1
|
Genome-Wide DNA Methylation Profiling in Early Stage I Lung Adenocarcinoma Reveals Predictive Aberrant Methylation in the Promoter Region of the Long Noncoding RNA PLUT: An Exploratory Study. J Thorac Oncol 2020; 15:1338-1350. [PMID: 32272161 DOI: 10.1016/j.jtho.2020.03.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Surgical procedure is the treatment of choice in early stage I lung adenocarcinoma. However, a considerable number of patients experience recurrence within the first 2 years after complete resection. Suitable prognostic biomarkers that identify patients at high risk of recurrence (who may probably benefit from adjuvant treatment) are still not available. This study aimed at identifying methylation markers for early recurrence that may become important tools for the development of new treatment modalities. METHODS Genome-wide DNA methylation profiling was performed on 30 stage I lung adenocarcinomas, comparing 14 patients with early metastatic recurrence with 16 patients with a long-term relapse-free survival period using methylated-CpG-immunoprecipitation followed by high-throughput next-generation sequencing. The differentially methylated regions between the two subgroups were validated for their prognostic value in two independent cohorts using the MassCLEAVE assay, a high-resolution quantitative methylation analysis. RESULTS Unsupervised clustering of patients in the discovery cohort on the basis of differentially methylated regions identified patients with shorter relapse-free survival (hazard ratio: 2.23; 95% confidence interval: 0.66-7.53; p = 0.03). In two validation cohorts, promoter hypermethylation of the long noncoding RNA PLUT was significantly associated with shorter relapse-free survival (hazard ratio: 0.54; 95% confidence interval: 0.31-0.93; p < 0.026) and could be reported as an independent prognostic factor in the multivariate Cox regression analysis. CONCLUSIONS Promoter hypermethylation of the long noncoding RNA PLUT is predictive in patients with early stage I adenocarcinoma at high risk for early recurrence. Further studies are needed to validate its role in carcinogenesis and its use as a biomarker to facilitate patient selection and risk stratification.
Collapse
|
2
|
Chong Y, Thakur N, Paik KY, Lee EJ, Kang CS. Prognostic significance of stem cell/ epithelial-mesenchymal transition markers in periampullary/pancreatic cancers: FGFR1 is a promising prognostic marker. BMC Cancer 2020; 20:216. [PMID: 32171280 PMCID: PMC7071628 DOI: 10.1186/s12885-020-6673-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/24/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Periampullary cancers (PAC) including pancreatic, ampulla of Vater (AOV), and common bile duct (CBD) cancers are highly aggressive with a lack of useful prognostic markers beyond T stage. However, T staging can be biased due to the anatomic complexity of this region. Recently, several markers related to cancer stem cells and epithelial-mesenchymal transition (EMT) such as octamer transcription factor-4 (Oct4) and fibroblast growth factor receptor 1 (FGFR1) respectively, have been proposed as new promising markers in other solid cancers. The aim of this study was to assess the expression and prognostic significance of stem cell/EMT markers in PACs. METHODS Formalin-fixed, paraffin-embedded tissues of surgically excised PACs from the laboratory archives from 1998 to 2014 were evaluated by immunohistochemical staining for stem cell/EMT markers using tissue microarray. The clinicopathologic parameters were documented and statistically analyzed with the immunohistochemical findings. Survival and recurrence data were collected and analyzed. RESULTS A total of 126 PAC cases were evaluated. The average age was 63 years, with 76 male and 50 female patient samples. Age less than 74 years, AOV cancers, lower T & N stage, lower tumor size, no lymphatic, vascular, perineural invasion and histologic well differentiation, intestinal type, no fibrosis, severe inflammation were significantly associated with the better overall survival High expression levels of FGFR1 as well as CK20, CDX2, and VEGF were significantly related to better overall survival, while other stem cell markers were not related. Similar findings were observed for tumor recurrence using disease-free survival. CONCLUSIONS In addition to other clinicopathologic parameters, severe fibrosis was related to frequent tumor recurrence, and high FGFR1 expression was associated with better overall survival. Histologic changes such as extensive fibrosis need to be investigated further in relation to EMT of PACs.
Collapse
Affiliation(s)
- Yosep Chong
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul, 07345 Republic of Korea
| | - Nishant Thakur
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul, 07345 Republic of Korea
| | - Kwang Yeol Paik
- Department of Surgery, College of Medicine, The Catholic University of Korea, Seoul, 07345 Republic of Korea
| | - Eun Jung Lee
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul, 07345 Republic of Korea
- Department of Pathology, Shinwon Medical Foundation, Soha-ro 109 beon-gil, Gwanmyeong-si, 14316 Gyeonggi-do Republic of Korea
| | - Chang Suk Kang
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul, 07345 Republic of Korea
- Department of Pathology, Samkwang Medical Laboratories, 57, Baumoe-ro 41-gil, Seocho-gu, Seoul, 06742 Republic of Korea
| |
Collapse
|
3
|
RNA cargos in extracellular vesicles derived from blood serum in pancreas associated conditions. Sci Rep 2020; 10:2800. [PMID: 32071328 PMCID: PMC7028741 DOI: 10.1038/s41598-020-59523-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 01/30/2020] [Indexed: 12/21/2022] Open
Abstract
Exosomes are extracellular vesicles which are released from healthy and tumor cells into blood circulation. Unique biomolecular cargos such as RNA and protein are loaded in these vesicles. These molecules may have biological functions such as signaling, cell communications and have the potential to be analyzed as biomarkers. In this initial study, we describe the analysis of exosomes in the serum of healthy subjects, intraductal papillary mucosal neoplasms and pancreatic ductal adenocarcinoma including the characterization of their RNA cargos by next generation sequencing (EXO-NGS). Results indicate the presence of a wide variety of RNAs including mRNA, miRNA, lincRNA, tRNA and piRNA in these vesicles. Based on the differential mRNA expression observed upon EXO-NGS analysis, we independently evaluated two protein coding genes, matrix metalloproteinase-8 (MMP-8) and transcription factor T-Box 3 (TBX3) by qRT-PCR for selective expression in the serum samples. Results indicate a variable expression pattern of these genes across serum samples between different study groups. Further, qRT-PCR analysis with the same serum exosomes processed for EXO-NGS, we observed two long non-coding RNAs, malat-1 and CRNDE to be variably expressed. Overall, our observations emphasize the potential value of different exosome components in distinguishing between healthy, premalignant and malignant conditions related to the pancreas.
Collapse
|
4
|
Gurevich LE, Proshchina AE, Voronkova IA, Ashevskaya VE, Korosteleva PA, Dolzhansky OV. [Differential diagnostic value of the expression of the transcription factor PDX-1 in neuroendocrine and non-neuroendocrine tumors of the pancreas and other organs]. Arkh Patol 2019; 81:11-21. [PMID: 31626200 DOI: 10.17116/patol20198105111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
An important role in the differentiation of tissues in different organs is played by transforming factors (TFs); pancreatic and duodenal homebox 1 (PDX-1) is one of the earliest factors for pancreatic cells. Many malignant tumors, including neuroendocrine tumors (NETs), are similar in structure, and therefore the actual problem of oncomorphology is to search for narrow-specific markers and TFs. AIM to comparatively analyze and assess the value of the expression of the TF PDX-1 in NETs and non-NETs of different localization and histogenetic origin. MATERIAL AND METHODS Anti-PDX-1 antibodies were used to study 528 tumors divided into 3 groups: Group 1 included 394 NETs, among them there were those of the pancreas (n=173), stomach (n=46), bowel (n=65), lung (n=40), thymus (n=8), kidney (n=6), Merkel's cell carcinomas (n=14), NETs of the breast (n=3), larynx (n=2), trachea (n=2), bladder (n=1), and metastatic NETs (n=34) of unknown primary site; Group 2 consisted of 16 tumors, of them there were paragangliomas (n=6), medullary thyroid cancers (MTC) (n=6) and adrenal pheochromocytomas (APCC) (n=4); Group 3 comprised 118 non-NETs, among them there were tumors of the pancreas (n=54), stomach (n=26), bowel (n=17), lung (n=11), breast (n=3), kidney (n=4), adrenal glands (n=2), and bladder (n=1). RESULTS PDX-1 was positive in 75.1% (130/173) of pancreatic NETs, all insulinomas (50/50), gastrinomas (11/11), somatostatinomas (3/3), ACTH-producing tumors (2/2); PDX-1 was positive in the non-functioning pancreatic NETs, all PPomas (19/19), 76.1% (35/46) of NETs without the hormone detected, 50% (2/4) of calcitoninomas, and 21.1% (8/38) of silent glucagonomas. PDX-1 was positive in 32.4% (11/34) of carcinoids and 50% (6/12) of neuroendocrine carcinomas, all duodenal NETs (18/18), 90% (9/10) of rectal carcinoids and 30.8% (4/13) colonic carcinoids, 37.5% (3/8) of thymic/mediastinal carcinoids, 66.7% (4/6) of kidney carcinoids, and 37.5% (9/24) of metastatic NETs of unknown primary site. PDX-1 was negative in all carcinoids of the colon and sigmoid (0/5), ileum and jejunum (0/24), lung (0/40), trachea (0/2), larynx (0/2), Merkel's cell carcinoma (0/14), breast (0/3), bladder (0/1), as well as MTC (0/6), APCC (0/4), and paragangliomas (0/6). PDX-1-positive non-NETs included 81.8% (18/22) of adenocarcinomas (AC) and all serous cystic, mucinous cystic, intraductal and acinar cell tumors of the pancreas (4/4, 3/3, 2/2, and 3/3), 57.1% of AC (8/14) and 83.3% of signet ring cell carcinomas of the stomach (10/12), 56.2% AC of the bowel (9/17), bladder cancer (1/1). PDX-1 was negative in all anaplastic cancers (0/2) and solid pseudopapillary tumors of the pancreas (0/20), cancers of the lung (0/11), kidney (0/4), breast (0/3), and adrenal glands (0/2). CONCLUSION The expression of PDX-1 is very specific for most digestive tract NETs and non-NETs. Pancreatic ductal and acinar cell tumors and gastric signet ring cell carcinomas are most commonly PDX-1-positive. Most tumors that do not originate from the digestive tract have a PDX-1 negative immunophenotype.
Collapse
Affiliation(s)
- L E Gurevich
- M.F.Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | | | - I A Voronkova
- M.F.Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | - V E Ashevskaya
- M.F.Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | - P A Korosteleva
- M.F.Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | - O V Dolzhansky
- M.F.Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| |
Collapse
|
5
|
Qian W, Li J, Chen K, Jiang Z, Cheng L, Zhou C, Yan B, Cao J, Ma Q, Duan W. Metformin suppresses tumor angiogenesis and enhances the chemosensitivity of gemcitabine in a genetically engineered mouse model of pancreatic cancer. Life Sci 2018; 208:253-261. [PMID: 30053447 DOI: 10.1016/j.lfs.2018.07.046] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/21/2018] [Accepted: 07/24/2018] [Indexed: 02/08/2023]
Abstract
AIMS Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant diseases and has few effective and reliable therapeutic strategies. The anti-tumor effect of metformin is widely known, however, there is only limited evidence regarding the anti-angiogenesis effect and chemosensitization of metformin and its underlying mechanisms in PDAC. MAIN METHODS In the present study, we adopted a spontaneous PDAC mouse model named LSL‑KrasG12D/+; Trp53fl/+; Pdx1‑Cre (KPC) mice to explore the mechanism of the modulation of tumor angiogenesis and chemosensitization of metformin by treating KPC mice with metformin, gemcitabine or a combination of the two. H&E staining, Masson staining and immunohistochemical staining were adopted to describe the histopathology and biomarkers of the KPC in different groups. KEY FINDINGS Metformin plus gemcitabine reduced tumorigenic potential of PDAC. Specifically, metformin showed an anti-pancreatic stellate cells (PSCs) effect via decreasing the expression of sonic hedgehog (SHH) and then sparked some downstream effects, for example, inhibiting the production of vascular endothelial growth factor (VEGF) in the tumor microenvironment, reducing the formation of tumor neovascularization, attenuating the desmoplastic reaction and enhancing the antitumor effect of gemcitabine. SIGNIFICANCE We concluded that metformin suppressed tumor angiogenesis and enhanced the chemosensitivity of gemcitabine via inactivating PSCs in PDAC of KPC mice.
Collapse
Affiliation(s)
- Weikun Qian
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Jie Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Ke Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Zhengdong Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Liang Cheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Bin Yan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Junyu Cao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
6
|
Verma RK, Yu W, Shrivastava A, Shankar S, Srivastava RK. α-Mangostin-encapsulated PLGA nanoparticles inhibit pancreatic carcinogenesis by targeting cancer stem cells in human, and transgenic (Kras(G12D), and Kras(G12D)/tp53R270H) mice. Sci Rep 2016; 6:32743. [PMID: 27624879 PMCID: PMC5021984 DOI: 10.1038/srep32743] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/02/2016] [Indexed: 12/25/2022] Open
Abstract
Activation of sonic hedgehog (Shh) in cancer stem cell (CSC) has been demonstrated with aggressiveness of pancreatic cancer. In order to enhance the biological activity of α-mangostin, we formulated mangostin-encapsulated PLGA nanoparticles (Mang-NPs) and examined the molecular mechanisms by which they inhibit human and KC mice (PdxCre;LSL-KrasG12D) pancreatic CSC characteristics in vitro, and pancreatic carcinogenesis in KPC (PdxCre;LSLKrasG12D;LSL-Trp53R172H) mice. Mang-NPs inhibited human and KrasG12D mice pancreatic CSC characteristics in vitro. Mang-NPs also inhibited EMT by up-regulating E-cadherin and inhibiting N-cadherin and transcription factors Slug, and pluripotency maintaining factors Nanog, c-Myc, and Oct4. Furthermore, Mang-NPs inhibited the components of Shh pathway and Gli targets. In vivo, Mang-NPs inhibited the progression of pancreatic intraneoplasia to pancreatic ductal adenocarcinoma and liver metastasis in KPC mice. The inhibitory effects of Mang-NPs on carcinogenesis in KPC mice were associated with downregulation of pluripotency maintaining factors (c-Myc, Nanog and Oct4), stem cell markers (CD24 and CD133), components of Shh pathway (Gli1, Gli2, Patched1/2, and Smoothened), Gli targets (Bcl-2, XIAP and Cyclin D1), and EMT markers and transcription factors (N-cadherin, Slug, Snail and Zeb1), and upregulation of E-cadherin. Overall, our data suggest that Mang-NPs can inhibit pancreatic cancer growth, development and metastasis by targeting Shh pathway.
Collapse
Affiliation(s)
- Raj Kumar Verma
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA
| | - Wei Yu
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA
| | - Anju Shrivastava
- St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Sharmila Shankar
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA.,Department of Pathology and Laboratory Medicine, University of Missouri Kansas City, MO, USA
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA.,Department of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| |
Collapse
|
7
|
Yang XJ. Liver cancer stem cells and new strategies for targeted therapy of hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2016; 24:3337-3346. [DOI: 10.11569/wcjd.v24.i22.3337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the past ten years, increasing studies show that liver cancer stem cells (LCSCs) are responsible not only for hepatocellular carcinoma (HCC) initiation and development but also for the generation of distant metastasis and relapse after therapy. Therefore, further research for LCSCs is considered a new avenue to explore the cause of HCC invasion and metastasis in order to formulate prevention and control strategies. Current traditional cancer therapies including chemotherapy and radiotherapy which primarily target rapidly dividing and most likely well differentiated tumor cells, would fail to eliminate LCSCs. After surgical removal of HCC mass, the remaining LCSCs still have the ability to differentiate, proliferate and even migrate to other places to form metastatic tumors. Therefore, to explore various kinds of targeted therapies for LCSCs is the only way to break through the "bottleneck" of HCC treatment. Strategies for targeted therapy of HCC include inhibiting LCSCs proliferation, inducing apoptosis and differentiation, increasing chemotherapy sensitivity and disrupting the tumor niche essential for CSC homeostasis.
Collapse
|
8
|
Klieser E, Swierczynski S, Mayr C, Jäger T, Schmidt J, Neureiter D, Kiesslich T, Illig R. Differential role of Hedgehog signaling in human pancreatic (patho-) physiology: An up to date review. World J Gastrointest Pathophysiol 2016; 7:199-210. [PMID: 27190692 PMCID: PMC4867399 DOI: 10.4291/wjgp.v7.i2.199] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/21/2015] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Since the discovery of the Hedgehog (Hh) pathway in drosophila melanogaster, our knowledge of the role of Hh in embryonic development, inflammation, and cancerogenesis in humans has dramatically increased over the last decades. This is the case especially concerning the pancreas, however, real therapeutic breakthroughs are missing until now. In general, Hh signaling is essential for pancreatic organogenesis, development, and tissue maturation. In the case of acute pancreatitis, Hh has a protective role, whereas in chronic pancreatitis, Hh interacts with pancreatic stellate cells, leading to destructive parenchym fibrosis and atrophy, as well as to irregular tissue remodeling with potency of initiating cancerogenesis. In vitro and in situ analysis of Hh in pancreatic cancer revealed that the Hh pathway participates in the development of pancreatic precursor lesions and ductal adenocarcinoma including critical interactions with the tumor microenvironment. The application of specific inhibitors of components of the Hh pathway is currently subject of ongoing clinical trials (phases 1 and 2). Furthermore, a combination of Hh pathway inhibitors and established chemotherapeutic drugs could also represent a promising therapeutic approach. In this review, we give a structured survey of the role of the Hh pathway in pancreatic development, pancreatitis, pancreatic carcinogenesis and pancreatic cancer as well as an overview of current clinical trials concerning Hh pathway inhibitors and pancreas cancer.
Collapse
|
9
|
Ko AH, LoConte N, Tempero MA, Walker EJ, Kelley RK, Lewis S, Chang WC, Kantoff E, Vannier MW, Catenacci DV, Venook AP, Kindler HL. A Phase I Study of FOLFIRINOX Plus IPI-926, a Hedgehog Pathway Inhibitor, for Advanced Pancreatic Adenocarcinoma. Pancreas 2016; 45:370-5. [PMID: 26390428 PMCID: PMC5908466 DOI: 10.1097/mpa.0000000000000458] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES In mouse models of pancreatic cancer, IPI-926, an oral Hedgehog inhibitor, increases chemotherapy delivery by depleting tumor-associated stroma. This multicenter phase Ib study evaluated IPI-926 in combination with FOLFIRINOX (5-fluorouracil, leucovorin, irinotecan, oxaliplatin) in patients with advanced pancreatic cancer. METHODS Patients were treated with once-daily IPI-926 plus FOLFIRINOX. A 3 + 3 dose escalation design was used, with cohort expansion at the maximum tolerated dose. A subset of patients underwent perfusion computed tomography to assess changes in tumor perfusion. RESULTS The maximum tolerated dose was identified 1 dose level below standard FOLFIRINOX. Common treatment-related adverse events included liver function test abnormalities, neuropathy, nausea/vomiting, and diarrhea. Objective response rate was high (67%), and patients receiving IPI-926 maintenance showed further declines in CA19-9 levels even after FOLFIRINOX discontinuation. Treatment did not result in consistent increases in tumor perfusion. The study closed early when a separate phase II trial of IPI-926 plus gemcitabine indicated detrimental effects of this combination. CONCLUSIONS This is the first study to demonstrate the feasibility of using FOLFIRINOX as the chemotherapeutic backbone in a clinical trial design. Although robust antitumor activity and acceptable safety were observed with the addition of IPI-926 to this regimen, future development of Hedgehog inhibitors in pancreatic cancer seems unlikely.
Collapse
Affiliation(s)
- Andrew H. Ko
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - Noelle LoConte
- Division of Hematology/Oncology, University of Wisconsin, Madison, WI
| | - Margaret A. Tempero
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - Evan J. Walker
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - R. Kate Kelley
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - Stephanie Lewis
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - Wei-Chou Chang
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Emily Kantoff
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | | | | | - Alan P. Venook
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - Hedy L. Kindler
- Division of Hematology/Oncology, University of Chicago, Chicago, IL
| |
Collapse
|
10
|
Zhou G, Yu J, Wang A, Liu SH, Sinnett-Smith J, Wu J, Sanchez R, Nemunaitis J, Ricordi C, Rozengurt E, Brunicardi FC. Metformin Restrains Pancreatic Duodenal Homeobox-1 (PDX-1) Function by Inhibiting ERK Signaling in Pancreatic Ductal Adenocarcinoma. Curr Mol Med 2016; 16:83-90. [PMID: 26695692 PMCID: PMC4994969 DOI: 10.2174/1566524016666151222145551] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 11/20/2015] [Accepted: 12/15/2015] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most potent and perilous diseases known, with a median survival rate of 3-5 months due to the combination of only advanced stage diagnosis and ineffective therapeutic options. Metformin (1,1-Dimethylbiguanide hydrochloride), the leading drug used for type 2 diabetes mellitus, emerges as a potential therapy for PDAC and other human cancers. Metformin exerts its anticancer action via a variety of adenosine monophosphate (AMP)-activated protein kinase (AMPK)- dependent and/or AMPK-independent mechanisms. We present data here showing that metformin downregulated pancreatic transcription factor pancreatic duodenal homeobox-1 (PDX-1), suggesting a potential novel mechanism by which metformin exerts its anticancer action. Metformin inhibited PDX-1 expression at both protein and mRNA levels and PDX-1 transactivity as well in PDAC cells. Extracellular signal-regulated kinase (ERK) was identified as a PDX-1-interacting protein by antibody array screening in GFP-PDX-1 stable HEK293 cells. Co-transfection of ERK1 with PDX-1 resulted in an enhanced PDX-1 expression in HEK293 cells in a dose-dependent manner. Immunoprecipitation/Western blotting analysis confirmed the ERK-PDX-1 interaction in PANC-1 cells stimulated by epidermal growth factor (EGF). EGF induced an enhanced PDX-1 expression in PANC-1 cells and this stimulation was inhibited by MEK inhibitor PD0325901. Metformin inhibited EGF-stimulated PDX-1 expression with an accompanied inhibition of ERK kinase activation in PANC- 1 cells. Taken together, our studies show that PDX-1 is a potential novel target for metformin in PDAC cells and that metformin may exert its anticancer action in PDAC by down-regulating PDX-1 via a mechanism involving inhibition of ERK signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - F C Brunicardi
- Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Zhou ZG, Zhang CY, Fei HX, Zhong LL, Bai Y. Phenolic alkaloids from Menispermum dauricum inhibits BxPC-3 pancreatic cancer cells by blocking of Hedgehog signaling pathway. Pharmacogn Mag 2015; 11:690-7. [PMID: 26600712 PMCID: PMC4621636 DOI: 10.4103/0973-1296.165548] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: The Hedgehog (Hh) signaling pathway plays an important role in pancreatic cancer (PC) cells. Phenolic alkaloids from Menispermum dauricum (PAMD), a traditional Chinese medicine used for the treatment of immune disorders, have been reported to have antitumor activity recently. Objective: To investigate the efficacy and mechanism of PAMD against PC cell BxPC-3. Materials and Methods: F assay was used to assess cell proliferation inhibition of PAMD; the apoptotic induction and cell cycle arrest was detected by flow cytometry; the BxPC-3 xenograft was established to evaluate the tumor growth inhibition of PAMD; hematoxylin-eosin staining was applied to analyze the pathological morphology of tumor tissues; immunohistochemistry (IHC) and Western blot was adopted to detect the protein levels; quantitative real-time polymerase chain reaction was used to determine the mRNA expressions. Results: PAMD shows time-and dose-dependent proliferation inhibition on the BxPC-3 cell, induced G0/G1 phase arrest and cell apoptosis in vitro. PAMD also showed better inhibition of tumor growth and a preferable safety profile compared with chemotherapeutic regimen 5-fluoro-2, 4 (1 H, 3 H) pyrimidinedione in BxPC-3 xenograft in vivo. Furthermore, PAMD directly decreases the protein and mRNA levels of Sonic Hedgehog (Shh) and its downstream transcription factor Gli-1 in the BxPC-3 tumor tissues. Conclusion: The treatment of PAMD displayed Hh signaling pathway blockade through decreasing the protein and mRNA levels of Shh and its downstream transcription factor Gli-1, suggesting a promising strategy in treating human PC.
Collapse
Affiliation(s)
- Zhong-Guang Zhou
- Research Institute of Traditional Chinese Medicine, Heilongjiang, China
| | - Chao-Ying Zhang
- The Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Hong-Xin Fei
- Research Institute of Traditional Chinese Medicine, Heilongjiang, China ; Department of Qiqihar Medical University, Basic Medicine, Heilongjiang, China
| | - Li-Li Zhong
- Department of Pathology, The First Affiliated Hospital, Heilongjiang, China
| | - Yun Bai
- Basic Medical College, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| |
Collapse
|
12
|
Xu X, Liu H, Zhang H, Dai W, Guo C, Xie C, Wei S, He S, Xu X. Sonic Hedgehog-GLI Family Zinc Finger 1 Signaling Pathway Promotes the Growth and Migration of Pancreatic Cancer Cells by Regulating the Transcription of Eukaryotic Translation Initiation Factor 5A2. Pancreas 2015; 44:1252-1258. [PMID: 26465952 DOI: 10.1097/mpa.0000000000000532] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The Hh (hedgehog) signaling pathway is still waiting for further studies because its downstream molecular mechanism remains elusive. Because EIF5A2 (eukaryotic translation initiation factor 5A2) gene was up-regulated upon Gli1 (GLI family zinc finger 1) in pancreatic cancer (PC) cells, we speculated that this pathway might promote tumor progression through regulating EIF5A2. METHODS We investigated regulation effect of Hh signaling pathway to EIF5A2 gene transcription by Gli1 knockdown or overexpression in PC cell lines first. Then, the regulation mechanism of Gli1 to EIF5A2 gene was studied at transcription level. Finally, we studied cancer-promoting effects of Gli1-dependent EIF5A2 in PC cells. RESULTS The data showed that Gli1 up-regulated expression of EIF5A2 by promoting transcription via cis-acting elements in PC cells. Moreover, vimentin gene was up-regulated significantly by sonic hedgehog (SHh)/Gli1 expression increasing, and E-cadherin was significantly reduced. The EIF5A2 knockdown partially reversed cell proliferation and migration induced by artificial SHh overexpression and inhibited epithelial mesenchymal transition process in PC cells with SHh overexpression (P < 0.05). CONCLUSIONS Our data establish a novel transcription mechanism of Gli1 to EIF5A2 gene in cis-regulatory manner in PC cells. Thus, EIF5A2 oncogene effect could be incorporated into cancer-promoting molecular network upon Hh signaling pathway.
Collapse
Affiliation(s)
- Xuanfu Xu
- From the *Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai; Departments of †Pathology and ‡Gastroenterology, The Second Hospital of Zhejiang University, Hangzhou, Zhejiang Province; and §Department of Hepato-Biliary and Pancreatic Oncology, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Bakry OA, Samaka RM, Shoeib MAM, Megahed DM. Immunolocalization of glioma-associated oncogene homolog 1 in non melanoma skin cancer. Ultrastruct Pathol 2014; 39:135-46. [PMID: 25350271 DOI: 10.3109/01913123.2014.970723] [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: 11/13/2022]
Abstract
Glioma-associated oncogene homolog (GLI)1 is involved in controlling cell proliferation and angiogenesis. The aim of this work was to explore its possible role in non-melanoma skin cancer pathogenesis through its immunohistochemical (IHC) expression in skin biopsies of these diseases and correlating this expression with the clinico-pathological parameters of the studied cases. Seventy-six cutaneous specimens were studied; 30 cases with basal cell carcinoma (BCC), 30 cases with squamous cell carcinoma (SCC) and 16 normal skin samples, from age- and gender-matched subjects, as a control group. GLI1 was expressed in all BCC cases and in 60% of SCC cases. All SCC cases showed cytoplasmic, while 70% of BCC cases showed nucleocytoplasmic immunoreactivity. It was over expressed in BCC and SCC compared to normal skin (p = 0.01 and 0.0006, respectively). Higher Histo (H) score in BCC cases was significantly associated with female gender (p = 0.04), multiple lesions, desmoplastic stromal reaction and stromal angiogenesis (p < 0.001 for all). Higher H score in SCC cases was significantly associated with scalp location, nodular type, recurrent lesions, high tumor grade, lymphovascular invasion (p = 0.004 for all), inflammatory stromal reaction (p = 0.01), lymph node involvement and absence of calcification (p = 0.001 for both). In conclusion, GLI1 may play a role in BCC pathogenesis through its role in cell proliferation, migration, and angiogenesis. Its upregulation and cytoplasmic localization in SCC may suggest that its role in tumor pathogenesis is through mechanisms other than Hedgehog pathway activation. Further studies are needed to clarify the exact molecular basis of its oncogenic action.
Collapse
Affiliation(s)
- Ola Ahmed Bakry
- Department of Dermatology, Andrology and STDs, Faculty of Medicine, Menoufiya University , Shibeen El Kom , Egypt and
| | | | | | | |
Collapse
|
14
|
Xu X, Su B, Xie C, Wei S, Zhou Y, Liu H, Dai W, Cheng P, Wang F, Xu X, Guo C. Sonic hedgehog-Gli1 signaling pathway regulates the epithelial mesenchymal transition (EMT) by mediating a new target gene, S100A4, in pancreatic cancer cells. PLoS One 2014; 9:e96441. [PMID: 25072505 PMCID: PMC4114558 DOI: 10.1371/journal.pone.0096441] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 04/08/2014] [Indexed: 01/05/2023] Open
Abstract
AIMS The hedgehog signaling pathway plays an important role in EMT of pancreatic cancer cells, but the precise mechanisms remain elusive. Because S100A4 as a key EMT moleculer marker was found to be upregulated upon Gli1 in pancreatic cancer cells, we focused on the relationship between Shh-Gli1 signals and S100 genes family. METHODS On the base of cDNA microarray data, we investigated regulating mechanism of Gli1 to some members of S100A genes family in pancreatic cancer cell lines firstly. Then, the regulation of Gli1 to S100A4 gene was studied by molecular biology assays and the pro-metastasis effection of Gli1-dependent S100A4 was investigated in vitro. Finally, the expressions of Shh, Gli1, S100A4 and E-cadherin in pancreatic cancer tissues were studied by using immunohistochemistry assays. RESULTS Five members of the S100 genes family, S100A2, S100A4, S100A6, S100A11, and S100A14 were found to be downregulated significantly upon Gli1 knockdown. Gli1 enhancer prediction combining with in vitro data demonstrated that Gli1 primarily regulates S100A family members via cis-acting elements. Indeed, the data indicate S100A4 and vimentin genes were upregulated significantly by Shh/Gli1-expression increasing and E-cadherin was significantly reduced at the same time. Migration of PC cells was increased significantly in a dose-dependent manner of Gli1 expression (P<0.05) and siS100A4 significantly reversed the response of PC cells induced by L-Shh transduction (P<0.01). CONCLUSION Our data establish a novel connection between Shh-Gli1 signaling and S100A4 regulation, which imply that S100A4 might be one of the key factors in EMT mediated by Shh-Gli1 signaling in pancreatic cancer.
Collapse
Affiliation(s)
- Xuanfu Xu
- Department of Gastroenterology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Bin Su
- Department of Endocrinology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Chuangao Xie
- Department of Gastroenterology, the Second Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Shumei Wei
- Department of Gastroenterology, the Second Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yingqun Zhou
- Department of Gastroenterology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Hua Liu
- Department of Gastroenterology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Weiqi Dai
- Department of Gastroenterology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Ping Cheng
- Department of Gastroenterology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Fan Wang
- Department of Gastroenterology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Xiaorong Xu
- Department of Gastroenterology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Chuanyong Guo
- Department of Gastroenterology, the Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| |
Collapse
|
15
|
Neureiter D, Jäger T, Ocker M, Kiesslich T. Epigenetics and pancreatic cancer: pathophysiology and novel treatment aspects. World J Gastroenterol 2014; 20:7830-7848. [PMID: 24976721 PMCID: PMC4069312 DOI: 10.3748/wjg.v20.i24.7830] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/07/2014] [Accepted: 03/12/2014] [Indexed: 02/06/2023] Open
Abstract
An improvement in pancreatic cancer treatment represents an urgent medical goal. Late diagnosis and high intrinsic resistance to conventional chemotherapy has led to a dismal overall prognosis that has remained unchanged during the past decades. Increasing knowledge about the molecular pathogenesis of the disease has shown that genetic alterations, such as mutations of K-ras, and especially epigenetic dysregulation of tumor-associated genes, such as silencing of the tumor suppressor p16(ink4a), are hallmarks of pancreatic cancer. Here, we describe genes that are commonly affected by epigenetic dysregulation in pancreatic cancer via DNA methylation, histone acetylation or miRNA (microRNA) expression, and review the implications on pancreatic cancer biology such as epithelial-mesenchymal transition, morphological pattern formation, or cancer stem cell regulation during carcinogenesis from PanIN (pancreatic intraepithelial lesions) to invasive cancer and resistance development. Epigenetic drugs, such as DNA methyltransferases or histone deactylase inhibitors, have shown promising preclinical results in pancreatic cancer and are currently in early phases of clinical development. Combinations of epigenetic drugs with established cytotoxic drugs or targeted therapies are promising approaches to improve the poor response and survival rate of pancreatic cancer patients.
Collapse
|
16
|
Li X, Wang Z, Ma Q, Xu Q, Liu H, Duan W, Lei J, Ma J, Wang X, Lv S, Han L, Li W, Guo J, Guo K, Zhang D, Wu E, Xie K. Sonic hedgehog paracrine signaling activates stromal cells to promote perineural invasion in pancreatic cancer. Clin Cancer Res 2014; 20:4326-38. [PMID: 24947933 DOI: 10.1158/1078-0432.ccr-13-3426] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE Pancreatic cancer is characterized by stromal desmoplasia and perineural invasion (PNI). We sought to explore the contribution of pancreatic stellate cells (PSC) activated by paracrine Sonic Hedgehog (SHH) in pancreatic cancer PNI and progression. EXPERIMENTAL DESIGN In this study, the expression dynamics of SHH were examined via immunohistochemistry, real-time PCR, and Western blot analysis in a cohort of carcinomatous and nonneoplastic pancreatic tissues and cells. A series of in vivo and in vitro assays was performed to elucidate the contribution of PSCs activated by paracrine SHH signaling in pancreatic cancer PNI and progression. RESULTS We show that SHH overexpression in tumor cells is involved in PNI in pancreatic cancer and is an important marker of biologic activity of pancreatic cancer. Moreover, the overexpression of SHH in tumor cells activates the hedgehog pathway in PSCs in the stroma instead of activating tumor cells. These activated PSCs are essential for the promotion of pancreatic cancer cell migration along nerve axons and nerve outgrowth to pancreatic cancer cell colonies in an in vitro three-dimensional model of nerve invasion in cancer. Furthermore, the coimplantation of PSCs activated by paracrine SHH induced tumor cell invasion of the trunk and nerve dysfunction along sciatic nerves and also promoted orthotropic xenograft tumor growth, metastasis, and PNI in in vivo models. CONCLUSIONS These results establish that stromal PSCs activated by SHH paracrine signaling in pancreatic cancer cells secrete high levels of PNI-associated molecules to promote PNI in pancreatic cancer.
Collapse
Affiliation(s)
- Xuqi Li
- Departments of Hepatobiliary Surgery, General Surgery, and
| | | | | | | | - Han Liu
- Departments of Hepatobiliary Surgery
| | | | | | - Jiguang Ma
- Oncology, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiu Wang
- Departments of Hepatobiliary Surgery
| | | | - Liang Han
- Departments of Hepatobiliary Surgery
| | - Wei Li
- Departments of Hepatobiliary Surgery
| | - Jian Guo
- Departments of Hepatobiliary Surgery
| | - Kun Guo
- Departments of Hepatobiliary Surgery
| | | | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota; and
| | - Keping Xie
- Department of Gastroenterology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
17
|
Wu J, Liu S, Yu J, Zhou G, Rao D, Jay CM, Kumar P, Sanchez R, Templeton N, Senzer N, Maples P, Nemunaitis J, Brunicardi FC. Vertically integrated translational studies of PDX1 as a therapeutic target for pancreatic cancer via a novel bifunctional RNAi platform. Cancer Gene Ther 2014; 21:48-53. [PMID: 24457987 DOI: 10.1038/cgt.2013.84] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 12/09/2013] [Indexed: 11/09/2022]
Abstract
RNA interference (RNAi) represents a powerful, new tool for scientific investigation as well as a promising new form of targeted gene therapy, with applications currently in clinical trials. Bifunctional short hairpin RNA (shRNA) are synthetic RNAi molecules, engineered to utilize multiple endogenous RNAi pathways to specifically silence target genes. Pancreatic and duodenal homeobox 1 (PDX1) is a key regulator of pancreatic development, β-cell differentiation, normal β-cell function and pancreatic cancer. Our aim is to review the process of identifying PDX1 as a specific, potential RNAi target in pancreatic cancer, as well as the underlying mechanisms and various forms of RNAi, with subsequent testing and development of PDX1-targeted bifunctional shRNA therapy.
Collapse
Affiliation(s)
- J Wu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - S Liu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - J Yu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - G Zhou
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - D Rao
- Gradalis, Carrollton, TX, USA
| | - C M Jay
- Gradalis, Carrollton, TX, USA
| | - P Kumar
- Gradalis, Carrollton, TX, USA
| | - R Sanchez
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - N Senzer
- 1] Gradalis, Carrollton, TX, USA [2] Mary Crowley Cancer Research Center, Dallas, TX, USA
| | | | - J Nemunaitis
- 1] Gradalis, Carrollton, TX, USA [2] Mary Crowley Cancer Research Center, Dallas, TX, USA
| | - F C Brunicardi
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
18
|
Quint K, Tonigold M, Di Fazio P, Montalbano R, Lingelbach S, Rückert F, Alinger B, Ocker M, Neureiter D. Pancreatic cancer cells surviving gemcitabine treatment express markers of stem cell differentiation and epithelial-mesenchymal transition. Int J Oncol 2012; 41:2093-2102. [PMID: 23026911 DOI: 10.3892/ijo.2012.1648] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/03/2012] [Indexed: 02/07/2023] Open
Abstract
Objective response rates to standard chemotherapeutic regimens remain low in pancreatic cancer. Subpopulations of cells have been identified in various solid tumors which express stem cell-associated markers and are associated with increased resistance against radiochemotherapy. We investigated the expression of stem cell genes and markers of epithelial-mesenchymal transition in pancreatic cancer cells that survived high concentrations of gemcitabine treatment. Capan-1 and Panc-1 cells were continuously incubated with 1 and 10 µM gemcitabine. Surviving cells were collected after 1, 3 and 6 days. Expression of PDX-1, SHH, CD24, CD44, CD133, EpCAM, CBX7, OCT4, SNAIL, SLUG, TWIST, Ki-67, E-cadherin, β-catenin and vimentin were quantified by qPCR or immunocytochemistry. Migration was assessed by wound‑healing assay. SHH was knocked down using RNA interference. Five primary pancreatic cancer cell lines were used to validate the qPCR results. All investigated genes were upregulated after 6 days of gemcitabine incubation. Highest relative expression levels were observed for OCT4 (13.4-fold), CD24 (47.3-fold) and EpCAM (15.9-fold) in Capan-1 and PDX-1 (13.3‑fold), SHH (24.1-fold), CD44 (17.4-fold), CD133 (20.2-fold) and SLUG (15.2-fold) in Panc-1 cells. Distinct upregulation patterns were observed in the primary cells. Migration was increased in Panc-1 cells and changes in the expression of E-cadherin and β-catenin were typical of epithelial-mesenchymal transition in both cell lines. SHH knockdown reduced IC(50) from 30.1 to 27.6 nM in Capan-1 while it strongly inhibited proli-feration in Panc-1 cells. Cells surviving high-dose gemcitabine treatment express increased levels of stem cell genes, show characteristics associated with epithelial-mesenchymal transition and retain their proliferative capacity.
Collapse
Affiliation(s)
- Karl Quint
- Institute for Surgical Research, Philipps University of Marburg, D-35043 Marburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ma J, Li J, Li H, Xiao X, Shen L, Fang L. Downregulation of pancreatic-duodenal homeobox 1 expression in breast cancer patients: a mechanism of proliferation and apoptosis in cancer. Mol Med Rep 2012; 6:983-8. [PMID: 22961564 DOI: 10.3892/mmr.2012.1067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 08/07/2012] [Indexed: 11/06/2022] Open
Abstract
Pancreatic-duodenal homeobox 1 (PDX-1) is a transcription factor that regulates embryological pancreas development and insulin expression in adult islets. The current study investigated the expression profile and potential role of PDX-1 in breast cancer. Immunohistochemistry was performed to determine the expression pattern of PDX-1 in breast cancer and adjacent benign breast tissues. In addition, cell proliferation and the cell cycle were evaluated following the transient inhibition of PDX-1 with antisense oligonucleotides in MCF-7 human breast cancer cells. Real-time PCR and western blotting were conducted to investigate the correlation between PDX-1, P53, Ki-67, Caspase 3 and Caspase 8. These experiments demonstrated that PDX-1 was downregulated in human breast cancer tissue compared with adjacent normal breast tissue. Knockdown of PDX-1 expression in vitro in MCF-7 breast cancer cells promoted cell proliferation and disrupted the cell cycle, as demonstrated by the overexpression of P53 and Ki-67 at the mRNA and protein levels. In conclusion, the current study shows that PDX-1 regulates cell proliferation and the cell cycle in human breast cancer cells by altering the expression of the cell cycle-related genes, P53 and Ki-67. These data suggest that PDX-1 is a putative tumor suppressor in breast cancer.
Collapse
Affiliation(s)
- Jie Ma
- Department of Breast and Thyroid Surgery, Tenth People's Hospital of Tongji University, Zhabei, Shanghai 200072, PR China
| | | | | | | | | | | |
Collapse
|
20
|
Neureiter D. New in Hedgehog signaling: a possible role in aging, and chronic degenerative and inflammatory diseases? (Comment on DOI 10.1002/bies.201200049). Bioessays 2012; 34:828-9. [PMID: 22903575 DOI: 10.1002/bies.201200107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University (PMU), Salzburger Landeskliniken (SALK), Salzburg, Austria.
| |
Collapse
|
21
|
Kemmerling R, Alinger B, Dietze O, Bösmüller HC, Ocker M, Wolkersdörfer GW, Berr F, Neureiter D, Kiesslich T. Association of stem cell marker expression pattern and survival in human biliary tract cancer. Int J Oncol 2012; 41:511-522. [PMID: 22614781 DOI: 10.3892/ijo.2012.1477] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/13/2012] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to investigate the molecular and protein expression pattern of markers of stemness phenotype and its clinicopathological significance in human biliary tract cancer (BTC). Human BTC cell lines (CCLP-1, Egi-1, MzChA-1, MzChA-2, SkChA-1, TFK-1 and GBC) were analyzed in vitro and in xenotransplanted animals for expression of markers of stemness and compared to tissue microarrays (TMA) of 34 cases of human BTC with complete pathomorphological and clinical data (survival). Molecular analyses on the mRNA and protein level included makers of stemness and progenitor (Bmi-1, Sox-2, Nestin, CD133, CD44 and Nanog), proliferation and differentiation (cell cycle proteins, intermediate filaments). The investigated BTC samples showed a low to moderate and partially significantly different expression pattern of the stem cell markers in vitro, in vivo and in TMA. Hierarchical cluster analysis identified subgroups with homogenous expression of stem cell markers significantly differing with respect to cytokeratin expression in xenografts and Ki67 proliferation marker in human TMA, respectively - thus indicating possible heterogeneous carcinogenesis pathways in BTC. Additionally, these stem cell markers could be linked to morphology and molecular markers of proliferation and differentiation on the mRNA and protein level. Finally, survival analysis identified the combination of CD133 and CD44 as an independent prognostic factor yet their value as prognostic factors need testing in prospective study design.
Collapse
Affiliation(s)
- Ralf Kemmerling
- Institute of Pathology, Paracelsus Medical University, Salzburger Landeskliniken, Salzburg, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Li Y, Maitah MY, Ahmad A, Kong D, Bao B, Sarkar FH. Targeting the Hedgehog signaling pathway for cancer therapy. Expert Opin Ther Targets 2012; 16:49-66. [PMID: 22243133 DOI: 10.1517/14728222.2011.617367] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Hedgehog (Hh) signaling pathway plays key roles in embryonic development, formation and maintenance of cancer stem cells (CSCs) and acquisition of epithelial-to-mesenchymal transition (EMT). Since CSCs and EMT are important biological factors responsible for cancer cell invasion, metastasis, drug resistance and tumor recurrence, the Hh signaling pathway is believed to be an important target for cancer therapy. AREAS COVERED In recent years, small-molecule inhibitors of Hh signaling have been synthesized for cancer treatment. Clinical trials using these inhibitors are being conducted to determine their toxicity profiles and efficacies. In addition, nutraceuticals (such as isoflavones, curcumin, vitamin D, etc) have been shown to inhibit cancer growth through downregulation of Hh signaling. EXPERT OPINION Inhibition of Hh signaling is important for suppression of cancer growth, invasion, metastasis and recurrence in cancer therapy. However, targeting only one molecule in Hh signaling may not be sufficient to kill cancer cells because cancers show deregulation of multiple signals. Therefore, utilizing new technologies to determine alterations in Hh and other signals for individuals and designing combination strategies with small-molecule Hh inhibitors, nutraceuticals and other chemotherapeutics in targeted personalized therapy could have a significant effect on improving the overall survival of patients with cancers.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
23
|
Combination of hedgehog signaling blockage and chemotherapy leads to tumor reduction in pancreatic adenocarcinomas. Pancreas 2012; 41:222-9. [PMID: 22076568 DOI: 10.1097/mpa.0b013e31822896dd] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Activation of the hedgehog signal transduction pathway, triggered by hedgehog binding to the transmembrane receptor patched 1 (PTCH1) or by mutations in the PTCH1 gene, plays an important role in the development of various tumors. METHODS To investigate whether the Hedgehog signaling pathway is also active in human pancreatic adenocarcinomas, we determined the expression levels of the known Hedgehog target genes PTCH1 and GLI-1 in pancreatic tumors. To determine whether alterations in the PTCH1 gene are responsible for this pathway activation, we screened pancreatic carcinomas for mutations in PTCH. To investigate the contribution of hedgehog signaling to the tumorigenicity of pancreatic tumor cells, we blocked the Hedgehog pathway in cultured tumor cells and xenografts using the steroidal alkaloid cyclopamine and the small-molecule Hedgehog inhibitor Hh-Antag. RESULTS We identified single nucleotide polymorphisms (SNPs) within the PTCH1 gene but no somatic PTCH1 mutations. Pathway-blockage resulted in a significant dose-dependent reduction of tumor cell growth in vitro and in vivo. Moreover, combined treatment with cyclopamine and the conventional antimetabolite gemcitabine revealed a synergistic effect on the reduction of tumor growth in pancreatic adenocarcinoma xenografts. CONCLUSIONS Inhibition of Hedgehog signaling could be a promising approach for the treatment of pancreatic adenocarcinomas.
Collapse
|
24
|
Kiesslich T, Neureiter D. Advances in targeting the Hedgehog signaling pathway in cancer therapy. Expert Opin Ther Targets 2012; 16:151-6. [PMID: 22233124 DOI: 10.1517/14728222.2012.652948] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The Hedgehog (Hh) pathway is one of the central developmental signaling mechanisms, which have recently been shown to contribute to malignant progression of a variety of human cancers. Additionally, the role of Hh and other embryonic signaling pathways in the regulation and maintenance of the tumorigenic cancer stem / -initiating subpopulation underlines the importance of this pathway in human malignancies. The review 'Targeting the Hedgehog signaling pathway for cancer therapy' by Li and coworkers has comprehensively described the potential of pharmacological targeting of Hh signaling. Here we provide an update on the current knowledge on i) the role of this pathway in human tumorigenesis and the rationale for therapeutic targeting, ii) the pharmacological approaches currently being investigated in preclinical and clinical studies, and iii) the outlook for future developments and efforts for establishing Hh antagonists as a valid approach in cancer treatment. Stratification of the tumor type according to Hh-specific expression patterns and clinicopathological characteristics, as well as investigation of possible tumor cell resistance against Hh antagonists, is the central area for further development.
Collapse
|
25
|
Abstract
Numerous signaling pathways are misregulated in pancreatic ductal adenocarcinoma (PDAC), a highly malignant type of cancer. One of these is the Hedgehog (HH) pathway, which is normally involved in patterning processes in the developing embryo. Expression of the main ligand Sonic Hedgehog is an early event in carcinogenesis and correlates with the mutation of the KRAS oncogene, the cardinal molecular feature of pancreatic cancer. Recent data establish a functional role for HH signaling primarily in the tumor microenvironment, where it is involved in myofibroblast differentiation and the induction of stroma-derived growth promoting molecules. Given the protumorigenic functions of the abundant stromal desmoplasia typically associated with pancreatic cancer, targeting the HH pathway might prove beneficial in the treatment of the disease. First data using small molecule antagonists of HH signaling in mouse models of pancreatic cancer are promising and reveal a substantial, yet transient, effect on the myofibroblastic stroma. In this review, we try to give an outline on the current knowledge about HH signaling in pancreatic cancer including a perspective of using pharmacological inhibitors of this pathway in the clinic.
Collapse
Affiliation(s)
- Matthias Lauth
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | | |
Collapse
|
26
|
Nuclear receptor liver receptor homologue 1 (LRH-1) regulates pancreatic cancer cell growth and proliferation. Proc Natl Acad Sci U S A 2011; 108:16927-31. [PMID: 21949357 DOI: 10.1073/pnas.1112047108] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
An essential regulator of gene transcription, nuclear receptor liver receptor homologue 1 (LRH-1) controls cell differentiation in the developing pancreas and maintains cholesterol homeostasis in adults. Recent genome-wide association studies linked mutations in the LRH-1 gene and its up-stream regulatory regions to development of pancreatic cancer. In this work, we show that LRH-1 transcription is activated up to 30-fold in human pancreatic cancer cells compared to normal pancreatic ductal epithelium. This activation correlates with markedly increased LRH-1 protein expression in human pancreatic ductal adenocarcinomas in vivo. Selective blocking of LRH-1 by receptor specific siRNA significantly inhibits pancreatic cancer cell proliferation in vitro. The inhibition is tracked in part to the attenuation of the receptor's transcriptional targets controlling cell growth, proliferation, and differentiation. Previously, LRH-1 was shown to contribute to formation of intestinal tumors. This study demonstrates the critical involvement of LRH-1 in development and progression of pancreatic cancer, suggesting the LRH-1 receptor as a plausible therapeutic target for treatment of pancreatic ductal adenocarcinomas.
Collapse
|
27
|
Solberg N, Machon O, Krauss S. Characterization and functional analysis of the 5'-flanking promoter region of the mouse Tcf3 gene. Mol Cell Biochem 2011; 360:289-99. [PMID: 21935611 DOI: 10.1007/s11010-011-1068-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 09/08/2011] [Indexed: 01/27/2023]
Abstract
Tcf3 is a nuclear mediator of canonical Wnt signaling which functions in many systems as a repressor of target gene transcription. In this study, we have cloned and characterized a 6.7 kb fragment of the 5'-flanking promoter region of the mouse Tcf3 gene. In silico analysis of the promoter sequence identified the existence of GC boxes and CpG islands, but failed to identify any TATA box. In addition, the promoter sequence contained putative binding sites for multiple transcription factors, including a few known to regulate the function of mTcf3. Of those, we confirmed functional binding sites for NFκB and Oct1 using a luciferase assay and ChIP. In vitro analysis revealed five potential transcription start sites which resulted in a 298 base pair 5'-untranslated region upstream of the mTcf3 translation start site ATG. Using a luciferase assay, we analyzed the activity of the cloned promoter fragment in embryonically derived neural stem cells. The luciferase activity of a 3.5 kb core promoter fragment (-3243/+211) showed up to 40-fold increased activity compared to the empty vector. Addition of sequences 5' to the 3.5 kb core promoter fragment resulted in a 20-fold drop in luciferase activity, indicating the presence of further upstream repressive elements. In vivo analysis of a 4.5 kb promoter fragment (-4303/+211) driving, the expression of EGFP in mouse embryos highly resembled endogenous expression of mTcf3.
Collapse
Affiliation(s)
- Nina Solberg
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Forskningsparken, Oslo, Norway.
| | | | | |
Collapse
|
28
|
Jamieson NB, Carter CR, McKay CJ, Oien KA. Tissue biomarkers for prognosis in pancreatic ductal adenocarcinoma: a systematic review and meta-analysis. Clin Cancer Res 2011; 17:3316-31. [PMID: 21444679 DOI: 10.1158/1078-0432.ccr-10-3284] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PURPOSE The management of pancreatic ductal adenocarcinoma (PDAC) continues to present a great challenge particularly with regard to prediction of outcome following pancreaticoduodenectomy. Molecular markers have been extensively investigated by numerous groups with the aim of enhancing prognostication; however, despite hundreds of studies that have sought to assess the potential prognostic value of molecular markers in predicting the clinical course following resection of PDAC, at this time, no molecular marker assay forms part of recommended clinical practice. EXPERIMENTAL DESIGN We conducted a systematic review and meta-analysis of the published literature for immunohistochemistry-based biomarkers of PDAC outcome. A dual search strategy was applied to the PubMed database on January 6, 2010, to identify cohort studies that reported associations between immunohistochemical biomarker expression and survival outcomes in PDAC, and conformed to the REMARK (REporting recommendations for tumor MARKer prognostic studies) criteria. RESULTS A total of 103 distinct proteins met all inclusion criteria. Promising markers that emerged for the prediction of overall survival included BAX (HR = 0.31, 95% CI: 0.71-0.56), Bcl-2 (HR = 0.41, 95% CI: 0.27-0.63), survivin (HR = 0.46, 95% CI: 0.29-0.73), Ki-67: (HR = 2.42, 95% CI: 1.87-3.14), COX-2 (HR = 1.39, 95% CI: 1.13-1.71), E-cadherin (HR = 1.80, 95% CI: 1.33-2.42), and S100 calcium-binding proteins, in particular S100A2 (HR = 3.23, 95% CI: 1.58-6.62). CONCLUSIONS We noted that that there was incomplete adherence to the REMARK guidelines with inadequate methodology reporting as well as failure to perform multivariate analysis. Addressing the persistent incomplete adoption of these criteria may eventually result in the incorporation of molecular marker assessment within PDAC management algorithms.
Collapse
Affiliation(s)
- Nigel B Jamieson
- West of Scotland Pancreatic Unit and Department of Pathology, Glasgow Royal Infirmary, Alexandra Parade, Glasgow, United Kingdom
| | | | | | | |
Collapse
|
29
|
Liu SH, Patel S, Gingras MC, Nemunaitis J, Zhou G, Chen C, Li M, Fisher W, Gibbs R, Brunicardi FC. PDX-1: demonstration of oncogenic properties in pancreatic cancer. Cancer 2011; 117:723-33. [PMID: 20886630 PMCID: PMC3017729 DOI: 10.1002/cncr.25629] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 08/06/2010] [Indexed: 01/13/2023]
Abstract
BACKGROUND Pancreatic-duodenal homeobox 1 (PDX-1) is a transcription factor that regulates embryologic pancreas development and insulin expression in the adult islet; however, it is overexpressed in many types of cancer, including pancreatic cancer. The purpose of this study was to investigate the role of PDX-1 in tumorigenesis in human cells. METHODS In vitro cell proliferation, invasion, and transformation were performed in human embryonic kidney cell line (HEK 293), pancreatic cancer cell line MIA PaCa2, and human pancreatic ductal epithelial (HPDE) cells transiently or stably expressing PDX-1 or green fluorescent protein (GFP) PDX-1, with or without cotransfection of PDX-1 short hairpin RNA (shRNA). In vivo tumor formation was carried out in severe combined immunodeficiency (SCID) mice with subcutaneous injection of HEK 293 and MIA PaCa2 stably transfected cells. Cell cycle was analyzed by Western blot or immunostaining. Microarray of RNA from pancreatic adenocarcinoma cells with and without PDX-1 shRNA was performed and analyzed. RESULTS Transient and stable expressing PDX-1 significantly increased cell proliferation and invasion in HEK 293, human pancreatic ductal epithelial (HPDE), and MIA PaCa2 cells versus controls (P < .05), human PDX-1 shRNA reversed these effects. Expression of PDX-1 significantly increased colony formation in HEK 293, HPDE, and MIA PaCa2 cells versus controls in vitro (P < .05). PDX-1 promoted HEK 293 and MIA PaCa2 tumor formation in SCID mice as compared with that of control (P < .05). PDX-1 overexpression disrupted cell cycles proteins. PDX-1 expression was confirmed by Western blot and tracked by viewing of GFP-PDX-1 expression. Microarray data support an oncogenic role of PDX-1 in pancreas cancer cells. CONCLUSIONS PDX-1 induced increased cell proliferation, invasion, and colony formation in vitro, and resulted in markedly increased HEK 293 and MIA PaCa2 tumor formation in SCID mice. These data suggest that PDX-1 is a potential oncogene that regulates tumorigenesis.
Collapse
Affiliation(s)
- Shi-He Liu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine Houston, TX
| | - Sanjeet Patel
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine Houston, TX
| | | | | | - Guisheng Zhou
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine Houston, TX
| | - Changyi Chen
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine Houston, TX
| | - Min Li
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine Houston, TX
| | - William Fisher
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine Houston, TX
| | - Richard Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine Houston, TX
| | | |
Collapse
|
30
|
Balentine CJ, Berger DH, Liu SH, Chen C, Nemunaitis J, Brunicardi FC. Defining the cancer master switch. World J Surg 2011; 35:1738-45. [PMID: 21286716 DOI: 10.1007/s00268-010-0941-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Recent research has focused on signaling cascades and their interactions yielding considerable insight into which genetic pathways are targeted and how they tend to be altered in tumors. Therapeutic interventions now can be designed based on the knowledge of pathways vital to tumor growth and survival. These critical targets for intervention, master switches for cancer, are termed so because the tumor attempts to "flip the switch" in a way that promotes its survival, whereas molecular therapy aims to "switch off" signals important for tumor-related processes. METHODS Literature review. CONCLUSIONS Defining useful targets for therapy depends on identifying pathways that are crucial for tumor growth, survival, and metastasis. Because not all signaling cascades are created equal, selecting master switches or targets for intervention needs to be done in a systematic fashion. This discussion proposes a set of criteria to define what it means to be a cancer master switch and provides examples to illustrate their application.
Collapse
|
31
|
Srivastava RK, Tang SN, Zhu W, Meeker D, Shankar S. Sulforaphane synergizes with quercetin to inhibit self-renewal capacity of pancreatic cancer stem cells. Front Biosci (Elite Ed) 2011; 3:515-28. [PMID: 21196331 DOI: 10.2741/e266] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
According to the cancer stem cell hypothesis, the aggressive growth and early metastasis of cancer may arise through dysregulation of self-renewal of stem cells. The objectives of this study were to examine the molecular mechanisms by which sulforaphane (SFN, an active compound in cruciferous vegetables) inhibits self-renewal capacity of pancreatic cancer stem cells (CSCs), and synergizes with quercetin, a major polyphenol and flavonoid commonly detected in many fruits and vegetables. Our data demonstrated that SFN inhibited self-renewal capacity of pancreatic CSCs. Inhibition of Nanog by lentiviral-mediated shRNA expression enhanced the inhibitory effects of sulforaphane on self-renewal capacity of CSCs. SFN induced apoptosis by inhibiting the expression of Bcl-2 and XIAP, phosphorylation of FKHR, and activating caspase-3. Moreover, SFN inhibited expression of proteins involved in the epithelial-mesenchymal transition (beta-catenin, vimentin, twist-1, and ZEB1), suggesting the blockade of signaling involved in early metastasis. Furthermore, the combination of quercetin with SFN had synergistic effects on self-renewal capacity of pancreatic CSCs. These data suggest that SFN either alone or in combination with quercetin can eliminate cancer stem cell-characteristics.
Collapse
Affiliation(s)
- Rakesh K Srivastava
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | | | | | | | |
Collapse
|
32
|
Chen M, Hildebrandt MAT, Clague J, Kamat AM, Picornell A, Chang J, Zhang X, Izzo J, Yang H, Lin J, Gu J, Chanock S, Kogevinas M, Rothman N, Silverman DT, Garcia-Closas M, Grossman HB, Dinney CP, Malats N, Wu X. Genetic variations in the sonic hedgehog pathway affect clinical outcomes in non-muscle-invasive bladder cancer. Cancer Prev Res (Phila) 2010; 3:1235-45. [PMID: 20858759 DOI: 10.1158/1940-6207.capr-10-0035] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Sonic hedgehog (Shh) pathway genetic variations may affect bladder cancer risk and clinical outcomes. Therefore, we genotyped 177 single-nucleotide polymorphisms (SNP) in 11 Shh pathway genes in a study including 803 bladder cancer cases and 803 controls. We assessed SNP associations with cancer risk and clinical outcomes in 419 cases of non-muscle-invasive bladder cancer (NMIBC) and 318 cases of muscle-invasive and metastatic bladder cancer (MiMBC). Only three SNPs (GLI3 rs3823720, rs3735361, and rs10951671) reached nominal significance in association with risk (P ≤ 0.05), which became nonsignificant after adjusting for multiple comparisons. Nine SNPs reached a nominally significant individual association with recurrence of NMIBC in patients who received transurethral resection (TUR) only (P ≤ 0.05), of which two (SHH rs1233560 and GLI2 rs11685068) were replicated independently in 356 TUR-only NMIBC patients, with P values of 1.0 × 10(-3) (SHH rs1233560) and 1.3 × 10(-3) (GLI2 rs11685068). Nine SNPs also reached a nominally significant individual association with clinical outcome of NMIBC patients who received Bacillus Calmette-Guérin (BCG; P ≤ 0.05), of which two, the independent GLI3 variants rs6463089 and rs3801192, remained significant after adjusting for multiple comparisons (P = 2 × 10(-4) and 9 × 10(-4), respectively). The wild-type genotype of either of these SNPs was associated with a lower recurrence rate and longer recurrence-free survival (versus the variants). Although three SNPs (GLI2 rs735557, GLI2 rs4848632, and SHH rs208684) showed nominal significance in association with overall survival in MiMBC patients (P ≤ 0.05), none remained significant after multiple-comparison adjustments. Germ-line genetic variations in the Shh pathway predicted clinical outcomes of TUR and BCG for NMIBC patients.
Collapse
Affiliation(s)
- Meng Chen
- Department of Epidemiology, The University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Marquardt J, Factor V, Thorgeirsson S. Epigenetic regulation of cancer stem cells in liver cancer: current concepts and clinical implications. J Hepatol 2010; 53:568-77. [PMID: 20646772 PMCID: PMC3492877 DOI: 10.1016/j.jhep.2010.05.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/04/2010] [Accepted: 05/05/2010] [Indexed: 12/21/2022]
Abstract
The two dominant models of carcinogenesis postulate stochastic (clonal evolution) or hierarchic organization of tumor (cancer stem cell model). According to the latter, at the germinal center of tumor evolution is a cancer stem cell (CSC) which, similar to normal adult stem cells, possesses the capacity of self-renewal and a differentiation potential. Over the past few years, compelling evidence has emerged in support of the hierarchic cancer model for many solid tumors including hepatocellular cancers. The CSCs are posited to be responsible not only for tumor initiation but also for the generation of distant metastasis and relapse after therapy. These characteristics are particularly relevant for a multi-resistant tumor entity like human hepatocellular carcinoma and may herald a paradigm shift in the management of this deadly disease. Identification and detailed characterization of liver CSCs is therefore imperative for improving prevention approaches, enhancing early detection, and extending the limited treatment options. Despite the current progress in understanding the contribution of CSCs to the generation of heterogeneity of tumors, the molecular complexity and exact regulation of CSCs is poorly understood. This review focuses on the genetic and epigenetic mechanisms that regulate and define the unique CSC properties with an emphasis on key regulatory pathways of liver CSCs and their clinical significance.
Collapse
MESH Headings
- AC133 Antigen
- Aldehyde Dehydrogenase/metabolism
- Antigens, CD/metabolism
- Antigens, Neoplasm/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/secondary
- Cell Adhesion Molecules/metabolism
- Cell Division
- Cell Separation/methods
- Epigenesis, Genetic
- Epithelial Cell Adhesion Molecule
- Genes, myc
- Glycoproteins/metabolism
- Hedgehog Proteins/metabolism
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- MicroRNAs/genetics
- Models, Biological
- Neoplasm Metastasis/genetics
- Neoplasm Metastasis/pathology
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/physiology
- Nuclear Proteins/genetics
- Peptides/metabolism
- Polycomb Repressive Complex 1
- Proto-Oncogene Proteins/genetics
- RNA, Neoplasm/genetics
- Receptors, Notch/metabolism
- Repressor Proteins/genetics
- Signal Transduction
- Thy-1 Antigens/metabolism
- Transforming Growth Factor beta/metabolism
- Wnt Proteins/metabolism
- beta Catenin/metabolism
Collapse
Affiliation(s)
- J.U. Marquardt
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, MD, USA
| | - V.M. Factor
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, MD, USA
| | - S.S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, MD, USA
| |
Collapse
|
34
|
Lauth M, Bergström A, Shimokawa T, Tostar U, Jin Q, Fendrich V, Guerra C, Barbacid M, Toftgård R. DYRK1B-dependent autocrine-to-paracrine shift of Hedgehog signaling by mutant RAS. Nat Struct Mol Biol 2010; 17:718-25. [PMID: 20512148 DOI: 10.1038/nsmb.1833] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 03/16/2010] [Indexed: 02/07/2023]
Abstract
Synergism between the RAS and Hedgehog (HH) pathways has been suggested for carcinogenesis in the pancreas, lung and colon. We investigated the molecular cross-talk between RAS and HH signaling and found that, although mutant RAS induces or enhances SHH expression and favors paracrine HH signaling, it antagonizes autocrine HH signal transduction. Activated RAS can be found in primary cilia, the central organelle of HH signal transduction, but functions in a cilium-independent manner and interferes with Gli2 function and Gli3 processing. In addition, the cell-autonomous negative regulation of HH signal transduction involves the RAS effector molecule dual specificity tyrosine phosphorylated and regulated kinase 1B (DYRK1B). In line with a redirection of autocrine toward paracrine HH signaling by a KRAS-DYRK1B network, we find high levels of GLI1 expression restricted to the stromal compartment and not to SHH-expressing tumor cells in human pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Matthias Lauth
- Karolinska Institutet, Center for Biosciences, Department of Biosciences and Nutrition, Huddinge, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Pancreatic cancer represents a major challenge for research studies and clinical management. No specific tumor marker for the diagnosis of pancreatic cancer exists. Therefore, extensive genomic, transcriptomic, and proteomic studies are being developed to identify candidate markers for use in high-throughput systems capable of large cohort screening. Understandably, the complex pathophysiology of pancreatic cancer requires sensitive and specific biomarkers that can improve both early diagnosis and therapeutic monitoring. The lack of a single diagnostic marker makes it likely that only a panel of biomarkers is capable of providing the appropriate combination of high sensitivity and specificity. Biomarker discovery using novel technology can improve prognostic upgrading and pinpoint new molecular targets for innovative therapy.
Collapse
|
36
|
Tanase CP, Neagu M, Albulescu R, Codorean E, Dima SO. Biomarkers in the diagnosis and early detection of pancreatic cancer. EXPERT OPINION ON MEDICAL DIAGNOSTICS 2009; 3:533-546. [PMID: 23495983 DOI: 10.1517/17530050903117256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pancreatic cancer, owing to its raising incidence and aggressiveness, is a major challenge, both for research and for clinical management. As pancreatic cancer has a complex pathophysiology, in addition to improving the methods of early diagnosis, sensitive and specific biomarkers are a prerequisite. OBJECTIVE As there is no specific tumor marker for pancreatic cancer diagnosis, extensive genomics/transcriptomics and proteomics studies have been developed with the aim of finding candidate markers and contributing to high-throughput systems for large cohort screening. METHODS A literature review was done to study these biomarkers in relation to diagnosis, prognosis and therapy targets in pancreatic cancer. RESULTS/CONCLUSION For early diagnosis improvement, only a panel of soluble biomarkers could provide the appropriate combination between high sensitivity and specificity. Prognostic upgrading would benefit from biomarker discovery and validation performed on tumor tissue. New technology could delineate molecular targets for innovative therapy in pancreatic cancer.
Collapse
Affiliation(s)
- Cristiana Pistol Tanase
- 'VICTOR BABES' National Institute of Pathology, 99-101 Splaiul Independentei, Bucharest, Romania +4021 319 45 28 ; +4021 319 45 28 ;
| | | | | | | | | |
Collapse
|
37
|
Abstract
As our understanding of pancreatic cancer evolves, evidence is growing to support a role for cancer stem cells in this devastating disease. Cancer stem cells constitute a distinct subpopulation in the tumor and are considered to drive both tumorigenesis and metastasis; these cells are thought to be highly resistant to standard treatment modalities. Here we review the current knowledge on pancreatic cancer stem cells and the implementation of cancer stem cell markers as prognostic or predictive biomarkers. We also discuss prospects for the use of cancer stem cells as targets for future therapeutic regimens in pancreatic cancer.
Collapse
|