151
|
Xu H, Yu S, Liu Q, Yuan X, Mani S, Pestell RG, Wu K. Recent advances of highly selective CDK4/6 inhibitors in breast cancer. J Hematol Oncol 2017; 10:97. [PMID: 28438180 PMCID: PMC5404666 DOI: 10.1186/s13045-017-0467-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/19/2017] [Indexed: 01/15/2023] Open
Abstract
Uncontrolled cell division is the hallmark of cancers. Full understanding of cell cycle regulation would contribute to promising cancer therapies. In particular, cyclin-dependent kinases 4/6 (CDK4/6), which are pivotal drivers of cell proliferation by combination with cyclin D, draw more and more attention. Subsequently, extensive studies were carried out to explore drugs inhibiting CDK4/6 and assess the efficacy and safety of these drugs in cancer, especially breast cancer. Due to the insuperable adverse events and the less activity observed in vivo, the drug development of the initial pan-CDK inhibitor flavopiridol was consequently discontinued, and then highly specific inhibitors were extensively researched and developed, including palbociclib (PD0332991), ribociclib (LEE011), and abemaciclib (LY2835219). Food and Drug Administration has approved palbociclib and ribociclib for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced or metastatic breast cancer, and recent clinical trial data suggest that palbociclib significantly improved clinical outcome when combined with letrozole or fulvestrant. Besides, the favorable effects of abemaciclib on prolonging survival of breast cancer patients have also been observed in clinical trials both for single-agent and combination strategy. In this review, we outline the preclinical and clinical advancement of these three orally bioavailable and highly selective CDK4/6 inhibitors in breast cancer.
Collapse
Affiliation(s)
- Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| | - Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| | - Sridhar Mani
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, NY 10461 USA
| | - Richard G. Pestell
- Pennsylvania Center for Cancer and Regenerative Medicine, Wynnewood, PA 19096 USA
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| |
Collapse
|
152
|
Zhang X, Zhao F, Zhang S, Song Y. Ectopic High Expression of E2-EPF Ubiquitin Carrier Protein Indicates a More Unfavorable Prognosis in Brain Glioma. Genet Test Mol Biomarkers 2017; 21:242-247. [PMID: 28384045 DOI: 10.1089/gtmb.2016.0281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Xiaohui Zhang
- Pathology Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fangbo Zhao
- School of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, China
| | - Shujun Zhang
- Pathology Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yichun Song
- Pathology Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
153
|
He BL, Xu N, Li YL, Pan CY, Cao R, Liao LB, Yin CX, Lan YQ, Lu ZY, Huang JX, Zhou HS, Liu QF, Liu XL. [Clinical analysis of adult Philadelphia chromosome-positive acute lymphoblastic leukemia with p16 gene deletion]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 38:204-209. [PMID: 28395443 PMCID: PMC7348375 DOI: 10.3760/cma.j.issn.0253-2727.2017.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Indexed: 12/01/2022]
Abstract
Objective: To investigate the clinical implications of p16 gene deletion in adult Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph(+) ALL) . Methods: Retrospective analysis of clinical, immunophenotypic, cytogenetics, molecular characteristics and prognosis of 80 newly diagnosed Ph(+) ALL patients with p16 deletion. Results: Of 80 adult Ph(+) ALL, the prevalence of p16 gene deletion was 31.3%. p16 gene deletion carriers frequently accompanied with high WBC counts (WBC≥30×10(9)/L) and CD20 expression. The incidence of complex chromosome abnormality in p16 gene deletion group was higher than that in non-deletion group, with alternations in chromosome 7, 8, 19 and der (22) more frequently observed. There was no difference occurred between patients with or without p16 gene deletion in complete remission (CR) rate following induction chemotherapy combined with tyrosine kinase inhibitors (TKIs) . However, after three cycles of chemotherapy, the MMR and CMR rate in the p16 gene deletion group was lower than patients with wild-type p16 gene (P=0.034, P=0.036) . The p16 gene deletion patients showed no significant differences in MMR, CMR and relapse rate between Imatinib or Dasatinib plus chemotherapy (P>0.05) . Deletion of p16 gene was significantly associated with poor outcomes including worse overall survival (OS) (37.1% vs 54.1%, P=0.037) , lower disease free-survival (DFS) (12.4% vs 45.9%, P=0.026) , and increased cumulative incidence of relapse (P=0.033) . Among the 25 patients with p16 deletion, 14 underwent allo-HSCT and the median survival was 21 months, better than that of patients received chemotherapy alone (12 months) (P=0.030) . Conclusion: This study indicated that deletion of p16 was associated with poor prognosis in adult Ph(+) ALL, and the utility of second-generation TKI (Dasatinib) does not necessarily have an edge on efficacy over Imatinib, but allo-HSCT has the potential of elongating life expectancy. It is an important significance to define the status of p16 in Ph(+) ALL for predicting prognosis and guiding therapy decision-making.
Collapse
Affiliation(s)
- B L He
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Mendez-Pena JE, Sadow PM, Nose V, Hoang MP. RNA chromogenic in situ hybridization assay with clinical automated platform is a sensitive method in detecting high-risk human papillomavirus in squamous cell carcinoma. Hum Pathol 2017; 63:184-189. [PMID: 28302536 DOI: 10.1016/j.humpath.2017.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 01/31/2017] [Accepted: 02/23/2017] [Indexed: 01/30/2023]
Abstract
Detection of active human papillomavirus (HPV) is clinically important because its presence has been shown to correlate with favorable clinical outcomes and better response to treatment in oropharyngeal squamous cell carcinomas. Using a clinical automated platform, we compared the performance of commercially available HPV DNA and RNA in situ hybridization (ISH) probes in archival tissues of 57 squamous cell carcinomas. Importantly, a clinical automated platform gives (1) consistent and reproducible results for HPV ISH and (2) better standardization across clinical laboratories. Compared with polymerase chain reaction results, RNA ISH exhibited 93% concordance versus 81% of DNA ISH. RNA ISH was more sensitive than DNA ISH (100% versus 88%) and more specific (87% versus 74%). When only accounting for 2+-3+ positivity, sensitivity was 92% for RNA ISH versus 73% for DNA ISH, highlighting the ease of interpretation. p16 exhibited 96% sensitivity, whereas specificity was only 55%. In 3 cases, both RNA and DNA ISH were positive, whereas polymerase chain reaction results were negative, suggesting that ISH methods might be a more sensitive method. Performing on a clinical automated platform, RNA ISH is sensitive in determining high-risk HPV status in formalin-fixed, paraffin-embedded tissues and has the potential of being a standalone clinical test.
Collapse
Affiliation(s)
| | - Peter M Sadow
- Massachusetts General Hospital, Boston, MA 02114, USA
| | - Vania Nose
- Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mai P Hoang
- Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
155
|
Kofler B, Borena W, Manzl C, Dudas J, Wegscheider AS, Jansen-Dürr P, Schartinger V, Riechelmann H. Sensitivity of tumor surface brushings to detect human papilloma virus DNA in head and neck cancer. Oral Oncol 2017; 67:103-108. [PMID: 28351563 DOI: 10.1016/j.oraloncology.2017.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/31/2017] [Accepted: 02/13/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Human papilloma virus (HPV) induced head and neck squamous cell carcinoma (HNSCC) represents a distinct tumor subset. We questioned how accurately a brushing from the tumor surface detects HPV in patients with HNSCC. MATERIALS AND METHODS Brushings from the tumor surface were compared with HPV DNA isolation from formalin-fixed and paraffin-embedded (FFPE) tumor biopsies, which served as the reference standard. In both matrices, HPV DNA was detected using a commercially available test kit. In addition, p16 was assessed in tumor biopsies by immunohistochemistry (IHC). The tumors were considered p16 positive if 70% or more of cancer cells expressed p16. RESULTS 93 patients with HNSCC were included. Sensitivity and specificity of the brush test were 83% (95%CI: 67-92%) and 85% (95%CI: 72-93%). Results of p16 IHC were concordant with FFPE samples DNA determinations in 73/93 patients. In 53 patients (57%) the tumor was located in the oropharynx and in 40 patients (43%) the tumor was located in the non-oropharynx region. Sensitivity and specificity of the brush test in patients with oropharyngeal cancer was higher with 86% (95%CI: 70-95%) and 89% (95%CI: 65-99%). CONCLUSION Superficial brushes from the tumor surface may be used to identify HPV positive HNSCC.
Collapse
Affiliation(s)
- Barbara Kofler
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria.
| | - Wegene Borena
- Division of Virology, Department of Hygiene, Microbiology, Social Medicine, Medical University of Innsbruck, Peter-Mayr-Strasse 4b, 6020 Innsbruck, Austria
| | - Claudia Manzl
- Department of Pathology, Medical University of Innsbruck, Müllerstrasse 44, 6020 Innsbruck, Austria
| | - Jozsef Dudas
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Anne-Sophie Wegscheider
- Department of Pathology, Medical University of Innsbruck, Müllerstrasse 44, 6020 Innsbruck, Austria
| | - Pidder Jansen-Dürr
- Institute for Biomedical Ageing Research, Medical University of Innsbruck, Rennweg 10, 6020 Innsbruck, Austria
| | - Volker Schartinger
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Herbert Riechelmann
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
156
|
Cai Z, Liu Q. Cell Cycle Regulation in Treatment of Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:251-270. [PMID: 29282688 DOI: 10.1007/978-981-10-6020-5_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell cycle progression and cell proliferation are under precise and orchestrated control in normal cells. However, uncontrolled cell proliferation caused by aberrant cell cycle progression is a crucial characteristic of cancer. Understanding cell cycle progression and its regulation sheds light on cancer treatment. Agents targeting cell cycle regulators (such as CDKs) have been considered as promising candidates in cancer treatment. Although the first-generation pan-CDK inhibitors failed in clinical trials because of their adverse events and low efficacy, new selective CDK 4/6 inhibitors showed potent efficacy with tolerable safety in preclinical and clinical studies. Here we will review the mechanisms of cell cycle regulation and targeting key cell cycle regulators (such as CDKs) in breast cancer treatment. Particularly, we will discuss the mechanism of CDK inhibitors in disrupting cell cycle progression, the use of selective CDK4/6 inhibitors in treatment of advanced, hormone receptor (HR)-positive postmenopausal breast cancer patients, and other clinical trials that aim to extend the utilization of these agents.
Collapse
Affiliation(s)
- Zijie Cai
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, Guangdong, China
| | - Qiang Liu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
157
|
Guan X, LaPak KM, Hennessey RC, Yu CY, Shakya R, Zhang J, Burd CE. Stromal Senescence By Prolonged CDK4/6 Inhibition Potentiates Tumor Growth. Mol Cancer Res 2016; 15:237-249. [PMID: 28039358 DOI: 10.1158/1541-7786.mcr-16-0319] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/06/2016] [Accepted: 12/08/2016] [Indexed: 12/20/2022]
Abstract
Senescent cells within the tumor microenvironment (TME) adopt a proinflammatory, senescence-associated secretory phenotype (SASP) that promotes cancer initiation, progression, and therapeutic resistance. Here, exposure to palbociclib (PD-0332991), a CDK4/6 inhibitor, induces senescence and a robust SASP in normal fibroblasts. Senescence caused by prolonged CDK4/6 inhibition is DNA damage-independent and associated with Mdm2 downregulation, whereas the SASP elicited by these cells is largely reliant upon NF-κB activation. Based upon these observations, it was hypothesized that the exposure of nontransformed stromal cells to PD-0332991 would promote tumor growth. Ongoing clinical trials of CDK4/6 inhibitors in melanoma prompted a validation of this hypothesis using a suite of genetically defined melanoma cells (i.e., Ras mutant, Braf mutant, and Ras/Braf wild-type). When cultured in the presence of CDK4/6i-induced senescent fibroblasts, melanoma cell lines exhibited genotype-dependent proliferative responses. However, in vivo, PD-0332991-treated fibroblasts enhanced the growth of all melanoma lines tested and promoted the recruitment of Gr-1-positive immune cells. These data indicate that prolonged CDK4/6 inhibitor treatment causes normal fibroblasts to enter senescence and adopt a robust SASP. Such senescent cells suppress the antitumor immune response and promote melanoma growth in immunocompetent, in vivo models.Implications: The ability of prolonged CDK4/6 inhibitor treatment to induce cellular senescence and a robust SASP in primary cells may hinder therapeutic efficacy and promote long-term, gerontogenic consequences that should be considered in clinical trials aiming to treat melanoma and other cancer types. Mol Cancer Res; 15(3); 237-49. ©2016 AACR.
Collapse
Affiliation(s)
- Xiangnan Guan
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
| | - Kyle M LaPak
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio.,Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Rebecca C Hennessey
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Christina Y Yu
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio.,Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Reena Shakya
- The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio
| | - Jianying Zhang
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Christin E Burd
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio. .,Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio.,The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio
| |
Collapse
|
158
|
Cocco E, Deng Y, Shapiro EM, Bortolomai I, Lopez S, Lin K, Bellone S, Cui J, Menderes G, Black JD, Schwab CL, Bonazzoli E, Yang F, Predolini F, Zammataro L, Altwerger G, de Haydu C, Clark M, Alvarenga J, Ratner E, Azodi M, Silasi DA, Schwartz PE, Litkouhi B, Saltzman WM, Santin AD. Dual-Targeting Nanoparticles for In Vivo Delivery of Suicide Genes to Chemotherapy-Resistant Ovarian Cancer Cells. Mol Cancer Ther 2016; 16:323-333. [PMID: 27956521 DOI: 10.1158/1535-7163.mct-16-0501] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 01/25/2023]
Abstract
Ovarian cancer is the most lethal gynecologic cancer. Claudin-3 and -4, the receptors for Clostridium perfringens enterotoxin (CPE), are overexpressed in more than 70% of these tumors. Here, we synthesized and characterized poly(lactic-co-glycolic-acid) (PLGA) nanoparticles (NPs) modified with the carboxy-terminal-binding domain of CPE (c-CPE-NP) for the delivery of suicide gene therapy to chemotherapy-resistant ovarian cancer cells. As a therapeutic payload, we generated a plasmid encoding for the diphtheria toxin subunit-A (DT-A) under the transcriptional control of the p16 promoter, a gene highly differentially expressed in ovarian cancer cells. Flow cytometry and immunofluorescence demonstrated that c-CPE-NPs encapsulating the cytomegalovirus (CMV) GFP plasmid (CMV GFP c-CPE-NP) were significantly more efficient than control NPs modified with a scrambled peptide (CMV GFP scr-NP) in transfecting primary chemotherapy-resistant ovarian tumor cell lines in vitro (P = 0.03). Importantly, c-CPE-NPs encapsulating the p16 DT-A vector (p16 DT-A c-CPE-NP) were significantly more effective than control p16 DT-A scr-NP in inducing ovarian cancer cell death in vitro (% cytotoxicity: mean ± SD = 32.9 ± 0.15 and 7.45 ± 7.93, respectively, P = 0.03). In vivo biodistribution studies demonstrated efficient transfection of tumor cells within 12 hours after intraperitoneal injection of CMV GFP c-CPE-NP in mice harboring chemotherapy-resistant ovarian cancer xenografts. Finally, multiple intraperitoneal injections of p16 DT-A c-CPE-NP resulted in a significant inhibition of tumor growth compared with control NP in chemotherapy-resistant tumor-bearing mice (P = 0.041). p16 DT-A c-CPE-NP may represent a novel dual-targeting therapeutic approach for the selective delivery of gene therapy to chemotherapy-resistant ovarian cancer cells. Mol Cancer Ther; 16(2); 323-33. ©2016 AACR.
Collapse
Affiliation(s)
- Emiliano Cocco
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Yang Deng
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Erik M Shapiro
- Department of Radiology, Michigan State University, East Lansing, Michigan
| | | | - Salvatore Lopez
- Division of Gynecologic Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Ken Lin
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stefania Bellone
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Jiajia Cui
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Gulden Menderes
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Jonathan D Black
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Carlton L Schwab
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Elena Bonazzoli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Fan Yang
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Federica Predolini
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Luca Zammataro
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Gary Altwerger
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Christopher de Haydu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Mitchell Clark
- Yale University Bridgeport Hospital, Bridgeport, Connecticut
| | - Julio Alvarenga
- Yale University Bridgeport Hospital, Bridgeport, Connecticut
| | - Elena Ratner
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Masoud Azodi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Dan-Arin Silasi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Peter E Schwartz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Babak Litkouhi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
159
|
Molecular Profiling of Thymoma and Thymic Carcinoma: Genetic Differences and Potential Novel Therapeutic Targets. Pathol Oncol Res 2016; 23:551-564. [PMID: 27844328 PMCID: PMC5487866 DOI: 10.1007/s12253-016-0144-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 10/26/2016] [Indexed: 01/30/2023]
Abstract
Thymoma and thymic carcinoma are thymic epithelial tumors (TETs). We performed a molecular profiling to investigate the pathogenesis of TETs and identify novel targets for therapy. We analyzed 37 thymomas (18 type A, 19 type B3) and 35 thymic carcinomas. The sequencing of 50 genes detected nonsynonymous mutations in 16 carcinomas affecting ALK, ATM, CDKN2A, ERBB4, FGFR3, KIT, NRAS and TP53. Only two B3 thymomas had a mutation in noncoding regions of the SMARCB1 and STK11 gene respectively. Three type A thymomas harbored a nonsynonymous HRAS mutation. Fluorescence in situ hybridization detected in 38 % of carcinomas a CDKN2A, in 32 % a TP53 and in 8 % an ATM gene deletion, whereas only one B3 thymoma exhibited a CDKNA deletion, and none of the type A thymomas showed a gene loss. Sequencing of the total miRNA pool of 5 type A thymomas and 5 thymic carcinomas identified the C19MC miRNA cluster as highly expressed in type A thymomas, but completely silenced in thymic carcinomas. Furthermore, the miRNA cluster C14MC was downregulated in thymic carcinomas. Among non-clustered miRNAs, the upregulation of miR-21, miR-9-3 and miR-375 and the downregulation of miR-34b, miR-34c, miR-130a and miR-195 in thymic carcinomas were most significant. The expression of ALK, HER2, HER3, MET, phospho-mTOR, p16INK4A, PDGFRA, PDGFRB, PD-L1, PTEN and ROS1 was investigated by immunohistochemistry. PDGFRA was increased in thymic carcinomas and PD-L1 in B3 thymomas and thymic carcinomas. In summary, our results reveal genetic differences between thymomas and thymic carcinomas and suggest potential novel targets for therapy.
Collapse
|
160
|
Seebauer CT, Brunner S, Glockzin G, Piso P, Ruemmele P, Schlitt HJ, Geissler EK, Fichtner-Feigl S, Kesselring R. Peritoneal carcinomatosis of colorectal cancer is characterized by structural and functional reorganization of the tumor microenvironment inducing senescence and proliferation arrest in cancer cells. Oncoimmunology 2016; 5:e1242543. [PMID: 28439450 DOI: 10.1080/2162402x.2016.1242543] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/21/2016] [Accepted: 09/23/2016] [Indexed: 12/19/2022] Open
Abstract
Background : Peritoneal carcinomatosis (PC) is a terminal evolution from primary colorectal cancer (pCRC) associated with poor patient survival. Impact of the immune cell infiltrate on PC pathogenesis is unknown. Therefore, we characterized the immunological tumor microenvironment regarding proliferation, senescence and neovascularization. Methods : Formalin-fixed and paraffin-embedded (FFPE) tissue of PC and pCRC was examined by immunohistochemistry. Cells infiltrating resected tissue were isolated and analyzed by flow cytometry. PCR arrays detected the expression of genes relevant for helper T (TH) cell responses, like TH1, TH2 and TH17 response. Results : PC tumor cells demonstrate significantly lower proliferation rates than pCRC, but show significantly more senescence. PC is surrounded by significantly increased numbers of cytotoxic active Natural Killer (NK) cells, follicular helper T cells (TFH) and B cells, whereas pCRC shows more CD4+ TH cells, CD8+ cytotoxic T (TC) cells, eosinophilic granulocytes, TH17 and regulatory T (Treg) cells. PC is characterized by significantly increased interferon-γ (IFNγ), an upregulation of tumor necrosis factor (TNF) and the NK cell-regulating cytokine interleukin-15 (IL-15). An upregulation of angiogenesis-related genes, like vascular endothelial growth factor-A (VEGF-A), leads to severe neovascularization in PC. Correlations of PC results reveal that elevated numbers of interleukin-17 (IL-17) positive cells are associated with high cancer cell proliferation, whereas high numbers of IFNγ positive cells correlate with more tumor cells in senescence. Conclusion : The cellular immune reaction is modified during metastasis, inducing senescence in PC tumor cells. Immune surveillance in PC is facilitated by NK cells and high levels of IFNγ and TNF. Counteracting this effect, TFH and B cells combined with VEGF-A enhancement promote neovascularization in PC (Illustration 1). During metastasis from primary CRC to PC the immune cell infiltrate changes, accompanied by the induction of senescence in PC cancer cells (marked red): In pCRC, the antitumor immune response is facilitated by CD4+TH cells, CD8+TC cells and PRG2+ eosinophilic granulocytes. The premetastatic niche development is promoted by Treg cells and TH17 cells producing systemic factors like VEGF-A, TGF-β and TNF. Along with TFH and B cells, as with a pro-tumor immune response, they support metastatic formation and lead to severe neovascularization in PC. This is counterbalanced by the IL-15-induced activation and proliferation of NK cells. The secreted cytokines IFNγ and TNF mediate immunosurveillance.
Collapse
Affiliation(s)
| | - Stefan Brunner
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Gabriel Glockzin
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Pompiliu Piso
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Petra Ruemmele
- Department of Pathology, University Medical Center Regensburg, Regensburg, Germany
| | - Hans-Juergen Schlitt
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | - Stefan Fichtner-Feigl
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany.,Regensburg Center of Interventional Immunology, Regensburg, Germany
| | - Rebecca Kesselring
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany.,Regensburg Center of Interventional Immunology, Regensburg, Germany
| |
Collapse
|
161
|
Cho-Vega JH. A diagnostic algorithm for atypical spitzoid tumors: guidelines for immunohistochemical and molecular assessment. Mod Pathol 2016; 29:656-70. [PMID: 27102343 DOI: 10.1038/modpathol.2016.70] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 02/28/2016] [Accepted: 03/06/2016] [Indexed: 11/09/2022]
Abstract
Atypical spitzoid tumors are a morphologically diverse group of rare melanocytic lesions most frequently seen in children and young adults. As atypical spitzoid tumors bear striking resemblance to Spitz nevus and spitzoid melanomas clinically and histopathologically, it is crucial to determine its malignant potential and predict its clinical behavior. To date, many researchers have attempted to differentiate atypical spitzoid tumors from unequivocal melanomas based on morphological, immonohistochemical, and molecular diagnostic differences. A diagnostic algorithm is proposed here to assess the malignant potential of atypical spitzoid tumors by using a combination of immunohistochemical and cytogenetic/molecular tests. Together with classical morphological evaluation, this algorithm includes a set of immunohistochemistry assays (p16(Ink4a), a dual-color Ki67/MART-1, and HMB45), fluorescence in situ hybridization (FISH) with five probes (6p25, 8q24, 11q13, CEN9, and 9p21), and an array-based comparative genomic hybridization. This review discusses details of the algorithm, the rationale of each test used in the algorithm, and utility of this algorithm in routine dermatopathology practice. This algorithmic approach will provide a comprehensive diagnostic tool that complements conventional histological criteria and will significantly contribute to improve the diagnosis and prediction of the clinical behavior of atypical spitzoid tumors.
Collapse
Affiliation(s)
- Jeong Hee Cho-Vega
- Department of Pathology, Dermatopathology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
162
|
Rodríguez-Santamarta T, Rodrigo JP, García-Pedrero JM, Álvarez-Teijeiro S, Ángeles Villaronga M, Suárez-Fernández L, Alvarez-Argüelles ME, Astudillo A, de Vicente JC. Prevalence of human papillomavirus in oral squamous cell carcinomas in northern Spain. Eur Arch Otorhinolaryngol 2016; 273:4549-4559. [PMID: 27342404 DOI: 10.1007/s00405-016-4152-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/14/2016] [Indexed: 01/19/2023]
Abstract
The aim of this study was to analyze the prevalence of high-risk HPV in oral squamous cell carcinoma (OSCC) in a northern Spanish population, as well as to ascertain the prognostic role of p16INK4a expression. The examination samples were collected from paraffin tissue blocks, from 125 patients surgically treated between 1996 and 2007. All cases were histologically evaluated, and the presence of HPV was assessed by p16 and p53immunohistochemistry followed by DNA detection by in situ hybridization (ISH) and polymerase chain reaction (PCR) amplification using the combination of consensus primers MY11/GP6 + . Fourteen cases (11 %) were p16-immunopositive, and p53 was scored positive in 73 cases (58 %). Five cases (4 %) showed a simultaneous p16-positive and p53-negative immunostaining. ISH was negative in all the cases. Among the p16INK4a-immunopositive cases, PCR amplification failed to reveal HPV DNA in any tumor samples. There were no statistically significant differences in any clinical or pathological characteristics of the patients regarding p16INK4a expression. T classification, neck-node metastasis, and clinical stage showed outcome relevance. However, no significant differences in cause-specific survival based on p16INK4a were observed. We did not find any high-risk HPV types in our patients, thus, are unlikely that HPV has an important role in the etiology of OSCC. p16INK4a protein was neither an accurate marker of HPV infection nor a prognosis marker in OSCC.
Collapse
Affiliation(s)
- Tania Rodríguez-Santamarta
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011, Oviedo, Asturias, Spain
| | - Juan Pablo Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011, Oviedo, Asturias, Spain.,Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias, Edificio Consultas Externas, Planta Baja Lab 2 ORL-IUOPA, C/Celestino Villamil, s/n, 33006, Oviedo, Asturias, Spain
| | - Juana M García-Pedrero
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias, Edificio Consultas Externas, Planta Baja Lab 2 ORL-IUOPA, C/Celestino Villamil, s/n, 33006, Oviedo, Asturias, Spain
| | - Saúl Álvarez-Teijeiro
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias, Edificio Consultas Externas, Planta Baja Lab 2 ORL-IUOPA, C/Celestino Villamil, s/n, 33006, Oviedo, Asturias, Spain
| | - M Ángeles Villaronga
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias, Edificio Consultas Externas, Planta Baja Lab 2 ORL-IUOPA, C/Celestino Villamil, s/n, 33006, Oviedo, Asturias, Spain
| | - Laura Suárez-Fernández
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias, Edificio Consultas Externas, Planta Baja Lab 2 ORL-IUOPA, C/Celestino Villamil, s/n, 33006, Oviedo, Asturias, Spain
| | - Marta E Alvarez-Argüelles
- Department of Microbiology, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011, Oviedo, Asturias, Spain
| | - Aurora Astudillo
- Department of Pathology, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011, Oviedo, Asturias, Spain
| | - Juan Carlos de Vicente
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011, Oviedo, Asturias, Spain. .,Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias, Edificio Consultas Externas, Planta Baja Lab 2 ORL-IUOPA, C/Celestino Villamil, s/n, 33006, Oviedo, Asturias, Spain. .,Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias, School of Medicine, c/Catedrático José Serrano s/n, 33006, Oviedo, Spain.
| |
Collapse
|
163
|
Selective impact of CDK4/6 suppression on patient-derived models of pancreatic cancer. Oncotarget 2016; 6:15788-801. [PMID: 26158861 PMCID: PMC4599237 DOI: 10.18632/oncotarget.3819] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/01/2015] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) harbors an exceedingly poor prognosis, and is generally considered a therapy-recalcitrant disease due to poor response to conventional chemotherapy coupled with non-actionable genetic drivers (e.g. KRAS mutations). However, PDA frequently loses p16ink4a, thereby leading to deregulation of CDK4/6. Surprisingly, in established cell models and xenografts, CDK4/6 inhibition has a modest effect on proliferation and resistance develops rapidly. To determine if such weak response was an intrinsic feature of PDA, we developed primary tumor explants that maintain the tumor environment and recapitulate feuture of primary PDA. The CDK4/6 inhibitor PD-0332991 was highly efficient at suppressing proliferation in 14 of the 15 explants. In the single resistant explant, we identified the rare loss of the RB tumor suppressor as the basis for resistance. Patient-derived xenografts (PDXs) were developed in parallel, and unlike the xenografts emerging from established cell lines, the PDXs maintained the histoarchitecture of the primary tumor. These PDXs were highly sensitive to CDK4/6 inhibition, yielding a complete suppression of PDA proliferation. Together, these data indicate that primary PDA is sensitive to CDK4/6 inhibition, that specific biomarkers can delineate intrinsic resistance, and that established cell line models may not represent an adequate means for evaluating therapeutic sensitivities.
Collapse
|
164
|
Mahajan A. Practical issues in the application of p16 immunohistochemistry in diagnostic pathology. Hum Pathol 2016; 51:64-74. [DOI: 10.1016/j.humpath.2015.12.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 11/25/2022]
|
165
|
Henrique T, José Freitas da Silveira N, Henrique Cunha Volpato A, Mioto MM, Carolina Buzzo Stefanini A, Bachir Fares A, Gustavo da Silva Castro Andrade J, Masson C, Verónica Mendoza López R, Daumas Nunes F, Paulo Kowalski L, Severino P, Tajara EH. HNdb: an integrated database of gene and protein information on head and neck squamous cell carcinoma. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2016; 2016:baw026. [PMID: 27013077 PMCID: PMC4806539 DOI: 10.1093/database/baw026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 02/19/2016] [Indexed: 12/19/2022]
Abstract
The total amount of scientific literature has grown rapidly in recent years. Specifically, there are several million citations in the field of cancer. This makes it difficult, if not impossible, to manually retrieve relevant information on the mechanisms that govern tumor behavior or the neoplastic process. Furthermore, cancer is a complex disease or, more accurately, a set of diseases. The heterogeneity that permeates many tumors is particularly evident in head and neck (HN) cancer, one of the most common types of cancer worldwide. In this study, we present HNdb, a free database that aims to provide a unified and comprehensive resource of information on genes and proteins involved in HN squamous cell carcinoma, covering data on genomics, transcriptomics, proteomics, literature citations and also cross-references of external databases. Different literature searches of MEDLINE abstracts were performed using specific Medical Subject Headings (MeSH terms) for oral, oropharyngeal, hypopharyngeal and laryngeal squamous cell carcinomas. A curated gene-to-publication assignment yielded a total of 1370 genes related to HN cancer. The diversity of results allowed identifying novel and mostly unexplored gene associations, revealing,for example, that processes linked to response to steroid hormone stimulus are significantly enriched in genes related to HN carcinomas. Thus, our database expands the possibilities for gene networks investigation, providing potential hypothesis to be tested. Database URL:http://www.gencapo.famerp.br/hndb.
Collapse
Affiliation(s)
- Tiago Henrique
- Department of Molecular Biology, School of Medicine of São José do Rio Preto, SP, Brazil Av Brigadeiro Faria Lima n° 5416 Vila Sao Pedro 15090-000 - São José do Rio Preto, SP - Brazil
| | - Nelson José Freitas da Silveira
- Institute of Exact Science, Federal University of Alfenas, MG, Brazil, Rua Gabriel Monteiro da Silva, 700 Centro 37130-000 - Alfenas, MG - Brazil
| | - Arthur Henrique Cunha Volpato
- Department of Molecular Biology, School of Medicine of São José do Rio Preto, SP, Brazil Av Brigadeiro Faria Lima n° 5416 Vila Sao Pedro 15090-000 - São José do Rio Preto, SP - Brazil
| | - Mayra Mataruco Mioto
- Department of Dermatological, Infectious, and Parasitic Diseases, School of Medicine of São José do Rio Preto, SP, Brazil Av Brigadeiro Faria Lima n° 5416 Vila Sao Pedro 15090-000 - São José do Rio Preto, SP - Brazil
| | - Ana Carolina Buzzo Stefanini
- Department of Molecular Biology, School of Medicine of São José do Rio Preto, SP, Brazil Av Brigadeiro Faria Lima n° 5416 Vila Sao Pedro 15090-000 - São José do Rio Preto, SP - Brazil Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil R. do Matão Butantã 05508-090 - São Paulo, SP, Brazil
| | - Adil Bachir Fares
- Department of Molecular Biology, School of Medicine of São José do Rio Preto, SP, Brazil Av Brigadeiro Faria Lima n° 5416 Vila Sao Pedro 15090-000 - São José do Rio Preto, SP - Brazil
| | - João Gustavo da Silva Castro Andrade
- Department of Molecular Biology, School of Medicine of São José do Rio Preto, SP, Brazil Av Brigadeiro Faria Lima n° 5416 Vila Sao Pedro 15090-000 - São José do Rio Preto, SP - Brazil
| | - Carolina Masson
- Department of Molecular Biology, School of Medicine of São José do Rio Preto, SP, Brazil Av Brigadeiro Faria Lima n° 5416 Vila Sao Pedro 15090-000 - São José do Rio Preto, SP - Brazil
| | - Rossana Verónica Mendoza López
- State of São Paulo Cancer Institute - ICESP, SP, Brazil Av. Dr. Arnaldo, 251 Pacaembu 01246-000 - São Paulo, SP - Brazil
| | - Fabio Daumas Nunes
- Department of Stomatology School of Dentistry, University of São Paulo, SP, Brazil Avenida Professor Lineu Prestes, 2227 Butantã 05508-000 - São Paulo, SP - Brazil
| | - Luis Paulo Kowalski
- Department of Head and Neck Surgery and Otorhinolaryngology, Cancer Hospital A.C. Camargo, SP, Brazil Rua Prof Antonio Prudente, 211 Liberdade 01509-010 - São Paulo, SP - Brazil and
| | - Patricia Severino
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, SP, Brazil Av. Albert Einstein, 627 Morumbi 05652-000 - São Paulo, SP - Brazil
| | - Eloiza Helena Tajara
- Department of Molecular Biology, School of Medicine of São José do Rio Preto, SP, Brazil Av Brigadeiro Faria Lima n° 5416 Vila Sao Pedro 15090-000 - São José do Rio Preto, SP - Brazil Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil R. do Matão Butantã 05508-090 - São Paulo, SP, Brazil
| |
Collapse
|
166
|
Franco J, Balaji U, Freinkman E, Witkiewicz AK, Knudsen ES. Metabolic Reprogramming of Pancreatic Cancer Mediated by CDK4/6 Inhibition Elicits Unique Vulnerabilities. Cell Rep 2016; 14:979-990. [PMID: 26804906 PMCID: PMC4757440 DOI: 10.1016/j.celrep.2015.12.094] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/23/2015] [Accepted: 12/19/2015] [Indexed: 12/22/2022] Open
Abstract
Due to loss of p16ink4a in pancreatic ductal adenocarcinoma (PDA), pharmacological suppression of CDK4/6 could represent a potent target for treatment. In PDA models, CDK4/6 inhibition had a variable effect on cell cycle but yielded accumulation of ATP and mitochondria. Pharmacological CDK4/6 inhibitors induce cyclin D1 protein levels; however, RB activation was required and sufficient for mitochondrial accumulation. CDK4/6 inhibition stimulated glycolytic and oxidative metabolism and was associated with an increase in mTORC1 activity. MTOR and MEK inhibitors potently cooperate with CDK4/6 inhibition in eliciting cell-cycle exit. However, MTOR inhibition fully suppressed metabolism and yielded apoptosis and suppression of tumor growth in xenograft models. The metabolic state mediated by CDK4/6 inhibition increases mitochondrial number and reactive oxygen species (ROS). Concordantly, the suppression of ROS scavenging or BCL2 antagonists cooperated with CDK4/6 inhibition. Together, these data define the impact of therapeutics on PDA metabolism and provide strategies for converting cytostatic response to tumor cell killing.
Collapse
Affiliation(s)
- Jorge Franco
- McDermott Center University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Uthra Balaji
- McDermott Center University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Elizaveta Freinkman
- Whitehead Institute, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Agnieszka K Witkiewicz
- McDermott Center University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Simmons Cancer Center, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA.
| | - Erik S Knudsen
- McDermott Center University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Simmons Cancer Center, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
167
|
Knudsen ES, O’Reilly EM, Brody JR, Witkiewicz AK. Genetic Diversity of Pancreatic Ductal Adenocarcinoma and Opportunities for Precision Medicine. Gastroenterology 2016; 150:48-63. [PMID: 26385075 PMCID: PMC5010785 DOI: 10.1053/j.gastro.2015.08.056] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 12/20/2022]
Abstract
Patients with pancreatic ductal adenocarcinoma (PDA) have a poor prognosis despite new treatments; approximately 7% survive for 5 years. Although there have been advances in systemic, primarily cytotoxic, therapies, it has been a challenge to treat patients with PDA using targeted therapies. Sequence analyses have provided a wealth of information about the genetic features of PDA and have identified potential therapeutic targets. Preclinical and early-phase clinical studies have found specific pathways could be rationally targeted; it might also be possible to take advantage of the genetic diversity of PDAs to develop therapeutic agents. The genetic diversity and instability of PDA cells have long been thought of as obstacles to treatment, but are now considered exploitable features. We review the latest findings in pancreatic cancer genetics and the promise of targeted approaches in PDA therapy.
Collapse
Affiliation(s)
- Erik S. Knudsen
- Simmons Cancer Center, University of Texas Southwestern Medical Center, TX,Department of Pathology, University of Texas Southwestern Medical Center, TX,CORRESPONDENCE, Erik Knudsen, PHD, UTSW, Dallas TX, , Agnieszka Witkiewicz, UTSW, Dallas TX,
| | - Eileen M. O’Reilly
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, NY
| | - Jonathan R. Brody
- Department of Surgery, Jefferson Pancreatic, Biliary, and Related Cancer Center, Thomas Jefferson University, PA
| | - Agnieszka K. Witkiewicz
- Simmons Cancer Center, University of Texas Southwestern Medical Center, TX,Department of Pathology, University of Texas Southwestern Medical Center, TX,CORRESPONDENCE, Erik Knudsen, PHD, UTSW, Dallas TX, , Agnieszka Witkiewicz, UTSW, Dallas TX,
| |
Collapse
|
168
|
Pacheco-Rivera R, Fattel-Fazenda S, Arellanes-Robledo J, Silva-Olivares A, Alemán-Lazarini L, Rodríguez-Segura M, Pérez-Carreón J, Villa-Treviño S, Shibayama M, Serrano-Luna J. Double staining of β-galactosidase with fibrosis and cancer markers reveals the chronological appearance of senescence in liver carcinogenesis induced by diethylnitrosamine. Toxicol Lett 2016; 241:19-31. [DOI: 10.1016/j.toxlet.2015.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 11/06/2015] [Accepted: 11/07/2015] [Indexed: 01/04/2023]
|
169
|
Franco J, Witkiewicz AK, Knudsen ES. CDK4/6 inhibitors have potent activity in combination with pathway selective therapeutic agents in models of pancreatic cancer. Oncotarget 2015; 5:6512-25. [PMID: 25156567 PMCID: PMC4171647 DOI: 10.18632/oncotarget.2270] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) has a poor prognosis, in part, due to the therapy-recalcitrant nature of the disease. Loss of the CDK4/6 inhibitor CDKN2A is a signature genetic event in PDA. Therefore, PDA may be amenable to treatment with pharmaceutical CDK4/6 inhibitors. Surprisingly, response to CDK4/6 inhibition was highly variable in PDA models, and associated with differential suppression of gene expression. Mitotic genes were repressed and FOXM1 was uniformly attenuated; however, genes involved in DNA replication were uniquely suppressed in sensitive models. Aberrant induction of Cyclin E1 was associated with resistance, and knockdown demonstrated synergistic suppression of the cell cycle with CDK4/6 inhibition. Combination therapies are likely required for the effective treatment of disease, and drug screening demonstrated additive/antagonistic interactions with CDK4/6 inhibitors. Agents dependent on mitotic progression (taxanes/PLK1 inhibitors) were antagonized by CDK4/6 inhibition, while the response to 5-FU and gemcitabine exhibited drug specific interactions. PI3K/MTOR and MEK inhibitors potently cooperated with CDK4/6 inhibition. These agents were synergistic with CDK4/6 inhibition, blocked the aberrant upregulation of Cyclin E1, and yielded potent inhibition of tumor cell growth. Together, these data identify novel mechanisms of resistance to CDK4/6 inhibitions and provide a roadmap for combination therapies in the treatment of PDA.
Collapse
Affiliation(s)
- Jorge Franco
- Department of Pathology , UT Southwestern, Dallas TX
| | - Agnieszka K Witkiewicz
- Department of Pathology , UT Southwestern, Dallas TX; Simmons Cancer Center, UT Southwestern, Dallas TX
| | - Erik S Knudsen
- Department of Pathology , UT Southwestern, Dallas TX; Simmons Cancer Center, UT Southwestern, Dallas TX
| |
Collapse
|
170
|
Wang S, Zheng W. Expression of p16 protein in infantile hemangioma. Oncol Lett 2015; 10:1589-1592. [PMID: 26622715 DOI: 10.3892/ol.2015.3418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 06/05/2015] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the expression and significance of p16 in the occurrence, development and regression of infantile hemangioma (IH). The expression of p16 was examined in proliferating, involuting hemangioma and normal tissues using immunohistochemical techniques. The expression of p16 was significantly lower in proliferating hemangioma than in involuting hemangioma, and was significantly lower in the involuting hemangioma than in normal tissues. Significant differences were found between the three groups (P<0.05). The results indicate that p16 may be important in the regression of IH endothelial cells and in anti-angiogenesis. There is a certain association between p16 expression and the regression of hemangioma. This provides a theoretical basis for the further study of the pathological mechanisms of p16 in hemangioma and potential gene therapies that may treat this disease.
Collapse
Affiliation(s)
- Shi Wang
- Department of Stomatology, The Third Central Hospital, Tianjin 300170, P.R. China
| | - Wei Zheng
- Department of Orthodontics, Tianjin Stomatological Hospital, Nankai University, Tianjin 300041, P.R. China
| |
Collapse
|
171
|
Abstract
Senescence was classically defined as an irreversible cell cycle arrest in G1 phase (G1 exit) triggered by eroded telomeres in aged primary cells. The molecular basis of this G1 arrest is thought to be due to a DNA damage response, resulting in accumulation of the cyclin dependent kinase (Cdk) inhibitors p21 and p16 that block the inactivating phosphorylation of the retinoblastoma tumor suppressor pRb, thereby preventing DNA replication. More than a decade ago, several studies showed that p21 also mediates permanent DNA damage-induced cell cycle arrest in G2 (G2 exit) by inhibiting mitotic Cdk complexes and pRb phosphorylation. The idea that the senescence program can also be launched after G2 arrest has gained support from several recent publications, including evidence for its existence in vivo.
Collapse
Affiliation(s)
- Véronique Gire
- a Centre de Recherche en Biologie Macromoléculaire (CRBM) ; CNRS UMR5237; Montpellier , France
| | | |
Collapse
|
172
|
CDK6-a review of the past and a glimpse into the future: from cell-cycle control to transcriptional regulation. Oncogene 2015; 35:3083-91. [PMID: 26500059 DOI: 10.1038/onc.2015.407] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 09/22/2015] [Accepted: 09/22/2015] [Indexed: 12/19/2022]
Abstract
The G1 cell-cycle kinase CDK6 has long been thought of as a redundant homolog of CDK4. Although the two kinases have very similar roles in cell-cycle progression, it has recently become apparent that they differ in tissue-specific functions and contribute differently to tumor development. CDK6 is directly involved in transcription in tumor cells and in hematopoietic stem cells. These functions point to a role of CDK6 in tissue homeostasis and differentiation that is partially independent of CDK6's kinase activity and is not shared with CDK4. We review the literature on the contribution of CDK6 to transcription in an attempt to link the new findings on CDK6's transcriptional activity to cell-cycle progression. Finally, we note that anticancer therapies based on the inhibition of CDK6 kinase activity fail to take into account its kinase-independent role in tumor development.
Collapse
|
173
|
Liu JQ, Zhang QH, Wang ZL. Clinicopathological significance of p16, cyclin D1, Rb and MIB-1 levels in skull base chordoma and chondrosarcoma. World J Otorhinolaryngol Head Neck Surg 2015; 1:50-56. [PMID: 29204540 PMCID: PMC5698503 DOI: 10.1016/j.wjorl.2015.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/18/2015] [Accepted: 09/29/2015] [Indexed: 11/25/2022] Open
Abstract
Objective To investigate the expression of p16, cyclin D1, retinoblastoma tumor suppressor protein (Rb) and MIB-1 in skull base chordoma and chondrosarcoma tissues, and to determine the clinicopathological significance of the above indexes in these diseases. Methods A total of 100 skull base chordoma, 30 chondrosarcoma, and 20 normal cartilage tissue samples were analyzed by immunohistochemistry. The expression levels of p16, cyclinD1, Rb and MIB-1 proteins were assessed for potential correlation with the clinicopathological features. Results As compared to normal cartilage specimen (control), there was decreased expression of p16, and increased expression of cyclin D1, Rb and MIB-1 proteins, in both skull base chordoma and chondrosarcoma specimens. MIB-1 LI levels were significantly increased in skull base chordoma specimens with negative expression of p16, and positive expression of cyclin D1 and Rb (P < 0.05). Significantly elevated MIB-1 LI was also detected in skull base chondrosarcoma tissues, while there was negative expression of p16, cyclin D1 and Rb (P < 0.05). In skull base chordoma, p16 negatively correlated with cyclin D1 and Rb, while cyclin D1 positively correlated with Rb. Additionally, p16, cyclin D1, Rb, or MIB-1 expression showed no correlation with age, gender, or pathological classification of patients with skull base chordoma (P > 0.05). However, p16 and MIB-1 levels correlated with the intradural invasion, and expression of p16, Rb and MIB-1 correlated with the number of tumor foci (P < 0.05). Further, the expression of p16 and MIB-1 appeared to correlate with the prognosis of patients with skull base chordoma. Conclusions The abnormal expression of p16, cyclin D1 and Rb proteins might be associated with the tumorigenesis of skull base chordoma and chondrosarcoma.
Collapse
Affiliation(s)
- Jun-Qi Liu
- Xuanwu Hospital Capital Medical University, China
| | | | | |
Collapse
|
174
|
Expression Analysis of Genes Involved in the RB/E2F Pathway in Astrocytic Tumors. PLoS One 2015; 10:e0137259. [PMID: 26317630 PMCID: PMC4552853 DOI: 10.1371/journal.pone.0137259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/13/2015] [Indexed: 02/08/2023] Open
Abstract
Astrocytic gliomas, which are derived from glial cells, are considered the most common primary neoplasias of the central nervous system (CNS) and are histologically classified as low grade (I and II) or high grade (III and IV). Recent studies have shown that astrocytoma formation is the result of the deregulation of several pathways, including the RB/E2F pathway, which is commonly deregulated in various human cancers via genetic or epigenetic mechanisms. On the basis of the assumption that the study of the mechanisms controlling the INK4/ARF locus can help elucidate the molecular pathogenesis of astrocytic tumors, identify diagnostic and prognostic markers, and help select appropriate clinical treatments, the present study aimed to evaluate and compare methylation patterns using bisulfite sequencing PCR and evaluate the gene expression profile using real-time PCR in the genes CDKN2A, CDKN2B, CDC6, Bmi-1, CCND1, and RB1 in astrocytic tumors. Our results indicate that all the evaluated genes are not methylated independent of the tumor grade. However, the real-time PCR results indicate that these genes undergo progressive deregulation as a function of the tumor grade. In addition, the genes CDKN2A, CDKN2B, and RB1 were underexpressed, whereas CDC6, Bmi-1, and CCND1 were overexpressed; the increase in gene expression was significantly associated with decreased patient survival. Therefore, we propose that the evaluation of the expression levels of the genes involved in the RB/E2F pathway can be used in the monitoring of patients with astrocytomas in clinical practice and for the prognostic indication of disease progression.
Collapse
|
175
|
Uddin MH, Choi MH, Kim WH, Jang JJ, Hong ST. Involvement of PSMD10, CDK4, and Tumor Suppressors in Development of Intrahepatic Cholangiocarcinoma of Syrian Golden Hamsters Induced by Clonorchis sinensis and N-Nitrosodimethylamine. PLoS Negl Trop Dis 2015; 9:e0004008. [PMID: 26313366 PMCID: PMC4551803 DOI: 10.1371/journal.pntd.0004008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/24/2015] [Indexed: 12/28/2022] Open
Abstract
Background Clonorchis sinensis is a group-I bio-carcinogen for cholangiocarcinoma (CCA). Although the epidemiological evidence links clonorchiasis and CCA, the underlying molecular mechanism involved in this process is poorly understood. In the present study, we investigated expression of oncogenes and tumor suppressors, including PSMD10, CDK4, p53 and RB in C. sinensis induced hamster CCA model. Methods Different histochemical/immunohistochemical techniques were performed to detect CCA in 4 groups of hamsters: uninfected control (Ctrl.), infected with C. sinensis (Cs), ingested N-nitrosodimethylamine (NDMA), and both Cs infected and NDMA introduced (Cs+NDMA). The liver tissues from all groups were analyzed for gene/protein expressions by quantitative PCR (qPCR) and western blotting. Principal Findings CCA was observed in all hamsters of Cs+NDMA group with well, moderate, and poorly differentiated types measured in 21.8% ± 1.5%, 13.3% ± 1.3%, and 10.8% ± 1.3% of total tissue section areas respectively. All CCA differentiations progressed in a time dependent manner, starting from the 8th week of infection. CCA stroma was characterized with increased collagen type I, mucin, and proliferative cell nuclear antigen (PCNA). The qPCR analysis showed PSMD10, CDK4 and p16INK4 were over-expressed, whereas p53 was under-expressed in the Cs+NDMA group. We observed no change in RB1 at mRNA level but found significant down-regulation of RB protein. The apoptosis related genes, BAX and caspase 9 were found downregulated in the CCA tissue. Gene/protein expressions were matched well with the pathological changes of different groups except the NDMA group. Though the hamsters in the NDMA group showed no marked pathological lesions, we observed over-expression of Akt/PKB and p53 genes proposing molecular interplay in this group which might be related to the CCA initiation in this animal model. Conclusions/Significance The present findings suggest that oncogenes, PSMD10 and CDK4, and tumor suppressors, p53 and RB, are involved in the carcinogenesis process of C. sinensis induced CCA in hamsters. Clonorchis sinensis is a helminth parasite and a carcinogenic agent for cholangiocarcinoma (CCA) or bile duct cancer in humans. Though a large and compelling body of evidence suggests an association between C. sinensis and CCA, the mechanism underlying at the genetic/proteomic level is little known. To explore the underlying molecular mechanism we investigated a number of genes/proteins in C. sinensis induced hamster CCA model. Here C. sinensis induced CCA successfully in all hamsters when introduced with N-nitrosodimethylamine. The histopathology confirmed the development of CCA and detected excessive collagen fibers, mucin and cell division related protein. The quantitative PCR analysis showed increased levels of oncogenes PSMD10, CDK4 and decreased level of tumor suppressor gene p53. The western blot analysis observed significant decrease of another tumor suppressor called RB protein. Genes/protein expressions were matched well with the pathological changes of CCA hamster. The present study suggests that oncogenes, PSMD10 and CDK4, and tumor suppressors gene p53 and protein RB, are involved in the carcinogenesis process of C. sinensis induced CCA in hamsters.
Collapse
Affiliation(s)
- Md. Hafiz Uddin
- Department of Parasitology and Tropical Medicine, Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Min-Ho Choi
- Department of Parasitology and Tropical Medicine, Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ja-June Jang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Tae Hong
- Department of Parasitology and Tropical Medicine, Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
176
|
Stewart CJR, Bharat C. Clinicopathological and immunohistological features of polypoid endometriosis. Histopathology 2015; 68:398-404. [DOI: 10.1111/his.12755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/05/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Colin JR Stewart
- Department of Pathology; King Edward Memorial Hospital; Perth WA Australia
- School of Women's and Infants' Health; University of Western Australia; Perth WA Australia
| | - Chrianna Bharat
- Centre for Applied Statistics; University of Western Australia; Perth WA Australia
- Department of Research; Sir Charles Gairdner Hospital; Perth WA Australia
| |
Collapse
|
177
|
Abstract
OBJECTIVE To credential Stathmin 1 (STMN1) and p16(INK4A) (p16) as adjunct markers for the diagnosis of serous tubal intraepithelial carcinoma (STIC), and to compare STMN1 and p16 expression in p53-positive and p53-negative STIC and invasive high-grade serous carcinoma (HGSC). METHODS Immunohistochemistry (IHC) was used to examine STMN1 and p16 expression in fallopian tube specimens (n=31) containing p53-positive and p53-negative STICs, invasive HGSCs, and morphologically normal FTE (fallopian tube epithelium). STMN1 and p16 expression was scored semiquantitatively by four individuals. The semiquantitative scores were dichotomized, and reported as positive or negative. Pooled siRNA was used to knockdown p53 in a panel of cell lines derived from immortalized FTE and HGSC. RESULTS STMN1 and p16 were expressed in the majority of p53-positive and p53-negative STICs and concomitant invasive HGSCs, but only scattered positive cells were present in morphologically normal FTE. Both proteins were expressed consistently across multiple STICs from the same patient and in concomitant invasive HGSC. Knockdown of p53 in immortalized FTE cells and in four HGSC-derived cell lines expressing different missense p53 mutations did not affect STMN1 protein levels. CONCLUSIONS This study demonstrates that STMN1 and p16 are sensitive and specific adjunct biomarkers that, when used with p53 and Ki-67, improve the diagnostic accuracy of STIC. The addition of STMN1 and p16 helps to compensate for practical limitations of p53 and Ki-67 that complicate the diagnosis in up to one third of STICs.
Collapse
|
178
|
Randall JM, Millard F, Kurzrock R. Molecular aberrations, targeted therapy, and renal cell carcinoma: current state-of-the-art. Cancer Metastasis Rev 2015; 33:1109-24. [PMID: 25365943 DOI: 10.1007/s10555-014-9533-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Renal cell carcinoma (RCC) is among the most prevalent malignancies in the USA. Most RCCs are sporadic, but hereditary syndromes associated with RCC account for 2-3 % of cases and include von Hippel-Lindau, hereditary leiomyomatosis, Birt-Hogg-Dube, tuberous sclerosis, hereditary papillary RCC, and familial renal carcinoma. In the past decade, our understanding of the genetic mutations associated with sporadic forms of RCC has increased considerably, with the most common mutations in clear cell RCC seen in the VHL, PBRM1, BAP1, and SETD2 genes. Among these, BAP1 mutations are associated with aggressive disease and decreased survival. Several targeted therapies for advanced RCC have been approved and include sunitinib, sorafenib, pazopanib, axitinib (tyrosine kinase inhibitors (TKIs) with anti-vascular endothelial growth factor (VEGFR) activity), everolimus, and temsirolimus (TKIs that inhibit mTORC1, the downstream part of the PI3K/AKT/mTOR pathway). High-dose interleukin 2 (IL-2) immunotherapy and the combination of bevacizumab plus interferon-α are also approved treatments. At present, there are no predictive genetic markers to direct therapy for RCC, perhaps because the vast majority of trials have been evaluated in unselected patient populations, with advanced metastatic disease. This review will focus on our current understanding of the molecular genetics of RCC, and how this may inform therapeutics.
Collapse
Affiliation(s)
- J Michael Randall
- Department of Medicine, Division of Hematology/Oncology, UCSD Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, #0987, La Jolla, CA, 92093-0987, USA,
| | | | | |
Collapse
|
179
|
Abstract
'Cellular senescence', a term originally defining the characteristics of cultured cells that exceed their replicative limit, has been broadened to describe durable states of proliferative arrest induced by disparate stress factors. Proposed relationships between cellular senescence, tumour suppression, loss of tissue regenerative capacity and ageing suffer from lack of uniform definition and consistently applied criteria. Here, we highlight caveats in interpreting the importance of suboptimal senescence-associated biomarkers, expressed either alone or in combination. We advocate that more-specific descriptors be substituted for the now broadly applied umbrella term 'senescence' in defining the suite of diverse physiological responses to cellular stress.
Collapse
Affiliation(s)
- Norman E Sharpless
- Department of Medicine and Genetics and The Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599-7295, USA
| | - Charles J Sherr
- Department of Tumor Cell Biology and The Howard Hughes Medical Institute, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-2794, USA
| |
Collapse
|
180
|
Expression of p16 in squamous cell carcinoma of the mobile tongue is independent of HPV infection despite presence of the HPV-receptor syndecan-1. Br J Cancer 2015; 113:321-6. [PMID: 26057450 PMCID: PMC4506391 DOI: 10.1038/bjc.2015.207] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/07/2015] [Accepted: 05/13/2015] [Indexed: 02/03/2023] Open
Abstract
Background: Tongue squamous cell carcinoma (TSCC) is increasing in incidence, especially among young patients and preferably females. Infection with human papilloma virus (HPV) has been suggested as a cause of SCC in the head and neck, and the proportion of oropharyngeal cancers caused by HPV has steadily increased. Methods: Samples from 109 patients with primary TSCC were analysed for the presence of HPV16 by in situ hybridisation and for expression of its surrogate marker p16 and the HPV receptor syndecan-1 by immunhistochemistry. Results: No evidence of HPV16 DNA was observed in the tumours, although one-third showed p16 staining. There was no difference in the expression of the primary HPV receptor, syndecan-1, between TSCC and a group of tonsil SCC. Conclusion: Whereas p16 is expressed in some TSCCs, HPV16 is undetectable, therefore, p16 cannot be used as a surrogate marker for high-risk HPV-infection in this tumour. Despite presence of the HPV-receptor syndecan-1 in TSCC, HPV prefers the tonsillar environment. Lack of p16 associates with worse prognosis primarily in patients aged ⩽40 years with tongue SCC. The improved prognosis seen in p16-positive TSCC can be due to induction of a senescent phenotype or an inherent radiosensitivity due to the ability of p16 to inhibit homologous recombination repair.
Collapse
|
181
|
Isayeva T, Xu J, Ragin C, Dai Q, Cooper T, Carroll W, Dayan D, Vered M, Wenig B, Rosenthal E, Grizzle W, Anderson J, Willey CD, Yang ES, Brandwein-Gensler M. The protective effect of p16(INK4a) in oral cavity carcinomas: p16(Ink4A) dampens tumor invasion-integrated analysis of expression and kinomics pathways. Mod Pathol 2015; 28:631-53. [PMID: 25523612 DOI: 10.1038/modpathol.2014.149] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/08/2014] [Accepted: 09/11/2014] [Indexed: 11/09/2022]
Abstract
A large body of evidence shows that p16(INK4a) overexpression predicts improved survival and increased radiosensitivity in HPV-mediated oropharyngeal squamous cell carcinomas.(OPSCC). Here we demonstrate that the presence of transcriptionally active HPV16 in oral cavity squamous cell carcinomas does not correlate with p16(INK4a) overexpression, enhanced local tumor immunity, or improved outcome. It is interesting that HPV-mediated oropharyngeal squamous cell carcinomas can be categorized as having a 'nonaggressive' invasion phenotype, whereas aggressive invasion phenotypes are more common in HPV-negative squamous cell carcinomas. We have developed primary cancer cell lines from resections with known pattern of invasion as determined by our validated risk model. Given that cell lines derived from HPV-mediated oropharyngeal squamous cell carcinomas are less invasive than their HPV-negative counterparts, we tested the hypothesis that viral oncoproteins E6, E7, and p16(INK4a) can affect tumor invasion. Here we demonstrate that p16(INK4a) overexpression in two cancer cell lines (UAB-3 and UAB-4), derived from oral cavity squamous cell carcinomas with the most aggressive invasive phenotype (worst pattern of invasion type 5 (WPOI-5)), dramatically decreases tumor invasiveness by altering expression of extracellular matrix remodeling genes. Pathway analysis integrating changes in RNA expression and kinase activities reveals different potential p16(INK4a)-sensitive pathways. Overexpressing p16(INK4a) in UAB-3 increases EGFR activity and increases MMP1 and MMP3 expression, possibly through STAT3 activation. Overexpressing p16(INK4a) in UAB-4 decreases PDGFR gene expression and reduces MMP1 and MMP3, possibly through STAT3 inactivation. Alternatively, ZAP70/Syk might increase MUC1 phosphorylation, leading to the observed decreased MMP1 expression.
Collapse
Affiliation(s)
- Tatyana Isayeva
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jie Xu
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Camille Ragin
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Temple University Health, Philadelphia, PA, USA
| | - Qian Dai
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tiffiny Cooper
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William Carroll
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dan Dayan
- The Maurice and Gabriela Goldschleger School of Dental Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Marilena Vered
- The Maurice and Gabriela Goldschleger School of Dental Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Bruce Wenig
- Department of Pathology, Beth Israel Medical Center, Continuum Health Partners, New York, NY, USA
| | - Eben Rosenthal
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William Grizzle
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joshua Anderson
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christopher D Willey
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eddy S Yang
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Margaret Brandwein-Gensler
- Departments of Pathology, Surgery, Medicine, Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
182
|
Khemthongcharoen N, Ruangpracha A, Sarapukdee P, Rattanavarin S, Jolivot R, Jarujareet U, Plaimas K, Bhattarakosol P, Patumraj S, Piyawattanametha W. Novel p16 binding peptide development for p16-overexpressing cancer cell detection using phage display. J Pept Sci 2015; 21:265-73. [PMID: 25754556 DOI: 10.1002/psc.2726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/24/2014] [Accepted: 11/24/2014] [Indexed: 12/31/2022]
Abstract
Protein p(16INK4a) (p16) is a well-known biomarker for diagnosis of human papillomavirus (HPV) related cancers. In this work, we identify novel p16 binding peptides by using phage display selection method. A random heptamer phage display library was screened on purified recombinant p16 protein-coated plates to elute only the bound phages from p16 surfaces. Binding affinity of the bound phages was compared with each other by enzyme-linked immunosorbent assay (ELISA), fluorescence imaging technique, and bioinformatic computations. Binding specificity and binding selectivity of the best candidate phage-displayed p16 binding peptide were evaluated by peptide blocking experiment in competition with p16 monoclonal antibody and fluorescence imaging technique, respectively. Five candidate phage-displayed peptides were isolated from the phage display selection method. All candidate p16 binding phages show better binding affinity than wild-type phage in ELISA test, but only three of them can discriminate p16-overexpressing cancer cell, CaSki, from normal uterine fibroblast cell, HUF, with relative fluorescence intensities from 2.6 to 4.2-fold greater than those of wild-type phage. Bioinformatic results indicate that peptide 'Ser-His-Ser-Leu-Leu-Ser-Ser' binds to p16 molecule with the best binding score and does not interfere with the common protein functions of p16. Peptide blocking experiment shows that the phage-displayed peptide 'Ser-His-Ser-Leu-Leu-Ser-Ser' can conceal p16 from monoclonal antibody interaction. This phage clone also selectively interacts with the p16 positive cell lines, and thus, it can be applied for p16-overexpressing cell detection.
Collapse
Affiliation(s)
- Numfon Khemthongcharoen
- NECTEC, National Science and Technology Development Agency (NSTDA), Pathumthani, 12120, Thailand; Advanced Imaging Research Center, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Kandefer-Gola M, Nowak M, Madej J, Dzimira S, Ciaputa R, Janus I. Useful immunohistochemical indicators in canine mast cell tumours. Acta Vet Hung 2015; 63:49-59. [PMID: 25655414 DOI: 10.1556/avet.2015.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Morphological and immunohistochemical analysis of 45 canine mast cell tumours was performed to determine whether the proteins examined are useful for a more precise description of tumour morphology and a more reliable determination of the prognosis in patients. Tissue sections were stained according to the standard haematoxylin and eosin (HE) technique and with toluidine blue to demonstrate cytoplasmic granules. Immunohistochemical studies were performed, using the cell markers CD117 (c-kit), p16 and von Willebrand factor (FVIII). In CD117 three different staining patterns were observed: (1) membranous reaction, (2) intense staining of cytoplasm, and (3) a diffuse, delicate cytoplasmic reaction. Von Willebrand antibody was evaluated on the basis of the number of blood vessels stained. p16 expression was evaluated by scoring positive nuclear reaction. Positive expression was demonstrated for all examined antigens, but their level of expression differed depending on the grades of tumour malignancy. Statistical analysis of the results documented a pronounced positive correlation between the markers studied and the grade of tumour malignancy (P < 0.001). It was shown that each of the cell markers examined represents a useful prognostic indicator for patients with mast cell tumours. The calculated correlation coefficients demonstrate a strong association between the expressions of CD117, FVIII and p16, and the histological malignancy of a tumour.
Collapse
Affiliation(s)
- Małgorzata Kandefer-Gola
- 1 Wroclaw University of Environmental and Life Sciences Department of Pathology, Faculty of Veterinary Medicine ul. C. K. Norwida 31 50-375 Wroclaw Poland
| | - Marcin Nowak
- 1 Wroclaw University of Environmental and Life Sciences Department of Pathology, Faculty of Veterinary Medicine ul. C. K. Norwida 31 50-375 Wroclaw Poland
| | - Janusz Madej
- 1 Wroclaw University of Environmental and Life Sciences Department of Pathology, Faculty of Veterinary Medicine ul. C. K. Norwida 31 50-375 Wroclaw Poland
| | - Stanisław Dzimira
- 1 Wroclaw University of Environmental and Life Sciences Department of Pathology, Faculty of Veterinary Medicine ul. C. K. Norwida 31 50-375 Wroclaw Poland
| | - Rafal Ciaputa
- 1 Wroclaw University of Environmental and Life Sciences Department of Pathology, Faculty of Veterinary Medicine ul. C. K. Norwida 31 50-375 Wroclaw Poland
| | - Izabela Janus
- 1 Wroclaw University of Environmental and Life Sciences Department of Pathology, Faculty of Veterinary Medicine ul. C. K. Norwida 31 50-375 Wroclaw Poland
| |
Collapse
|
184
|
Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov 2015; 14:130-46. [PMID: 25633797 PMCID: PMC4480421 DOI: 10.1038/nrd4504] [Citation(s) in RCA: 1313] [Impact Index Per Article: 131.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cancer represents a pathological manifestation of uncontrolled cell division; therefore, it has long been anticipated that our understanding of the basic principles of cell cycle control would result in effective cancer therapies. In particular, cyclin-dependent kinases (CDKs) that promote transition through the cell cycle were expected to be key therapeutic targets because many tumorigenic events ultimately drive proliferation by impinging on CDK4 or CDK6 complexes in the G1 phase of the cell cycle. Moreover, perturbations in chromosomal stability and aspects of S phase and G2/M control mediated by CDK2 and CDK1 are pivotal tumorigenic events. Translating this knowledge into successful clinical development of CDK inhibitors has historically been challenging, and numerous CDK inhibitors have demonstrated disappointing results in clinical trials. Here, we review the biology of CDKs, the rationale for therapeutically targeting discrete kinase complexes and historical clinical results of CDK inhibitors. We also discuss how CDK inhibitors with high selectivity (particularly for both CDK4 and CDK6), in combination with patient stratification, have resulted in more substantial clinical activity.
Collapse
Affiliation(s)
- Uzma Asghar
- Breakthrough Breast Cancer Research Centre, Chester Beatty Laboratories, Institute of Cancer Research, London, SW3 6JB, UK
| | - Agnieszka K Witkiewicz
- Simmons Cancer Center and Department of Pathology, University of Texas Southwestern, Dallas, USA
| | - Nicholas C Turner
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust Breast Cancer Unit, London, SW3 6JJ, UK
| | - Erik S Knudsen
- Simmons Cancer Center and Department of Pathology, University of Texas Southwestern, Dallas, USA
| |
Collapse
|
185
|
Liu Y, Zhong X, Wan S, Zhang W, Lin J, Zhang P, Li Y. p16(INK4a) expression in retinoblastoma: a marker of differentiation grade. Diagn Pathol 2014; 9:180. [PMID: 25499675 PMCID: PMC4300043 DOI: 10.1186/s13000-014-0180-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 09/07/2014] [Indexed: 05/05/2025] Open
Abstract
Background The tumor suppressor protein p16INK4a has been extensively studied in many tumors with very different results, ranging from its loss to its clear overexpression, which may be associated with degree of tumor differentiation and prognosis. However, its expression remains unclear in human retinoblastoma (RB), a common malignant tumor of retina in childhood. The aim of this study was to explore the expression pattern of p16INK4a in RB, and the correlation between p16INK4a expression and histopathological features of RB. Methods Sixty-five cases of RB were retrospectively analyzed. Paraffin-embedded blocks were retrieved from the archives of ocular pathology department at Zhongshan Ophthalmic Center of Sun Yat-sen University, China. Serial sections were cut and subjected to hematoxylin and eosin staining. Immunohistochemical staining was further done with antibodies p16INK4a, CRX and Ki67. The correlation of p16 INK4a expression with CRX and Ki67 and clinicopathological features of RB were analyzed. Results RB tumor histologically consists of various differentiation components including undifferentiated (UD) cells, Homer-Wright rosettes (HWR) or Flexner-Winterstein rosettes (FWR) and fleurettes characteristic of photoreceptor differentiation or Retinocytoma (RC). p16INK4a expression was negative in both fleurette region and the residual retinal tissue adjacent to the tumor, weakly to moderately positive in FWR, strongly positive in both HWR and UD region. However, CRX had the reverse expression patterns in comparison with p16INK4a. It was strongly positive in photoreceptor cells within the residual retina and fleurettes, but weakly to moderately positive in UD area. Together with Ki67 staining, high p16INK4a expression was associated with poor histological differentiation of RB tumors, which had higher risk features with the optic nerve invasion and uveal invasion. Conclusions p16INK4a expression increased with the decreasing level of cell differentiation of RBs. RB tumors extensively expressing p16INK4a tended to have higher risk features with poor prognosis. This study suggested that p16INK4a would be a valuable molecular marker of RB to distinguish its histological phenotypes and to serve as a predictor of its prognosis. Virtual Slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/13000_2014_180
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 S Xianlie Rd, Guangzhou, 510060, China.
| | - Xiufeng Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 S Xianlie Rd, Guangzhou, 510060, China.
| | - Shangtao Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 S Xianlie Rd, Guangzhou, 510060, China.
| | - Wenxin Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 S Xianlie Rd, Guangzhou, 510060, China.
| | - Jianxian Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 S Xianlie Rd, Guangzhou, 510060, China.
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 S Xianlie Rd, Guangzhou, 510060, China.
| | - Yongping Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 S Xianlie Rd, Guangzhou, 510060, China.
| |
Collapse
|
186
|
Hsu CK, Chen YC, Yang WL, Hsu KF, Chao SC, Lee JYY. Bowen's disease with features resembling myrmecia wart. J Dermatol 2014; 42:90-3. [PMID: 25387725 DOI: 10.1111/1346-8138.12691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 09/29/2014] [Indexed: 11/28/2022]
Abstract
We report the clinical and pathological findings of two cases of Bowen's disease (BD) with features resembling myrmecia wart, and tried to find evidence of human papillomavirus (HPV) infection in such lesions by immunohistological staining, genotyping systems, polymerase chain reaction (PCR) and electron microscopy. Both cases manifested unique barnacle-like hyperkeratotic nodules or plaques clinically, and microscopically proliferation of atypical keratinocytes involving the entire thickness of the epidermis, hypergranulosis with eosinophilic and/or basophilic inclusion bodies, features that mimicked myrmecia wart. Electron microscopy revealed myrmecia inclusion-like large intranuclear and cytoplasmic electron-dense bodies. Immunohistological staining with anti-HPV antibody, genotyping systems for HPV infection and specific PCR designed to detect HPV-1 L1 sequences failed to detect evidence of HPV infection. P16(INK4a) was overexpressed in the atypical keratinocytes of both cases. This finding suggests that the pathogenesis of these two BD may involve certain unknown or undetectable HPV, or reflect disturbances of the Rb signaling pathway unrelated to HPV infection. The unique "myrmecioid" clinicopathological features in our cases suggest that this type of lesion may be a new variant of BD.
Collapse
Affiliation(s)
- Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan; Institute of Clinical Medicine, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan; International Research Center of Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
187
|
Llanos AA, Dumitrescu RG, Brasky TM, Liu Z, Mason JB, Marian C, Makambi KH, Spear SL, Kallakury BVS, Freudenheim JL, Shields PG. Relationships among folate, alcohol consumption, gene variants in one-carbon metabolism and p16INK4a methylation and expression in healthy breast tissues. Carcinogenesis 2014; 36:60-7. [PMID: 25344837 DOI: 10.1093/carcin/bgu219] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
p16(INK4a) is a tumor suppressor gene, frequently hypermethylated in breast cancer; this epigenetic silencing of p16(INK4a) occurs early in carcinogenesis. The risk factors and functional consequences of p16(INK4a) methylation are unknown. Alcohol consumption, a breast cancer risk factor, impedes folate metabolism and may thereby alter gene methylation since folate plays a pivotal role in DNA methylation. In a cross-sectional study of 138 women with no history of breast cancer who underwent reduction mammoplasty, we studied breast cancer risk factors, plasma and breast folate concentrations, variation in one-carbon metabolism genes, p16(INK4a) promoter methylation and P16 protein expression. Logistic regression was used to estimate multivariable-adjusted odds ratios (OR) and 95% confidence intervals (CI). p16(INK4a) methylation was negatively correlated with P16 expression (r = -0.28; P = 0.002). Alcohol consumption was associated with lower breast folate (P = 0.03), higher p16(INK4a) promoter methylation (P = 0.007) and less P16 expression (P = 0.002). Higher breast folate concentrations were associated with lower p16(INK4a) promoter methylation (P = 0.06). Genetic variation in MTRR (rs1801394) and MTHFD1 (rs1950902) was associated with higher p16 (INK4a) promoter methylation (OR = 2.66, 95% CI: 1.11-6.42 and OR = 2.72, 95% CI: 1.12-6.66, respectively), whereas variation in TYMS (rs502396) was associated with less P16 protein expression (OR = 0.22, 95% CI: 0.05-0.99). Given that this is the first study to indicate that alcohol consumption, breast folate and variation in one-carbon metabolism genes are associated with p16(INK4a) promoter methylation and P16 protein expression in healthy tissues; these findings require replication.
Collapse
Affiliation(s)
- Adana A Llanos
- Division of Population Sciences, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43201, USA, Department of Epidemiology, RBHS-School of Public Health and Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08903, USA
| | - Ramona G Dumitrescu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA, Department of Medical Genetics and Epidemiology, Basic Sciences Program, Saba University School of Medicine, Saba, Dutch Caribbean, The Netherlands
| | - Theodore M Brasky
- Division of Cancer Prevention and Control, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Zhenhua Liu
- Human Nutrition Research Center, Tufts University, Boston, MA 02111, USA, Department of Nutrition, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Joel B Mason
- Human Nutrition Research Center, Tufts University, Boston, MA 02111, USA
| | - Catalin Marian
- Division of Cancer Prevention and Control, College of Medicine, The Ohio State University, Columbus, OH 43210, USA, Department of Biochemistry and Pharmacology, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Kepher H Makambi
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC 20057, USA
| | - Scott L Spear
- Department of Plastic Surgery, Georgetown University, Washington, DC 20057, USA
| | | | - Jo L Freudenheim
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Peter G Shields
- Division of Cancer Prevention and Control, College of Medicine, The Ohio State University, Columbus, OH 43210, USA,
| |
Collapse
|
188
|
Shimizu A, Kato M, Takeuchi Y, Sano T, Kaira K, Uezato H, Ishikawa O. Detection of human papillomavirus (
HPV
) in patients with squamous cell carcinoma and the clinical characteristics of
HPV
‐positive cases. Br J Dermatol 2014; 171:779-85. [DOI: 10.1111/bjd.13234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2014] [Indexed: 12/12/2022]
Affiliation(s)
- A. Shimizu
- Department of Dermatology Gunma University Graduate School of Medicine 3‐39‐22 Showa‐machi Maebashi Gunma371‐8511 Japan
| | - M. Kato
- Department of Dermatology Gunma University Graduate School of Medicine 3‐39‐22 Showa‐machi Maebashi Gunma371‐8511 Japan
| | - Y. Takeuchi
- Department of Dermatology Gunma University Graduate School of Medicine 3‐39‐22 Showa‐machi Maebashi Gunma371‐8511 Japan
| | - T. Sano
- Department of Diagnostic Pathology Gunma University Graduate School of Medicine 3‐39‐22 Showa‐machi Maebashi Gunma371‐8511 Japan
| | - K. Kaira
- Department of Oncology Clinical Development Gunma University Graduate School of Medicine 3‐39‐22 Showa‐machi Maebashi Gunma371‐8511 Japan
| | - H. Uezato
- Department of Dermatology Graduate School of Medicine University of the Ryukyus 207 Uehara Nishihara Okinawa 903‐0215 Japan
| | - O. Ishikawa
- Department of Dermatology Gunma University Graduate School of Medicine 3‐39‐22 Showa‐machi Maebashi Gunma371‐8511 Japan
| |
Collapse
|
189
|
Witkiewicz AK, Cox D, Knudsen ES. CDK4/6 inhibition provides a potent adjunct to Her2-targeted therapies in preclinical breast cancer models. Genes Cancer 2014; 5:261-72. [PMID: 25221644 PMCID: PMC4162138 DOI: 10.18632/genesandcancer.24] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 08/06/2014] [Indexed: 01/27/2023] Open
Abstract
In spite of the efficacy of Her2-targeted therapies, recurrence and progression remain a challenge for treatment of Her2 positive breast cancer. CDK4/6 controls pathway downstream of Her2, Inhibition of these kinases could represent an important therapeutic approach to augment the effectiveness of standard therapies. In models of acquired resistance to Her2-targeted therapies, Cyclin D1 was inappropriately activated and CDK4/6 inhibition was effective at blocking proliferation by targeting this common pathway associated with resistance. These data were recapitulated in Her2 positive xenografts. Furthermore, in a series of 35 primary breast tumor explants, treatment with PD-0332991 resulted in a greater than 4-fold suppression of the Ki67. The effects of CDK4/6 inhibition were dependent on an intact RB-pathway, and consonantly, loss of RB and high-levels of p16 were associated with resistance to CDK4/6 inhibition. Combination studies illustrated that CDK4/6 inhibition is cooperative with multiple Her2-targeted agents and provides a complementary mechanism of action to T-DM1 to efficiently suppresses the proliferation of residual Her2-positive tumor cell populations that survive T-DM1. Together, these data indicate CDK4/6 is a viable therapeutic target that functions downstream of Her2, and tissue based markers are available to direct rational utilization of CDK4/6 inhibitors in combination with Her2-targeted agents.
Collapse
Affiliation(s)
- Agnieszka K Witkiewicz
- Department of Pathology, Simmons Cancer Center, Dalls, TX ; Department of Pathology, UT Southwestern, Dallas, TX
| | - Derek Cox
- Department of Pathology, Simmons Cancer Center, Dalls, TX
| | - Erik S Knudsen
- Department of Pathology, Simmons Cancer Center, Dalls, TX ; Department of Pathology, UT Southwestern, Dallas, TX
| |
Collapse
|
190
|
Guan C, Shi H, Wang H, Zhang J, Ni W, Chen B, Hou S, Yang X, Shen A, Ni R. CtBP2 contributes to malignant development of human esophageal squamous cell carcinoma by regulation of p16INK4A. J Cell Biochem 2014; 114:1343-54. [PMID: 23255392 DOI: 10.1002/jcb.24475] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 11/28/2012] [Indexed: 12/26/2022]
Abstract
C-terminal binding protein-2 (CtBP2), as a transcriptional co-repressor, has been shown to mediate the repression of p16(INK4A) , a tumor suppressor gene product, in primary human cells. Here we aimed to investigate how the correlation between CtBP2 and p16(INK4A) influenced the development of esophageal squamous cell carcinoma (ESCC). Immunohistochemistry of ESCC tissue sections indicated that the CtBP2 and p16(INK4A) expressions were inversely correlated to each other with a linear regression coefficient of -0.747 (P < 0.05), and Western blot analysis revealed that CtBP2 was higher expressed in tumorous tissues than in adjacent non-tumorous tissues. Either CtBP2 or p16(INK4A) expression was significantly related to histological differentiation (P = 0.016 or 0.001) and to the expression of Ki-67, a proliferating marker (P = 0.006 or 0.02), and patients with higher CtBP2 and lower p16(INK4A) expressions had shorter overall survival. We also observed that CtBP2 modulated the cell proliferation and cell cycle in ECA109 cells, an ESCC cell line, by inhibiting p16(INK4A) . Overexpression or knockdown of CtBP2 in ECA109 cells was found to inhibit or activate the mRNA or protein expression of p16(INK4A) , which in turn altered the cell proliferation and cell cycle in ECA109 cells, as measured by flow cytometry and cell count assay. Additionally, after ECA109 cells silenced for CtBP2 were treated with cisplatin (an anti-ESCC agent), the p16(INK4A) expression was up-regulated, and the cell apoptosis was promoted, thus confirming the repression of p16(INK4A) by CtBP2. Collectively, all results suggested that CtBP2 might contribute to the progression of ESCC through a negative transcriptional regulation of p16(INK4A).
Collapse
Affiliation(s)
- Chengqi Guan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Wu G, Fan H, Huang Y, Zheng C, Ye J, Liu X. Duhuo Jisheng Decoction‑containing serum promotes proliferation of interleukin‑1β‑induced chondrocytes through the p16‑cyclin D1/CDK4‑Rb pathway. Mol Med Rep 2014; 10:2525-34. [PMID: 25189115 DOI: 10.3892/mmr.2014.2527] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 08/06/2014] [Indexed: 11/06/2022] Open
Abstract
Duhuo Jisheng Decoction (DHJSD) is a traditional Chinese herbal medicine that has multiple uses, including as a treatment for osteoarthritis (OA). However, the molecular mechanisms underlying the therapeutic effects of DHJSD on OA remain unknown. In the present study, a serum pharmacological method was applied to investigate the effects of DHJSD on the proliferation of chondrocytes treated with interleukin‑1β (IL‑1β) in vitro. This is a cell model commonly used to reproduce the mechanisms involved in degenerative arthropathies, including OA. The most effective intervention conditions of DHJSD serum were examined by MTT assay. The degenerative chondrocyte model was established by IL‑1β‑culture for 24 h, and was verified by optical microscopy and immunohistochemical analyses. Following the successful establishment of the degenerative chondrocyte model, the chondrocytes were subsequently randomly divided into two groups: The blank serum group and the DHJSD treatment group. Subsequent to treatment with the corresponding serum, cell proliferation was detected by MTT assay and DNA staining followed by FACS analysis, and the mRNA and protein expression levels of cyclin D1, cyclin‑dependent kinase 4 (CDK4), retinoblastoma tumor suppressor protein (Rb) and p16 were measured by reverse transcription polymerase chain reaction and western blotting, respectively. The results indicated that the most effective condition for the promotion of chondrocyte proliferation was 10% concentration of DHJSD 2‑h serum, and the degenerative chondrocyte model was successfully reproduced by IL‑1β‑treatment for 24 h. The mRNA and protein expression levels of cyclin D1, CDK4 and Rb in the DHJSD serum‑treated cells were significantly increased compared with those in the blank serum group, whereas p16 expression was significantly downregulated. These results indicate that treatment of cells with DHJSD‑containing serum is able to promote IL‑1β‑induced chondrocyte proliferation by promoting G1/S phase transition via modulating the expressions of cyclin D1, CDK4, Rb and p16, which contribute to the clinical efficacy of DHJSD in OA.
Collapse
Affiliation(s)
- Guangwen Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou 350122, P.R. China
| | - Huailing Fan
- Academy of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou 350122, P.R. China
| | - Yuanpeng Huang
- Fujian Sports Vocational Education and Technical College, Fujian, Fuzhou 350003, P.R. China
| | - Chunsong Zheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou 350122, P.R. China
| | - Jinxia Ye
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou 350122, P.R. China
| | - Xianxiang Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou 350122, P.R. China
| |
Collapse
|
192
|
Milea A, George SHL, Matevski D, Jiang H, Madunic M, Berman HK, Gauthier ML, Gallie B, Shaw PA. Retinoblastoma pathway deregulatory mechanisms determine clinical outcome in high-grade serous ovarian carcinoma. Mod Pathol 2014; 27:991-1001. [PMID: 24336157 DOI: 10.1038/modpathol.2013.218] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/01/2013] [Accepted: 10/06/2013] [Indexed: 01/08/2023]
Abstract
Alterations in the retinoblastoma pathway are frequent in ovarian/tubal high-grade serous cancers, but the mechanism of deregulation and the impact on patient outcome are poorly understood. A cohort of 334 high-grade serous carcinomas was studied by immunohistochemical analysis of RB1, p16, cyclin D1, cyclin E1, and Ki67. Additional detailed analyses including RB1 allelic deletion (n=42), mutation (n=75), methylation (n=31), and SNP array analyses (n=75) were performed on cases with clinical parameters, including age, debulking status, treatment, and clinical outcome. p16/RB1 expression results yielded three distinct clinically relevant subgroups upon multivariable analysis controlling for stage, debulking status, and treatment types: p16 homogeneous/RB1+ with the shortest progression-free survival (median 15 months (95% CI: 13-18); P=0.016) compared with the p16 heterogeneous/RB1+ subgroup (median 22 months (95% CI: 16-32)) and the p16 homogeneous/RB1- subgroup (median 20 months (95% CI: 15-24)). Patients in the p16 homo/RB1- subgroup showed a significant increase in overall survival (>60 months; P=0.013), which suggests an increase in sensitivity to cytotoxic agents. Analyses of Rb pathway mechanistic differences among these groups revealed frequent RB1 genomic alterations such as RB1 allelic loss and/or large spanning deletions (83%) in the p16 homo/RB1- subgroups, also indicating that RB1 deletions are frequent in high-grade serous carcinoma. CCNE1 gene gains/amplifications were frequent in the p16 homogeneous/RB1+ subgroup (68%) and cyclin D1 protein overexpression was predominantly characteristic of the p16 heterogeneous/RB1+ subgroup. These subcategories occur early in tumor progression and are seen with similar frequency in the cancer precursor lesion, serous tubal intra-epithelial carcinoma. Overall, this study uniquely identifies multiple non-synonymous mechanisms of retinoblastoma pathway deregulation that correlate with significantly different clinical outcomes. Furthermore, deregulations identified in precursor lesions suggest a key role of this pathway in serous tumor development. Recognition of these categories may identify patients with increased sensitivity to chemotherapy and new opportunities for novel therapeutics.
Collapse
Affiliation(s)
- Anca Milea
- 1] Campbell Family Institute for Breast Cancer Research, Toronto, ON, Canada [2] Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada [3] Princess Margaret Cancer Centre, Toronto, ON, Canada [4] Department of Pathology, University Health Network, Toronto, ON, Canada
| | - Sophia H L George
- 1] Campbell Family Institute for Breast Cancer Research, Toronto, ON, Canada [2] Princess Margaret Cancer Centre, Toronto, ON, Canada [3] Department of Pathology, University Health Network, Toronto, ON, Canada
| | - Donco Matevski
- 1] Department of Pathology, University Health Network, Toronto, ON, Canada [2] Impact Genetics, Toronto, ON, Canada
| | - Haiyan Jiang
- 1] Princess Margaret Cancer Centre, Toronto, ON, Canada [2] Department of Pathology, University Health Network, Toronto, ON, Canada
| | - Mary Madunic
- 1] Princess Margaret Cancer Centre, Toronto, ON, Canada [2] Department of Pathology, University Health Network, Toronto, ON, Canada
| | - Hal K Berman
- 1] Campbell Family Institute for Breast Cancer Research, Toronto, ON, Canada [2] Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada [3] Princess Margaret Cancer Centre, Toronto, ON, Canada [4] Department of Pathology, University Health Network, Toronto, ON, Canada
| | - Mona L Gauthier
- 1] Campbell Family Institute for Breast Cancer Research, Toronto, ON, Canada [2] Princess Margaret Cancer Centre, Toronto, ON, Canada [3] Department of Pathology, University Health Network, Toronto, ON, Canada [4] Department of Medical Biophyics, University of Toronto, Toronto, ON, Canada
| | - Brenda Gallie
- 1] Princess Margaret Cancer Centre, Toronto, ON, Canada [2] Department of Pathology, University Health Network, Toronto, ON, Canada [3] Impact Genetics, Toronto, ON, Canada [4] Department of Medical Biophyics, University of Toronto, Toronto, ON, Canada [5] Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Patricia A Shaw
- 1] Campbell Family Institute for Breast Cancer Research, Toronto, ON, Canada [2] Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada [3] Princess Margaret Cancer Centre, Toronto, ON, Canada [4] Department of Pathology, University Health Network, Toronto, ON, Canada
| |
Collapse
|
193
|
Witkiewicz AK, Knudsen ES. Retinoblastoma tumor suppressor pathway in breast cancer: prognosis, precision medicine, and therapeutic interventions. Breast Cancer Res 2014; 16:207. [PMID: 25223380 PMCID: PMC4076637 DOI: 10.1186/bcr3652] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A series of recent studies have demonstrated that the retinoblastoma tumor suppressor (RB) pathway plays a critical role in multiple clinically relevant aspects of breast cancer biology, spanning early stage lesions to targeted treatment of metastatic disease. In ductal carcinoma in situ, multiple groups have shown that dysregulation of the RB pathway is critically associated with recurrence and disease progression. Functional models have similarly illustrated key roles for RB in regulating epithelial–mesenchymal transition and other features contributing to aggressive disease. Invasive breast cancers are treated in distinct fashions, and heterogeneity within the RB pathway relates to prognosis and response to commonly used therapeutics. Luminal B breast cancers that have a poor prognosis amongst estrogen receptor-positive disease are defined based on the expression of RB-regulated genes. Such findings have led to clinical interventions that directly target the RB pathway through CDK4/6 inhibition which have promise in both estrogen receptor-positive and Her2-positive disease. In contrast, RB loss results in improved response to chemotherapy in triple-negative breast cancer, where ongoing research is attempting to define intrinsic vulnerabilities for targeted intervention. These findings support a wide-reaching impact of the RB pathway on disease that could be harnessed for improved clinical interventions.
Collapse
|
194
|
Dimopoulos K, Gimsing P, Grønbæk K. The role of epigenetics in the biology of multiple myeloma. Blood Cancer J 2014; 4:e207. [PMID: 24786391 PMCID: PMC4042299 DOI: 10.1038/bcj.2014.29] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/12/2014] [Accepted: 03/31/2014] [Indexed: 12/19/2022] Open
Abstract
Several recent studies have highlighted the biological complexity of multiple myeloma (MM) that arises as a result of several disrupted cancer pathways. Apart from the central role of genetic abnormalities, epigenetic aberrations have also been shown to be important players in the development of MM, and a lot of research during the past decades has focused on the ways DNA methylation, histone modifications and noncoding RNAs contribute to the pathobiology of MM. This has led to, apart from better understanding of the disease biology, the development of epigenetic drugs, such as histone deacetylase inhibitors that are already used in clinical trials in MM with promising results. This review will present the role of epigenetic abnormalities in MM and how these can affect specific pathways, and focus on the potential of novel 'epidrugs' as future treatment modalities for MM.
Collapse
Affiliation(s)
- K Dimopoulos
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - P Gimsing
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - K Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
195
|
A genotoxic stress-responsive miRNA, miR-574-3p, delays cell growth by suppressing the enhancer of rudimentary homolog gene in vitro. Int J Mol Sci 2014; 15:2971-90. [PMID: 24566139 PMCID: PMC3958894 DOI: 10.3390/ijms15022971] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 02/13/2014] [Indexed: 12/17/2022] Open
Abstract
MicroRNA (miRNA) is a type of non-coding RNA that regulates the expression of its target genes by interacting with the complementary sequence of the target mRNA molecules. Recent evidence has shown that genotoxic stress induces miRNA expression, but the target genes involved and role in cellular responses remain unclear. We examined the role of miRNA in the cellular response to X-ray irradiation by studying the expression profiles of radio-responsive miRNAs and their target genes in cultured human cell lines. We found that expression of miR-574-3p was induced in the lung cancer cell line A549 by X-ray irradiation. Overexpression of miR-574-3p caused delayed growth in A549 cells. A predicted target site was detected in the 3′-untranslated region of the enhancer of the rudimentary homolog (ERH) gene, and transfected cells showed an interaction between the luciferase reporter containing the target sequences and miR-574-3p. Overexpression of miR-574-3p suppressed ERH protein production and delayed cell growth. This delay was confirmed by knockdown of ERH expression. Our study suggests that miR-574-3p may contribute to the regulation of the cell cycle in response to X-ray irradiation via suppression of ERH protein production.
Collapse
|
196
|
Bian Z, Yu Y, Yang T, Quan C, Sun W, Fu S. Effect of tumor suppressor gene cyclin-dependent kinase inhibitor 2A wild-type and A148T mutant on the cell cycle of human ovarian cancer cells. Oncol Lett 2014; 7:1229-1232. [PMID: 24944698 PMCID: PMC3961237 DOI: 10.3892/ol.2014.1867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 01/15/2014] [Indexed: 01/23/2023] Open
Abstract
Single-base substitution may affect the function of genes. This study identified a single-base substitution of G for A in codon 148 of cyclin-dependent kinase inhibitor 2A (CDKN2A/p16) by sequencing human ovarian cancer cell line UACC-1598. As a tumor suppressor gene, the expression of CDKN2A/p16 should be strictly controlled. In order to control CDKN2A/p16 gene expression, an inducible pTUNE vector system was selected. Using recombinant DNA technology, a CDKN2A/p16-A148T and CDKN2A/p16-wild-type gene expression system was successfully constructed to investigate whether this single-base substitution affects the function of CDKN2A/p16. For the wild-type and the mutant, expression of CDKN2A/p16-green fluorescent protein fusion protein increased markedly following isopropyl-β-D-thiogalactoside induction, and was accompanied by significant G1 arrest in the transfected human ovarian cancer SKOV3 cell line. The inducible vectors used in this study, CDKN2A/p16-wild-type and CDKN2A/p16-A148T open reading frame, may be useful for further investigation into whether this somatic mutation could alter the function of CDKN2A/p16 as a tumor suppressor gene. In summary, CDKN2A/p16-A148T was identified in ovarian cancer cells, and this single-base substitution did not affect the ability of CDKN2A/p16 to arrest the cell cycle.
Collapse
Affiliation(s)
- Zehua Bian
- Laboratory of Medical Genetics, Harbin Medical University, Heilongjiang Higher Education Institutions, Harbin, Heilongjiang 150081, P.R. China
| | - Yang Yu
- Laboratory of Medical Genetics, Harbin Medical University, Heilongjiang Higher Education Institutions, Harbin, Heilongjiang 150081, P.R. China
| | - Terigele Yang
- Laboratory of Medical Genetics, Harbin Medical University, Heilongjiang Higher Education Institutions, Harbin, Heilongjiang 150081, P.R. China
| | - Chao Quan
- Laboratory of Medical Genetics, Harbin Medical University, Heilongjiang Higher Education Institutions, Harbin, Heilongjiang 150081, P.R. China
| | - Wenjing Sun
- Laboratory of Medical Genetics, Harbin Medical University, Heilongjiang Higher Education Institutions, Harbin, Heilongjiang 150081, P.R. China
| | - Songbin Fu
- Laboratory of Medical Genetics, Harbin Medical University, Heilongjiang Higher Education Institutions, Harbin, Heilongjiang 150081, P.R. China ; Key Laboratory of Medical Genetics (Harbin Medical University), Heilongjiang Higher Education Institutions, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
197
|
von Knebel Doeberitz M, Reuschenbach M, Schmidt D, Bergeron C. Biomarkers for cervical cancer screening: the role of p16INK4ato highlight transforming HPV infections. Expert Rev Proteomics 2014; 9:149-63. [DOI: 10.1586/epr.12.13] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
198
|
Piaton E, Carré C, Advenier AS, Decaussin-Petrucci M, Mège-Lechevallier F, Lantier P, Granier G, Ruffion A. p16 INK4a overexpression and p16/Ki-67 dual labeling versus conventional urinary cytology in the evaluation of urothelial carcinoma. Cancer Cytopathol 2013; 122:211-20. [PMID: 24302621 DOI: 10.1002/cncy.21376] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 10/18/2013] [Accepted: 10/23/2013] [Indexed: 11/07/2022]
Abstract
BACKGROUND Taking into consideration the known overexpression of p16(INK4a) in histologically demonstrated high-grade urothelial malignancies, the objective of the current study was to examine the value of p16(INK4a) overexpression and of p16/Ki-67 dual labeling versus urinary cytology in the detection of urothelial lesions. METHODS Immunolabeling was performed on demounted and destained Papanicolaou slides after liquid-based ThinPrep processing. Actual diagnoses were ascertained by cystoscopy controls and histopathology. Negative cases, papillary urothelial neoplasia of low malignant potential/low-grade tumor, and high-grade lesions were considered separately. RESULTS A total of 216 urine samples were collected from new patients with symptoms who were referred for cystoscopy (92 cases) or patients who were being followed after conservative treatment for lesions involving the bladder (117 cases) or the upper urinary tract (7 cases). p16(INK4a) positivity was assessed in 171 of the 216 cases (79.2%) and in 93 of 99 high-grade cases with positive cytology (93.9%). Coexpression of p16/Ki-67 in the same cells was observed in 119 of 216 cases (55.1%) and was noted in 18 of 51 cases of negative or papillary urothelial neoplasia of low malignant potential/low-grade tumor (35.3%) and in 80 of 101 high-grade tumors (79.2%) (P < .0001). Thirteen of 14 high-grade intraurothelial lesions (92.8%) were dual labeled. When high-grade tumors, disease progression (increased grade, muscle infiltration, and extension into the upper urinary tract), and cancer-related death were grouped together as an endpoint, dual labeling demonstrated a sensitivity that was slightly higher than that of urinary cytology (82.5% vs 80.8%; P = .8), with 94.9% overall specificity. CONCLUSIONS When applied to the search for high-grade and aggressive disease, p16/Ki-67 dual labeling and urinary cytology appear to demonstrate comparable performance.
Collapse
Affiliation(s)
- Eric Piaton
- Hospices Civils de Lyon, Centre de Biologie et Pathologie Est, Hôpital Femme-Mère-Enfant, Lyon Bron, France; Université Claude Bernard Lyon 1, Lyon, France
| | | | | | | | | | | | | | | |
Collapse
|
199
|
Kawakami H, Okamoto I, Terao K, Sakai K, Suzuki M, Ueda S, Tanaka K, Kuwata K, Morita Y, Ono K, Nishio K, Nishimura Y, Doi K, Nakagawa K. Human papillomavirus DNA and p16 expression in Japanese patients with oropharyngeal squamous cell carcinoma. Cancer Med 2013; 2:933-41. [PMID: 24403267 PMCID: PMC3892398 DOI: 10.1002/cam4.151] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 09/18/2013] [Accepted: 09/18/2013] [Indexed: 12/31/2022] Open
Abstract
Human papillomavirus (HPV) is a major etiologic factor for oropharyngeal squamous cell carcinoma (OPSCC). However, little is known about HPV-related OPSCC in Japan. During the study, formalin-fixed, paraffin-embedded OPSCC specimens from Japanese patients were analyzed for HPV DNA by the polymerase chain reaction (PCR) and for the surrogate marker p16 by immuno-histochemistry. For HPV DNA-positive, p16-negative specimens, the methylation status of the p16 gene promoter was examined by methylation-specific PCR. Overall survival was calculated in relation to HPV DNA and p16 status and was subjected to multivariate analysis. OPSCC cell lines were examined for sensitivity to radiation or cisplatin in vitro. The study results showed that tumor specimens from 40 (38%) of the 104 study patients contained HPV DNA, with such positivity being associated with tumors of the tonsils, lymph node metastasis, and nonsmoking. Overall survival was better for OPSCC patients with HPV DNA than for those without it (hazard ratio, 0.214; 95% confidence interval, 0.074–0.614; P = 0.002). Multivariate analysis revealed HPV DNA to be an independent prognostic factor for overall survival (P = 0.015). Expression of p16 was associated with HPV DNA positivity. However, 20% of HPV DNA-positive tumors were negative for p16, with most of these tumors manifesting DNA methylation at the p16 gene promoter. Radiation or cisplatin sensitivity did not differ between OPSCC cell lines positive or negative for HPV DNA. Thus, positivity for HPV DNA identifies a distinct clinical subset of OPSCC with a more favorable outcome in Japanese.
Collapse
Affiliation(s)
- Hisato Kawakami
- Department of Medical Oncology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Al-Khalaf HH, Mohideen P, Nallar SC, Kalvakolanu DV, Aboussekhra A. The cyclin-dependent kinase inhibitor p16INK4a physically interacts with transcription factor Sp1 and cyclin-dependent kinase 4 to transactivate microRNA-141 and microRNA-146b-5p spontaneously and in response to ultraviolet light-induced DNA damage. J Biol Chem 2013; 288:35511-25. [PMID: 24163379 DOI: 10.1074/jbc.m113.512640] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
p16(INK4a) is a tumor suppressor protein involved in several stress-related cellular responses, including apoptosis. Recent lines of evidence indicate that p16(INK4a) is also a modulator of gene expression. However, the molecular mechanisms underlying this novel function are still obscure. Here, we present clear evidence that p16(INK4a) modulates the levels of various microRNAs, with marked positive effect on miR-141 and miR-146b-5p. This effect is mediated through the formation of the p16-CDK4-Sp1 heterocomplex, which binds to Sp1 consensus-binding motifs present in the promoters of miR-141 and miR-146b-5p, and it enables their transcription. In addition, we have shown that p16(INK4a) interacts with Sp1 through the fourth ankyrin repeat, which is crucial for Sp1 binding to the miR-141 and miR-146b-5p promoters and their transcriptional activation. The physiological importance of this association was revealed by the inability of cancer-related p16(INK4a) mutants to interact with Sp1. Moreover, we have shown p16-CDK4-Sp1-dependent up-regulation of miR-141 and miR-146b-5p following UV light-induced DNA damage and the role of these two microRNAs in mediating p16-related induction of apoptosis in response to this genotoxic stress. Together, these results indicate that p16(INK4a) associates with CDK4 not only to inhibit the cell cycle but also to enable the transcription of two important onco-microRNAs, which act as downstream effectors.
Collapse
Affiliation(s)
- Huda H Al-Khalaf
- From the Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, MBC 03, P. O. Box 3354, Riyadh 11211, Kingdom of Saudi Arabia
| | | | | | | | | |
Collapse
|