1
|
Ralte L, Sailo H, Kumar R, Khiangte L, Kumar NS, Singh YT. Identification of novel AKT1 inhibitors from Sapria himalayana bioactive compounds using structure-based virtual screening and molecular dynamics simulations. BMC Complement Med Ther 2024; 24:116. [PMID: 38454426 PMCID: PMC10921764 DOI: 10.1186/s12906-024-04415-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/27/2024] [Indexed: 03/09/2024] Open
Abstract
Through the experimental and computational analyses, the present study sought to elucidate the chemical composition and anticancer potential of Sapria himalayana plant extract (SHPE). An in vitro analysis of the plant extract was carried out to determine the anticancer potential. Further, network pharmacology, molecular docking, and molecular dynamic simulation were employed to evaluate the potential phytochemical compounds for cervical cancer (CC) drug formulations. The SHPE exhibited anti-cancerous potential through inhibition properties against cancer cell lines. The LC-MS profiling showed the presence of 14 compounds in SHPE. Using network pharmacology analysis, AKT1 (AKT serine/threonine kinase 1) is identified as the possible potential target, and EGFR (Epidermal Growth Factor Receptor) is identified as the possible key signal pathway. The major targets were determined to be AKT1, EGFR by topological analysis and molecular docking. An in silico interaction of phytoconstituents employing molecular docking demonstrated a high binding inclination of ergoloid mesylate and Ergosta-5,7,9(11),22-tetraen-3-ol, (3.beta.,22E)- with binding affinities of -15.5 kcal/mol, and -11.3 kcal/mol respectively. Further, MD simulation and PCA analyses showed that the phytochemicals possessed significant binding efficacy with CC protein. These results point the way for more investigation into SHPE compound's potential as CC treatment.
Collapse
Affiliation(s)
- Laldinfeli Ralte
- Department of Botany, Mizoram University, Aizawl, Mizoram, 796004, India
| | - Hmingremhlua Sailo
- Department of Botany, Mizoram University, Aizawl, Mizoram, 796004, India
| | - Rakesh Kumar
- Department of Botany, Mizoram University, Aizawl, Mizoram, 796004, India
| | | | | | - Yengkhom Tunginba Singh
- Department of Botany, Mizoram University, Aizawl, Mizoram, 796004, India.
- Department of Life Sciences (Botany), Manipur University, Imphal, Manipur, 795003, India.
| |
Collapse
|
2
|
Upadhyay L, Hartzell M, Parikh AR, Strickland MR, Klempner S, Malla M. Recent Advances in the Management of Anal Cancer. Healthcare (Basel) 2023; 11:3010. [PMID: 38063578 PMCID: PMC10706124 DOI: 10.3390/healthcare11233010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 08/29/2024] Open
Abstract
The incidence and mortality of squamous cell carcinoma of the anus (SCCA) is on the rise, which highlights the unmet need for advances in treatment options. The landscape of treatment for this cancer is rapidly evolving with novel combination strategies including immunotherapy, radiation therapy and biomarker-guided therapy. This review article features an overview of recent advancements in both locoregional and metastatic SCCA. The recent focus on locoregional SCCA management is to tailor treatment according to tumor burden and minimize treatment-related toxicities. Mitomycin plus either infusional 5-fluorouracil (5-FU) or capecitabine is used for first-line chemoradiotherapy (CRT), and intensity-modulated radiotherapy (IMRT) is the preferred modality for radiation for locoregional anal cancer. Locally recurrent disease is managed with surgical resection. Systemic treatment is first-line for metastatic SCCA and immunotherapy with nivolumab and pembrolizumab being included as second-line agents. Current and future clinical trials are evaluating treatments for SCCA including immunotherapy alone or in combination regimens, radiotherapies, targeted treatments and novel agents. Another critical aspect of current research in SCCA is the personalization of CRT and immunotherapies based on molecular characterization and biomarkers such as the programmed death-ligand 1 (PD-L1), epidermal growth factor receptor (EGFR) and circulating tumor DNA.
Collapse
Affiliation(s)
- Laxmi Upadhyay
- Department of Medicine, West Virginia University, Morgantown, WV 26506, USA; (L.U.); (M.H.)
| | - Michelle Hartzell
- Department of Medicine, West Virginia University, Morgantown, WV 26506, USA; (L.U.); (M.H.)
| | - Aparna R. Parikh
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; (A.R.P.); (M.R.S.); (S.K.)
| | - Matthew R. Strickland
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; (A.R.P.); (M.R.S.); (S.K.)
| | - Samuel Klempner
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; (A.R.P.); (M.R.S.); (S.K.)
| | - Midhun Malla
- O’Neal Comprehensive Cancer Center, The University of Alabama, Birmingham, AL 35294, USA
| |
Collapse
|
3
|
Liu Y, Zheng P, Jiao T, Zhang M, Wu Y, Zhang X, Wang S, Zhao Z. Paiteling induces apoptosis of cervical cancer cells by down-regulation of the E6/E7-Pi3k/Akt pathway: A network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2023; 305:116062. [PMID: 36535331 DOI: 10.1016/j.jep.2022.116062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/29/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Human papillomavirus (HPV) infection is considered to be the main pathogen causing intraepithelial neoplasia. Paiteling (PTL) has been used to treat intraepithelial neoplasia caused by human papillomavirus (HPV) infection for more than 20 years in China, but its specific mechanism of action is not very clear, and further research is still needed. OBJECTIVE This study designed a comprehensive strategy to study the pharmacological mechanism of paiteling in regulating cervical cancer cell apoptosis by integrating LC-MS/MS, network pharmacology and pharmacological experiments. METHODS We used liquid chromatography-tandem mass spectrometry to detect the active substances in PTL and performed protein-protein interaction analysis on the intersection of the targets of these key compounds and the targets of intraepithelial neoplasia. Additionally, by using Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes (KEGG), the potential pathway of PTL against HPV-induced intraepithelial neoplasia was predicted. Finally, we used HeLa and Ect1/E6E7 cells for experimental verification. RESULTS The protein-protein interaction network predicted that AKT1, TP53, MYC, STAT3, MTOR, and MAPK were pivotal targets for PTL to inhibit epithelial neoplasia. KEGG enrichment analysis showed that the Pi3k/Akt pathway and HPV infection had scientific significance. Compared to the control group, after PTL diluent stimulated HeLa and Ect1/E6E7 cells for 24 h, cell viability, migration, and invasion capabilities were significantly reduced, and cell apoptosis was significantly increased, conforming to a dose-effect relationship and time-effect relationship. PCR, cellular immunohistochemistry, and western blot experiments showed that PTL reduced the expression of E6, Pi3k, E7, Akt, Bcl-xl, while increasing the expression of Bad in HeLa and Ect1/E6E7 cells. CONCLUSION PTL can induce cervical cancer cell apoptosis by inhibiting the E6/E7-Pi3k/Akt signaling pathway. It may provide an effective alternative strategy of traditional Chinese medicine for the treatment of epithelial neoplasia caused by HPV infection.
Collapse
Affiliation(s)
- Yunhua Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tingting Jiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Mengmeng Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yingjie Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xinjiang Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuyue Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zongjiang Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
4
|
Elasbali AM, Al-Soud WA, Mousa Elayyan AE, Al-Oanzi ZH, Alhassan HH, Mohamed BM, Alanazi HH, Ashraf MS, Moiz S, Patel M, Patel M, Adnan M. Integrating network pharmacology approaches for the investigation of multi-target pharmacological mechanism of 6-shogaol against cervical cancer. J Biomol Struct Dyn 2023; 41:14135-14151. [PMID: 36943780 DOI: 10.1080/07391102.2023.2191719] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/02/2023] [Indexed: 03/23/2023]
Abstract
Traditional treatment of cancer has been plagued by a number of obstacles, such as multiple drug resistance, toxicity and financial constraints. In contrast, phytochemicals that modulate a variety of molecular mechanisms are garnering increasing interest in complementary and alternative medicine. Therefore, an approach based on network pharmacology was used in the present study to explore possible regulatory mechanisms of 6-shogaol as a potential treatment for cervical cancer (CC). A number of public databases were screened to collect information on the target genes of 6-shogaol (SuperPred, Targetnet, Swiss target prediction and PharmMapper), while targets pertaining to CC were taken from disease databases (DisGeNet and Genecards) and gene expression omnibus (GEO) provided expression datasets. With STRING and Cytoscape, protein-protein interactions (PPI) were generated and topology analysis along with CytoNCA were used to identify the Hub genes. The Gene Ontology (GO) database Enrichr was used to annotate the target proteins, while, using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, signaling pathway enrichment analysis was conducted. Molecular docking and survival analysis for the Hub genes revealed four genes (HSP90AA1, HRAS, ESR1 and EGFR) with lowest binding energy and majority of the Hub genes (EGFR, SRC, CASP-3, HSP90AA1, MTOR, MAPK-1, MDM2 and ESR1) were linked with the overall survival of CC patients. In conclusion, the present study provides the scientific evidence which strongly supports the use of 6-shogoal as an inhibitor of cellular proliferation, growth, migration as well as inducer of apoptosis via targeting the hub genes involved in the growth of CC.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Waleed Abu Al-Soud
- Department of Clinical Laboratory Science, College of Applied Sciences-Sakaka, Jouf University, Saudi Arabia
| | - Afnan Elayyan Mousa Elayyan
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Ziad H Al-Oanzi
- Department of Clinical Laboratory Science, College of Applied Sciences-Sakaka, Jouf University, Saudi Arabia
| | - Hassan H Alhassan
- Department of Clinical Laboratory Science, College of Applied Sciences-Sakaka, Jouf University, Saudi Arabia
| | - Bashir M Mohamed
- Trinity St. James's Cancer Institute, Dublin, Ireland
- Department of Histopathology, Trinity College of Dublin, Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin, Ireland
- Department of Obstetrics and Gynecology, Trinity College of Dublin, Dublin, Ireland
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Mohammad Saquib Ashraf
- Department of Medical Laboratory Science, College of Applied Medical Sciences, Riyadh ELM University (REU), Riyadh, Saudi Arabia
| | - Shadman Moiz
- Department of Biotechnology, Lalit Narayan Mithila University, Darbhanga, Bihar, India
| | - Mitesh Patel
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, India
| | - Mirav Patel
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| |
Collapse
|
5
|
Lin Z, Zhao Y, Li Q, Ci X, Ye X, Chen G, Tu Q, Feng W, Jiang P, Zhu S, Xue X, Saunders NA, Zhang L, Zhu X, Zhao KN. OUP accepted manuscript. Carcinogenesis 2022; 43:479-493. [PMID: 35134836 DOI: 10.1093/carcin/bgac010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/19/2022] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Zhongmin Lin
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Yu Zhao
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital and Yuyin Children Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Qijia Li
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital and Yuyin Children Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xingyuan Ci
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital and Yuyin Children Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xiaoxian Ye
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Guorong Chen
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Quanmei Tu
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Weixu Feng
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Pengfei Jiang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Shanli Zhu
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xiangyang Xue
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Nicholas A Saunders
- Diamantina Institute for Cancer Immunology and Metabolic Medicine, The University of Queensland, TRI, Woolloongabba, Queensland, Australia
| | - Lifang Zhang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital and Yuyin Children Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Kong-Nan Zhao
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital and Yuyin Children Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
6
|
Armstrong SA, Malley R, Wang H, Lenz HJ, Arguello D, El-Deiry WS, Xiu J, Gatalica Z, Hwang JJ, Philip PA, Shields AF, Marshall JL, Salem ME, Weinberg BA. Molecular characterization of squamous cell carcinoma of the anal canal. J Gastrointest Oncol 2021; 12:2423-2437. [PMID: 34790403 DOI: 10.21037/jgo-20-610] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background Squamous cell carcinoma of the anal canal (SCCA) is an uncommon malignancy with limited therapeutic options. Nivolumab and pembrolizumab show promising results in patients with SCCA. Human papillomavirus (HPV)-negative tumors are frequently TP53-mutated (TP53-MT) and often resistant to therapy. Methods We present a large molecularly-profiled cohort of SCCA, exploring the underlying biology of SCCA, differences between TP53-wild type (TP53-WT) and TP53-MT tumors, and differences between local and metastatic tumors. SCCA specimens (n=311) underwent multiplatform testing with immunohistochemistry (IHC), in situ hybridization (ISH) and next-generation sequencing (NGS). Tumor mutational burden (TMB) was calculated using only somatic nonsynonymous missense mutations. Chi-square testing was used for comparative analyses. Results The most frequently mutated genes included PIK3CA (28.1%), KMT2D (19.5%), FBXW7 (12%), TP53 (12%) and PTEN (10.8%). The expression of PD-1 was seen in 68.8% and PD-L1 in 40.5% of tumors. High TMB was present in 6.7% of specimens. HER2 IHC was positive in 0.9%, amplification by chromogenic in situ hybridization (CISH) was seen 1.3%, and mutations in ERBB2 were present in 1.8% of tumors. The latter mutation has not been previously described in SCCA. When compared with TP53-WT tumors, TP53-MT tumors had higher rates of CDKN2A, EWSR1, JAK1, FGFR1 and BRAF mutations. PD-1 and PD-L1 expression were similar, and high TMB did not correlate with PD-1 (P=0.50) or PD-L1 (P=0.52) expression. Conclusions Molecular profiling differences between TP53-MT and TP53-WT SCCA indicate different carcinogenic pathways which may influence response to therapy. Low frequency mutations in several druggable genes may provide therapeutic opportunities for patients with SCCA.
Collapse
Affiliation(s)
- Samantha A Armstrong
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Rita Malley
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Hongkun Wang
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, Los Angeles, CA, USA
| | | | | | | | | | - Jimmy J Hwang
- Department of Hematology/Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Philip A Philip
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Anthony F Shields
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - John L Marshall
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Mohamed E Salem
- Department of Hematology/Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Benjamin A Weinberg
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
7
|
Carr RM, Jin Z, Hubbard J. Research on Anal Squamous Cell Carcinoma: Systemic Therapy Strategies for Anal Cancer. Cancers (Basel) 2021; 13:cancers13092180. [PMID: 34062753 PMCID: PMC8125190 DOI: 10.3390/cancers13092180] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Anal cancer is rare with an estimated 9000 new cases predicted to occur in the United States in 2021. However, rates of new anal cancer cases and deaths from the disease are increasing by about 2% and 3% per year respectively. In light of these trends it is critical to better understand the nature of this disease and progress in its management. The present review focuses on the history and development of the role of systemic therapy in the treatment of anal cancer. Major trials establishing the role of chemotherapy in the management of locoregional and metastatic anal cancer are summarized. In addition, the rapidly evolving role of immunotherapy is discussed. Finally, major insights into the molecular pathobiology of anal cancer and opportunities for advancement in precision medicine in treatment of the disease. Abstract Anal squamous cell carcinoma (ASCC) is a rare malignancy, with most cases associated with human papilloma virus and an increased incidence in immunocompromised patients. Progress in management of ASCC has been limited not only due to its rarity, but also the associated lack of research funding and social stigma. Historically, standard of care for invasive ASCC has been highly morbid surgical resection, requiring a permanent colostomy. Surgery was associated with disease recurrence in approximately half of the patients. However, the use of chemotherapy (5-fluorouracil and mitomycin C) concomitantly with radiation in the 1970s resulted in disease regression, curing a subset of patients and sparing them from morbid surgery. Validation of the use of systemic therapy in prospective trials was not achieved until approximately 20 years later. In this review, advancements and shortcomings in the use of systemic therapy in the management of ASCC will be discussed. Not only will standard-of-care systemic therapies for locoregional and metastatic disease be reviewed, but the evolving role of novel treatment strategies such as immune checkpoint inhibitors, HPV-based vaccines, and molecularly targeted therapies will also be covered. While advances in ASCC treatment have remained largely incremental, with increased biological insight, an increasing number of promising systemic treatment modalities are being explored.
Collapse
|
8
|
Reddy D, Kumavath R, Tan TZ, Ampasala DR, Kumar AP. Peruvoside targets apoptosis and autophagy through MAPK Wnt/β-catenin and PI3K/AKT/mTOR signaling pathways in human cancers. Life Sci 2019; 241:117147. [PMID: 31830480 DOI: 10.1016/j.lfs.2019.117147] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022]
Abstract
AIM To investigate the cytotoxic effect of Peruvoside and mechanism of action in human cancers. MAIN METHODS Cell viability was measured by MTT assay and the cell cycle arrest was identified by FACS. Real-time qPCR and western blotting studies were performed to identify important gene and protein expressions in the different pathways leading to apoptosis. Immunofluorescence was performed to understand protein localization and molecular docking studies were performed to identify protein-ligand interactions. KEY FINDINGS Peruvoside showed significant anti-proliferative activities against human breast, lung, and liver cancer cells in dose-dependent manner. The anti-cancer mechanism was further confirmed by DNA damage and cell cycle arrest at the G0/G1 phase. Dysregulation of Wnt/β-catenin signaling with Peruvoside treatment resulted in inhibition of cyclin D1 and c-Myc also observed in this study. Furthermore, we identified that Peruvoside can inhibit autophagy by PI3K/AKT/mTOR signaling and through downregulating MEK1. Moreover, Peruvoside has the ability to modulate the expressions of key proteins from the cell cycle, MAPK, NF-kB, and JAK-STAT signaling. In silico studies revealed that Peruvoside has the ability to interact with crucial proteins from different biochemical signaling pathways. SIGNIFICANCE Our results demonstrated that Peruvoside has the ability to inhibit cancer cell proliferation by modulating the expression of various key proteins involved in cell cycle arrest, apoptosis, and autophagic cell death. Clinical data generated from the present study might provide a novel impetus for targeting several human cancers. Conclusively, our findings suggest that the Peruvoside possesses a broad spectrum of anticancer activity in breast, lung, and liver cancers, which provides an impetus for further investigation of the anticancer potentiality of this biomolecule.
Collapse
Affiliation(s)
- Dhanasekhar Reddy
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (P.O), Kasaragod, Kerala 671320, India
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (P.O), Kasaragod, Kerala 671320, India.
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Dinakara Rao Ampasala
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
9
|
Chauhan N, Maher DM, Yallapu MM, B Hafeez B, Singh MM, Chauhan SC, Jaggi M. A triphenylethylene nonsteroidal SERM attenuates cervical cancer growth. Sci Rep 2019; 9:10917. [PMID: 31358785 PMCID: PMC6662837 DOI: 10.1038/s41598-019-46680-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/28/2019] [Indexed: 11/09/2022] Open
Abstract
Selective estrogen receptor modulator drug molecules of triphenylethylene family have gained considerable attention as anti-cancer agents. Despite recent advances in screening and development of HPV vaccines, cervical cancer remains one of the deadliest malignancies as advanced stage metastatic disease is mostly untreatable, thus warrants newer therapeutic strategies. Ormeloxifene (ORM) is a well-known SERM of triphenylethylene family that has been approved for human use, thus represents an ideal molecule for repurposing. In this study, we for the first time have demonstrated the anti-cancerous properties of ormeloxifene in cervical cancer. Ormeloxifene efficiently attenuated tumorigenic and metastatic properties of cervical cancer cells via arresting cell cycle at G1-S transition, inducing apoptosis, decreasing PI3K and Akt phosphorylation, mitochondrial membrane potential, and modulating G1-S transition related proteins (p21, cyclin E and Cdk2). Moreover, ORM repressed the expression of HPV E6/ E7 oncoproteins and restored the expression of their downstream target tumor suppressor proteins (p53, Rb and PTPN 13). As a result, ormeloxifene induces radio-sensitization in cervical cancer cells and caused potent tumor growth inhibition in orthotopic mouse model. Taken together, ormeloxifene represents an alternative therapeutic modality for cervical cancer which may have rapid clinical translation as it is already proven safe for human use.
Collapse
Affiliation(s)
- Neeraj Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 38163, Memphis, TN, USA.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, 78504, McAllen, TX, USA
| | - Diane M Maher
- Sanford Research Center, USD, 57104, Sioux Falls, SD, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 38163, Memphis, TN, USA.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, 78504, McAllen, TX, USA
| | - Bilal B Hafeez
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 38163, Memphis, TN, USA.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, 78504, McAllen, TX, USA
| | - Man M Singh
- Saraswati Dental College, Lucknow, Uttar Pradesh, India
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 38163, Memphis, TN, USA. .,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, 78504, McAllen, TX, USA.
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 38163, Memphis, TN, USA. .,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, 78504, McAllen, TX, USA.
| |
Collapse
|
10
|
Chiarini A, Liu D, Rassu M, Armato U, Eccher C, Dal Prà I. Over Expressed TKTL1, CIP-2A, and B-MYB Proteins in Uterine Cervix Epithelium Scrapings as Potential Risk Predictive Biomarkers in HR-HPV-Infected LSIL/ASCUS Patients. Front Oncol 2019; 9:213. [PMID: 31001477 PMCID: PMC6456695 DOI: 10.3389/fonc.2019.00213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 03/11/2019] [Indexed: 01/11/2023] Open
Abstract
High oncogenic risk human papillomaviruses (HR-HPVs) promote cervical carcinoma development, the fourth most common feminine cancer. A slow oncodevelopmental phase—defined histopathologically as Cervical Intraepithelial Neoplasia (CIN) grades 1–3, or cytologically as Low- or High-grade Squamous Intraepithelial Lesions (LSIL or HSIL)—precedes the malignancy. Cervical carcinoma screenings through HR-HPV genotyping and Pap smears are regularly performed in Western countries. Faulty cytology screening or genotyping or patients' non-compliance with follow-ups can let slip an oncoprogression diagnosis. Novel biomarker tests flanking HR-HPV genotyping and cytology could objectively predict the risk of disease progression thus helping triage LSIL/ASCUS patients. Here, anonymized leftovers of fresh cervical epithelium scrapings from twice (LSIL/ASCUS and HR-HPV DNA)-positive and twice (Pap smear- and HR-HPV DNA)-negative (control) patients in a proteome-preserving solution served to assess the biomarker worth of three cervical carcinoma-related proteins, i.e., B-MYB (or MYBL2), Cancerous Inhibitor of PP2A (CIP-2a), and transketolase-like1 (TKTL1). Leftovers anonymity was strictly kept and storage at −80°C, protein extraction, immunoblotting, and band densitometry were blindly performed. Only after tests completion, the anonymous yet code-corresponding HR-HPV-genotyping and cytology data allowed to assign each sample to the twice-positive or twice-negative group. Descriptive statistics showed that the three proteins levels significantly increased in the twice-positive vs. twice-negative scrapings. Diagnostic ROC curve analysis identified each protein's Optimal Decision Threshold (OTD) showing that TKTL1 and CIP-2a are stronger risk predictive biomarkers (Sensitivity, 0.91–0.93; Specificity, 0.77–0.83) than B-MYB. Logistic Regression coupled with Likelihood-Ratio Tests confirmed that a highly significant relation links increasing TKTL1/CIP-2a/B-MYB protein levels in twice-positive cervical scrapings to the risk of HR-HPV-driven oncoprogression. Finally, a 3 year clinical follow-up showed that 13 patients (50% of total) of the twice-positive group with biomarker values over OTDs compliantly underwent scheduled colposcopy and biopsy. Of these, 11 (i.e., 84.7%) received a positive histological diagnosis, i.e., CIN1 (n = 5; 38.5%) or CIN2/CIN2+ (n = 6; 46,2%). Therefore, TKTL1/CIP-2a/B-MYB protein levels could objectively predict oncoprogression risk in twice (HR-HPV- and Pap smear)-positive women. Further studies will assess the translatability of these findings into clinical settings.
Collapse
Affiliation(s)
- Anna Chiarini
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Daisong Liu
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy.,Plastic Surgery Department, Xiangya Third Hospital, Central South University, Changsha, China
| | - Mario Rassu
- Microbiology and Virology Unit, San Bortolo Hospital, Vicenza, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | | | - Ilaria Dal Prà
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| |
Collapse
|
11
|
Marref I, Reichling C, Vendrely V, Mouillot T. Prise en charge du cancer du canal anal en 2018. ONCOLOGIE 2018. [DOI: 10.3166/onco-2018-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
12
|
Glynne-Jones R, Harrison M. Cetuximab in the Context of Current Treatment of Squamous Cell Carcinoma of the Anus. J Clin Oncol 2017; 35:699-701. [PMID: 28068173 DOI: 10.1200/jco.2016.70.9394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Rob Glynne-Jones
- Rob Glynne-Jones and Mark Harrison, Centre for Cancer Treatment, Mount Vernon Hospital, Northwood, United Kingdom
| | - Mark Harrison
- Rob Glynne-Jones and Mark Harrison, Centre for Cancer Treatment, Mount Vernon Hospital, Northwood, United Kingdom
| |
Collapse
|
13
|
|
14
|
Phase II Study of Capecitabine in Substitution of 5-FU in the Chemoradiotherapy Regimen for Patients with Localized Squamous Cell Carcinoma of the Anal Canal. J Gastrointest Cancer 2016; 47:75-81. [PMID: 26691173 DOI: 10.1007/s12029-015-9790-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
UNLABELLED This was a phase II study of capecitabine in substitution of 5-fluorouracil (5-FU) in the chemoradiotherapy regimen for patients with localized squamous cell carcinoma of the anal canal. BACKGROUND Combined chemoradiation with infusional 5-FU and mitomycin is the standard treatment for localized squamous cell carcinoma (SCC) of the anal canal. Capecitabine is an oral fluoropirimidine that has been shown to be equally effective to 5-FU in many solid tumors. However, the efficacy of the substitution of 5-FU for capecitabine in anal SCC needs confirmation. METHODS Patients with SCC of anal cancer T2-4N0M0 or T (any) N1-3M0, with good performance status and normal blood and renal function, were treated with capecitabine 825 mg/m(2) bid during radiotherapy associated with a single dose of mitomycin 15 mg/m(2) on day 1. The primary objective was local control rate at 6 months determined by clinical examination and radiological assessment. Sample size was calculated using the Fleming single-stage design. RESULTS From November 2010 to February 2014, N = 51 patients were initially included; however, 43 patients were assessed. Seventeen patients (39.5%) were stage II, 11 patients (25.6%) stage IIIA, and 15 patients (34.9%) stage IIIB. Four patients (9.3%) were HIV positive. With a median follow-up of 23.1 months (range 4 to 44.4 months), 3 patients (7%) presented partial response, 37 (86%) had complete response, and 3 patients developed progression of the disease (7%) at 6 months. The colostomy rate was 18.6%. It was observed a locoregional control of 86% in 6 months (CI 95% 0.72-0.94). The main grade 3-4 toxicities were grade 3 radiodermitis in 10 patients (23.2%), grade 3 lymphopenia in 5 patients (11.6%), and grade 3 neutropenia in 2 patients (6.9%). One HIV-positive patient had septic shock, pneumonia, herpetic encephalitis, atrial fibrillation, and macrophage activation syndrome. CONCLUSIONS Capecitabine can safely substitute infusional 5-FU in the standard chemoradiation regimen for SCC of the anal cancer, with a locoregional control of 86% in 6 months (CI 95% 0.72-0.94).
Collapse
|
15
|
Glynne-Jones R, Saleem W, Harrison M, Mawdsley S, Hall M. Background and Current Treatment of Squamous Cell Carcinoma of the Anus. Oncol Ther 2016; 4:135-172. [PMID: 28261646 PMCID: PMC5315080 DOI: 10.1007/s40487-016-0024-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Indexed: 12/19/2022] Open
Abstract
In this review, a summary of our current understanding of squamous cell carcinoma of the anus (SCCA) and the advances in our knowledge of SCCA regarding screening, prevention, the role of the immune system, current treatment and the potential for novel targets are discussed. The present standard of care in terms of treatment is 5-fluorouracil (5-FU) and mitomycin C (MMC) concurrently with radiation, which results in a high level of disease control for small early cancers. Preservation of the anal sphincter is achieved in the majority, although anorectal function is often impaired. Although evidence from prospective studies to support a change in the treatment strategy is lacking, patients with HPV-negative SCCA appear to be less responsive to chemoradiation (CRT) and relapse more frequently. In contrast, HPV-positive tumours usually fare better, but oncological outcomes are modified by smoking and immune incompetence. There is current interest in escalating the radiotherapy dose for larger, more advanced tumours, and de-escalating treatment for HPV-positive tumours. The use of novel immunological treatments to target the underlying different molecular pathways of HPV-positive cancers is exciting.
Collapse
Affiliation(s)
- Rob Glynne-Jones
- Centre for Cancer Treatment, Mount Vernon Hospital, Northwood, Middlesex UK
| | - Waqar Saleem
- Centre for Cancer Treatment, Mount Vernon Hospital, Northwood, Middlesex UK
| | - Mark Harrison
- Centre for Cancer Treatment, Mount Vernon Hospital, Northwood, Middlesex UK
| | - Suzy Mawdsley
- Centre for Cancer Treatment, Mount Vernon Hospital, Northwood, Middlesex UK
| | - Marcia Hall
- Centre for Cancer Treatment, Mount Vernon Hospital, Northwood, Middlesex UK
| |
Collapse
|
16
|
Mutational analysis of anal cancers demonstrates frequent PIK3CA mutations associated with poor outcome after salvage abdominoperineal resection. Br J Cancer 2016; 114:1387-94. [PMID: 27219019 PMCID: PMC4984471 DOI: 10.1038/bjc.2016.144] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/19/2016] [Accepted: 04/27/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A better understanding of the molecular profile of anal squamous cell carcinomas (ASCCs) is necessary to consider new therapeutic approaches, and the identification of prognostic and predictive factors for response to treatment. METHODS We retrospectively analysed tumours from ASCC patients for mutational analysis of KRAS, NRAS, HRAS, BRAF, PIK3CA, MET, TP53 and FBXW7 genes by HRM and Sanger sequencing analysis. RESULTS Specimens from 148 patients were analysed: 96 treatment-naive tumours and 52 recurrences after initial radiotherapy (RT) or chemoradiotherapy (CRT). Mutations of KRAS, PIK3CA, FBXW7 and TP53 genes were present in 3 (2.0%), 30 (20.3%), 9 (6.1%) and 7 tumours (4.7%), respectively. The distribution of the mutations was similar between treatment-naive tumours and recurrences, except for TP53 mutations being more frequent in recurrences (P=0.0005). In patients treated with abdominoperineal resection (APR) after relapse (n=38, median follow-up of 18.2 years), overall survival (OS) was significantly correlated with HPV16 status (P=0.048), gender (P=0.045) and PIK3CA mutation (P=0.037). The PIK3CA status retained its prognostic significance in Cox multivariate regression analysis (P=0.025). CONCLUSIONS Our study identified PIK3CA mutation as an independent prognostic factor in patients who underwent APR for ASCC recurrence, suggesting a potential benefit from adjuvant treatment and the evaluation of targeted therapies with PI3K/Akt/mTor inhibitors in PIK3CA-mutated patients.
Collapse
|
17
|
Smaglo BG, Tesfaye A, Halfdanarson TR, Meyer JE, Wang J, Gatalica Z, Reddy S, Arguello D, Boland PM. Comprehensive multiplatform biomarker analysis of 199 anal squamous cell carcinomas. Oncotarget 2015; 6:43594-604. [PMID: 26498363 PMCID: PMC4791253 DOI: 10.18632/oncotarget.6202] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/09/2015] [Indexed: 02/01/2023] Open
Abstract
Anal squamous cell carcinoma (ASCC) is a rare, HPV-associated malignancy typically diagnosed in early stages and definitively treated with chemoradiation. In situations where patients exhibit metastatic or recurrent disease, treatment options are severely limited. In this study, molecular alterations were identified that could be used to aid in therapeutic decisions for patients with metastatic or recurrent anal squamous cell carcinoma. Specimens from patients with this cancer were tested via a multiplatform profiling service (Caris Life Sciences, Phoenix, AZ) consisting of gene sequencing, protein expression by immunohistochemistry, and gene amplification with in situ hybridization. Utilizing these techniques, novel treatment strategies that could be explored were identified, including potential benefit with anti-EGFR therapies, immune checkpoint inhibitors, topoisomerase inhibitors, and taxanes. The frequency of overexpression of proteins that mark resistance to chemotherapeutic drugs, such as MRP1 (chemotherapy efflux pump), ERCC1 (resistance to platinum-based chemotherapy), and thymidylate synthase (resistance to fluoropyrimidines) were also identified, suggesting a lack of benefit. This multiplatform strategy could be explored for its potential to generate a personalized treatment selection for patients with advanced ASCC, provide a guide for future therapeutic development for this cancer, and be extended to other rare cancer types as well.
Collapse
Affiliation(s)
- Brandon G. Smaglo
- The Ruesch Center for the Cure of GI Cancers, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Anteneh Tesfaye
- Departments of Hematology/Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | | | - Joshua E. Meyer
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jue Wang
- Division of Oncology, University of Arizona Cancer Center, Phoenix, AZ, USA
| | - Zoran Gatalica
- Department of Pathology, Caris Life Sciences, Phoenix, AZ, USA
| | - Sandeep Reddy
- Department of Pathology, Caris Life Sciences, Phoenix, AZ, USA
| | - David Arguello
- Department of Pathology, Caris Life Sciences, Phoenix, AZ, USA
| | - Patrick M. Boland
- Department of Medicine, GI Center, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
18
|
Molecular biology of anal squamous cell carcinoma: implications for future research and clinical intervention. Lancet Oncol 2015; 16:e611-21. [DOI: 10.1016/s1470-2045(15)00292-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/20/2015] [Accepted: 08/27/2015] [Indexed: 12/18/2022]
|
19
|
Immune Checkpoint Modulation in Colorectal Cancer: What's New and What to Expect. J Immunol Res 2015; 2015:158038. [PMID: 26605342 PMCID: PMC4641952 DOI: 10.1155/2015/158038] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC), as one of the most prevalent types of cancer worldwide, is still a leading cause of cancer related mortality. There is an urgent need for more efficient therapies in metastatic disease. Immunotherapy, a rapidly expanding field of oncology, is designed to boost the body's natural defenses to fight cancer. Of the many approaches currently under study to improve antitumor immune responses, immune checkpoint inhibition has thus far been proven to be the most effective. This review will outline the treatments that take advantage of our growing understanding of the role of the immune system in cancer, with a particular emphasis on immune checkpoint molecules, involved in CRC pathogenesis.
Collapse
|
20
|
Zhang L, Wu J, Ling MT, Zhao L, Zhao KN. The role of the PI3K/Akt/mTOR signalling pathway in human cancers induced by infection with human papillomaviruses. Mol Cancer 2015; 14:87. [PMID: 26022660 PMCID: PMC4498560 DOI: 10.1186/s12943-015-0361-x] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/06/2015] [Indexed: 01/08/2023] Open
Abstract
Infection with Human papillomaviruses (HPVs) leads to the development of a wide-range of cancers, accounting for 5% of all human cancers. A prominent example is cervical cancer, one of the leading causes of cancer death in women worldwide. It has been well established that tumor development and progression induced by HPV infection is driven by the sustained expression of two oncogenes E6 and E7. The expression of E6 and E7 not only inhibits the tumor suppressors p53 and Rb, but also alters additional signalling pathways that may be equally important for transformation. Among these pathways, the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signalling cascade plays a very important role in HPV-induced carcinogenesis by acting through multiple cellular and molecular events. In this review, we summarize the frequent amplification of PI3K/Akt/mTOR signals in HPV-induced cancers and discuss how HPV oncogenes E6/E7/E5 activate the PI3K/Akt/mTOR signalling pathway to modulate tumor initiation and progression and affect patient outcome. Improvement of our understanding of the mechanism by which the PI3K/Akt/mTOR signalling pathway contributes to the immortalization and carcinogenesis of HPV-transduced cells will assist in devising novel strategies for preventing and treating HPV-induced cancers.
Collapse
Affiliation(s)
- Lifang Zhang
- Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, 325035 , Zhejiang, PR China.
| | - Jianhong Wu
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, 4102, QLD, Australia.
- Current address: Department of Gastric Cancer and Soft Tissue Sarcomas Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China.
| | - Ming Tat Ling
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, 4102, QLD, Australia.
| | - Liang Zhao
- The University of Queensland, Brisbane, 4072, QLD, Australia.
| | - Kong-Nan Zhao
- Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, 325035 , Zhejiang, PR China.
- Centre for Kidney Disease Research-Venomics Research, The University of Queensland School of Medicine, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, QLD, Australia.
| |
Collapse
|
21
|
Elimova E, Wadhwa R, Estrella JS, Sudo K, Shiozaki H, Blum MA, Chen Q, Song S, Das P, Ajani JA. Anal canal cancer: biology and therapy. Expert Opin Orphan Drugs 2014. [DOI: 10.1517/21678707.2014.870469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Sun ZJ, Zhang L, Zhang W, Hall B, Bian Y, Kulkarni AB. Inhibition of mTOR reduces anal carcinogenesis in transgenic mouse model. PLoS One 2013; 8:e74888. [PMID: 24124460 PMCID: PMC3790781 DOI: 10.1371/journal.pone.0074888] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/06/2013] [Indexed: 01/09/2023] Open
Abstract
The molecular mechanism of human anal squamous cell carcinoma (ASCC) is unclear, and the accumulating evidence indicate association of ASCC with the activation of the Akt/mTOR pathway. Here we describe a mouse model with spontaneous anal squamous cell cancer, wherein a combined deletion of Tgfbr1 and Pten in stratified squamous epithelia was induced using inducible K14-Cre. Histopathologic analyses confirmed that 33.3% of the mice showed increased susceptibility to ASCC and precancerous lesions. Biomarker analyses demonstrated that the activation of the Akt pathway in ASCC of the Tgfbr1 and Pten double knockout (2cKO) mouse was similar to that observed in human anal cancer. Chemopreventive experiments using mTOR inhibitor-rapamycin treatment significantly delayed the onset of the ASCC tumors and reduced the tumor burden in 2cKO mice by decreasing the phosphorylation of Akt and S6. This is the first conditional knockout mouse model used for investigating the contributions of viral and cellular factors in anal carcinogenesis without carcinogen-mediated induction, and it would provide a platform for assessing new therapeutic modalities for treating and/or preventing this type of cancer.
Collapse
Affiliation(s)
- Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wei Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bradford Hall
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yansong Bian
- Head and Neck Surgery Branch, National Institute of Deafness and Other Communicative Disorders, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ashok B. Kulkarni
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
23
|
Ghosh A, Lai C, McDonald S, Suraweera N, Sengupta N, Propper D, Dorudi S, Silver A. HSP27 expression in primary colorectal cancers is dependent on mutation of KRAS and PI3K/AKT activation status and is independent of TP53. Exp Mol Pathol 2012; 94:103-8. [PMID: 22982087 DOI: 10.1016/j.yexmp.2012.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 09/02/2012] [Indexed: 11/19/2022]
Abstract
Colorectal adenomas display features of senescence, but these are often lost upon progression to carcinoma, indicating that oncogene induced senescence (OIS) could be a roadblock in colorectal cancer (CRC) development. Heat shock proteins (HSPs) have been implicated in the prognosis of CRC and HSP based therapy is a current interest for drug development. Recent cell culture studies have suggested that in the absence of a TP53 mutation, OIS mediated by PI3K/AKT activation can be circumvented by high expression of HSPs. Furthermore, while PI3K/AKT activation and KRAS mutations are independent inducers of OIS, PI3K/AKT activation can suppress KRAS-induced OIS when both are present in cultured cells. As KRAS mutations, PI3K/AKT activation and TP53 mutations are all common features of CRC, it is possible that the requirement for HSP to inhibit OIS in CRC is dependent on the mutation spectrum of a tumour. However, work on HSP that utilised mutation profiled human tumour tissues has been limited. Here, we characterised the expression of two major HSP proteins (HSP27 and 72) by immunohistochemistry (IHC), the mutation status of TP53, KRAS and PIK3CA genes by direct sequencing and the activation status of AKT by IHC in a cohort of unselected primary CRC (n=74). We compare our data with findings generated from cell-based studies. Expression of HSP27 and HSP72 was correlated to clinicopathological and survival data but no significant association was found. We also established the mutation status of TP53, KRAS and PIK3CA genes and the activation status of AKT in our CRC panel. We did not detect any associations between HSP27 or HSP72 expression with TP53 mutation status. However, HSP27 expression in CRCs was strongly associated with the co-presence of wildtype KRAS and activated PI3K/AKT (p=0.004), indicating a possible role of HSP27 in overcoming PI3K/AKT induced OIS in tumours. Our studies suggest a role for using archival tissues in validating hypotheses generated from cell culture based investigations.
Collapse
Affiliation(s)
- Anil Ghosh
- Centre for Digestive Diseases, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark St, Whitechapel, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Oliveira S, Teixeira L, Hoff PM, de Gramont A, Tournigand C. Squamous-cell carcinoma of the anal canal: room for improvement with targeted therapy. Clin Res Hepatol Gastroenterol 2012; 36:209-13. [PMID: 22138064 DOI: 10.1016/j.clinre.2011.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/25/2011] [Accepted: 10/27/2011] [Indexed: 02/04/2023]
Abstract
Carcinoma of the anal canal is a rare disease accounting for 1-5% of gastrointestinal tract malignancies. However, its incidence is increasing worldwide. Chemoradiation is the standard treatment for most patients with squamous-cell carcinoma of the anal canal and was first described by Nigro et al. Since then, no other effective treatment was developed. Patients with metastatic disease should be considered candidates for clinical trials. New treatment strategies, including molecular target therapies, are warranted in order to improve disease control. Despite the rarity of this disease, it is urgent to improve its treatment by introducing targeted therapy in the arena.
Collapse
Affiliation(s)
- Suilane Oliveira
- Instituto do Cancer do Estado de São Paulo, 251 avenida Doutor Arnaldo, 01246-000 São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
25
|
Yhim HY, Lee NR, Song EK, Kwak JY, Lee ST, Kim JH, Kim JS, Park HS, Chung IJ, Shim HJ, Hwang JE, Kim HR, Nam TK, Park MR, Shim H, Park HS, Kim HS, Yim CY. The prognostic significance of tumor human papillomavirus status for patients with anal squamous cell carcinoma treated with combined chemoradiotherapy. Int J Cancer 2011; 129:1752-60. [DOI: 10.1002/ijc.25825] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 11/19/2010] [Indexed: 11/09/2022]
|
26
|
Abstract
Human anal cancers are associated with high-risk human papillomaviruses (HPV) that cause other anogenital cancers and head and neck cancers. As with other cancers, HPV16 is the most common high-risk HPV in anal cancers. We describe the generation and characterization of a mouse model for human anal cancer. This model makes use of K14E6 and K14E7 transgenic mice in which the HPV16 E6 and E7 genes are directed in their expression to stratified squamous epithelia. HPV16 E6 and E7 possess oncogenic properties including, but not limited to, their capacity to inactivate the cellular tumor suppressors p53 and pRb, respectively. Both E6 and E7 were found to be functionally expressed in the anal epithelia of K14E6/K14E7 transgenic mice. To assess the susceptibility of these mice to anal cancer, mice were treated topically with dimethylbenz[a]anthracene (DMBA), a chemical carcinogen that is known to induce squamous cell carcinomas in other sites. Nearly 50% of DMBA-treated HPV16 E6/E7 transgenic mice showed overt signs of tumors, whereas none of the like-treated nontransgenic mice showed tumors. Histopathologic analyses confirmed that the HPV16 transgenic mice were increased in their susceptibility to anal cancers and precancerous lesions. Biomarker analyses demonstrated that these mouse anal cancers exhibit properties that are similar to those observed in HPV-positive precursors to human anal cancer. This is the first mouse model for investigating the contributions of viral and cellular factors in anal carcinogenesis, and should provide a platform for assessing new therapeutic modalities for treating and/or preventing this type of cancer.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene/toxicity
- Animals
- Anus Neoplasms/chemically induced
- Anus Neoplasms/pathology
- Anus Neoplasms/virology
- Blotting, Western
- Carcinogens/toxicity
- Carcinoma, Squamous Cell/chemically induced
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/virology
- Cell Transformation, Neoplastic
- Disease Models, Animal
- Female
- Humans
- Immunoenzyme Techniques
- Immunoprecipitation
- Male
- Mice
- Mice, Transgenic
- Oncogene Proteins, Viral/physiology
- Papillomaviridae/genetics
- Papillomavirus E7 Proteins/physiology
- Phenotype
- Repressor Proteins/physiology
Collapse
Affiliation(s)
- Marie K. Stelzer
- University of Wisconsin School of Medicine and Public Health, Madison WI, Department of Surgery
- University of Wisconsin School of Medicine and Public Health, Madison WI, Department of Oncology and McArdle Laboratory for Cancer Research
| | - Henry C. Pitot
- University of Wisconsin School of Medicine and Public Health, Madison WI, Department of Oncology and McArdle Laboratory for Cancer Research
| | - Amy Liem
- University of Wisconsin School of Medicine and Public Health, Madison WI, Department of Oncology and McArdle Laboratory for Cancer Research
| | | | | | - Paul F. Lambert
- University of Wisconsin School of Medicine and Public Health, Madison WI, Department of Oncology and McArdle Laboratory for Cancer Research
| |
Collapse
|
27
|
Molecular biomarkers correlate with disease-free survival in patients with anal canal carcinoma treated with chemoradiation. Dig Dis Sci 2010; 55:1098-105. [PMID: 19399614 DOI: 10.1007/s10620-009-0812-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 04/01/2009] [Indexed: 12/31/2022]
Abstract
Large primary tumor and clinical nodal involvement in patients with anal carcinoma treated with chemoradiation are associated with poor disease-free survival (DFS). However, the outcome in individual patient is unpredictable. We hypothesized that biomarkers related to chemotherapy and/or radiation resistance would be associated with DFS. We analyzed clinical and biomarker data in 30 patients with anal carcinoma who had chemoradiation. Patient selection was based on the availability of untreated cancer for biomarkers, completion of prescribed chemoradiation, and patient outcomes (~50% disease-free) nonrepresentative of published cohorts but conducive to biomarker discovery. Ten biomarkers, Ki67, human telomerase (hTERT), epidermal growth factor receptor (EGFR), p53, p16, Bcl-2, vascular endothelial growth factor (VEGF), nuclear factor kappa-B (NF-kappaB), SHH, and Gli-1, were studied. Raw data as continuous variable (only EGFR was trichotomized) were analyzed. Univariate and multivariate Cox models were utilized to assess relationship between DFS and biomarkers. Twenty-three of 30 patients were women, tumor diameter was >5 cm in 30, and 37% had clinically positive nodes. Fourteen (30%) patients had a DFS event after chemoradiation. In univariate analysis, NF-kappaB (P = 0.01), SHH (P = 0.02), Gli-1 (P = 0.02), and tumor diameter (P = 0.03) were significantly associated with DFS, and Ki67 (P = 0.07) was marginally significant. In multivariate analysis, tumor diameter (P = 0.003), Ki67 (P = 0.005), NF-kappaB (P = 0.002), SHH (P = 0.02), and Gli-1 (P = 0.02) were significantly associated with DFS. Our data, albeit preliminary, suggest that several biomarkers (Ki67, NF-kappaB, SHH, and Gli-1) are associated with DFS. Upon further expansion and validation, these results may provide a biomarker-based understanding of heterogeneous clinical biology of patients with anal carcinoma.
Collapse
|