1
|
AbuQeis I, Zou Y, Ba YC, Teeti AA. Neuroscience of cancer: Research progress and emerging of the field. IBRAIN 2024; 10:305-322. [PMID: 39346791 PMCID: PMC11427805 DOI: 10.1002/ibra.12172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 10/01/2024]
Abstract
Cancer cells immediately expand and penetrate adjoining tissues, as opposed to metastasis, that is the spread of cancer cells through the circulatory or lymphatic systems to more distant places via the invasion process. We found that a lack of studies discussed tumor development with the nervous system, by the aspects of cancer-tissue invasion (biological) and chemical modulation of growth that cascades by releasing neural-related factors from the nerve endings via chemical substances known as neurotransmitters. In this review, we aimed to carefully demonstrate and describe the cancer invasion and interaction with the nervous system, as well as reveal the research progress and the emerging neuroscience of cancer. An initial set of 160 references underwent systematic review and summarization. Through a meticulous screening process, these data were refined, ultimately leading to the inclusion of 98 studies that adhered to predetermined criteria. The outcomes show that one formidable challenge in the realm of cancer lies in its intrinsic heterogeneity and remarkable capacity for rapid adaptation. Despite advancements in genomics and precision medicine, there is still a need to identify new molecular targets. Considering cancer within its molecular and cellular environment, including neural components, is crucial for addressing this challenge. In conclusion, this review provides good referential data for direct, indirect, biological, and chemical interaction for nerve tissue-tumor interaction, suggesting the establishment of new therapy techniques and mechanisms by controlling and modifying neuron networks that supply signals to tumors.
Collapse
Affiliation(s)
- Issam AbuQeis
- Department of Radiology Palestinian Ministry of Health Ramallah Palestine
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Yu Zou
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Ying-Chun Ba
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Abeer A Teeti
- Department of Chemistry, School of Science Hebron University Hebron Palestine
- Department of Epidemiology, School of Public Health Kunming Medical University Kunming China
| |
Collapse
|
2
|
Li J, Du J, Li Y, Meng M, Hang J, Shi H. A nomogram based on CT texture features to predict the response of patients with advanced pancreatic cancer treated with chemotherapy. BMC Gastroenterol 2023; 23:274. [PMID: 37563572 PMCID: PMC10416463 DOI: 10.1186/s12876-023-02902-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE This study aimed to evaluate the predictive value of computed tomography (CT) texture features in the treatment response of patients with advanced pancreatic cancer (APC) receiving palliative chemotherapy. METHODS This study enrolled 84 patients with APC treated with first-line chemotherapy and conducted texture analysis on primary pancreatic tumors. 59 patients and 25 were randomly assigned to the training and validation cohorts at a ratio of 7:3. The treatment response to chemotherapy was evaluated according to the Response Evaluation Criteria in Solid Tumors (RECIST1.1). The patients were divided into progressive and non-progressive groups. The least absolute shrinkage selection operator (LASSO) was applied for feature selection in the training cohort and a radiomics signature (RS) was calculated. A nomogram was developed based on a multivariate logistic regression model incorporating the RS and carbohydrate antigen 19-9 (CA19-9), and was internally validated using the C-index and calibration plot. We performed the decision curve analysis (DCA) and clinical impact curve analysis to reflect the clinical utility of the nomogram. The nomogram was further externally confirmed in the validation cohort. RESULTS The multivariate logistic regression analysis indicated that the RS and CA19-9 were independent predictors (P < 0.05), and a trend was found for chemotherapy between progressive and non-progressive groups. The nomogram incorporating RS, CA19-9 and chemotherapy showed favorable discriminative ability in the training (C-index = 0.802) and validation (C-index = 0.920) cohorts. The nomogram demonstrated favorable clinical utility. CONCLUSION The RS of significant texture features was significantly associated with the early treatment effect of patients with APC treated with chemotherapy. Based on the RS, CA19-9 and chemotherapy, the nomogram provided a promising way to predict chemotherapeutic effects for APC patients.
Collapse
Affiliation(s)
- Jingjing Li
- Graduate College, Dalian Medical University, Dalian, China
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Jiadi Du
- Department of Computer Science, Missouri University of Science and Technology, Rolla, MO, U.S
| | - Yuying Li
- Graduate College, Dalian Medical University, Dalian, China
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Mingzhu Meng
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Junjie Hang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 518116, Shenzhen, China.
- Department of Oncology, the Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, China.
| | - Haifeng Shi
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China.
| |
Collapse
|
3
|
Huo J, Guan J, Li Y. Metabolism reprogramming signature associated with stromal cells abundance in tumor microenvironment improve prognostic risk classification for gastric cancer. BMC Gastroenterol 2022; 22:364. [PMID: 35907819 PMCID: PMC9338655 DOI: 10.1186/s12876-022-02451-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 07/26/2022] [Indexed: 11/30/2022] Open
Abstract
Background Stromal cells play an important role in the process of tumor progression, but the relationship between stromal cells and metabolic reprogramming is not very clear in gastric cancer (GC). Methods Metabolism-related genes associated with stromal cells were identified in The Cancer Genome Atlas (TCGA) and GSE84437 datasets, and the two datasets with 804 GC patients were integrated into a training cohort to establish the prognostic signature. Univariate Cox regression analysis was used to screen for prognosis-related genes. A risk score was constructed by LASSO regression analysis combined with multivariate Cox regression analysis. The patients were classified into groups with high and low risk according to the median value. Two independent cohorts, GSE62254 (n = 300) and GSE15459 (n = 191), were used to externally verify the risk score performance. The CIBERSORT method was applied to quantify the immune cell infiltration of all included samples. Results A risk score consisting of 24 metabolic genes showed good performance in predicting the overall survival (OS) of GC patients in both the training (TCGA and GSE84437) and testing cohorts (GSE62254 and GSE15459). As the risk score increased, the patients’ risk of death increased. The risk score was an independent prognostic indicator in both the training and testing cohorts suggested by the univariate and multivariate Cox regression analyses. The patients were clustered into four subtypes according to the quantification of 22 kinds of immune cell infiltration (ICI). The proportion of ICI Cluster C with the best prognosis in the low-risk group was approximately twice as high as that in the high-risk group, and the risk score of ICI Cluster C was significantly lower than that of the other three subtypes. Conclusion Our study proposed the first scheme for prognostic risk classification of GC from the perspective of tumor stromal cells and metabolic reprogramming, which may contribute to the development of therapeutic strategies for GC. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02451-2.
Collapse
Affiliation(s)
- Junyu Huo
- The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266003, China
| | - Jing Guan
- Department of General Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, 266035, Shandong, China
| | - Yankun Li
- Department of Critical Care Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, 266035, Shandong, China.
| |
Collapse
|
4
|
Tanaka C, Furihata K, Naganuma S, Ogasawara M, Yoshioka R, Taniguchi H, Furihata M, Taniuchi K. Establishment of a mouse model of pancreatic cancer using human pancreatic cancer cell line S2-013-derived organoid. Hum Cell 2022; 35:735-744. [PMID: 35150409 PMCID: PMC8866361 DOI: 10.1007/s13577-022-00684-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/03/2022] [Indexed: 12/26/2022]
Abstract
A well-established preclinical model of pancreatic cancer needs to be established to facilitate research on new therapeutic targets. Recently established animal models of pancreatic cancer, including patient-derived tumor models and organoid models, are used for pre-clinical drug testing and biomarker discovery. These models have useful characteristics over conventional xenograft mouse models based on cell lines in preclinical studies, but still cannot accurately predict the clinical outcomes of new treatments and have not yet been broadly implemented in research. We employed pancreatic cancer organoid culture methods using the pancreatic cancer cell line S2-013, and performed pathological and immunohistochemical analyses to characterize tumor xenografts obtained from a mouse model implanted with S2-013 cell line-derived organoids. Serum levels of the pancreatic cancer tumor marker CA19-9 were measured by ELISA. We generated human pancreatic cancer organoids using a co-culture of S2-013 cells, human endothelial cells derived from human umbilical vein endothelial cells, and human mesenchymal stem cells, and established a mouse model with subcutaneously transplanted human pancreatic cancer organoids (S2-013-organoid model). Although blood clotting crater-like formation developed in the middle of subcutaneous xenografts in the S2-013-conventional model, created by subcutaneously injecting S2-013 cells into the right flank of nude mice, the size of xenografts in the S2-013-organoid model gradually increased without crater-like formation. Importantly, tumor xenografts obtained from the S2-013-organoid model exhibited a clinical human pancreatic cancer tissue-like cellular morphology, tissue architecture, and polarity, and actively formed cancer stroma containing mature blood vessels with the high expression of the vascular tight junction marker CD31. In subcutaneous xenografts of S2-013-conventional mice, no blood vessel density or widely expanding areas of necrotic regions were present. Consequently, serum levels of CA19-9 in the S2-013-organoid model correlated with tumor volumes. In addition, epithelial–mesenchymal transition, the conversion of epithelial cells to the mesenchymal phenotype, was observed in tumor xenografts of the S2-013-organoid model. The S2-013-organoid model provides tumor xenografts consisting of clinical human pancreatic cancer-like tissue formation with the effective development of vascularized stroma, and may be valuable for facilitating studies on pre-clinical drug testing and biomarker discovery.
Collapse
Affiliation(s)
- Chiharu Tanaka
- Department of Pathology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Kaoru Furihata
- Department of Pathology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Seiji Naganuma
- Department of Pathology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Mitsunari Ogasawara
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Reiko Yoshioka
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Hideki Taniguchi
- Department of Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Mutsuo Furihata
- Department of Pathology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Keisuke Taniuchi
- Department of Gastroenterology and Hepatology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan.
| |
Collapse
|
5
|
Patched 1-interacting Peptide Represses Fibrosis in Pancreatic Cancer to Augment the Effectiveness of Immunotherapy. J Immunother 2021; 43:121-133. [PMID: 31834207 DOI: 10.1097/cji.0000000000000305] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is resistant to immunotherapy. As a factor of resistance, the dense fibrosis of this cancer acts as a barrier to inhibit immune cell infiltration into a tumor. We examined the influence of a Hedgehog signal inhibitor, Patched 1-interacting peptide, on fibrosis, infiltration of immune cells, and immunotherapeutic effects on PDAC. We found that this peptide inhibited proliferation and migration of cancer-associated fibroblasts and cancer cells. Furthermore, this peptide reduced the production of extracellular matrix and transforming growth factor β1 in cancer-associated fibroblasts and induced expression of HLA-ABC in PDAC cells and interferon-γ in lymphocytes. In vivo, the peptide suppressed fibrosis of PDAC and increased immune cell infiltration into tumors. The combination of this peptide and an anti-programmed death-1 antibody augmented the antitumor effect, and this combination showed the same effect in experiments using cancer cells and autologous lymphocytes. These results indicate that, in addition to the direct effect of tumor suppression, the Patched 1-interacting peptide increases the infiltration of immune cells by reducing fibrosis of PDAC and consequently enhances the effect of immunotherapy. Therefore, treatment with this peptide may be a novel therapy with 2 different mechanisms: direct tumor suppression and enhancing the immune response against PDAC.
Collapse
|
6
|
Lin Q, Qian Z, Jusko WJ, Mager DE, Ma WW, Straubinger RM. Synergistic Pharmacodynamic Effects of Gemcitabine and Fibroblast Growth Factor Receptor Inhibitors on Pancreatic Cancer Cell Cycle Kinetics and Proliferation. J Pharmacol Exp Ther 2021; 377:370-384. [PMID: 33753538 PMCID: PMC9885358 DOI: 10.1124/jpet.120.000412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/16/2021] [Indexed: 02/02/2023] Open
Abstract
Median survival of pancreatic ductal adenocarcinoma cancer (PDAC) is 6 months, with 9% 5-year survival. Standard-of-care gemcitabine (Gem) provides only modest survival benefits, and combination therapies integrating novel targeted agents could improve outcomes. Fibroblast growth factor (FGF) receptors (FGFRs) play important roles in PDAC growth and invasion. Therefore, FGFR inhibitors (FGFRi) merit further investigation. Efficacy of Gem combined with NVP-BGJ398, a pan-FGFRi, was investigated in multiple PDAC cell lines exposed to the drugs alone and combined. Cell cycle distribution and cell numbers were quantified over time. Two pharmacodynamic models were developed to investigate Gem/BGJ398 interactions quantitatively: a drug-mediated cell proliferation/death model, and a drug-perturbed cell cycle progression model. The models captured temporal changes in cell numbers, cell cycle progression, and cell death during drug exposure. Simultaneous fitting of all data provided reasonable parameter estimates. Therapeutic efficacy was then evaluated in a PDAC mouse model. Compared with Gem alone, combined Gem + FGFRi significantly downregulated ribonucleotide-diphosphate reductase large subunit 1 (RRM1), a gemcitabine resistance (GemR) biomarker, suggesting the FGFRi inhibited GemR emergence. The cell proliferation/death pharmacodynamic model estimated the drug interaction coefficient ψ death = 0.798, suggesting synergistic effects. The mechanism-based cell cycle progression model estimated drug interaction coefficient ψ cycle = 0.647, also suggesting synergy. Thus, FGFR inhibition appears to synergize with Gem in PDAC cells and tumors by sensitizing cells to Gem-mediated inhibition of proliferation and cell cycle progression. SIGNIFICANCE STATEMENT: An integrated approach of quantitative modeling and experimentation was employed to investigate the nature of fibroblast growth factor receptor inhibitor (FGFRi)/gemcitabine (Gem) interaction, and to identify mechanisms by which FGFRi exposure reverses Gem resistance in pancreatic cancer cells. The results show that FGFRi interacts synergistically with Gem to sensitize pancreatic cancer cells and tumors to Gem-mediated inhibition of proliferation and cell cycle progression. Thus, addition of FGFRi to standard-of-care Gem treatment could be a clinically deployable approach to enhance therapeutic benefit to pancreatic cancer patients.
Collapse
Affiliation(s)
- Qingxiang Lin
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.M.S.; Z.Q., W.J.J., D.E.M.); Departments of Cell Stress Biology (Q.L., R.M.S.) and Pharmacology and Therapeutics (R.M.S.), Roswell Park Comprehensive Cancer Center, Buffalo, New York; and Department of Medicine, Mayo Clinic, Rochester, Minnesota (W.W.M.)
| | - Zhicheng Qian
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.M.S.; Z.Q., W.J.J., D.E.M.); Departments of Cell Stress Biology (Q.L., R.M.S.) and Pharmacology and Therapeutics (R.M.S.), Roswell Park Comprehensive Cancer Center, Buffalo, New York; and Department of Medicine, Mayo Clinic, Rochester, Minnesota (W.W.M.)
| | - William J Jusko
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.M.S.; Z.Q., W.J.J., D.E.M.); Departments of Cell Stress Biology (Q.L., R.M.S.) and Pharmacology and Therapeutics (R.M.S.), Roswell Park Comprehensive Cancer Center, Buffalo, New York; and Department of Medicine, Mayo Clinic, Rochester, Minnesota (W.W.M.)
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.M.S.; Z.Q., W.J.J., D.E.M.); Departments of Cell Stress Biology (Q.L., R.M.S.) and Pharmacology and Therapeutics (R.M.S.), Roswell Park Comprehensive Cancer Center, Buffalo, New York; and Department of Medicine, Mayo Clinic, Rochester, Minnesota (W.W.M.)
| | - Wen Wee Ma
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.M.S.; Z.Q., W.J.J., D.E.M.); Departments of Cell Stress Biology (Q.L., R.M.S.) and Pharmacology and Therapeutics (R.M.S.), Roswell Park Comprehensive Cancer Center, Buffalo, New York; and Department of Medicine, Mayo Clinic, Rochester, Minnesota (W.W.M.)
| | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.M.S.; Z.Q., W.J.J., D.E.M.); Departments of Cell Stress Biology (Q.L., R.M.S.) and Pharmacology and Therapeutics (R.M.S.), Roswell Park Comprehensive Cancer Center, Buffalo, New York; and Department of Medicine, Mayo Clinic, Rochester, Minnesota (W.W.M.)
| |
Collapse
|
7
|
Jiang Z, Hou Z, Liu W, Yu Z, Liang Z, Chen S. circ-Keratin 6c Promotes Malignant Progression and Immune Evasion of Colorectal Cancer through microRNA-485-3p/Programmed Cell Death Receptor Ligand 1 Axis. J Pharmacol Exp Ther 2021; 377:358-367. [PMID: 33771844 DOI: 10.1124/jpet.121.000518] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/08/2021] [Indexed: 12/24/2022] Open
Abstract
Recently, circular RNA was reported to be a significant participant in the development of tumorigenesis, including colorectal cancer. Therefore, we aimed to clarify the precise role of circ-keratin 6C (circ-KRT6C) in colorectal cancer progression. The relative expression levels of circ-KRT6C, microRNA-485-3p (miR-485-3p), and programmed cell death receptor ligand 1 (PDL1) were analyzed by real-time quantitative polymerase chain reaction and Western blot assays. The proliferation was assessed by cell count kit 8 and colony-forming assays. The apoptotic cells were determined by flow cytometry assay. The migration and invasion were analyzed by transwell assay. Colorectal cancer cells were cocultured with peripheral blood mononuclear cells or cytokine-induced killer cells to assess immune response. The interaction relationships among circ-KRT6C, miR-485-3p, and PDL1 were examined by dual-luciferase reporter assay. The effects of circ-KRT6C inhibition in vivo were analyzed by an animal experiment. circ-KRT6C was overexpressed in colorectal cancer tissues and cells, and its level was associated with overall survival time of patients with colorectal cancer. The suppression of circ-KRT6C suppressed growth, migration, invasion, and immune escape while stimulating apoptosis in colorectal cancer cells, which was abolished by shortage of miR-485-3p. In addition, overexpression of miR-485-3p repressed malignant progression and immune evasion of colorectal cancer by targeting PDL1, implying that PDL1 was a functional target of miR-485-3p. A xenograft experiment also suggested that circ-KRT6C inhibition could repress tumor growth in vivo. circ-KRT6C could increase PDL1 expression by functioning as an miR-485-3p sponge, which promoted malignant progression and immune evasion of colorectal cancer cells. SIGNIFICANCE STATEMENT: circ-keratin 6c could increase programmed cell death receptor ligand 1 expression by functioning as a microRNA-16-5p sponge, which promoted malignant progression and immune evasion of colorectal cancer.
Collapse
Affiliation(s)
- Zhipeng Jiang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, Guangdong, P.R.China
| | - Zehui Hou
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, Guangdong, P.R.China
| | - Wei Liu
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, Guangdong, P.R.China
| | - Zhuomin Yu
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, Guangdong, P.R.China
| | - Zhiqiang Liang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, Guangdong, P.R.China
| | - Shuang Chen
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, Guangdong, P.R.China
| |
Collapse
|
8
|
Wang J, Wu Q, Wang Y, Xiang L, Feng J, Zhou Z, Fu Q, Zhang L. Collagenase-loaded pH-sensitive nanocarriers efficiently remodeled tumor stroma matrixes and improved the enrichment of nanomedicines. NANOSCALE 2021; 13:9402-9414. [PMID: 34002757 DOI: 10.1039/d1nr00950h] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The dense extracellular matrix (ECM) in tumor tissue severely hinders the penetration and enrichment of antitumor nanomedicines, which could significantly affect their efficiency. In this study, we used pH-sensitive nanocarriers loaded with collagenase (Col) to remold the tumor microenvironment (TME). Furthermore, we combined the collagenase delivery system with a nanomedicine to improve its penetration and enrichment in the tumor, thereby improving efficacy. We synthesized acetalated dextran (Ace-DEX) with an ideal pH-sensitivity as the carrier material of collagenase. Under mild preparation conditions, collagenase was loaded into Ace-DEX nanoparticles (NPs) with a high loading capacity (>4%) and remained highly active (>90%). Col-carrying NPs (Col-NPs) significantly reduced the tumor collagen content by 15.1%. Pretreatment with Col-NPs increased the accumulation of doxorubicin (DOX)-loaded liposome (DOX-Lipo) in the tumor by 2.8-fold. There were no safety concerns as the Col-NP showed no significant toxicity and reduced Col-induced damage to healthy tissues. Additionally, the number of circulating tumor cells remained unchanged after Col-NP treatment, suggesting no increased risk of tumor metastasis. Because the Col-NP acts essentially independent of the subsequent treatment, it has considerable potential for enhancing many existing delivery systems and drugs for cancer treatment. It may also be used for treating other collagen-related diseases.
Collapse
Affiliation(s)
- Jiading Wang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
| | - Qingsi Wu
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuanfang Wang
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ling Xiang
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiaxing Feng
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhaojie Zhou
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
| | - Qiang Fu
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
9
|
Koltai T, Reshkin SJ, Carvalho TMA, Cardone RA. Targeting the Stromal Pro-Tumoral Hyaluronan-CD44 Pathway in Pancreatic Cancer. Int J Mol Sci 2021; 22:3953. [PMID: 33921242 PMCID: PMC8069142 DOI: 10.3390/ijms22083953] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies. Present-day treatments have not shown real improvements in reducing the high mortality rate and the short survival of the disease. The average survival is less than 5% after 5 years. New innovative treatments are necessary to curtail the situation. The very dense pancreatic cancer stroma is a barrier that impedes the access of chemotherapeutic drugs and at the same time establishes a pro-proliferative symbiosis with the tumor, thus targeting the stroma has been suggested by many authors. No ideal drug or drug combination for this targeting has been found as yet. With this goal in mind, here we have explored a different complementary treatment based on abundant previous publications on repurposed drugs. The cell surface protein CD44 is the main receptor for hyaluronan binding. Many malignant tumors show over-expression/over-activity of both. This is particularly significant in pancreatic cancer. The independent inhibition of hyaluronan-producing cells, hyaluronan synthesis, and/or CD44 expression, has been found to decrease the tumor cell's proliferation, motility, invasion, and metastatic abilities. Targeting the hyaluronan-CD44 pathway seems to have been bypassed by conventional mainstream oncological practice. There are existing drugs that decrease the activity/expression of hyaluronan and CD44: 4-methylumbelliferone and bromelain respectively. Some drugs inhibit hyaluronan-producing cells such as pirfenidone. The association of these three drugs has never been tested either in the laboratory or in the clinical setting. We present a hypothesis, sustained by hard experimental evidence, suggesting that the simultaneous use of these nontoxic drugs can achieve synergistic or added effects in reducing invasion and metastatic potential, in PDAC. A non-toxic, low-cost scheme for inhibiting this pathway may offer an additional weapon for treating pancreatic cancer.
Collapse
Affiliation(s)
| | - Stephan Joel Reshkin
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (R.A.C.)
| | - Tiago M. A. Carvalho
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (R.A.C.)
| | - Rosa A. Cardone
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (R.A.C.)
| |
Collapse
|
10
|
Suhail Y, Afzal J. Evolved Resistance to Placental Invasion Secondarily Confers Increased Survival in Melanoma Patients. J Clin Med 2021; 10:jcm10040595. [PMID: 33562461 PMCID: PMC7915120 DOI: 10.3390/jcm10040595] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 12/25/2022] Open
Abstract
Mammals exhibit large differences in rates of cancer malignancy, even though the tumor formation rates may be similar. In placental mammals, rates of malignancy correlate with the extent of placental invasion. Our Evolved Levels of Invasibility (ELI) framework links these two phenomena identifying genes that potentially confer resistance in stromal fibroblasts to limit invasion, from trophoblasts in the endometrium, and from disseminating melanoma in the skin. Herein, using patient data from The Cancer Genome Atlas (TCGA), we report that these anti-invasive genes may be crucial in melanoma progression in human patients, and that their loss is correlated with increased cancer spread and lowered survival. Our results suggest that, surprisingly, these anti-invasive genes, which have lower expression in humans compared to species with non-invasive placentation, may potentially prevent stromal invasion, while a further reduction in their levels increases the malignancy and lethality of melanoma. Our work links evolution, comparative biology, and cancer progression across tissues, indicating new avenues for using evolutionary medicine to prognosticate and treat human cancers.
Collapse
Affiliation(s)
- Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA;
- Cancer Systems Biology (CaSB@Yale), Yale West Campus, West Haven, CT 06477, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA
| | - Junaid Afzal
- Department of Cardiology, University of California, San Francisco, CA 94143, USA;
| |
Collapse
|
11
|
Ghidini M, Lampis A, Mirchev MB, Okuducu AF, Ratti M, Valeri N, Hahne JC. Immune-Based Therapies and the Role of Microsatellite Instability in Pancreatic Cancer. Genes (Basel) 2020; 12:33. [PMID: 33383713 PMCID: PMC7823781 DOI: 10.3390/genes12010033] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/25/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is one of the most aggressive malignancies with limited treatment options thus resulting in high morbidity and mortality. Among all cancers, with a five-year survival rates of only 2-9%, pancreatic cancer holds the worst prognostic outcome for patients. To improve the overall survival, an earlier diagnosis and stratification of cancer patients for personalized treatment options are urgent needs. A minority of pancreatic cancers belong to the spectrum of Lynch syndrome-associated cancers and are characterized by microsatellite instability (MSI). MSI is a consequence of defective mismatch repair protein functions and it has been well characterized in other gastrointestinal tumors such as colorectal and gastric cancer. In the latter, high levels of MSI are linked to a better prognosis and to an increased benefit to immune-based therapies. Therefore, the same therapies could offer an opportunity of treatment for pancreatic cancer patients with MSI. In this review, we summarize the current knowledge about immune-based therapies and MSI in pancreatic cancer.
Collapse
Affiliation(s)
- Michele Ghidini
- Division of Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London SM25NG, UK; (A.L.); (M.R.); (N.V.)
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM25NG, UK
| | - Milko B. Mirchev
- Clinic of Gastroenterology, Medical University, 9002 Varna, Bulgaria;
| | | | - Margherita Ratti
- Division of Molecular Pathology, The Institute of Cancer Research, London SM25NG, UK; (A.L.); (M.R.); (N.V.)
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM25NG, UK
- Medical Department, Division of Oncology, ASST di Cremona, Ospedale di Cremona, 26100 Cremona, Italy
| | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, London SM25NG, UK; (A.L.); (M.R.); (N.V.)
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM25NG, UK
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London SM25NG, UK
| | - Jens C. Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London SM25NG, UK; (A.L.); (M.R.); (N.V.)
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM25NG, UK
| |
Collapse
|
12
|
The Interplay between MicroRNAs and the Components of the Tumor Microenvironment in B-Cell Malignancies. Int J Mol Sci 2020; 21:ijms21093387. [PMID: 32403283 PMCID: PMC7246984 DOI: 10.3390/ijms21093387] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/22/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
An increased focus is being placed on the tumorigenesis and contexture of tumor microenvironment in hematopoietic and solid tumors. Despite recent clinical revolutions in adoptive T-cell transfer approaches and immune checkpoint blockade, tumor microenvironment is a major obstacle to tumor regression in B-cell malignancies. A transcriptional alteration of coding and non-coding RNAs, such as microRNAs (miRNAs), has been widely demonstrated in the tumor microenvironment of B-cell malignancies. MiRNAs have been associated with different clinical-biological forms of B-cell malignancies and involved in the regulation of B lymphocyte development, maturation, and function, including B-cell activation and malignant transformation. Additionally, tumor-secreted extracellular vesicles regulate recipient cell functions in the tumor microenvironment to facilitate metastasis and progression by delivering miRNA contents to neighboring cells. Herein, we focus on the interplay between miRNAs and tumor microenvironment components in the different B-cell malignancies and its impact on diagnosis, proliferation, and involvement in treatment resistance.
Collapse
|
13
|
Zinger A, Koren L, Adir O, Poley M, Alyan M, Yaari Z, Noor N, Krinsky N, Simon A, Gibori H, Krayem M, Mumblat Y, Kasten S, Ofir S, Fridman E, Milman N, Lübtow MM, Liba L, Shklover J, Shainsky-Roitman J, Binenbaum Y, Hershkovitz D, Gil Z, Dvir T, Luxenhofer R, Satchi-Fainaro R, Schroeder A. Collagenase Nanoparticles Enhance the Penetration of Drugs into Pancreatic Tumors. ACS NANO 2019; 13:11008-11021. [PMID: 31503443 PMCID: PMC6837877 DOI: 10.1021/acsnano.9b02395] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Overexpressed extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDAC) limits drug penetration into the tumor and is associated with poor prognosis. Here, we demonstrate that a pretreatment based on a proteolytic-enzyme nanoparticle system disassembles the dense PDAC collagen stroma and increases drug penetration into the pancreatic tumor. More specifically, the collagozome, a 100 nm liposome encapsulating collagenase, was rationally designed to protect the collagenase from premature deactivation and prolonged its release rate at the target site. Collagen is the main component of the PDAC stroma, reaching 12.8 ± 2.3% vol in diseased mice pancreases, compared to 1.4 ± 0.4% in healthy mice. Upon intravenous injection of the collagozome, ∼1% of the injected dose reached the pancreas over 8 h, reducing the level of fibrotic tissue to 5.6 ± 0.8%. The collagozome pretreatment allowed increased drug penetration into the pancreas and improved PDAC treatment. PDAC tumors, pretreated with the collagozome followed by paclitaxel micelles, were 87% smaller than tumors pretreated with empty liposomes followed by paclitaxel micelles. Interestingly, degrading the ECM did not increase the number of circulating tumor cells or metastasis. This strategy holds promise for degrading the extracellular stroma in other diseases as well, such as liver fibrosis, enhancing tissue permeability before drug administration.
Collapse
Affiliation(s)
- Assaf Zinger
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Lilach Koren
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Omer Adir
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Maria Poley
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Mohammed Alyan
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Zvi Yaari
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Nadav Noor
- The School for Molecular Cell Biology and Biotechnology and the Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv 6997800, Israel
| | - Nitzan Krinsky
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Assaf Simon
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Hadas Gibori
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997800, Israel
| | - Majd Krayem
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Yelena Mumblat
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Shira Kasten
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Sivan Ofir
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Eran Fridman
- Department of Otolaryngology Head and Neck Surgery, Rambam Healthcare Campus, Technion-Israel Institute of Technology, Haifa 3200000, Israel
| | - Neta Milman
- Department of Otolaryngology Head and Neck Surgery, Rambam Healthcare Campus, Technion-Israel Institute of Technology, Haifa 3200000, Israel
| | - Michael M. Lübtow
- Functional Polymer Materials, Lehrstuhl für Chemische Technologie der Materialsynthese, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg 97070, Germany
| | - Lior Liba
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Jeny Shklover
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Janna Shainsky-Roitman
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Yoav Binenbaum
- Department of Otolaryngology Head and Neck Surgery, Rambam Healthcare Campus, Technion-Israel Institute of Technology, Haifa 3200000, Israel
| | - Dov Hershkovitz
- Department of Pathology, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997800, Israel
| | - Ziv Gil
- Department of Otolaryngology Head and Neck Surgery, Rambam Healthcare Campus, Technion-Israel Institute of Technology, Haifa 3200000, Israel
| | - Tal Dvir
- The School for Molecular Cell Biology and Biotechnology and the Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv 6997800, Israel
| | - Robert Luxenhofer
- Functional Polymer Materials, Lehrstuhl für Chemische Technologie der Materialsynthese, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg 97070, Germany
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997800, Israel
| | - Avi Schroeder
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
- Corresponding author: (AS)
| |
Collapse
|
14
|
Qi D, Wu E. Cancer prognosis: Considering tumor and its microenvironment as a whole. EBioMedicine 2019; 43:28-29. [PMID: 31010748 PMCID: PMC6558022 DOI: 10.1016/j.ebiom.2019.04.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 02/03/2023] Open
Affiliation(s)
- Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 78508, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 78508, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M University College of Medicine, Temple, TX 76504, USA; Department of Pharmaceutical Sciences, Texas A & M University College of Pharmacy, College Station, TX 77843, USA; LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
15
|
Cheng SH, Cheng YJ, Jin ZY, Xue HD. Unresectable pancreatic ductal adenocarcinoma: Role of CT quantitative imaging biomarkers for predicting outcomes of patients treated with chemotherapy. Eur J Radiol 2019; 113:188-197. [PMID: 30927946 DOI: 10.1016/j.ejrad.2019.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/04/2019] [Accepted: 02/10/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The primary aim of this study was to determine if computed tomographic (CT) texture analysis measurements of the tumor are independently associated with progression-free survival (PFS) and overall survival (OS) in patients with unresectable pancreatic ductal adenocarcinoma (PDAC), including both unresectable locally advanced and metastatic PDAC, who were treated with chemotherapy. METHODS After an institutional review board waiver was obtained, contrast material-enhanced CT studies in 41 patients with unresectable PDAC who underwent contrast-enhanced CT before chemotherapy between 2014 and 2017 were analyzed in terms of tumor texture, with quantification of mean gray-level intensity (Mean), entropy, mean of positive pixels (MPP), kurtosis, standard deviation (SD), and skewness for fine to coarse textures (spatial scaling factor (SSF) 0-6, respectively). The association between pretreatment and posttreatment texture parameters, as well as Δ value (difference between posttreatment and pretreatment texture parameters), and survival time was assessed by using Cox proportional hazards models and Kaplan-Meier analysis. RESULTS Findings from the multivariate Cox model indicated that tumor size, tumor SD (HR, 0.942; 95% CI: 0.898, 0.988) and skewness (HR, 0.407; 95% CI: 0.172, 0.962) measurements with SSF = 3, and tumor SD (HR, 0.958; 95% CI: 0.92, 0.997) measurements with SSF = 4 were significantly and independently associated with PFS, while tumor size and tumor SD (HR, 0.928; 95% CI: 0.882, 0.976) measurements with SSF = 3 were significantly and independently associated with OS. None of the post-therapy texture parameters or Δ value had a significant association with OS or PFS in multivariate Cox regression models. Medium SD (SSF = 3) of more than 38.38 and coarse SD (SSF = 4) of more than 40.67 were associated with longer PFS after chemotherapy (for SSF = 3, median PFS was 10.0 vs 6.0 months [P = 0.024], and for SSF = 4, median PFS was 12.0 vs 6.0 months [P = 0.003]). SD of 38.38 or greater (SSF = 3) as a dichotomized variable was a significant positive prognostic factor for OS (median OS, 20.0 vs 9.0 months [P = 0.04]). Survival models that included a combination of pretreatment SD (SSF = 3) with tumor size, had the potential to perform better than SD alone, while having no statistical significance in this study (area under the ROC curve, 0.756 vs 0.715 [P = 0.066]). CONCLUSIONS Pretreatment CT quantitative imaging biomarkers from texture analysis are associated with PFS and OS in patients with unresectable PDAC who were treated with chemotherapy, and the combination of pretreatment texture parameters and tumor size have the potential to perform better in survival models than imaging biomarker alone.
Collapse
Affiliation(s)
- Si-Hang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yue-Juan Cheng
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zheng-Yu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Hua-Dan Xue
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
16
|
Costanza B, Rademaker G, Tiamiou A, De Tullio P, Leenders J, Blomme A, Bellier J, Bianchi E, Turtoi A, Delvenne P, Bellahcène A, Peulen O, Castronovo V. Transforming growth factor beta-induced, an extracellular matrix interacting protein, enhances glycolysis and promotes pancreatic cancer cell migration. Int J Cancer 2019; 145:1570-1584. [PMID: 30834519 DOI: 10.1002/ijc.32247] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/02/2019] [Accepted: 02/25/2019] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a deadly malignancy with no efficient therapy available up-to-date. Glycolysis is the main provider of energetic substrates to sustain cancer dissemination of PDAC. Accordingly, altering the glycolytic pathway is foreseen as a sound approach to trigger pancreatic cancer regression. Here, we show for the first time that high transforming growth factor beta-induced (TGFBI) expression in PDAC patients is associated with a poor outcome. We demonstrate that, although usually secreted by stromal cells, PDAC cells synthesize and secrete TGFBI in quantity correlated with their migratory capacity. Mechanistically, we show that TGFBI activates focal adhesion kinase signaling pathway through its binding to integrin αVβ5, leading to a significant enhancement of glycolysis and to the acquisition of an invasive phenotype. Finally, we show that TGFBI silencing significantly inhibits PDAC tumor development in a chick chorioallantoic membrane assay model. Our study highlights TGFBI as an oncogenic extracellular matrix interacting protein that bears the potential to serve as a target for new anti-PDAC therapeutic strategies.
Collapse
Affiliation(s)
- Brunella Costanza
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Gilles Rademaker
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Assia Tiamiou
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Pascal De Tullio
- Center for Interdisciplinary Research on Medicines, Metabolomics Group, University of Liège, Liège, Belgium
| | - Justine Leenders
- Center for Interdisciplinary Research on Medicines, Metabolomics Group, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Justine Bellier
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Elettra Bianchi
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Laboratory, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Philippe Delvenne
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium.,Laboratory of Experimental Pathology, GIGA Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| |
Collapse
|
17
|
Liu SL, Cao SG, Li Y, Sun B, Chen D, Wang DS, Zhou YB. Pancreatic stellate cells facilitate pancreatic cancer cell viability and invasion. Oncol Lett 2018; 17:2057-2062. [PMID: 30675272 PMCID: PMC6341873 DOI: 10.3892/ol.2018.9816] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/12/2018] [Indexed: 12/26/2022] Open
Abstract
The biological features of pancreatic cancer and the associated hypoxic environment around the cancer cells often lead to resistance to radiotherapy and chemotherapy. The present study was performed in order to explore the effect pancreatic stellate cells (PSCs) have on the proliferation of pancreatic cancer cells. In the present study, PSCs from human pancreatic cancer tissues were isolated, and the PSCs markers α-smooth muscle actin and desmin were overexpressed in the cytoplasm of PSCs. An MTT assay revealed that PSCs promoted the viability of pancreatic cancer cells. However, the viability of pancreatic cancer cells promoted by PSCs was partially blocked by SB525334. Cellular invasion analysis demonstrated that PSCs promoted the invasion ability of pancreatic cancer cells. An apoptosis assay indicated that PSCs decreased the level of apoptosis induced by gemcitabine. In vivo experiments consisting of mice bearing MIA-PaCa-2 and PSCs demonstrated an increase in the rate of tumor growth compared with MIA-PaCA-2 alone, whereas SB525334 may delay the tumor progression induced by PSCs. The present findings indicated that PSCs promoted the viability and invasion of pancreatic cancer cells, and decreased the apoptosis of pancreatic cancer cells induced by gemcitabine.
Collapse
Affiliation(s)
- Shang-Long Liu
- Department of General Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Shou-Gen Cao
- Department of General Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ying Li
- Department of Blood Transfusion, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Bo Sun
- Department of Blood Transfusion, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Dong Chen
- Department of General Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Dong-Sheng Wang
- Department of General Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yan-Bing Zhou
- Department of General Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
18
|
Yun G, Kim YH, Lee YJ, Kim B, Hwang JH, Choi DJ. Tumor heterogeneity of pancreas head cancer assessed by CT texture analysis: association with survival outcomes after curative resection. Sci Rep 2018; 8:7226. [PMID: 29740111 PMCID: PMC5940761 DOI: 10.1038/s41598-018-25627-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 04/25/2018] [Indexed: 12/12/2022] Open
Abstract
The value of image based texture features as a powerful method to predict prognosis and assist clinical management in cancer patients has been established recently. However, texture analysis using histograms and grey-level co-occurrence matrix in pancreas cancer patients has rarely been reported. We aimed to analyze the association of survival outcomes with texture features in pancreas head cancer patients. Eighty-eight pancreas head cancer patients who underwent preoperative CT images followed by curative resection were included. Texture features using different filter values were obtained. The texture features of average, contrast, correlation, and standard deviation with no filter, and fine to medium filter values as well as the presence of nodal metastasis were significantly different between the recurred (n = 70, 79.5%) and non-recurred group (n = 18, 20.5%). In the multivariate Cox regression analysis, lower standard deviation and contrast and higher correlation with lower average value representing homogenous texture were significantly associated with poorer DFS (disease free survival), along with the presence of lymph node metastasis. Texture parameters from routinely performed pre-operative CT images could be used as an independent imaging tool for predicting the prognosis in pancreas head cancer patients who underwent curative resection.
Collapse
Affiliation(s)
- Gabin Yun
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea
| | - Young Hoon Kim
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea.
| | - Yoon Jin Lee
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea
| | - Bohyoung Kim
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea.,Hankuk University of Foreign Studies, Division of Biomedical Engineering, Yongin, 17035, Korea
| | - Jin-Hyeok Hwang
- Seoul National University Bundang Hospital, Department of Internal Medicine, Seongnam, 13620, Korea
| | - Dong Joon Choi
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea
| |
Collapse
|
19
|
Jiang Z, Zhou C, Cheng L, Yan B, Chen K, Chen X, Zong L, Lei J, Duan W, Xu Q, Li X, Wang Z, Ma Q, Ma J. Inhibiting YAP expression suppresses pancreatic cancer progression by disrupting tumor-stromal interactions. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:69. [PMID: 29587800 PMCID: PMC5870346 DOI: 10.1186/s13046-018-0740-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/16/2018] [Indexed: 12/21/2022]
Abstract
Background Hippo/YAP pathway is known to be important for development, growth and organogenesis, and dysregulation of this pathway leads to tumor progression. We and others find that YAP is up-regulated in pancreatic ductal adenocarcinoma (PDAC) and associated with worse prognosis of patients. Activated pancreatic stellate cells (PSCs) forming the components of microenvironment that enhance pancreatic cancer cells (PCs) invasiveness and malignance. However, the role and mechanism of YAP in PDAC tumor-stromal interaction is largely unknown. Methods The expression of YAP in Pancreatic cancer cell lines and PDAC samples was examined by Western blot and IHC. The biological role of YAP on cancer cell proliferation, epithelial-mesenchymal transition (EMT) and invasion were evaluated by MTT, Quantitative real-time PCR analysis, Western blot analysis and invasion assay. The effect of YAP on PSC activation was evaluated by PC-PSC co-culture conditions and xenograft PDAC mouse model. Results Firstly, knockdown of YAP inhibits PDAC cell proliferation and invasion in vitro. In addition, YAP modulates the PC and PSC interaction via reducing the production of connective tissue growth factor (CTGF) from PCs, inhibits paracrine-mediated PSC activation under PC-PSC co-culture conditions and in turn disrupts TGF-β1-mediated tumor-stromal interactions. Lastly, inhibiting YAP expression prevents tumor growth and suppresses desmoplastic reaction in vivo. Conclusions These results demonstrate that YAP contributes to the proliferation and invasion of PC and the activation of PSC via tumor-stromal interactions and that targeting YAP may be a promising therapeutic strategy for PDAC treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-0740-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhengdong Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Liang Cheng
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Bin Yan
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Ke Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Liang Zong
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Qinhong Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Xuqi Li
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| | - Jiguang Ma
- Department of Anesthesiology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| |
Collapse
|
20
|
Zong L, Chen K, Jiang Z, Chen X, Sun L, Ma J, Zhou C, Xu Q, Duan W, Han L, Lei J, Li X, Ma Q, Wang Z. Lipoxin A4 reverses mesenchymal phenotypes to attenuate invasion and metastasis via the inhibition of autocrine TGF-β1 signaling in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:181. [PMID: 29228980 PMCID: PMC5725800 DOI: 10.1186/s13046-017-0655-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022]
Abstract
Background Pancreatic cancer is a lethal disease in part because of its potential for aggressive invasion and metastasis. Lipoxin A4 (LXA4) is one of the metabolites that is derived from arachidonic acid and that is catalyzed by 15-lipoxygenase (15-LOX), and it has recently been reported to exhibit anti-cancer effects. However, the role of LXA4 in pancreatic cancer remains to be elucidated. Methods Pancreatic cell lines were treated with vehicle or LXA4, and the invasive capacity was then assessed by Transwell assays. The expression of epithelial and mesenchymal markers was determined by western blotting and immunofluorescence. Anti-TGF-β1 neutralizing antibody and exogenous recombinant human TGF-β1 (rhTGF-β1) were used to study the effect of LXA4 on the TGF-β signaling. A liver metastasis model was applied to investigate the effect of LXA4 in vivo. The correlation between the Lipoxin effect score (LES) and the clinical-pathological features of pancreatic cancer was also analyzed. Results We found that in patients with pancreatic cancer, low LES was correlated with aggressive metastatic potential. The LXA4 activity, which was mediated by the LXA4 receptor FPRL1, could significantly suppress invasion capacity and mesenchymal phenotypes. The expression and autocrine signaling pathway activity of TGF-β1 were also downregulated by LXA4. In the liver metastasis model in nude mice, the stable analog of LXA4, BML-111, could inhibit the metastasis of pancreatic cancer cells. Conclusion Our results demonstrated that LXA4 could reverse mesenchymal phenotypes, which attenuated invasion and metastasis via the inhibition of autocrine TGF-β1 signaling in pancreatic cancer, which may provide a new strategy to prevent the metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Liang Zong
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.,Department of Emergency, Peking Union Medical College Hospital, 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China
| | - Ke Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Zhengdong Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Liankang Sun
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Jiguang Ma
- Department of Anesthesiology, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Qinhong Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Xuqi Li
- Department of General Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| |
Collapse
|
21
|
Widder M, Lemke K, Kekeç B, Förster T, Grodrian A, Gastrock G. A modified 384-well-device for versatile use in 3D cancer cell (co-)cultivation and screening for investigations of tumor biology in vitro. Eng Life Sci 2017; 18:132-139. [PMID: 29610566 PMCID: PMC5873453 DOI: 10.1002/elsc.201700008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 06/28/2017] [Accepted: 10/10/2017] [Indexed: 01/12/2023] Open
Abstract
Pancreatic cancer exhibits a worst prognosis owed to an aggressive tumor progression i.a. driven by chemoresistance or tumor‐stroma‐interactions. The identification of candidate genes, which promote this progression, can lead to new therapeutic targets and might improve patient's outcome. The identification of these candidates in a plethora of genes requires suitable screening protocols. The aim of the present study was to establish a universally usable device which ensures versatile cultivation, screening and handling protocols of cancer cells with the 3D spheroid model, an approved model to study tumor biology. By surface modification and alternative handling of a commercial 384‐well plate, a modified device enabling (i) 3D cultivation either by liquid overlay or by a modified hanging drop method for (ii) screening of substances as well as for tumor‐stroma‐interactions (iii) either with manual or automated handling was established. The here presented preliminary results of cell line dependent dose‐response‐relations and a stromal‐induced spheroid‐formation of the pancreatic cancer cells demonstrate the proof‐of‐principle of the versatile functionality of this device. By adapting the protocols to automation, a higher reproducibility and the ability for high‐throughput analyses were ensured.
Collapse
Affiliation(s)
- Miriam Widder
- Department of Bioprocess Engineering Institute for Bioprocessing and Analytical Measurement Techniques e.V. Rosenhof Heilbad Heiligenstadt Germany
| | - Karen Lemke
- Department of Bioprocess Engineering Institute for Bioprocessing and Analytical Measurement Techniques e.V. Rosenhof Heilbad Heiligenstadt Germany
| | - Bünyamin Kekeç
- Department of Bioprocess Engineering Institute for Bioprocessing and Analytical Measurement Techniques e.V. Rosenhof Heilbad Heiligenstadt Germany
| | - Tobias Förster
- Department of Bioprocess Engineering Institute for Bioprocessing and Analytical Measurement Techniques e.V. Rosenhof Heilbad Heiligenstadt Germany
| | - Andreas Grodrian
- Department of Bioprocess Engineering Institute for Bioprocessing and Analytical Measurement Techniques e.V. Rosenhof Heilbad Heiligenstadt Germany
| | - Gunter Gastrock
- Department of Bioprocess Engineering Institute for Bioprocessing and Analytical Measurement Techniques e.V. Rosenhof Heilbad Heiligenstadt Germany
| |
Collapse
|
22
|
Tang D, Zhang J, Yuan Z, Zhang H, Chong Y, Huang Y, Wang J, Xiong Q, Wang S, Wu Q, Tian Y, Lu Y, Ge X, Shen W, Wang D. PSC-derived Galectin-1 inducing epithelial-mesenchymal transition of pancreatic ductal adenocarcinoma cells by activating the NF-κB pathway. Oncotarget 2017; 8:86488-86502. [PMID: 29156810 PMCID: PMC5689700 DOI: 10.18632/oncotarget.21212] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023] Open
Abstract
Galectin-1 has previously been shown to be strongly expressed in activated pancreatic stellate cells (PSCs) and promote the development and metastasis of pancreatic ductal adenocarcinoma (PDAC). However, the molecular mechanisms by which Galectin-1 promotes the malignant behavior of pancreatic cancer cells remain unclear. In this study, we examined the effects of Galectin-1 knockdown or overexpression in PSCs co-cultured with pancreatic cancer (PANC-1) cells. Immunohistochemical analysis showed expression of epithelial-mesenchymal transition (EMT) markers and MMP9 were positively associated with the expression of Galectin-1 in 66 human PDAC tissues. In addition, our in vitro studies showed PSC-derived Galectin-1 promoted the proliferation, invasion, and survival (anti-apoptotic effects) of PANC-1 cells. We also showed PSC-derived Galectin-1 induced EMT of PANC-1 cells and activated the NF-кB pathway in vitro. Our mixed (PSCs and PANC-1 cells) mouse orthotopic xenograft model indicated that overexpression of Galectin-1 in PSCs significantly promoted the proliferation, growth, invasion, and liver metastasis of the transplanted tumor. Moreover, Galectin-1 overexpression in PSCs was strongly associated with increased expression of EMT markers in both the orthotopic xenograft tumor in the pancreas and in metastatic lesions of naked mice. We conclude that PSC-derived Galectin-1 promotes the malignant behavior of PDAC by inducing EMT via activation of the NF-κB pathway. Our results suggest that targeting Galectin-1 in PSCs could represent a promising therapeutic strategy for PDAC progression and metastasis.
Collapse
Affiliation(s)
- Dong Tang
- 1 Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Jingqiu Zhang
- 1 Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Zhongxu Yuan
- 2 Department of General Surgery, Anhui No. 2 Provincial People’s Hospital, Hefei, Anhui Province, P.R. China
| | - Hongpeng Zhang
- 1 Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Yang Chong
- 1 Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Yuqin Huang
- 1 Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Jie Wang
- 1 Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Qingquan Xiong
- 1 Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Sen Wang
- 3 Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Qi Wu
- 4 Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Ying Tian
- 4 Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Yongdie Lu
- 4 Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Xiao Ge
- 4 Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Wenjing Shen
- 4 Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Daorong Wang
- 1 Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| |
Collapse
|
23
|
Aguilera KY, Huang H, Du W, Hagopian MM, Wang Z, Hinz S, Hwang TH, Wang H, Fleming JB, Castrillon DH, Ren X, Ding K, Brekken RA. Inhibition of Discoidin Domain Receptor 1 Reduces Collagen-mediated Tumorigenicity in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2017; 16:2473-2485. [PMID: 28864681 DOI: 10.1158/1535-7163.mct-16-0834] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/04/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022]
Abstract
The extracellular matrix (ECM), a principal component of pancreatic ductal adenocarcinoma (PDA), is rich in fibrillar collagens that facilitate tumor cell survival and chemoresistance. Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase that specifically binds fibrillar collagens and has been implicated in promoting cell proliferation, migration, adhesion, ECM remodeling, and response to growth factors. We found that collagen-induced activation of DDR1 stimulated protumorigenic signaling through protein tyrosine kinase 2 (PYK2) and pseudopodium-enriched atypical kinase 1 (PEAK1) in pancreatic cancer cells. Pharmacologic inhibition of DDR1 with an ATP-competitive orally available small-molecule kinase inhibitor (7rh) abrogated collagen-induced DDR1 signaling in pancreatic tumor cells and consequently reduced colony formation and migration. Furthermore, the inhibition of DDR1 with 7rh showed striking efficacy in combination with chemotherapy in orthotopic xenografts and autochthonous pancreatic tumors where it significantly reduced DDR1 activation and downstream signaling, reduced primary tumor burden, and improved chemoresponse. These data demonstrate that targeting collagen signaling in conjunction with conventional cytotoxic chemotherapy has the potential to improve outcome for pancreatic cancer patients. Mol Cancer Ther; 16(11); 2473-85. ©2017 AACR.
Collapse
Affiliation(s)
- Kristina Y Aguilera
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Huocong Huang
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Wenting Du
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Moriah M Hagopian
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Zhen Wang
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Stefan Hinz
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Tae Hyun Hwang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Huamin Wang
- Department of Pathology, UT MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Surgical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Diego H Castrillon
- Department of Clinical Science, UT Southwestern Medical Center, Dallas, Texas
| | - Xiaomei Ren
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Ke Ding
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Rolf A Brekken
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas. .,Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
24
|
Giordano G, Pancione M, Olivieri N, Parcesepe P, Velocci M, Di Raimo T, Coppola L, Toffoli G, D’Andrea MR. Nano albumin bound-paclitaxel in pancreatic cancer: Current evidences and future directions. World J Gastroenterol 2017; 23:5875-5886. [PMID: 28932079 PMCID: PMC5583572 DOI: 10.3748/wjg.v23.i32.5875] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/03/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PDAC) is an aggressive and chemoresistant disease, representing the fourth cause of cancer related deaths in western countries. Majority of patients have unresectable, locally advanced or metastatic disease at time of diagnosis and the 5-year survival rate in these conditions is extremely low. For more than a decade gemcitabine has been the cornerstone of metastatic PDAC treatment, although survival benefit was very poor. PDAC cells are surrounded by an intense desmoplastic reaction that may create a barrier to the drugs penetration within the tumor. Recently PDAC stroma has been addressed as a potential therapeutic target. Nano albumin bound (Nab)-paclitaxel is an innovative molecule depleting tumor stroma, through interaction between albumin and secreted protein acidic and rich in cysteine. Addition of nab-paclitaxel to gemcitabine has showed activity and efficacy in metastatic PDAC first-line treatment improving survival and overall response rate vs gemcitabine alone in the MPACT phase III study. This combination represents one of the standards of care in advanced PDAC therapy and is suitable to a broader spectrum of patients compared to other schedules. Nab-paclitaxel is under investigation as a backbone of chemotherapy in novel combinations with target agents or immunotherapy in locally advanced or metastatic PDAC. In this article, we provide an updated and critical overview about the role of nab-paclitaxel in PDAC treatment based on the latest advances in preclinical and clinical research. Furthermore, we focus on the use of nab-paclitaxel within the context of metastatic PDAC treatment landscape and we discuss about future implications in the light of current clinical ongoing trials.
Collapse
Affiliation(s)
- Guido Giordano
- Medical Oncology Unit, San Filippo Neri Hospital, 00135 Roma, Italy
- CRO Aviano National Cancer Institute, 33081 Aviano, Italy
| | - Massimo Pancione
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University, 28040 Madrid, Spain
| | - Nunzio Olivieri
- Department of Biology, University of Naples, Federico II, Via Mezzocannone, 80134 Napoli, Italy
| | - Pietro Parcesepe
- Department of Pathology and Diagnostics, University of Verona Strada, 37134 Verona, Italy
| | - Marianna Velocci
- Medical Oncology Unit, San Filippo Neri Hospital, 00135 Roma, Italy
| | - Tania Di Raimo
- Medical Oncology Unit, San Filippo Neri Hospital, 00135 Roma, Italy
| | - Luigi Coppola
- Anatomic Pathology Unit, San Filippo Neri, 00135 Roma, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO-National Cancer Institute Via F, 33081 Aviano (Pordenone), Italy
| | | |
Collapse
|
25
|
Kwegyir-Afful AK, Murigi FN, Purushottamachar P, Ramamurthy VP, Martin MS, Njar VCO. Galeterone and its analogs inhibit Mnk-eIF4E axis, synergize with gemcitabine, impede pancreatic cancer cell migration, invasion and proliferation and inhibit tumor growth in mice. Oncotarget 2016; 8:52381-52402. [PMID: 28881737 PMCID: PMC5581036 DOI: 10.18632/oncotarget.14154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/19/2016] [Indexed: 12/22/2022] Open
Abstract
Survival rate for pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) is poor, with about 80% of patients presenting with the metastatic disease. Gemcitabine, the standard chemotherapeutic agent for locally advanced and metastatic PDAC has limited efficacy, attributed to innate/acquired resistance and activation of pro-survival pathways. The Mnk1/2-eIF4E and NF-κB signaling pathways are implicated in PDAC disease progression/metastasis and also associated with gemcitabine-induced resistance in PDAC. Galeterone (gal), a multi-target, agent in phase III clinical development for prostate cancer has also shown effects on the aforementioned pathways. We show for the first time, that gal/analogs (VNPT55, VNPP414 and VNPP433-3β) profoundly inhibited cell viability of gemcitabine-naive/resistance PDAC cell lines and strongly synergized with gemcitabine in gemcitabine-resistant PDAC cells. In addition, to inducing G1 cell cycle arrest, gal/analogs induced caspase 3-mediated cell-death of PDAC cells. Gal/analogs caused profound downregulation of Mnk1/2, peIF4E and NF-κB (p-p65), metastatic inducing factors (N-cadherin, MMP-1/-2/-9, Slug, Snail and CXCR4) and putative stem cell factors, (β-Catenin, Nanog, BMI-1 and Oct-4). Gal/analog also depleted EZH2 and upregulated E-Cadherin. These effects resulted in significant inhibition of PDAC cell migration, invasion and proliferation. Importantly, we also observed strong MiaPaca-2 tumor xenograft growth inhibition (61% to 92%). Collectively, these promising findings strongly support further development of gal/analogs as novel therapeutics for PDAC.
Collapse
Affiliation(s)
- Andrew K Kwegyir-Afful
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Francis N Murigi
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Vidya P Ramamurthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Marlena S Martin
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Current Address: Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, VA 20147, USA
| | - Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| |
Collapse
|
26
|
Calle AS, Nair N, Oo AK, Prieto-Vila M, Koga M, Khayrani AC, Hussein M, Hurley L, Vaidyanath A, Seno A, Iwasaki Y, Calle M, Kasai T, Seno M. A new PDAC mouse model originated from iPSCs-converted pancreatic cancer stem cells (CSCcm). Am J Cancer Res 2016; 6:2799-2815. [PMID: 28042501 PMCID: PMC5199755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/06/2016] [Indexed: 06/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most representative form of pancreatic cancers. PDAC solid tumours are constituted of heterogeneous populations of cells including cancer stem cells (CSCs), differentiated cancer cells, desmoplastic stroma and immune cells. The identification and consequent isolation of pancreatic CSCs facilitated the generation of genetically engineered murine models. Nonetheless, the current models may not be representative for the spontaneous tumour occurrence. In the present study, we show the generation of a novel pancreatic iPSC-converted cancer stem cell lines (CSCcm) as a cutting-edge model for the study of PDAC. The CSCcm lines were achieved only by the influence of pancreatic cancer cell lines conditioned medium and were not subjected to any genetic manipulation. The xenografts tumours from CSCcm lines displayed histopathological features of ADM, PanIN and PDAC lesions. Further molecular characterization from RNA-sequencing analysis highlighted primary culture cell lines (1st CSCcm) as potential candidates to represent the pancreatic CSCs and indicated the establishment of the pancreatic cancer molecular pattern in their subsequent progenies 2nd CSCcm and 3rd CSCcm. In addition, preliminary RNA-seq SNPs analysis showed that the distinct CSCcm lines did not harbour single point mutations for the oncogene Kras codon 12 or 13. Therefore, PDAC-CSCcm model may provide new insights about the actual occurrence of the pancreatic cancer leading to develop different approaches to target CSCs and abrogate the progression of this fatidic disease.
Collapse
Affiliation(s)
- Anna Sanchez Calle
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Neha Nair
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Aung KoKo Oo
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Marta Prieto-Vila
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Megumi Koga
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Apriliana Cahya Khayrani
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Maram Hussein
- Department of Chemistry, Faculty of Science, Menoufia UniversityShebin El-Koam 32511, Egypt
| | - Laura Hurley
- Cancer Biology Graduate Program, School of Medicine, Wayne State University10 E Warren, Avenue, Suite 2215, Detroit, Michigan 48201, USA
| | - Arun Vaidyanath
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Akimasa Seno
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Yoshiaki Iwasaki
- Department of Gastroenterology and Hepatology, Okayama University, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama 700-8558, Japan
| | - Malu Calle
- Department of Systems Biology, University of VicVic, Barcelona 08500, Spain
| | - Tomonari Kasai
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Masaharu Seno
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
27
|
Komura D, Isagawa T, Kishi K, Suzuki R, Sato R, Tanaka M, Katoh H, Yamamoto S, Tatsuno K, Fukayama M, Aburatani H, Ishikawa S. CASTIN: a system for comprehensive analysis of cancer-stromal interactome. BMC Genomics 2016; 17:899. [PMID: 27829362 PMCID: PMC5103609 DOI: 10.1186/s12864-016-3207-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 10/25/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Cancer microenvironment plays a vital role in cancer development and progression, and cancer-stromal interactions have been recognized as important targets for cancer therapy. However, identifying relevant and druggable cancer-stromal interactions is challenging due to the lack of quantitative methods to analyze whole cancer-stromal interactome. RESULTS We present CASTIN (CAncer-STromal INteractome analysis), a novel framework for the evaluation of cancer-stromal interactome from RNA-Seq data using cancer xenograft models. For each ligand-receptor interaction which is derived from curated protein-protein interaction database, CASTIN summarizes gene expression profiles of cancer and stroma into three evaluation indices. These indices provide quantitative evaluation and comprehensive visualization of interactome, and thus enable to identify critical cancer-microenvironment interactions, which would be potential drug targets. We applied CASTIN to the dataset of pancreas ductal adenocarcinoma, and successfully characterized the individual cancer in terms of cancer-stromal relationships, and identified both well-known and less-characterized druggable interactions. CONCLUSIONS CASTIN provides comprehensive view of cancer-stromal interactome and is useful to identify critical interactions which may serve as potential drug targets in cancer-microenvironment. CASTIN is available at: http://github.com/tmd-gpat/CASTIN .
Collapse
Affiliation(s)
- Daisuke Komura
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takayuki Isagawa
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuki Kishi
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.,Graduate School of Interdisciplinary Information Studies, The University of Tokyo, Tokyo, Japan
| | - Ryohei Suzuki
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.,Graduate School of Information and Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Reiko Sato
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mariko Tanaka
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroto Katoh
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shogo Yamamoto
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kenji Tatsuno
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
28
|
Wang T, Notta F, Navab R, Joseph J, Ibrahimov E, Xu J, Zhu CQ, Borgida A, Gallinger S, Tsao MS. Senescent Carcinoma-Associated Fibroblasts Upregulate IL8 to Enhance Prometastatic Phenotypes. Mol Cancer Res 2016; 15:3-14. [PMID: 27678171 DOI: 10.1158/1541-7786.mcr-16-0192] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/30/2016] [Accepted: 09/14/2016] [Indexed: 11/16/2022]
Abstract
Carcinoma-associated fibroblasts (CAF) represent a significant component of pancreatic cancer stroma and are biologically implicated in tumor progression. However, evidence of both cancer-promoting and -restraining properties amongst CAFs suggests the possibility of multiple phenotypic subtypes. Here, it is demonstrated that senescent CAFs promote pancreatic cancer invasion and metastasis compared with nonsenescent control CAFs using in vitro Transwell invasion models and in vivo xenograft mouse models. Screening by gene expression microarray and cytokine ELISA assays revealed IL8 to be upregulated in senescent CAFs. Experimental modulation through IL8 overexpression or receptor inhibition implicates the IL8 pathway as a mediator of the proinvasive effects of senescent CAFs. In a cohort of human pancreatic cancer cases, more abundant stromal senescence as indicated by p16 immunohistochemistry correlated with decreased survival in patients with early-stage disease. These data support senescent fibroblasts as a pathologically and clinically relevant feature of pancreatic cancer. The inhibition of senescent stroma-cancer signaling pathways has the potential to restrain pancreatic cancer progression. IMPLICATIONS Findings show that senescent cancer-associated fibroblasts secret excess IL8 to promote pancreatic cancer invasion and metastasis; thus, senescent CAFs represent a phenotypic subtype, challenging conventional assumptions that CAFs are a homogeneous population. Mol Cancer Res; 15(1); 3-14. ©2016 AACR.
Collapse
Affiliation(s)
- Tao Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Pathology, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Faiyaz Notta
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Roya Navab
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Joella Joseph
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Emin Ibrahimov
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Ontario Institute for Cancer Research, Toronto, Canada
| | - Jing Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Chang-Qi Zhu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Ayelet Borgida
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Canada
| | - Steven Gallinger
- Department of General Surgery, University Health Network, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada. .,Department of Pathology, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
29
|
Yoshida M, Miyasaka Y, Ohuchida K, Okumura T, Zheng B, Torata N, Fujita H, Nabae T, Manabe T, Shimamoto M, Ohtsuka T, Mizumoto K, Nakamura M. Calpain inhibitor calpeptin suppresses pancreatic cancer by disrupting cancer-stromal interactions in a mouse xenograft model. Cancer Sci 2016; 107:1443-1452. [PMID: 27487486 PMCID: PMC5084662 DOI: 10.1111/cas.13024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 12/29/2022] Open
Abstract
Desmoplasia contributes to the aggressive behavior of pancreatic cancer. However, recent clinical trials testing several antifibrotic agents on pancreatic cancer have not shown clear efficacy. Therefore, further investigation of desmoplasia‐targeting antifibrotic agents by another mechanism is needed. Calpeptin, an inhibitor of calpains, suppressed fibroblast function and inhibited fibrosis. In this study, we investigated the anticancer effects of calpeptin on pancreatic cancer. We investigated whether calpeptin inhibited tumor progression using a mouse xenograft model. We used quantitative RT‐PCR to evaluate the expression of calpain‐1 and calpain‐2 mRNA in pancreatic cancer cells (PCCs) and pancreatic stellate cells (PSCs). We also undertook functional assays, including proliferation, migration, and invasion, to evaluate the inhibitory effects of calpeptin on PCCs and PSCs. Quantitative RT‐PCR indicated that PCCs and PSCs expressed calpain‐2 mRNA. Calpeptin reduced tumor volume (P = 0.0473) and tumor weight (P = 0.0471) and inhibited the tumor desmoplastic reaction (P < 0.001) in xenograft tumors in nude mice. Calpeptin also inhibited the biologic functions of PCCs and PSCs including proliferation (P = 0.017), migration (P = 0.027), and invasion (P = 0.035) in vitro. Furthermore, calpeptin reduced the migration of PCCs and PSCs by disrupting the cancer–stromal interaction (P = 0.0002). Our findings indicate that calpeptin is a promising antitumor agent for pancreatic cancer, due not only to its suppressive effect on PCCs and PSCs but also its disruption of the cancer–stromal interaction.
Collapse
Affiliation(s)
- Masaki Yoshida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Miyasaka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. .,Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Takashi Okumura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Biao Zheng
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuhiro Torata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hayato Fujita
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshinaga Nabae
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuya Manabe
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Takao Ohtsuka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
30
|
Hirakawa T, Yashiro M, Doi Y, Kinoshita H, Morisaki T, Fukuoka T, Hasegawa T, Kimura K, Amano R, Hirakawa K. Pancreatic Fibroblasts Stimulate the Motility of Pancreatic Cancer Cells through IGF1/IGF1R Signaling under Hypoxia. PLoS One 2016; 11:e0159912. [PMID: 27487118 PMCID: PMC4972430 DOI: 10.1371/journal.pone.0159912] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 07/11/2016] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by its hypovascularity, with an extremely poor prognosis because of its highly invasive nature. PDAC proliferates with abundant stromal cells, suggesting that its invasive activity might be controlled by intercellular interactions between cancer cells and fibroblasts. Using four PDAC cell lines and two pancreas cancer-associated fibroblasts (CAFs), the expression of insulin-like growth factor-1 (IGF1) and IGF1 receptor (IGF1R) was evaluated by RT-PCR, FACScan, western blot, or ELISA. Correlation between IGF1R and the hypoxia marker carbonic anhydrase 9 (CA9) was examined by immunohistochemical staining of 120 pancreatic specimens. The effects of CAFs, IGF1, and IGF1R inhibitors on the motility of cancer cells were examined by wound-healing assay or invasion assay under normoxia (20% O2) and hypoxia (1% O2). IGF1R expression was significantly higher in RWP-1, MiaPaCa-2, and OCUP-AT cells than in Panc-1 cells. Hypoxia increased the expression level of IGF1R in RWP-1, MiaPaCa-2, and OCUP-AT cells. CA9 expression was correlated with IGF1R expression in pancreatic specimens. CAFs produced IGF1 under hypoxia, but PDAC cells did not. A conditioned medium from CAFs, which expressed αSMA, stimulated the migration and invasion ability of MiaPaCa-2, RWP-1, and OCUP-AT cells. The motility of all PDAC cells was greater under hypoxia than under normoxia. The motility-stimulating ability of CAFs was decreased by IGF1R inhibitors. These findings might suggest that pancreas CAFs stimulate the invasion activity of PDAC cells through paracrine IGF1/IGF1R signaling, especially under hypoxia. Therefore the targeting of IGF1R signaling might represent a promising therapeutic approach in IGF1R-dependent PDAC.
Collapse
Affiliation(s)
- Toshiki Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
- * E-mail:
| | - Yosuke Doi
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Haruhito Kinoshita
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tamami Morisaki
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tatsunari Fukuoka
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tsuyoshi Hasegawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kenjiro Kimura
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ryosuke Amano
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
31
|
Vallerand D, Massonnet G, Kébir F, Gentien D, Maciorowski Z, De la Grange P, Sigal-Zafrani B, Richardson M, Humbert S, Thuleau A, Assayag F, de Plater L, Nicolas A, Scholl S, Marangoni E, Weigand S, Roman-Roman S, Savina A, Decaudin D. Characterization of Breast Cancer Preclinical Models Reveals a Specific Pattern of Macrophage Polarization. PLoS One 2016; 11:e0157670. [PMID: 27388901 PMCID: PMC4936680 DOI: 10.1371/journal.pone.0157670] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 06/02/2016] [Indexed: 12/23/2022] Open
Abstract
Drug discovery efforts have focused on the tumor microenvironment in recent years. However, few studies have characterized the stroma component in patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMs). In this study, we characterized the stroma in various models of breast cancer tumors in mice. We performed transcriptomic and flow cytometry analyses on murine populations for a series of 25 PDXs and the two most commonly used GEMs (MMTV-PyMT and MMTV-erBb2). We sorted macrophages from five models. We then profiled gene expression in these cells, which were also subjected to flow cytometry for phenotypic characterization. Hematopoietic cell composition, mostly macrophages and granulocytes, differed between tumors. Macrophages had a specific polarization phenotype related to their M1/M2 classification and associated with the expression of genes involved in the recruitment, invasion and metastasis processes. The heterogeneity of the stroma component of the models studied suggests that tumor cells modify their microenvironment to satisfy their needs. Our observations suggest that such models are of relevance for preclinical studies.
Collapse
Affiliation(s)
- David Vallerand
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
- Institut Roche, Boulogne-Billancourt, France
| | - Gérald Massonnet
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | - Fatima Kébir
- Department of Pathology, Institut Curie, Paris, France
| | - David Gentien
- Platform of Molecular Biology Facilities, Institut Curie, PSL University, Paris, France
| | - Zofia Maciorowski
- Flow Cytometry Core Facility, Institut Curie, PSL University, Paris, France
| | | | - Brigitte Sigal-Zafrani
- Department of Pathology, Institut Curie, Paris, France
- Inserm, U830, Institut Curie, PSL University, Paris, France
| | | | - Sandrine Humbert
- CNRS UMR3306, INSERM U1005, Institut Curie, PSL University, Orsay, France
| | - Aurélie Thuleau
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | - Franck Assayag
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | - Ludmilla de Plater
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | - André Nicolas
- Department of Pathology, Institut Curie, Paris, France
| | - Suzy Scholl
- Department of Medical Oncology, Institut Curie, Institut Curie, Paris, France
| | - Elisabetta Marangoni
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
| | | | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, PSL University, Paris, France
| | | | - Didier Decaudin
- Translational Research Department, Laboratory of Preclinical Investigation, Institut Curie, PSL University, Paris, France
- Department of Medical Oncology, Institut Curie, Institut Curie, Paris, France
- Translational Research Department, Institut Curie, PSL University, Paris, France
- * E-mail:
| |
Collapse
|
32
|
CAF cellular glycolysis: linking cancer cells with the microenvironment. Tumour Biol 2016; 37:8503-14. [PMID: 27075473 DOI: 10.1007/s13277-016-5049-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/01/2016] [Indexed: 02/06/2023] Open
Abstract
Cancers have long being hallmarked as cells relying heavily on their glycolysis for energy generation in spite of having functional mitochondria. The metabolic status of the cancer cells have been revisited time and again to get better insight into the overall carcinogenesis process which revealed the apparent crosstalks between the cancer cells with the fibroblasts present in the tumour microenvironment. This review focuses on the mechanisms of transformations of normal fibroblasts to cancer-associated fibroblasts (CAF), the participation of the CAF in tumour progression with special interest to the role of CAF cellular glycolysis in the overall tumorigenesis. The fibroblasts, when undergoes the transformation process, distinctly switches to a more glycolytic phenotype in order to provide the metabolic intermediates necessary for carrying out the mitochondrial pathways of ATP generation in cancer cells. This review will also discuss the molecular mechanisms responsible for this metabolic make over promoting glycolysis in CAF cells. A thorough investigation of the pathways and molecules involved will not only help in understanding the process of activation and metabolic reprogramming in CAF cells but also might open up new targets for cancer therapy.
Collapse
|
33
|
Walmsley GG, Maan ZN, Hu MS, Atashroo DA, Whittam AJ, Duscher D, Tevlin R, Marecic O, Lorenz HP, Gurtner GC, Longaker MT. Murine Dermal Fibroblast Isolation by FACS. J Vis Exp 2016. [PMID: 26780559 DOI: 10.3791/53430] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Fibroblasts are the principle cell type responsible for secreting extracellular matrix and are a critical component of many organs and tissues. Fibroblast physiology and pathology underlie a spectrum of clinical entities, including fibroses in multiple organs, hypertrophic scarring following burns, loss of cardiac function following ischemia, and the formation of cancer stroma. However, fibroblasts remain a poorly characterized type of cell, largely due to their inherent heterogeneity. Existing methods for the isolation of fibroblasts require time in cell culture that profoundly influences cell phenotype and behavior. Consequently, many studies investigating fibroblast biology rely upon in vitro manipulation and do not accurately capture fibroblast behavior in vivo. To overcome this problem, we developed a FACS-based protocol for the isolation of fibroblasts from the dorsal skin of adult mice that does not require cell culture, thereby preserving the physiologic transcriptional and proteomic profile of each cell. Our strategy allows for exclusion of non-mesenchymal lineages via a lineage negative gate (Lin(-)) rather than a positive selection strategy to avoid pre-selection or enrichment of a subpopulation of fibroblasts expressing specific surface markers and be as inclusive as possible across this heterogeneous cell type.
Collapse
Affiliation(s)
- Graham G Walmsley
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Zeshaan N Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine
| | - Michael S Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine; Department of Surgery, John A. Burns School of Medicine, University of Hawai'i
| | - David A Atashroo
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine
| | - Alexander J Whittam
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine
| | - Dominik Duscher
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine
| | - Ruth Tevlin
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine
| | - Owen Marecic
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine
| | - H Peter Lorenz
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine
| | - Geoffrey C Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine;
| |
Collapse
|
34
|
Boonstra MC, Prakash J, Van De Velde CJH, Mesker WE, Kuppen PJK, Vahrmeijer AL, Sier CFM. Stromal Targets for Fluorescent-Guided Oncologic Surgery. Front Oncol 2015; 5:254. [PMID: 26636036 PMCID: PMC4653299 DOI: 10.3389/fonc.2015.00254] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/05/2015] [Indexed: 12/17/2022] Open
Abstract
Pre-operative imaging techniques are essential for tumor detection and diagnosis, but offer limited help during surgery. Recently, the applicability of imaging during oncologic surgery has been recognized, using near-infrared fluorescent dyes conjugated to targeting antibodies, peptides, or other vehicles. Image-guided oncologic surgery (IGOS) assists the surgeFon to distinguish tumor from normal tissue during operation, and can aid in recognizing vital structures. IGOS relies on an optimized combination of a dedicated fluorescent camera system and specific probes for targeting. IGOS probes for clinical use are not widely available yet, but numerous pre-clinical studies have been published and clinical trials are being established or prepared. Most of the investigated probes are based on antibodies or peptides against proteins on the membranes of malignant cells, whereas others are directed against stromal cells. Targeting stroma cells for IGOS has several advantages. Besides the high stromal content in more aggressive tumor types, the stroma is often primarily located at the periphery/invasive front of the tumor, which makes stromal targets particularly suited for imaging purposes. Moreover, because stroma up-regulation is a physiological reaction, most proteins to be targeted on these cells are “universal” and not derived from a specific genetic variation, as is the case with many upregulated proteins on malignant cancer cells.
Collapse
Affiliation(s)
- Martin C Boonstra
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | - Jai Prakash
- Department of Biomaterial Science and Technology, Targeted Therapeutics, University of Twente , Enschede , Netherlands
| | | | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands ; Antibodies for Research Applications BV , Gouda , Netherlands
| |
Collapse
|
35
|
Prognosis Relevance of Serum Cytokines in Pancreatic Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:518284. [PMID: 26346854 PMCID: PMC4539422 DOI: 10.1155/2015/518284] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/05/2015] [Accepted: 01/12/2015] [Indexed: 02/06/2023]
Abstract
The overall survival of patients with pancreatic ductal adenocarcinoma is extremely low. Although gemcitabine is the standard used chemotherapy for this disease, clinical outcomes do not reflect significant improvements, not even when combined with adjuvant treatments. There is an urgent need for prognosis markers to be found. The aim of this study was to analyze the potential value of serum cytokines to find a profile that can predict the clinical outcome in patients with pancreatic cancer and to establish a practical prognosis index that significantly predicts patients' outcomes. We have conducted an extensive analysis of serum prognosis biomarkers using an antibody array comprising 507 human cytokines. Overall survival was estimated using the Kaplan-Meier method. Univariate and multivariate Cox's proportional hazard models were used to analyze prognosis factors. To determine the extent that survival could be predicted based on this index, we used the leave-one-out cross-validation model. The multivariate model showed a better performance and it could represent a novel panel of serum cytokines that correlates to poor prognosis in pancreatic cancer. B7-1/CD80, EG-VEGF/PK1, IL-29, NRG1-beta1/HRG1-beta1, and PD-ECGF expressions portend a poor prognosis for patients with pancreatic cancer and these cytokines could represent novel therapeutic targets for this disease.
Collapse
|
36
|
Valcz G, Sipos F, Tulassay Z, Molnar B, Yagi Y. Republished: Importance of carcinoma-associated fibroblast-derived proteins in clinical oncology. Postgrad Med J 2015; 91:1026-31. [PMID: 25976496 DOI: 10.1136/postgradmedj-2014-202561rep] [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] [Indexed: 02/05/2023]
Abstract
Carcinoma-associated fibroblast (CAF) as prominent cell type of the tumour microenvironment has complex interaction with both the cancer cells and other non-neoplastic surrounding cells. The CAF-derived regulators and extracellular matrix proteins can support cancer progression by providing a protective microenvironment for the cancer cells via reduction of chemotherapy sensitivity. On the other hand, these proteins may act as powerful prognostic markers as well as potential targets of anticancer therapy. In this review, we summarise the clinical importance of the major CAF-derived signals influencing tumour behaviour and determining the outcome of chemotherapy.
Collapse
Affiliation(s)
- Gabor Valcz
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ferenc Sipos
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Bela Molnar
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Yukako Yagi
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Teague A, Lim KH, Wang-Gillam A. Advanced pancreatic adenocarcinoma: a review of current treatment strategies and developing therapies. Ther Adv Med Oncol 2015; 7:68-84. [PMID: 25755680 DOI: 10.1177/1758834014564775] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pancreatic adenocarcinoma is one of the deadliest solid malignancies. A large proportion of patients are diagnosed with locally advanced or metastatic disease at the time of presentation and, unfortunately, this severely limits the number of patients who can undergo surgical resection, which offers the only chance for cure. Recent therapeutic advances for patients with advanced pancreatic cancer have extended overall survival, but prognosis still remains grim. Given that traditional chemotherapy is ineffective in curing advanced pancreatic adenocarcinoma, current research is taking a multidirectional approach in the hopes of developing more effective treatments. This article reviews the major clinical trial data that is the basis for the current chemotherapy regimens used as first- and second-line treatments for advanced pancreatic adenocarcinoma. We also review the current ongoing clinical trials, which include the use of agents targeting the oncogenic network signaling of K-Ras, agents targeting the extracellular matrix, and immune therapies.
Collapse
Affiliation(s)
- Andrea Teague
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Medicine, Campus Box 8056, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
38
|
Xu Q, Wang Z, Chen X, Duan W, Lei J, Zong L, Li X, Sheng L, Ma J, Han L, Li W, Zhang L, Guo K, Ma Z, Wu Z, Wu E, Ma Q. Stromal-derived factor-1α/CXCL12-CXCR4 chemotactic pathway promotes perineural invasion in pancreatic cancer. Oncotarget 2015; 6:4717-32. [PMID: 25605248 PMCID: PMC4467110 DOI: 10.18632/oncotarget.3069] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/17/2014] [Indexed: 12/22/2022] Open
Abstract
Perineural invasion (PNI) is considered as an alternative route for the metastatic spread of pancreatic cancer cells; however, the molecular changes leading to PNI are still poorly understood. In this study, we show that the CXCL12/CXCR4 axis plays a pivotal role in the neurotropism of pancreatic cancer cells to local peripheral nerves. Immunohistochemical staining results revealed that CXCR4 elevation correlated with PNI in 78 pancreatic cancer samples. Both in vitro and in vivo PNI models were applied to investigate the function of the CXCL12/CXCR4 signaling in PNI progression and pathogenesis. The results showed that the activation of the CXCL12/CXCR4 axis significantly increased pancreatic cancer cells invasion and promoted the outgrowth of the dorsal root ganglia. CXCL12 derived from the peripheral nerves stimulated the invasion and chemotactic migration of CXCR4-positive cancer cells in a paracrine manner, eventually leading to PNI. In vivo analyses revealed that the abrogation of the activated signaling inhibited tumor growth and invasion of the sciatic nerve toward the spinal cord. These data indicate that the CXCL12/CXCR4 axis may be a novel therapeutic target to prevent the perineural dissemination of pancreatic cancer.
Collapse
Affiliation(s)
- Qinhong Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Liang Zong
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Xuqi Li
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Liang Sheng
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Jiguang Ma
- Department of Oncology, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Wei Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Lun Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Kun Guo
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhenhua Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| |
Collapse
|
39
|
Walmsley GG, Rinkevich Y, Hu MS, Montoro DT, Lo DD, McArdle A, Maan ZN, Morrison SD, Duscher D, Whittam AJ, Wong VW, Weissman IL, Gurtner GC, Longaker MT. Live fibroblast harvest reveals surface marker shift in vitro. Tissue Eng Part C Methods 2014; 21:314-21. [PMID: 25275778 DOI: 10.1089/ten.tec.2014.0118] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Current methods for the isolation of fibroblasts require extended ex vivo manipulation in cell culture. As a consequence, prior studies investigating fibroblast biology may fail to adequately represent cellular phenotypes in vivo. To overcome this problem, we describe a detailed protocol for the isolation of fibroblasts from the dorsal dermis of adult mice that bypasses the need for cell culture, thereby preserving the physiological, transcriptional, and proteomic profiles of each cell. Using the described protocol we characterized the transcriptional programs and the surface expression of 176 CD markers in cultured versus uncultured fibroblasts. The differential expression patterns we observed highlight the importance of a live harvest for investigations of fibroblast biology.
Collapse
Affiliation(s)
- Graham G Walmsley
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine , Stanford, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Valcz G, Sipos F, Tulassay Z, Molnar B, Yagi Y. Importance of carcinoma-associated fibroblast-derived proteins in clinical oncology. J Clin Pathol 2014; 67:1026-31. [PMID: 25135950 DOI: 10.1136/jclinpath-2014-202561] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Carcinoma-associated fibroblast (CAF) as prominent cell type of the tumour microenvironment has complex interaction with both the cancer cells and other non-neoplastic surrounding cells. The CAF-derived regulators and extracellular matrix proteins can support cancer progression by providing a protective microenvironment for the cancer cells via reduction of chemotherapy sensitivity. On the other hand, these proteins may act as powerful prognostic markers as well as potential targets of anticancer therapy. In this review, we summarise the clinical importance of the major CAF-derived signals influencing tumour behaviour and determining the outcome of chemotherapy.
Collapse
Affiliation(s)
- Gabor Valcz
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ferenc Sipos
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Bela Molnar
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Yukako Yagi
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
41
|
Wang Z, Li J, Chen X, Duan W, Ma Q, Li X. Disrupting the balance between tumor epithelia and stroma is a possible therapeutic approach for pancreatic cancer. Med Sci Monit 2014; 20:2002-6. [PMID: 25327552 PMCID: PMC4211416 DOI: 10.12659/msm.892523] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a type of highly lethal malignant tumor. PDAC is locally invasive and is surrounded by a dense desmoplasia or fibrosis, which can involve adjacent vital structures. Previously, the effect of pancreatic stellate cells (PSCs) of stroma in the progression of PDAC has received more attention, and most in vitro and in vivo studies revealed that PSCs appear to confer biological aggressiveness. However, clinical trials targeting desmoplasia or PSCs showed disappointing results. Recent studies found that stromal components, especially activated PSCs, are able to inhibit the occurrence and progression of PDAC. Inhibition of the stroma or desmoplasia through genetic regulations or drugs accelerates the formation and progression of PDAC. Thus, we hypothesized that in various times and spaces, there is a balance between the tumor epithelia and stroma; once the balance is upset, the tumor traits may undergo certain changes. Therefore, finding the key changing points of this relationship to corrupt or influence it, instead of blindly inhibiting the stroma motivation or simply maintaining stroma activation, will destroy the cooperation or promote the competition and antagonism among cells. This approach may render tumors more vulnerable and thus unable to resist anti-cancer therapies.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Jiahui Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Xin Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Xuqi Li
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
42
|
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: 112] [Impact Index Per Article: 11.2] [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
|
43
|
Al-Batran SE, Geissler M, Seufferlein T, Oettle H. Nab-paclitaxel for metastatic pancreatic cancer: clinical outcomes and potential mechanisms of action. Oncol Res Treat 2014; 37:128-34. [PMID: 24685917 DOI: 10.1159/000358890] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 01/22/2014] [Indexed: 11/19/2022]
Abstract
For almost 15 years there has been stagnation in the systemic treatment of patients with pancreatic ductal adenocarcinoma (PDAC). Recently, several developments seem to indicate clinically relevant improvements in the treatment of patients with metastatic disease. One of these developments is the introduction of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) into the firstline treatment of metastatic disease. In this review, underlying preclinical and clinical data are discussed, with a special focus on mechanisms of action, the potential interaction with albumin and calcium-binding matricellular glycoproteins, such as the secreted protein acidic and rich in cysteine (SPARC), as well as the clinical outcome associated with the use of nab-paclitaxel.
Collapse
Affiliation(s)
- Salah-Eddin Al-Batran
- Krankenhaus Nordwest, UCT University Cancer Center Frankfurt, Frankfurt/Main, Germany
| | | | | | | |
Collapse
|
44
|
Arcangeli A, Crociani O, Bencini L. Interaction of tumour cells with their microenvironment: ion channels and cell adhesion molecules. A focus on pancreatic cancer. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130101. [PMID: 24493749 DOI: 10.1098/rstb.2013.0101] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cancer must be viewed as a 'tissue', constituted of both transformed cells and a heterogeneous microenvironment, the 'tumour microenvironment' (TME). The TME undergoes a complex remodelling during the course of multistep tumourigenesis, hence strongly contributing to tumour progression. Ion channels and transporters (ICTs), being expressed on both tumour cells and in the different cellular components of the TME, are in a strategic position to sense and mediate signals arising from the TME. Often, this transmission is mediated by integrin adhesion receptors, which are the main cellular receptors capable of mediating cell-to-cell and cell-to-matrix bidirectional signalling. Integrins can often operate in conjunction with ICT because they can behave as functional partners of ICT proteins. The role of integrin receptors in the crosstalk between tumour cells and the TME is particularly relevant in the context of pancreatic cancer (PC), characterized by an overwhelming TME which actively contributes to therapy resistance. We discuss the possibility that this occurs through integrins and ICTs, which could be exploited as targets to overcome chemoresistance in PC.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, , Viale G.B. Morgagni, 50, 50134 Firenze, Italy
| | | | | |
Collapse
|
45
|
Wei G, Chang Y, Zheng J, He S, Chen N, Wang X, Sun X. Notch1 silencing inhibits proliferation and invasion in SGC‑7901 gastric cancer cells. Mol Med Rep 2014; 9:1153-8. [PMID: 24469571 DOI: 10.3892/mmr.2014.1920] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/20/2014] [Indexed: 11/05/2022] Open
Abstract
Downregulation of Notch1 has been shown to exert antineoplastic effects in vivo and in vitro. However, the role of the Notch1 gene in the proliferative and invasive ability of gastric cancer cells is not clear. In this study, we investigated the effect of Notch1 gene silencing on the proliferation and invasion of gastric cancer SGC‑7901 cells. Small interfering RNA (siRNA) targeting Notch1 was transfected into SGC‑7901 cells using Lipofectamine 2000. Proliferation of SGC‑7901 cells was then determined by the MTT assay. Notch1 mRNA expression was determined by reverse transcription‑polymerase chain reaction (RT‑PCR). Invasion of the SGC‑7901 cells was detected by the Transwell assay. The protein levels of cyclin D1, cyclin A1 and cyclin-dependent kinase 2 (CDK2) were determined by western blotting. The mRNA levels of matrix metalloproteinase‑2 (MMP‑2) and cyclooxygenase‑2 (COX‑2) were determined by RT‑PCR. Compared to the control group, the Notch1 mRNA level was significantly decreased following transfection. The growth and invasion rates of SGC‑7901 cells were significantly reduced after Notch1 silencing. Additionally, the expression of cyclin D1 and cyclin A1 proteins and of the MMP‑2 and COX‑2 mRNAs was markedly attenuated. From these results, it was concluded that Notch1 gene silencing inhibits the proliferation of gastric SGC‑7901 cells by decreasing the expression of cyclins D1 and A1, and reduces the invasive ability of SGC‑7901 cells through the downregulation of MMP‑2 and COX‑2 genes. Thus, silencing of the Notch1 pathway may be a novel approach in the treatment of gastrointestinal cancer.
Collapse
Affiliation(s)
- Guangbing Wei
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuanhong Chang
- Department of Gastroenterology, Xi'an No. 4 Hospital, Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jianbao Zheng
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Sai He
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Nanzheng Chen
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaolong Wang
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
46
|
Nikfarjam M, Yeo D, He H, Baldwin G, Fifis T, Costa P, Tan B, Yang E, Wen SW, Christophi C. Comparison of two syngeneic orthotopic murine models of pancreatic adenocarcinoma. J INVEST SURG 2013; 26:352-9. [PMID: 23957638 DOI: 10.3109/08941939.2013.797057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic adenocarcinoma has an extremely poor prognosis. The use of appropriate in vivo models is essential in devising methods to improve treatment outcomes. METHODS A pancreatic adenocarcinoma model based on tumor injection into the pancreatic head was compared with a pancreatic tail injection model in C57/BL6 mice. The murine pancreatic adenocarcinoma cell line PAN02, dispersed in Matrigel™, was used for tumor induction. RESULTS Tumors developed in all animals in both models. Tumor size was more consistent within the pancreatic tail group at 20 days following induction, with no evidence of metastatic disease. Animals in the pancreatic head injection group showed signs of reduced health by 20 days following injection and developed jaundice. Microscopic liver metastases were noted in some of these animals at this time point. The overall survival of animals at 40 days following tumor induction was significantly lower in the pancreatic head injection group (0% vs. 35%; p < .001). Multiple liver metastases were noted in five of 10 (50%) animals in the head injection group, without evidence of peritoneal metastases. In the pancreatic tail injection group, 18 of 20 (90%) animals had multiple peritoneal metastases, and nine of 20 (45%) animals had evidence of isolated liver deposits. Tumors in both regions of the pancreas had similar histologic characteristics, with a dense fibrotic stroma at the interface between the tumor and the normal pancreas. CONCLUSION Pancreatic head and tail orthotopic cancer models produce consistent tumors, but the patterns of tumor spread and survival differ according to the site of injection.
Collapse
Affiliation(s)
- Mehrdad Nikfarjam
- 1 Department of Surgery, Austin Health, University of Melbourne, Austin Health, LTB8, Heidleberg, Victoria, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Pedersen SF, Hoffmann EK, Novak I. Cell volume regulation in epithelial physiology and cancer. Front Physiol 2013; 4:233. [PMID: 24009588 PMCID: PMC3757443 DOI: 10.3389/fphys.2013.00233] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 08/09/2013] [Indexed: 12/21/2022] Open
Abstract
The physiological function of epithelia is transport of ions, nutrients, and fluid either in secretory or absorptive direction. All of these processes are closely related to cell volume changes, which are thus an integrated part of epithelial function. Transepithelial transport and cell volume regulation both rely on the spatially and temporally coordinated function of ion channels and transporters. In healthy epithelia, specific ion channels/transporters localize to the luminal and basolateral membranes, contributing to functional epithelial polarity. In pathophysiological processes such as cancer, transepithelial and cell volume regulatory ion transport are dys-regulated. Furthermore, epithelial architecture and coordinated ion transport function are lost, cell survival/death balance is altered, and new interactions with the stroma arise, all contributing to drug resistance. Since altered expression of ion transporters and channels is now recognized as one of the hallmarks of cancer, it is timely to consider this especially for epithelia. Epithelial cells are highly proliferative and epithelial cancers, carcinomas, account for about 90% of all cancers. In this review we will focus on ion transporters and channels with key physiological functions in epithelia and known roles in the development of cancer in these tissues. Their roles in cell survival, cell cycle progression, and development of drug resistance in epithelial cancers will be discussed.
Collapse
Affiliation(s)
- Stine F Pedersen
- Department of Biology, University of Copenhagen Copenhagen, Denmark
| | | | | |
Collapse
|
48
|
Tang D, Wang D, Yuan Z, Xue X, Zhang Y, An Y, Chen J, Tu M, Lu Z, Wei J, Jiang K, Miao Y. Persistent activation of pancreatic stellate cells creates a microenvironment favorable for the malignant behavior of pancreatic ductal adenocarcinoma. Int J Cancer 2013; 132:993-1003. [PMID: 22777597 DOI: 10.1002/ijc.27715] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/20/2012] [Accepted: 06/28/2012] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most common malignant tumors with poor prognosis due to extremely high malignancy, low rate of eligibility for surgical resection and chemoradiation resistance. Increasing evidence indicate that the interaction between activated pancreatic stellate cells (PSCs) and PDAC cells plays an important role in the development of PDAC. By producing high levels of cytokines, chemotactic factors, growth factors and excessive extracellular matrix (ECM), PSCs create desmoplasia and a hypoxic microenvironment that promote the initiation, development, evasion of immune surveillance, invasion, metastasis and resistance to chemoradiation of PDAC. Therefore, targeting the interaction between PSCs and PDAC cells may represent a novel therapeutic approach to advanced PDAC, especially therapies that target PSCs of the pancreatic tumor microenvironment.
Collapse
Affiliation(s)
- Dong Tang
- Department of Gastrointestinal Surgery, Subei People's Hospital of Jiangsu Province (Clinical Medical College of Yangzhou University), Yangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Brentnall TA. Arousal of cancer-associated stromal fibroblasts: palladin-activated fibroblasts promote tumor invasion. Cell Adh Migr 2012; 6:488-94. [PMID: 23076142 PMCID: PMC3547892 DOI: 10.4161/cam.21453] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer-associated fibroblasts (CAF), comprised of activated fibroblasts or myofibroblasts, are found in stroma surrounding solid tumors; these myofibroblasts promote invasion and metastasis of cancer cells. Activation of stromal fibroblasts into myofibroblasts is induced by expression of cystoskeleton protein, palladin, at early stages in tumorigenesis and increases with neoplastic progression. Expression of palladin in fibroblasts is triggered by paracrine signaling from adjacent k-ras-expressing epithelial cells. Three-dimensional co-cultures of palladin-expressing fibroblasts and pancreatic cancer cells reveals that the activated fibroblasts lead the invasion by creating tunnels through the extracellular matrix through which the cancer cells follow. Invasive tunneling occurs as a result of the development of invadopodia-like cellular protrusions in the palladin-activated fibroblasts and the addition of a wounding/inflammatory trigger. Abrogation of palladin reduces the invasive capacity of these cells. CAF also play a role in cancer resistance and immuno-privilege, making the targeting of activators of these cells of interest for oncologists.
Collapse
Affiliation(s)
- Teresa A Brentnall
- Department of Medicine, University of Washington Medical Center, Seattle, WA USA.
| |
Collapse
|
50
|
Niu H, Jiang H, Cheng B, Li X, Dong Q, Shao L, Liu S, Wang X. Stromal proteome expression profile and muscle-invasive bladder cancer research. Cancer Cell Int 2012; 12:39. [PMID: 22920603 PMCID: PMC3489783 DOI: 10.1186/1475-2867-12-39] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/17/2012] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND To globally characterize the cancer stroma expression profile of muscle-invasive transitional cell carcinoma and to discuss the cancer biology as well as biomarker discovery from stroma. Laser capture micro dissection was used to harvest purified muscle-invasive bladder cancer stromal cells and normal urothelial stromal cells from 4 paired samples. Two-dimensional liquid chromatography tandem mass spectrometry was used to identify the proteome expression profile. The differential proteins were further analyzed using bioinformatics tools and compared with the published literature. RESULTS We identified 868/872 commonly expressed proteins and 978 differential proteins from 4 paired cancer and normal stromal samples using laser capture micro dissection coupled with two-dimensional liquid chromatography tandem mass spectrometry. 487/491 proteins uniquely expressed in cancer/normal stroma. Differential proteins were compared with the entire list of the international protein index (IPI), and there were 42/42 gene ontology (GO) terms exhibited as enriched and 8/5 exhibited as depleted in cellular Component, respectively. Significantly altered pathways between cancer/normal stroma mainly include metabolic pathways, ribosome, focal adhesion, etc. Finally, descriptive statistics show that the stromal proteins with extremes of PI and MW have the same probability to be a biomarker. CONCLUSIONS Based on our results, stromal cells are essential component of the cancer, biomarker discovery and network based multi target therapy should consider neoplastic cells itself and corresponding stroma as whole one.
Collapse
Affiliation(s)
- Haitao Niu
- Department of Urology, The Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Haiping Jiang
- Department of Oncology, The Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Bo Cheng
- Department of Urology, The Central Hospital of Shengli Oil Field, Dondying, China
| | - Xinhui Li
- Department of Urology, The Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Qian Dong
- Department of Pediatric Surgery, The Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Leping Shao
- Department of Nephrology, The Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Shiguo Liu
- Gout Laboratory, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Xinsheng Wang
- Department of Urology, The Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| |
Collapse
|