ratio, whereas 99mTc-L4 showed the highest tumor/muscle ratio. The micro-SPECT/CT study showed that complex 99mTc-L4 had visible uptake at the tumor site, and the accumulation was clearly reduced in the image after pretreatment with palbociclib, further indicating CDK4/6 specificity. All the results showed that the 99mTc-labeled complexes in this work have the potential for tumor imaging.
Collapse
Affiliation(s)
- Xiaoqing Song
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry , Beijing Normal University , Beijing 100875 , P. R. China
| | - Qianqian Gan
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry , Beijing Normal University , Beijing 100875 , P. R. China
| | - Xuran Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry , Beijing Normal University , Beijing 100875 , P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry , Beijing Normal University , Beijing 100875 , P. R. China
| |
Collapse
|
216
|
Xiao X, Zhang Y, Lin Q, Zhong K. The better effects of microbubble ultrasound transfection of miR-940 on cell proliferation inhibition and apoptosis promotion in human cervical cancer cells. Onco Targets Ther 2019; 12:6813-6824. [PMID: 31686839 PMCID: PMC6709033 DOI: 10.2147/ott.s209692] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/31/2019] [Indexed: 01/18/2023] Open
Abstract
Purpose Cervical cancer is the second leading cause of women’s cancer-related death. MiR-940 has been reported as a critical factor in various cancers. Based on the high transfection efficiency and low side effect, the clinical application of microbubble ultrasound contrast agent in gene treatment has attracted a widespread attention. In this study, we determined the mechanism of miR-940 inhibiting cell proliferation and cycle procession, and promoting cell apoptosis in cervical cancer Hela cells. In addition, we compared the effects of different transfection methods, including liposome, microbubble, ultrasound, and microbubble coupled with ultrasound. Patients and methods MTT assay, PI staining, and Annexin-Ⅴ/PI staining assays were, respectively, performed to evaluate cell proliferation status, cell cycle progression, and apoptosis status. RT-PCR and Western blot were conducted to measure the levels of cell cycle- and apoptosis-related factors, and the phosphorylation levels of PI3K and Akt. Results Results showed that the overexpression of miR-940 inhibited cell proliferation, blocked cell cycle, and promoted apoptosis by regulating cell cycle-related factors (such as inhibited Cyclin D1 and CDK4) and apoptosis-related factors (such as promoted Puma and Bax, inhibited Bcl-2 and Cleaved caspase9), and inhibiting the phosphorylation and activation of PI3K/AKT pathway. Among all of them, miR-940 transfected with microbubble and ultrasound showed the greatest changes. Conclusion It provides evidence that miR-940 could be a wonderful biomarker and treatment agent for cervical cancer, and microbubble ultrasound would have more wide application in the clinical treatment of cancers.
Collapse
Affiliation(s)
- Xiaojun Xiao
- Department of Ultrasound, Shenzhen People's Hospital, Shenzhen, Guangdong Province 518020, People's Republic of China
| | - Yujuan Zhang
- Department of Ultrasound, Shenzhen People's Hospital, Shenzhen, Guangdong Province 518020, People's Republic of China
| | - Qi Lin
- Department of Ultrasound, Shenzhen People's Hospital, Shenzhen, Guangdong Province 518020, People's Republic of China
| | - Keli Zhong
- Department of Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong Province 518020, People's Republic of China
| |
Collapse
|
217
|
Zhang X, Fan T, Li S, Guan F, Zhang J, Liu H. C-Phycocyanin elicited antitumor efficacy via cell-cycle arrest, apoptosis induction, and invasion inhibition in esophageal squamous cell carcinoma. J Recept Signal Transduct Res 2019; 39:114-121. [PMID: 31322033 DOI: 10.1080/10799893.2019.1638400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objectives: Mounting evidence has demonstrated that C-Phycocyanin (C-PC) exhibits marked antitumor activity in a wide type of tumors, such as pancreas cancer, breast carcinoma, lung cancer, and colon cancer. The current study aimed to confirm the antitumor efficacy of C-PC in esophageal squamous cell carcinoma (ESCC). Methods: The efficacy of C-PC was evaluated against the proliferation of ESCC cell lines EC9706 and EC1 by CCK-8 kit and in a mice model of ESCC EC9706. Cell cycle and apoptosis were investigated by flow cytometry, and cell invasion was determined via transwell chamber. Protein expression was examined by Western blots. Results: We found that C-PC exhibited anti-proliferation ability in a time-dependent manner and a dose-dependent manner in ESCC EC9706 and EC1 cells. Besides, C-PC markedly arrested cell cycle in the G0/G1 phase, induced cell apoptosis and suppressed cell invasion ability in both EC9706 and EC1 cells (p < .01). Notably, C-PC evoked the elevations of Bax, PARP, and cleaved-caspase-3 protein, but reduced cyclin D1, CDK4, Bcl-2, MMP-2, and MMP-9 expression levels. Further investigation from in vivo experiment revealed that C-PC displayed significant antitumor efficacy in the xenografted EC9706 model. Conclusions: Our data presented herein suggest C-PC exerts antitumor efficacy in ESCC.
Collapse
Affiliation(s)
- Xiaqing Zhang
- a College of Life Sciences of Zhengzhou University , Zhengzhou , China
| | - Tianli Fan
- b Department of Pharmacology, School of Basic Medicine, Zhengzhou University , Zhengzhou , China
| | - Shenglei Li
- c Department of Pathology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Fangxia Guan
- a College of Life Sciences of Zhengzhou University , Zhengzhou , China
| | - Jianying Zhang
- d Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University , Zhengzhou , China
| | - Hongtao Liu
- a College of Life Sciences of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
218
|
Wang X, Peng P, Pan Z, Fang Z, Lu W, Liu X. Psoralen inhibits malignant proliferation and induces apoptosis through triggering endoplasmic reticulum stress in human SMMC7721 hepatoma cells. Biol Res 2019; 52:34. [PMID: 31277690 PMCID: PMC6612100 DOI: 10.1186/s40659-019-0241-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Psoralen is a coumarin-like and coumarin-related benzofuran glycoside, which is a commonly used traditional Chinese medicine to treat patients with kidney and spleen-yang deficiency symptom. Psoralen has been reported to show estrogen-like activity, antioxidant activity, osteoblastic proliferation accelerating activity, antitumor effects and antibacterial activity. However, the antitumor mechanism of psoralen is not fully understood. This study aimed to investigate the therapeutic efficacy of psoralen in human hepatoma cell line SMMC7721 and the mechanism of antitumor effects. RESULTS Psoralen inhibited proliferation of SMMC7721 in a dose- and time-dependent manner, and promoted apoptosis. Further, psoralen activated the ER stress signal pathway, including the expansion of endoplasmic reticulum, increasing the mRNA levels of GRP78, DDIT3, ATF4, XBP1, GADD34 and the protein levels of GDF15, GRP78, IRE1α, XBP-1s in a time-dependent manner. Psoralen induces cell cycle arrest at G1 phase by enhancing CyclinD1 and reducing CyclinE1 expression. Moreover, TUDC couldn't inhibit the psoralen-induced ER stress in SMMC7721 cells. CONCLUSIONS Psoralen can inhibit the proliferation of SMMC7721 cells and induce ER stress response to induce cell apoptosis, suggesting that psoralen may represent a novel therapeutic option for the prevention and treatment hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiaomin Wang
- Basic Medical School of Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Peike Peng
- Basic Medical School of Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Zhiqiang Pan
- Basic Medical School of Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Zhaoqin Fang
- Basic Medical School of Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Wenli Lu
- Basic Medical School of Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Xiaomei Liu
- Basic Medical School of Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| |
Collapse
|
219
|
Validation and functional analysis of the critical proteins in combination with taurine, epigallocatechin gallate and genistein against liver fibrosis in rats. Biomed Pharmacother 2019; 115:108975. [DOI: 10.1016/j.biopha.2019.108975] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/22/2022] Open
|
220
|
Betulinic Acid Induces ROS-Dependent Apoptosis and S-Phase Arrest by Inhibiting the NF- κB Pathway in Human Multiple Myeloma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5083158. [PMID: 31281581 PMCID: PMC6590575 DOI: 10.1155/2019/5083158] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/08/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023]
Abstract
Betulinic acid (BA), as a prospective natural compound, shows outstanding antitumor bioactivities against many solid malignancies. However, its mechanism against multiple myeloma (MM) remains elusive. Herein, for the first time, we studied the antitumor activity of BA against MM both in vivo and in vitro. We showed that BA mediated cytotoxicity in MM cells through apoptosis, S-phase arrest, mitochondrial membrane potential (MMP) collapse, and overwhelming reactive oxygen species (ROS) accumulation. Moreover, when the ROS scavenger N-acetyl cysteine (NAC) effectively abated elevated ROS, the BA-induced apoptosis was partially reversed. Our results revealed that BA-mediated ROS overproduction played a pivotal role in anticancer activity. Molecularly, we found that BA resulted in marked inhibition of the aberrantly activated NF-κB pathway in MM. As demonstrated by using the NF-κB pathway-specific activator TNF-α and the inhibitor BAY 11-7082, BA-mediated inhibition of the NF-κB pathway directly promoted the overproduction of ROS and, ultimately, cell death. Furthermore, BA also exerted enormous tumor-inhibitory effects via repressing proliferation and inhibiting the NF-κB pathway in our xenograft model. Overall, by blocking the NF-κB pathway that breaks redox homeostasis, BA, as a potent NF-κB inhibitor, is a promising therapeutic alternative for MM.
Collapse
|
221
|
Indovina P, Pentimalli F, Conti D, Giordano A. Translating RB1 predictive value in clinical cancer therapy: Are we there yet? Biochem Pharmacol 2019; 166:323-334. [PMID: 31176618 DOI: 10.1016/j.bcp.2019.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022]
Abstract
The retinoblastoma RB1 gene has been identified in the 80s as the first tumor suppressor. RB1 loss of function, as well alterations in its pathway, occur in most human cancers and often have prognostic value. RB1 has a key role in restraining cell cycle entry and, along with its family members, regulates a myriad of cellular processes and affects cell response to a variety of stimuli, ultimately determining cell fate. Consistently, RB1 status is a crucial determinant of the cell response to antitumoral therapies, impacting on the outcome of both traditional and modern anti-cancer strategies, including precision medicine approaches, such as kinase inhibitors, and immunotherapy. Despite many efforts however, the predictive value of RB1 status in the clinical practice is still underused, mainly owing to the complexity of RB1 function, to differences depending on the cellular context and on the therapeutic strategies, and, not-lastly, to technical issues. Here, we provide an overview of studies analyzing the role of RB1 in response to conventional cytotoxic and cytostatic therapeutic agents in different cancer types, including hormone dependent ones. We also review RB1 predictive value in the response to the last generation CDK4/6 inhibitors, other kinase inhibitors, and immunotherapy and discuss new emerging non-canonical roles of RB1 that could impact on the response to antitumoral treatments.
Collapse
Affiliation(s)
- Paola Indovina
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Francesca Pentimalli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli 80131, Italy
| | - Daniele Conti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy.
| |
Collapse
|
222
|
Overexpression of zinc finger protein 384 (ZNF 384), a poor prognostic predictor, promotes cell growth by upregulating the expression of Cyclin D1 in Hepatocellular carcinoma. Cell Death Dis 2019; 10:444. [PMID: 31168049 PMCID: PMC6551341 DOI: 10.1038/s41419-019-1681-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous, multigene-driven malignant tumor. ZNF384 is an overexpressed gene with a high frequency of alteration in HCC, but research on the function of ZNF384 in HCC is lacking. In this study, the expression level of ZNF384 in HCC was analyzed through immunohistochemical (IHC) staining, Western blot analysis and qRT-PCR. We also generated ZNF384 knockdown and knockout HCC cell lines using short hairpin RNA (shRNA) and CRISPR/Cas9 systems. MTS, colony formation, and 5-ethynyl-20-deoxyuridine (EdU) assays; flow cytometry; and a xenograft mouse model were used to evaluate the effects of ZNF384 on cell proliferation. Western blot analysis, a dual luciferase reporter assay and a ChIP assay were performed to explore the potential mechanism. We found that overexpression of ZNF384 in HCC and elevated expression of ZNF384 in HCC tissues was significantly correlated with tumor recurrence (P = 0.0097). Kaplan-Meier survival analysis revealed that high expression levels of ZNF384 were correlated with poor overall survival (P = 0.0386). Downregulation of ZNF384 expression suppressed HCC cell proliferation by inhibiting the expression of Cyclin D1. These findings suggest that ZNF384 tends to act as an oncogene in the development of HCC. ZNF384 promotes the proliferation of HCC cells by directly upregulating the expression of Cyclin D1 and might serve as a prognostic predictive factor for HCC patients.
Collapse
|
223
|
Qie S, Yoshida A, Parnham S, Oleinik N, Beeson GC, Beeson CC, Ogretmen B, Bass AJ, Wong KK, Rustgi AK, Diehl JA. Targeting glutamine-addiction and overcoming CDK4/6 inhibitor resistance in human esophageal squamous cell carcinoma. Nat Commun 2019; 10:1296. [PMID: 30899002 PMCID: PMC6428878 DOI: 10.1038/s41467-019-09179-w] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 02/20/2019] [Indexed: 02/08/2023] Open
Abstract
The dysregulation of Fbxo4-cyclin D1 axis occurs at high frequency in esophageal squamous cell carcinoma (ESCC), where it promotes ESCC development and progression. However, defining a therapeutic vulnerability that results from this dysregulation has remained elusive. Here we demonstrate that Rb and mTORC1 contribute to Gln-addiction upon the dysregulation of the Fbxo4-cyclin D1 axis, which leads to the reprogramming of cellular metabolism. This reprogramming is characterized by reduced energy production and increased sensitivity of ESCC cells to combined treatment with CB-839 (glutaminase 1 inhibitor) plus metformin/phenformin. Of additional importance, this combined treatment has potent efficacy in ESCC cells with acquired resistance to CDK4/6 inhibitors in vitro and in xenograft tumors. Our findings reveal a molecular basis for cancer therapy through targeting glutaminolysis and mitochondrial respiration in ESCC with dysregulated Fbxo4-cyclin D1 axis as well as cancers resistant to CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Shuo Qie
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Akihiro Yoshida
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Stuart Parnham
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Natalia Oleinik
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Gyda C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Craig C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Adam J Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kwok-Kin Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Anil K Rustgi
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - J Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
224
|
Bisphenol S promotes the cell cycle progression and cell proliferation through ERα-cyclin D-CDK4/6-pRb pathway in MCF-7 breast cancer cells. Toxicol Appl Pharmacol 2019; 366:75-82. [DOI: 10.1016/j.taap.2019.01.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/15/2019] [Accepted: 01/19/2019] [Indexed: 11/20/2022]
|
225
|
Abdel-Razeq H. Current frontline endocrine treatment options for women with hormone receptor-positive, Human Epidermal Growth Factor Receptor 2 (HER2)-negative advanced-stage breast cancer. Hematol Oncol Stem Cell Ther 2019; 12:1-9. [DOI: 10.1016/j.hemonc.2018.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 04/14/2018] [Accepted: 04/27/2018] [Indexed: 01/28/2023] Open
|
226
|
Zhang G, Ma F, Li L, Li J, Li P, Zeng S, Sun H, Li E. Palbociclib triggers apoptosis in bladder cancer cells by Cdk2-induced Rad9-mediated reorganization of the Bak.Bcl-xl complex. Biochem Pharmacol 2019; 163:133-141. [PMID: 30772267 DOI: 10.1016/j.bcp.2019.02.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/13/2019] [Indexed: 12/16/2022]
Abstract
Palbociclib is a Cdk4/6 inhibitor approved for metastatic estrogen receptor-positive breast cancer. The drug is also under clinical evaluation for metastatic urothelial cancer and other solid tumors. Preclinical studies from multiple tumor types suggest that other factors also affect the sensitivity of individual tumors to Cdk4/6 inhibitor. We show here that Cdk2 has an essential role in palbociclib antitumor effect against bladder cancers. We found that palbociclib induced apoptosis instead of cell cycle arrest to exhibit its anticancer activity in T24 cells, as was evidenced by membrane blebbing, caspase-3 activation and AIF release from mitochondria. Cdk2 activation was important to palbociclib-induced apoptotic triggering activity, since depletion of Cdk2 significantly inhibited caspase-3 activation and cell apoptosis. Cdk2 activation caused p-Rad9 translocation to the mitochondria and subsequently interaction with Bcl-xl, leading to conformational activation of Bak and cell apoptosis. The anticancer activity and Cdk2 activation of palbociclib-treated mice were finally validated in a T24 xenograft model. Collectively, these results together demonstrate that palbociclib exerts its anticancer effect in T24 cells mainly through Cdk2 activation. Our findings provide new insights into the molecular interactions and anticancer mechanisms of Cdk4/6 inhibitors.
Collapse
Affiliation(s)
- Guohai Zhang
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China.
| | - Feng'e Ma
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Liangping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Jingjing Li
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Pingping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Shulan Zeng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Hongbin Sun
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Erguang Li
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
227
|
He Q, Xue S, Tan Y, Zhang L, Shao Q, Xing L, Li Y, Xiang T, Luo X, Ren G. Dual inhibition of Akt and ERK signaling induces cell senescence in triple-negative breast cancer. Cancer Lett 2019; 448:94-104. [PMID: 30742942 DOI: 10.1016/j.canlet.2019.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/05/2019] [Accepted: 02/01/2019] [Indexed: 12/31/2022]
Abstract
Activated Akt and ERK signaling pathways are closely related to breast cancer progression, and Akt or ERK inhibition induces cell senescence. However, the crosstalk between the Akt and ERK signaling pathways in cell senescence and how to simultaneously suppress Akt and ERK signaling in triple-negative breast cancer (TNBC) are undefined. In this study, we found that norcantharidin (NCTD) effectively induced cell senescence and cell cycle arrest in TNBC in vitro, which was accompanied by a decline in phosphorylated Akt and ERK1/2 and a rise in p21 and p16. The inhibitors LY294002 and U0126 imitated the effect of NCTD when these two inhibitors were combined regardless of crosstalk between these two signaling pathways. In addition, NCTD inhibited the growth of xenografts via downregulation of phosphorylated Akt and ERK1/2 and upregulation of p21 in vivo. However, NCTD upregulated the level of soluble signaling factors of the senescence-associated secretory phenotype (SASP) in a NF-κB-independent manner. Collectively, these findings demonstrate that NCTD induced cell senescence and cell cycle arrest mainly by simultaneously blocking Akt and ERK signaling in TNBC, suggesting that NCTD may be used as a potential adjuvant therapy in TNBC.
Collapse
Affiliation(s)
- Qiang He
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Yiqing Tan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Zhang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Shao
- Breast Center, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Lei Xing
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunhai Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinrong Luo
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
228
|
Belső N, Gubán B, Manczinger M, Kormos B, Bebes A, Németh I, Veréb Z, Széll M, Kemény L, Bata-Csörgő Z. Differential role of D cyclins in the regulation of cell cycle by influencing Ki67 expression in HaCaT cells. Exp Cell Res 2019; 374:290-303. [DOI: 10.1016/j.yexcr.2018.11.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 11/30/2022]
|
229
|
Gottesman SRS, Somma J, Tsiperson V, Dresner L, Govindarajulu U, Patel P, Blain SW. Tyrosine Phosphorylation of p27Kip1 Correlates with Palbociclib Responsiveness in Breast Cancer Tumor Cells Grown in Explant Culture. Mol Cancer Res 2018; 17:669-675. [PMID: 30559257 DOI: 10.1158/1541-7786.mcr-18-0188] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 10/19/2018] [Accepted: 12/12/2018] [Indexed: 12/30/2022]
Abstract
Cdk4-targeting drugs, such as palbociclib, are approved for metastatic ER/PR+, Her2- breast cancer. However, other than loss of retinoblastoma, which is very rare in this subset, there are no biomarkers to predict response. Cyclin D or cdk4 levels are not by themselves indicative, because p27Kip1 is required for cyclin D-cdk4 complex activation. Tyrosine phosphorylation of p27, including modification on residue Y88 (pY88), activates DK4-p27, and the pY88 level correlates with palbociclib responsiveness in cell lines. We developed dual IHC staining for p27 and pY88, and found that benign breast epithelium was negative, while breast cancer biopsies (of varied hormonal status) could be stratified for pY88 status. Lack of pY88 suggested that DK4 was inactive, and that these samples would not have the target required for palbociclib response. Tumor resection material was grown in explant culture, treated with palbociclib, and stained with Ki67 as a marker of response. Explants from the no pY88 group were nonresponsive, while explants from the low or high pY88 group responded to drug. IMPLICATIONS: Use of the pY88 biomarker, as a surrogate for cdk4 activity, may identify patients responsive to cdk4-targeting drugs and expand use of this therapy.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/17/3/669/F1.large.jpg.
Collapse
Affiliation(s)
- Susan R S Gottesman
- Departments of Pathology and Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York
| | - Jonathan Somma
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Vladislav Tsiperson
- Departments of Pediatrics and Cell Biology, SUNY Downstate Medical Center, Brooklyn, NewYork
| | - Lisa Dresner
- Department of Surgery, SUNY Downstate Medical Center, Brooklyn, New York
| | - Usha Govindarajulu
- Department of Epidemiology and Biostatistics, SUNY Downstate Medical Center, Brooklyn, New York
| | - Priyank Patel
- School of Graduate Studies, SUNY Downstate Medical Center, Brooklyn, New York
| | - Stacy W Blain
- Departments of Pediatrics and Cell Biology, SUNY Downstate Medical Center, Brooklyn, NewYork.
| |
Collapse
|
230
|
Geng B, Liang M, Qin L, Zhao W, Wang H, Wang L, Pan X, Chen X. An TRIM59-CDK6 axis regulates growth and metastasis of lung cancer. J Cell Mol Med 2018; 23:1458-1469. [PMID: 30515965 PMCID: PMC6349187 DOI: 10.1111/jcmm.14052] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/16/2018] [Accepted: 11/02/2018] [Indexed: 12/17/2022] Open
Abstract
Lung cancer (LC) is a devastating malignancy with no effective treatments, due to its complex genomic profile. Using bioinformatics analysis and immunohistochemical of lung carcinoma tissues, we show that TRIM59 as a critical oncoprotein relating to LC proliferation and metastasis. In this study, high TRIM59 expression was significantly correlated with lymph node metastasis, distant metastasis, and tumour stage. Furthermore, up‐regulation of TRIM59 expression correlated with poorer outcomes in LC patients. Mechanistically, TRIM59 play a key role in promoting LC growth and metastasis through regulation of extracellular‐signal regulated protein kinase (ERK) signalling pathway and epithelial‐to‐mesenchymal transition (EMT)‐markers, as validated by loss‐of‐function studies. In‐depth bioinformatics analysis showed that there is preliminary evidence of co‐expression of TRIM59 and cyclin dependent kinase 6 (CDK6) in LC. Notably, CDK6 expression significantly decreased when TRIM59 was knocked down in the LC cells. In contrast, exogenous up‐regulation of TRIM59 expression also induced significant increases in the expression of CDK6. Moreover, the expression of CDK6 was also inhibited by the ERK signalling inhibitor, U0126. The results of both loss‐ and gain‐of‐function studies showed that TRIM59 could regulate the expression of CDK6. Collectively, these data provide evidence that TRIM59 is involved in lung carcinoma growth and progression possibly through the induction of CDK6 expression and EMT process by activation of ERK pathway.
Collapse
Affiliation(s)
- Biao Geng
- Department of Respiratory Medicine, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Manman Liang
- Department of Infectious Diseases, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Lilong Qin
- Department of Respiratory Medicine, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Wenying Zhao
- Department of Medical Oncology, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Hanli Wang
- Department of Respiratory Medicine, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Lijing Wang
- Department of Respiratory Medicine, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Xianhui Pan
- Department of Respiratory Medicine, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Xingwu Chen
- Department of Respiratory Medicine, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
231
|
Huang IT, Dhungel B, Shrestha R, Bridle KR, Crawford DHG, Jayachandran A, Steel JC. Spotlight on Bortezomib: potential in the treatment of hepatocellular carcinoma. Expert Opin Investig Drugs 2018; 28:7-18. [PMID: 30474444 DOI: 10.1080/13543784.2019.1551359] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION This study reviews the evidence for the use of Bortezomib (BZB), a first-in-class proteasome inhibitor in advanced Hepatocellular carcinoma (HCC). This review aims to delineate the role of BZB within the management of non-surgical and metastatic HCC, either as an alternative or as an adjunct to the current treatment paradigm. AREAS COVERED In addition to BZB pharmacology and mechanism of action, safety and tolerance profiles of the drug obtained from clinical trials are explored. The utility of BZB as a therapeutic agent either alone or in combination with other therapies against HCC, including its application in both preclinical and clinical settings has been reviewed. In particular, we highlight the importance of preclinical evaluation of BZB as a combinatorial agent in synergism with other therapies for the use in the management of HCC. EXPERT OPINION There has been much interest surrounding the use of BZB, a first-in-class proteasome inhibitor for HCC therapy. The discernment of outcomes of BZB clinical trials for HCC need to take into consideration the disease-specific factors that can affect survival outcomes including patient selection and aetiological differences. Further preclinical testing of BZB in combination with other therapeutic modalities can be important for eliciting enhanced anti-HCC effects.
Collapse
Affiliation(s)
- I-Tao Huang
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Bijay Dhungel
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Ritu Shrestha
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Kim R Bridle
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Darrell H G Crawford
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Aparna Jayachandran
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,b Gallipoli Medical Research Institute , Greenslopes Private Hospital , Brisbane , Australia
| | - Jason C Steel
- a School of Clinical Medicine , The University of Queensland , Brisbane , Australia.,c School of Health, Medical and Applied Sciences , CQ University , Rockhampton , Australia
| |
Collapse
|
232
|
Wen J, Hu Y, Liu Q, Ling Y, Zhang S, Luo K, Xie X, Fu J, Yang H. miR-424 coordinates multilayered regulation of cell cycle progression to promote esophageal squamous cell carcinoma cell proliferation. EBioMedicine 2018; 37:110-124. [PMID: 30361064 PMCID: PMC6284509 DOI: 10.1016/j.ebiom.2018.10.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Dysregulation of the cell cycle has been implicated in esophageal squamous cell carcinoma (ESCC) progression. This study aimed to evaluate the role of miR-424 in cell cycle regulation and ESCC proliferation. METHODS The role of miR-424 in cell proliferation was evaluated in vitro and in vivo. Transcriptional activation of miR-424 was determined using chromatin immunoprecipitation, and binding of miR-424 to targets was verified using miRNA ribonucleoprotein complex immunoprecipitation. FINDINGS miR-424 was upregulated and correlated with poor survival in ESCC patients. Repression or overexpression of miR-424 respectively decreased or increased ESCC cell proliferation in vitro and in vivo. miR-424 expression is transcriptionally regulated by E2F1 and increased during G1/S transition. Knockdown or overexpression of miR-424 respectively inhibited or promoted both G1/S and G2/M cell cycle transitions in ESCC cells, and these effects were mediated by two newly identified miR-424 targets, PRKCD and WEE1, respectively. Consequently, elevation of PRKCD by miR-424 knockdown led to enhanced stability of the p21Cip1 protein via increased activation of PRKCD and downstream p38 MAPK and JNK signaling to block CDK2 activation and G1/S transition, while elevated WEE1 maintained CDC2 in an inactive state to block G2/M transition. However, circLARP4 could sponge the binding of miR-424 to PRKCD, thus compromising the regulation of G1/S progression by miR-424. INTERPRETATION miR-424 coordinates a previously unknown, multilayered regulation of ESCC cell cycle progression to promote ESCC proliferation, and may be used as a novel prognostic marker and an effective therapeutic target for ESCCs. FUND: National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yi Hu
- Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Qianwen Liu
- Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yihong Ling
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Shuishen Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou 510080, China
| | - Kongjia Luo
- Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Xiuying Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Jianhua Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Hong Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China.
| |
Collapse
|
233
|
He G, Ma Y, Zhu Y, Yong L, Liu X, Wang P, Liang C, Yang C, Zhao Z, Hai B, Pan X, Liu Z, Liu X, Mao C. Cross Talk Between Autophagy and Apoptosis Contributes to ZnO Nanoparticle-Induced Human Osteosarcoma Cell Death. Adv Healthc Mater 2018; 7:e1800332. [PMID: 29900694 PMCID: PMC6310009 DOI: 10.1002/adhm.201800332] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/18/2018] [Indexed: 12/13/2022]
Abstract
Killing osteosarcoma cells by zinc oxide nanoparticles (NPs) and its underlying subcellular mechanism are never studied. Here, it is found that the NPs induce cross talk between apoptosis and autophagy, which leads to osteosarcoma cell death. Specifically, the NP uptake promotes autophagy by inducing accumulation of autophagosomes along with impairment of lysosomal functions. The autophagy further causes the uptaken NPs to release zinc ions by promoting their dissolution. These intracellular zinc ions, together with those that are originally released from the extracellular NPs and flowed into the cells, collectively target and damage mitochondria to produce reactive oxygen species (ROS). Then the ROS inhibit cell proliferation by arresting S phase and trigger apoptosis by extrinsic and intrinsic pathways, ultimately leading to cell death. More importantly, suppression of the early stage autophagy restores cell viability by abolishing apoptosis whereas blockade of the late stage autophagy inversely enhances apoptosis. In contrast, inhibition of apoptosis shows a limited ability to restore cell viability but obviously enhance autophagy. Notably, cell viability is strongly ameliorated by the combination of inhibitors for both the late stage autophagy and the apoptosis. These findings provide a mechanistic understanding of the NP-directed autophagy and apoptosis in osteosarcoma cells.
Collapse
Affiliation(s)
- Guanping He
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yunlong Ma
- The Center for Pain Medicine, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Ye Zhu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Lei Yong
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiao Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Peng Wang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chen Liang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chenlong Yang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhigang Zhao
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Bao Hai
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiaoyu Pan
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhongjun Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiaoguang Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, USA
| |
Collapse
|
234
|
Gong Y, Mao J, Wu D, Wang X, Li L, Zhu L, Song R. Circ-ZEB1.33 promotes the proliferation of human HCC by sponging miR-200a-3p and upregulating CDK6. Cancer Cell Int 2018; 18:116. [PMID: 30123094 PMCID: PMC6090603 DOI: 10.1186/s12935-018-0602-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/23/2018] [Indexed: 12/23/2022] Open
Abstract
Background Accumulating data indicated that circRNA plays important roles in regulating many biological processes of the tumor, the present study is designated for exploring roles of the circ-ZEB1.33-miR-200a-3p-CDK6 regulating axis in human hepatocellular carcinoma (HCC). Methods The regulation axis as predicted by using online tool circNet, the expression and correlation of circ-ZEB1.33-miR-200a-3p-CDK6 was verified in human HCC. The diagnostic value of both tumor and serum circ-ZEB1.33 was estimated by using clinical samples. The roles of circ-ZEB1.33-miR-200a-3p-CDK6 in regulating cell cycle were explored by using in vitro studies. Results Overexpression of circ-ZEB1.33 and CDK6, downregulation of miR-200a-3p were detected in human HCC tissues, negative correlation between circ-ZEB1.33 and miR-200a-3p, positive correlation between circ-ZEB1.33 and CDK6 were confirmed in human HCC tissues. Tissue and serum circ-ZEB1.33 were related to different TMN stages and prognosis in HCC patients. RNA pull-down assay implied that circ-ZEB1.33 could decrease miR-200a-3p by sponging miR-200a-3p, and the luciferase reporter assay indicated that miR-200a-3p could downregulate CDK6 transcription by targeting its 3′UTR. The in vitro assays indicated that circ-ZEB1.33 could promote the proliferation of HCC cells by increasing the percentage of S phase regulated by CDK6/Rb. Conclusion Proliferation promotion roles of the circ-ZEB1.33-miR-200a-3p-CDK6 regulating axis are existed and verified in human HCC, both tumor and serum circ-ZEB1.33 can serve as an indicator for the prognosis of HCC patients. Electronic supplementary material The online version of this article (10.1186/s12935-018-0602-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuhua Gong
- Department of Clinical Laboratory, The Third People's Hospital of Zhenjiang, 300 Daijiamen, Zhenjiang, 212005 Jiangsu China
| | - Jinzhong Mao
- Department of Radiology, The Third People's Hospital of Zhenjiang, 300 Daijiamen, Zhenjiang, 212005 Jiangsu China
| | - Di Wu
- Department of Hepatosis Inpatient, The Third People's Hospital of Zhenjiang, 300 Daijiamen, Zhenjiang, 212005 Jiangsu China
| | - Xuemei Wang
- Department of Clinical Laboratory, The Third People's Hospital of Zhenjiang, 300 Daijiamen, Zhenjiang, 212005 Jiangsu China
| | - Long Li
- Department of Clinical Laboratory, The Third People's Hospital of Zhenjiang, 300 Daijiamen, Zhenjiang, 212005 Jiangsu China
| | - Liang Zhu
- Department of Clinical Laboratory, The Third People's Hospital of Zhenjiang, 300 Daijiamen, Zhenjiang, 212005 Jiangsu China
| | - Rong Song
- Department of Clinical Laboratory, The Third People's Hospital of Zhenjiang, 300 Daijiamen, Zhenjiang, 212005 Jiangsu China
| |
Collapse
|
235
|
Li K, Ma YB, Zhang Z, Tian YH, Xu XL, He YQ, Xu L, Gao Y, Pan WT, Song WJ, He X, Wei L. Upregulated IQUB promotes cell proliferation and migration via activating Akt/GSK3β/β-catenin signaling pathway in breast cancer. Cancer Med 2018; 7:3875-3888. [PMID: 29968965 PMCID: PMC6089180 DOI: 10.1002/cam4.1568] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 04/18/2018] [Accepted: 04/30/2018] [Indexed: 12/28/2022] Open
Abstract
Breast cancer was the highest incidence of tumor in women, which seriously threaten women's health. Our previous study found that the expression of IQUB (IQ motif and ubiquitin domain containing) was significantly increased in the development of breast cancer by transcriptome sequencing. However, there were no studies on the mechanism of IQUB in tumorigenesis. Further study showed that IQUB expression was significantly increased in breast cancer, which had a significantly positive correlation with pathological differentiation of breast cancer by tissue microarray analysis. Furthermore, we also discovered that IQUB overexpression could obviously promote the proliferation and migration of MCF‐7 cells and increase the proportion of MCF‐7 cells in S and G2/M phase in vitro study, while knockdown of IQUB caused inhibition of cell proliferation and migration in MDA‐MB‐231 cells and increased the proportion of MDA‐MB‐231 cells in G1 phase. Furthermore, IQUB overexpression or knockdown combined with treatment of Licl or MG‐132 showed that IQUB activated Akt to promote GSK3β phosphorylation, which in turn activated Wnt/β‐catenin signaling pathway in breast cancer cells. Taken together, these results indicated that upregulated IQUB promoted breast cancer cell proliferation and migration via activating Akt/GSK3β/β‐catenin signaling pathway, which played an important part in the tumorigenesis and development of breast cancer.
Collapse
Affiliation(s)
- Kai Li
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yan-Bin Ma
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Zun Zhang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yi-Hao Tian
- Department of Anatomy, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiao-Long Xu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yan-Qi He
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Liu Xu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yang Gao
- Hubei Key Laboratory of Tumor Biological Behaviors, Department of Breast and Thyroid Surgery, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Wen-Ting Pan
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Wen-Jing Song
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xin He
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Lei Wei
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
236
|
Abstract
Molecular imaging using 16α-[18F]fluoro-17β-estradiol (FES) and 18F-fluoro-furanyl-norprogesterone PET can assess in vivo function of steroid hormone receptors in breast cancer. These experimental agents have been tested in many single-center clinical trials and show promise to elucidate prognosis and predict endocrine therapy response. The current multicenter trial of FES-PET imaging will help bring this radiotracer closer to clinical use. There is tremendous potential for these tracers to advance drug development, enhance understanding of estrogen receptor-positive tumor biology, and personalize treatment.
Collapse
Affiliation(s)
- Hannah M Linden
- Department of Medical Oncology, Seattle Cancer Care Alliance, UWMC, 825 Eastlake Avenue East, Valley Building LV-200, Seattle, WA 98109-1023, USA
| | - Lanell M Peterson
- Department of Medical Oncology, Seattle Cancer Care Alliance, UWMC, 825 Eastlake Avenue East, Valley Building LV-200, Seattle, WA 98109-1023, USA.
| | - Amy M Fowler
- Department of Radiology, University of Wisconsin, School of Medicine and Public Health, E3/366 Clinical Science Center, 600 Highland Avenue, Madison, WI 53792-3252, USA
| |
Collapse
|
237
|
Xiao W, Li B, Sun X, Yu D, Xie Y, Wu H, Chang S, Zhou Y, Wang H, Lan X, Xu Z, Shi J, Zhu W. DCZ3301, a novel aryl-guanidino inhibitor, induces cell apoptosis and cell cycle arrest via suppressing the PI3K/AKT pathway in T-cell leukemia/lymphoma. Acta Biochim Biophys Sin (Shanghai) 2018; 50:643-650. [PMID: 29688264 DOI: 10.1093/abbs/gmy047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Indexed: 12/25/2022] Open
Abstract
DCZ3301, a novel aryl-guanidino compound, was previously found to have potent anti-tumor activity in myeloma and B-cell lymphoma. In the present study, we investigated the effects of DCZ3301 on T-cell leukemia/lymphoma cells both in vitro and in vivo via cell proliferation, cell cycle analysis, apoptosis assay, mitochondrial membrane potential (MMP) assay, western blot analysis and tumor xenograft models. We found that DCZ3301 inhibited the viability of T-cell leukemia/lymphoma cells in a dose- and time-dependent manner. DCZ3301-induced G2/M cell cycle arrest, associated with downregulation of CDK1, cyclin B1, and cdc25C. DCZ3301 also induced cell apoptosis by decreasing MMP in T-cell leukemia/lymphoma cells, but had no significant pro-apoptotic effect on normal peripheral blood mononuclear cells (PBMCs). In addition, DCZ3301-induced apoptosis may be mediated by the caspase-dependent pathway and suppressing the phosphoinositide 3-kinase (PI3K)/AKT pathway. Finally, we showed that DCZ3301 treatment effectively inhibited tumor growth, with no significant side effects, in xenograft mouse models. In conclusion, these results suggest that DCZ3301 may be regarded as a new therapeutic strategy for T-cell leukemia/lymphoma patients.
Collapse
Affiliation(s)
- Wenqin Xiao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bo Li
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xi Sun
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dandan Yu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongsheng Xie
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huiqun Wu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuaikang Chang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yunfei Zhou
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Houcai Wang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiucai Lan
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jumei Shi
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
238
|
Lynce F, Shajahan-Haq AN, Swain SM. CDK4/6 inhibitors in breast cancer therapy: Current practice and future opportunities. Pharmacol Ther 2018; 191:65-73. [PMID: 29933034 DOI: 10.1016/j.pharmthera.2018.06.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dysregulation of the cyclin dependent kinase pathway in luminal breast cancer creates a new therapeutic opportunity for estrogen receptor positive breast cancer. Initial pan-CDK inhibitors were associated with extensive toxicities but in recent years, the development of potent specific CDK inhibitors with favorable tolerability has driven renewed interests in this class of targeted therapies. Palbociclib, ribociclib and abemaciclib are specific CDK4/6 inhibitors that have been approved by the U.S. Food and Drug Administration for use in combination with endocrine therapy for women with advanced hormone receptor positive breast cancer. These three anticancer therapeutics were approved based on progression free survival benefit seen on phase III trials with the most common grade 3 treatment-related side effects being neutropenia, fatigue, nausea and diarrhea. Except for estrogen receptor positivity, no biomarkers predictive of response to CDK4/6 inhibitors have been identified to date. Based on mechanistic insights here described, CDK4/6 inhibitors are currently being explored in combination with other agents, including targeted therapies, immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Filipa Lynce
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Ayesha N Shajahan-Haq
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Sandra M Swain
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
239
|
Li K, Xu X, He Y, Tian Y, Pan W, Xu L, Ma Y, Gao Y, Gao J, Qi Y, Wei L, Zhang J. P21-activated kinase 7 (PAK7) interacts with and activates Wnt/β-catenin signaling pathway in breast cancer. J Cancer 2018; 9:1821-1835. [PMID: 29805709 PMCID: PMC5968771 DOI: 10.7150/jca.24934] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 02/22/2018] [Indexed: 12/30/2022] Open
Abstract
Background: Breast cancer is the highest incidence of tumor in women, which seriously threaten women's health. The occurrence and progression of breast cancer is linked to inactivation or downregulation of tumor suppressors, and activation or upregulation of oncogenes. However, the mechanism of PAK7 involving in the occurrence and progression of breast cancer is not yet fully understood. Methods: PAK7 expression was analyzed by RT-qPCR and immunohistochemistry and correlated with clinicopatholgical parameters in breast cancer tissue microarray. The effects of PAK7 on breast cancer cells were detected by CCK-8 assay, colon formation assay, wound healing and transwell assays, and flow cytometry. The relationship between PAK7 and Wnt/β-catenin signaling pathway was determined by western blotting, TOP/FOP flash, co-Immunoprecipitation and co-localization assays. Results: PAK7 expression was significantly increased in breast cancer tissues and positively correlated with pathological differentiation and TNM stage of breast cancer. Overexpression of PAK7 could significantly promote proliferation and migration of breast cancer cells, and inhibit apoptosis. In contrast, PAK7 knockdown significantly inhibited the proliferation and migration of breast cancer cells and promoted apoptosis. In addition, PAK7 could activate Wnt/β-catenin signaling pathway in breast cancer cells. Further study found that PAK7 could directly bind to GSK3β and β-catenin, and regulate β-catenin degradation by phosphorylating GSK3β. Conclusions: Our study demonstrated that PAK7, as an oncogene, involved in breast cancer progression by activating the Wnt/β-catenin signaling pathway, suggesting that the potential applicability of PAK7 as a target for breast cancer treatment.
Collapse
Affiliation(s)
- Kai Li
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Xiaolong Xu
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yanqi He
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yihao Tian
- Department of Anatomy, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Wenting Pan
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Liu Xu
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yanbin Ma
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yang Gao
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan 430071, Hubei, China
| | - Jingbo Gao
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan 430071, Hubei, China
| | - Yuwen Qi
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan 430071, Hubei, China
| | - Lei Wei
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Jingwei Zhang
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
240
|
Russo I, Carrizzo A, Bochicchio S, Piazza O, Lamberti G, Barba AA, Vecchione C, Zeppa P, Iovino P, Bucci C, Santonicola A, Ciacci C. siRNA Delivery for Control of Cyclin D1 and E2F1 Expression in Crohn's Disease. Transl Med UniSa 2018; 17:25-33. [PMID: 30083520 PMCID: PMC6067069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Evidence in inflammatory bowel diseases (IBD) supports a connection between inflammation and cancer due to the alteration of the cell cycle with loss of control at the G1/S checkpoint. In this study, we analyze the expression and modulation of CyD1 and E2F1 in colon explants from Crohn's disease (CD) patients. We used ex vivo culture of colon explants from 4 CD patients and 2 healthy controls, stimulated with lipopolysaccharide from Escherichia Coli (EC-LPS). Commercial siRNAs for CyD1 and E2F1 inhibition were encapsulated in Invivofectamine® and in purposely produced nanoliposomal vectors to silencing CyD1 and E2F1 expression. Western blot analysis was used to investigate the effect of siRNA on CyD1, E2F1 and cyclooxygenase 2 (COX-2) expression. In CD patients colon explants, CyD1 and E2F1 increased after the inflammatory stimulus but siRNA silencing attenuated their expression and controlled the COX-2 expression too. These data represent a prelimiary exploration of in vitro siRNA use.
Collapse
Affiliation(s)
- Ilaria Russo
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Albino Carrizzo
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Sabrina Bochicchio
- Department of Industrial Engineering, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) – ITALY,Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) - ITALY
| | - Ornella Piazza
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Gaetano Lamberti
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) - ITALY
| | - Anna Angela Barba
- Department of Industrial Engineering, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) – ITALY
| | - Carmine Vecchione
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Pio Zeppa
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Paola Iovino
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Cristina Bucci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Antonella Santonicola
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Carolina Ciacci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| |
Collapse
|
241
|
Russo I, Carrizzo A, Bochicchio S, Piazza O, Lamberti G, Barba AA, Vecchione C, Zeppa P, Iovino P, Bucci C, Santonicola A, Ciacci C. siRNA Delivery for Control of Cyclin D1 and E2F1 Expression in Crohn's Disease. Transl Med UniSa 2018; 17:22-30. [PMID: 30050877 PMCID: PMC6056255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Evidence in inflammatory bowel diseases (IBD) supports a connection between inflammation and cancer due to the alteration of the cell cycle with loss of control at the G1/S checkpoint. In this study, we analyze the expression and modulation of CyD1 and E2F1 in colon explants from Crohn's disease (CD) patients. We used ex vivo culture of colon explants from 4 CD patients and 2 healthy controls, stimulated with lipopolysaccharide from Escherichia Coli (EC-LPS). Commercial siRNAs for CyD1 and E2F1 inhibition were encapsulated in Invivofectamine® and in purposely produced nanoliposomal vectors to silencing CyD1 and E2F1 expression. Western blot analysis was used to investigate the effect of siRNA on CyD1, E2F1 and cyclooxygenase 2 (COX-2) expression. In CD patients colon explants, CyD1 and E2F1 increased after the inflammatory stimulus but siRNA silencing attenuated their expression and controlled the COX-2 expression too. These data represent a prelimiary exploration of in vitro siRNA use.
Collapse
Affiliation(s)
- Ilaria Russo
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Albino Carrizzo
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Sabrina Bochicchio
- Department of Industrial Engineering, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) – ITALY,Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) - ITALY
| | - Ornella Piazza
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Gaetano Lamberti
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) - ITALY
| | - Anna Angela Barba
- Department of Industrial Engineering, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) – ITALY
| | - Carmine Vecchione
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Pio Zeppa
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Paola Iovino
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Cristina Bucci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Antonella Santonicola
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Carolina Ciacci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| |
Collapse
|
242
|
Nakano K, Takahashi S. Current Molecular Targeted Therapies for Bone and Soft Tissue Sarcomas. Int J Mol Sci 2018; 19:E739. [PMID: 29510588 PMCID: PMC5877600 DOI: 10.3390/ijms19030739] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/01/2018] [Accepted: 03/03/2018] [Indexed: 12/16/2022] Open
Abstract
Systemic treatment options for bone and soft tissue sarcomas remained unchanged until the 2000s. These cancers presented challenges in new drug development partly because of their rarity and heterogeneity. Many new molecular targeting drugs have been tried in the 2010s, and some were approved for bone and soft tissue sarcoma. As one of the first molecular targeted drugs approved for solid malignant tumors, imatinib's approval as a treatment for gastrointestinal stromal tumors (GISTs) has been a great achievement. Following imatinib, other tyrosine kinase inhibitors (TKIs) have been approved for GISTs such as sunitinib and regorafenib, and pazopanib was approved for non-GIST soft tissue sarcomas. Olaratumab, the monoclonal antibody that targets platelet-derived growth factor receptor (PDGFR)-α, was shown to extend the overall survival of soft tissue sarcoma patients and was approved in 2016 in the U.S. as a breakthrough therapy. For bone tumors, new drugs are limited to denosumab, a receptor activator of nuclear factor κB ligand (RANKL) inhibitor, for treating giant cell tumors of bone. In this review, we explain and summarize the current molecular targeting therapies approved and in development for bone and soft tissue sarcomas.
Collapse
Affiliation(s)
- Kenji Nakano
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo 135-8550, Japan.
| | - Shunji Takahashi
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo 135-8550, Japan.
| |
Collapse
|
243
|
Solute carrier family 34 member 2 overexpression contributes to tumor growth and poor patient survival in colorectal cancer. Biomed Pharmacother 2018; 99:645-654. [PMID: 29653487 DOI: 10.1016/j.biopha.2018.01.124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/14/2018] [Accepted: 01/24/2018] [Indexed: 02/07/2023] Open
Abstract
Solute carrier family 34 member 2 (SLC34A2) is a well-known sodium-dependent phosphate transporter that has recently been linked to cancer development. However, its specific oncogenic role remains controversial in numerous human malignancies, and is currently unknown in colorectal cancer (CRC). Therefore, in this study we firstly used Oncomine database to determine its expression in cancer tissues and found it is overexpressed in thyroid, ovarian and renal cancer, while it is opposite in lung, breast and pancreas cancer. Using qRT-PCR and western blot, we then demonstrated its overexpression in CRC tissues as compared with adjacent normal tissues (n = 20). In a retrospective cohort enrolling 190 CRC patients, we proved its expression was significantly correlated with N stage. Furthermore, high SLC34A2 expression is associated with higher postoperative metastasis rate and serves as an independent adverse factor affecting patient prognosis. In subgroup analysis, SLC34A2 expression could stratify the patient prognosis in stage II and III CRC, but failed in stage IV CRC. In cellular assays in vitro, knockdown of SLC34A2 dramatically inhibited the proliferation and colony formation, induced the apoptosis and arrests the cell cycle progression of HCT-116 CRC cells. In cellular assays in vivo, knockdown of SLC34A2 significantly inhibited the growth of xenografts, decreasing Ki-67 and proliferating cell nuclear antigen (PCNA) expression and increasing apoptosis rate. Taken together, our study indicates SLC34A2 plays a crucial promoting role in CRC development and therefore has great potential to be further developed as a reliable biomarker for CRC diagnosis and treatment.
Collapse
|
244
|
Zha C, Deng W, Fu Y, Tang S, Lan X, Ye Y, Su Y, Jiang L, Chen Y, Huang Y, Ding J, Geng M, Huang M, Wan H. Design, synthesis and biological evaluation of tetrahydronaphthyridine derivatives as bioavailable CDK4/6 inhibitors for cancer therapy. Eur J Med Chem 2018; 148:140-153. [PMID: 29459274 DOI: 10.1016/j.ejmech.2018.02.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 10/18/2022]
Abstract
CDK4/6 pathway is an attractive chemotherapeutic target for antitumor drug discovery and development. Herein, we reported the structure-based design and synthesis of a series of novel tetrahydronaphthyridine analogues as selective CDK4/6 inhibitors. Compound 5 was identified as a hit and then systematically structure optimization study was conducted. These efforts led to compound 28, which exhibited excellent in vitro potencies against CDK4/6 enzymatic activity with high selectivity over CDK1, and against Colo-205 cell growth. The compound demonstrated favorable in vitro metabolic and robust mice pharmacokinetic properties. In Colo-205 xenograft models, compound 28 showed potent tumor growth inhibition with acceptable toxic effects, which could serve as a novel anticancer agent for further preclinical study.
Collapse
Affiliation(s)
- Chuantao Zha
- Shanghai HaiHe Pharmaceutical, Co. Ltd, No. 421 Newton Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Wenjia Deng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Fu
- Shanghai HaiHe Pharmaceutical, Co. Ltd, No. 421 Newton Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Shuai Tang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Xiaojing Lan
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Yan Ye
- Shanghai HaiHe Pharmaceutical, Co. Ltd, No. 421 Newton Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Yi Su
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Lei Jiang
- Shanghai HaiHe Pharmaceutical, Co. Ltd, No. 421 Newton Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Yi Chen
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Ying Huang
- Shanghai HaiHe Pharmaceutical, Co. Ltd, No. 421 Newton Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Jian Ding
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Meiyu Geng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China
| | - Min Huang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China.
| | - Huixin Wan
- Shanghai HaiHe Pharmaceutical, Co. Ltd, No. 421 Newton Road, Zhangjiang Hi-tech Park, Pudong New Area, Shanghai 201203, China.
| |
Collapse
|
245
|
Bharate SB, Kumar V, Jain SK, Mintoo MJ, Guru SK, Nuthakki VK, Sharma M, Bharate SS, Gandhi SG, Mondhe DM, Bhushan S, Vishwakarma RA. Discovery and Preclinical Development of IIIM-290, an Orally Active Potent Cyclin-Dependent Kinase Inhibitor. J Med Chem 2018; 61:1664-1687. [PMID: 29370702 DOI: 10.1021/acs.jmedchem.7b01765] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rohitukine (1), a chromone alkaloid isolated from Indian medicinal plant Dysoxylum binectariferum, has inspired the discovery of flavopiridol and riviciclib, both of which are bioavailable only via intravenous route. With the objective to address the oral bioavailability issue of this scaffold, four series of rohitukine derivatives were prepared and screened for Cdk inhibition and cellular antiproliferative activity. The 2,6-dichloro-styryl derivative IIIM-290 (11d) showed strong inhibition of Cdk-9/T1 (IC50 1.9 nM) kinase and Molt-4/MIAPaCa-2 cell growth (GI50 < 1.0 μM) and was found to be highly selective for cancer cells over normal fibroblast cells. It inhibited the cell growth of MIAPaCa-2 cells via caspase-dependent apoptosis. It achieved 71% oral bioavailability with in vivo efficacy in pancreatic, colon, and leukemia xenografts at 50 mg/kg, po. It did not have CYP/efflux-pump liability, was not mutagenic/genotoxic or cardiotoxic, and was metabolically stable. The preclinical data presented herein indicates the potential of 11d for advancement in clinical studies.
Collapse
Affiliation(s)
- Sandip B Bharate
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India.,Academy of Scientific & Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Vikas Kumar
- Academy of Scientific & Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India.,Preformulation Laboratory, PK-PD Toxicology & Formulation Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Shreyans K Jain
- Natural Products Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Mubashir J Mintoo
- Academy of Scientific & Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Santosh K Guru
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Vijay K Nuthakki
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Mohit Sharma
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Sonali S Bharate
- Preformulation Laboratory, PK-PD Toxicology & Formulation Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Sumit G Gandhi
- Academy of Scientific & Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India.,Plant Biotechnology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Dilip M Mondhe
- Academy of Scientific & Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Shashi Bhushan
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India.,Indian Pharmacopeia Commission , Sec-23, Raj Nagar, Ghaziabad-201002, India
| | - Ram A Vishwakarma
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India.,Academy of Scientific & Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| |
Collapse
|
246
|
He L, Yang H, Zhou S, Zhu H, Mao H, Ma Z, Wu T, Kumar AK, Kathera C, Janardhan A, Pan F, Hu Z, Yang Y, Luo L, Guo Z. Synergistic antitumor effect of combined paclitaxel with FEN1 inhibitor in cervical cancer cells. DNA Repair (Amst) 2018; 63:1-9. [PMID: 29358095 DOI: 10.1016/j.dnarep.2018.01.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/06/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
Abstract
Studies on cervical cancer are urgently required to improve clinical outcomes. As a major anticancer drug for cervical cancer, paclitaxel has been used for many years in clinical therapy but its therapeutic efficacy is limited by common obstacle from cancer cells. The enhanced DNA repair pathways of cancer cells have been proved to survive DNA damage induced by chemotherapeutic drug. Inhibitors of specific DNA repair pathway can sensitize cancer cells to the treatment of chemotherapeutic drugs. In this paper we found that the effect of paclitaxel can be significantly improved when used in combination with FEN1 inhibitor SC13, suggesting a synergistic mechanism between the two compounds. Our studies suggest that FEN1 inhibition could be a novel strategy of tumor-targeting therapy for cervical cancer. Our work also revealed that paclitaxel demonstrates stronger synergistic effect with SC13 than other common used chemical drugs such as doxorubicin, carboplatin or camptothecin on cervical cancer cells.
Collapse
Affiliation(s)
- Lingfeng He
- Changzhou No. 7 People's Hospital, China; Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Huan Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Shiying Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Hong Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Huiwen Mao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Zhuang Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Ting Wu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Alagamuthu Karthick Kumar
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Chandrasekhar Kathera
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Avilala Janardhan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | | | - Libo Luo
- Changzhou No. 7 People's Hospital, China.
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China.
| |
Collapse
|
247
|
Abstract
During the last decades, much has been learned about with cyclin-dependent kinases (CDK) playing a pivotal role in the cell cycle regulation. CDK4/6 is the key regulator of the G1-S transition. Palbociclib (PD 0332991, Ibrance®) is the first oral CDK4/6 inhibitor showing a substantially improved median progression-free survival (PFS) in advanced estrogen receptor (ER) positive and human epidermal growth factor receptor 2 (HER2) negative breast cancer. This PFS prolongation was seen both with letrozole as first-line therapy (24.8 vs. 14.5 months [PALOMA 2]) and with fulvestrant in endocrine pretreated patients (9.2 vs. 3.8 months [PALOMA-3]). The main toxicity is neutropenia due to cell cycle arrest which can be easily managed with dose interruption or dose reduction leading to a favorable safety profile with delayed deterioration of global quality of life (QoL). Palbociclib is approved by the Federal Drug Administration (FDA) and the European Medicines Agency (EMA) for ER-positive/HER2-negative advanced breast cancer. Despite the well-understood mode of action of palbociclib, predictive biomarkers are not yet defined. In conclusion, inhibition of CDK4/6 using palbociclib in combination with endocrine therapy is an efficient and well-tolerated treatment option in ER-positive/HER2-negative advanced breast cancer. Ongoing clinical trials are investigating the role of palbociclib in early breast cancer as well as in other types of cancer.
Collapse
Affiliation(s)
- Marcus Schmidt
- Department of Obstetrics and Gynecology, University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Martin Sebastian
- Department of Hematology/Oncology, Rheumatology, HIV, J.W. Goethe University, Frankfurt, Germany
| |
Collapse
|
248
|
Abstract
OBJECTIVE The goals of this review are to provide background information on the definitions and applications of the general term "biomarker" and to highlight the specific roles of breast imaging biomarkers in research and clinical breast cancer care. A search was conducted of the main electronic biomedical databases (PubMed, Cochrane, Embase, MEDLINE [Ovid], Scopus, and Web of Science). The search was focused on review literature in general radiology and biomedical sciences and on reviews and primary research articles on biomarkers in breast imaging over the 15 years ending in June 2017. The keywords included "biomarker," "trial endpoints," "breast imaging," "breast cancer," "radiomics," and "precision medicine" in the titles and abstracts of the papers. CONCLUSION Clinical breast care and breast cancer-related research rely on imaging biomarkers for decision support. In the era of precision medicine and big data, the practice of radiology is likely to change. A closer integration of breast imaging with related biomedical fields and the creation of large integrated and shareable databases of clinical, molecular, and imaging biomarkers should allow the field to continue guiding breast cancer care and research.
Collapse
|
249
|
Kosovec JE, Zaidi AH, Omstead AN, Matsui D, Biedka MJ, Cox EJ, Campbell PT, Biederman RW, Kelly RJ, Jobe BA. CDK4/6 dual inhibitor abemaciclib demonstrates compelling preclinical activity against esophageal adenocarcinoma: a novel therapeutic option for a deadly disease. Oncotarget 2017; 8:100421-100432. [PMID: 29245989 PMCID: PMC5725031 DOI: 10.18632/oncotarget.22244] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/16/2017] [Indexed: 12/21/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is a deadly disease with limited therapeutic options. In the present study, we determined the preclinical efficacy of CDK4/6 inhibitor abemaciclib for treatment of EAC. In vitro, apoptosis, proliferation, and pathway regulation were evaluated in OE19, OE33, and FLO1 EAC cell lines. In vivo, esophagojejunostomy was performed on rats to induce EAC. At 36 weeks post-surgery, MRI and endoscopic biopsy established baseline tumor volume and molecular correlates, respectively. Next, the study animals were randomized to 26mg/kg intraperitoneal abemaciclib treatment or vehicle control for 28 days. Pre and post treatment MRIs, histopathology, and qRT-PCR were utilized to determine response. Our results demonstrated treatment with abemaciclib lead to increased apoptosis, and decreased proliferation in OE19 (p=0.185), OE33 (p=0.048), and FLO1 (p=0.043) with anticipated downstream molecular inhibition. In vivo, 78.9% of treatment animals demonstrated >20% tumor volume decrease (placebo 0%). Mean tumor volume changed in the treatment arm by -65.5% (placebo +133.5%) (p<0.01), and prevalence changed by -37.5% (placebo +16.7%) (p<0.01). Pre vs post treatment qRT-PCR demonstrated significant inhibition of all downstream molecular correlates. Overall our findings suggest potent antitumor efficacy of abemaciclib against EAC with evident molecular pathway inhibition and reasonable safety, establishing the rationale for future clinical development.
Collapse
Affiliation(s)
- Juliann E. Kosovec
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ali H. Zaidi
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ashten N. Omstead
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Daisuke Matsui
- Department of Gastroenterological Surgery, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Mark J. Biedka
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Erin J. Cox
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Patrick T. Campbell
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | | | - Ronan J. Kelly
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Blair A. Jobe
- Esophageal and Lung Institute, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
250
|
Mills CC, Kolb EA, Sampson VB. Recent Advances of Cell-Cycle Inhibitor Therapies for Pediatric Cancer. Cancer Res 2017; 77:6489-6498. [PMID: 29097609 DOI: 10.1158/0008-5472.can-17-2066] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/24/2017] [Accepted: 09/19/2017] [Indexed: 12/21/2022]
Abstract
This review describes the pivotal roles of cell-cycle and checkpoint regulators and discusses development of specific cell-cycle inhibitors for therapeutic use for pediatric cancer. The mechanism of action as well as the safety and tolerability of drugs in pediatric patients, including compounds that target CDK4/CDK6 (palbociclib, ribociclib, and abemaciclib), aurora kinases (AT9283 and MLN8237), Wee1 kinase (MK-1775), KSP (ispinesib), and tubulin (taxanes, vinca alkaloids), are presented. The design of mechanism-based combinations that exploit the cross-talk of signals activated by cell-cycle arrest, as well as pediatric-focused drug development, are critical for the advancement of drugs for rare childhood diseases. Cancer Res; 77(23); 6489-98. ©2017 AACR.
Collapse
Affiliation(s)
| | - E A Kolb
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Valerie B Sampson
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware.
| |
Collapse
|