1
|
Beig Parikhani A, Dehghan R, Talebkhan Y, Bayat E, Biglari A, Shokrgozar MA, Ahangari Cohan R, Mirabzadeh E, Ajdary S, Behdani M. A novel nanobody-based immunocytokine of a mutant interleukin-2 as a potential cancer therapeutic. AMB Express 2024; 14:19. [PMID: 38337114 PMCID: PMC10857990 DOI: 10.1186/s13568-023-01648-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 11/26/2023] [Indexed: 02/12/2024] Open
Abstract
The immunotherapeutic application of interleukin-2 (IL-2) in cancer treatment is limited by its off-target effects on different cell populations and insufficient activation of anti-tumor effector cells at the site of the tumor upon tolerated doses. Targeting IL-2 to the tumor microenvironment by generating antibody-cytokine fusion proteins (immunocytokine) would be a promising approach to increase efficacy without associated toxicity. In this study, a novel nanobody-based immunocytokine is developed by the fusion of a mutant (m) IL-2 with a decreased affinity toward CD25 to an anti-vascular endothelial growth factor receptor-2 (VEGFR2) specific nanobody, denoted as VGRmIL2-IC. The antigen binding, cell proliferation, IFN-γ-secretion, and cytotoxicity of this new immunocytokine are evaluated and compared to mIL-2 alone. Furthermore, the pharmacokinetic properties are analyzed. Flow cytometry analysis shows that the VGRmIL2-IC molecule can selectively target VEGFR2-positive cells. The results reveal that the immunocytokine is comparable to mIL-2 alone in the stimulation of Primary Peripheral Blood Mononuclear Cells (PBMCs) and cytotoxicity in in vitro conditions. In vivo studies demonstrate improved pharmacokinetic properties of VGRmIL2-IC in comparison to the wild or mutant IL-2 proteins. The results presented here suggest VGRmIL2-IC could be considered a candidate for the treatment of VEGFR2-positive tumors.
Collapse
Affiliation(s)
- Arezoo Beig Parikhani
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Rada Dehghan
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Yeganeh Talebkhan
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Bayat
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Biglari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Esmat Mirabzadeh
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
Cao Y, Sun C, Huo G, Wang H, Wu Y, Wang F, Liu S, Zhai S, Zhang X, Zhao H, Hu M, Gu W, Yang Y, Wang S, Liang C, Lyu J, Lu T, Wang Y, Xie L, Fan C. Novel hKDR mouse model depicts the antiangiogenesis and apoptosis-promoting effects of neutralizing antibodies targeting vascular endothelial growth factor receptor 2. Cancer Sci 2022; 114:115-128. [PMID: 36114822 PMCID: PMC9807522 DOI: 10.1111/cas.15594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 01/07/2023] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2)/KDR plays a critical role in tumor growth, diffusion, and invasion. The amino acid sequence homology of KDR between mouse and human in the VEGF ligand-binding domain was low, thus the WT mice could not be used to evaluate Abs against human KDR, and the lack of a suitable mouse model hindered both basic research and drug developments. Using the CRISPR/Cas9 technique, we successfully inserted different fragments of the human KDR coding sequence into the chromosomal mouse Kdr exon 4 locus to obtain an hKDR humanized mouse that can be used to evaluate the marketed Ab ramucirumab. In addition, the humanized mAb VEGFR-HK19 was developed, and a series of comparative assays with ramucirumab as the benchmark revealed that VEGFR-HK19 has higher affinity and superior antiproliferation activity. Moreover, VEGFR-HK19 selectively inhibited tumor growth in the hKDR mouse model but not in WT mice. The most important binding epitopes of VEGFR2-HK19 are D257, L313, and T315, located in the VEGF binding region. Therefore, the VEGFR2-HK19 Ab inhibits tumor growth by blocking VEGF-induced angiogenesis, inflammation, and promoting apoptosis. To our best knowledge, this novel humanized KDR mouse fills the gaps both in an animal model and the suitable in vivo evaluation method for developing antiangiogenesis therapies in the future, and the newly established humanized Ab is expected to be a drug candidate possibly benefitting tumor patients.
Collapse
Affiliation(s)
- Yuan Cao
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Chunyun Sun
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Guitao Huo
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Huiyu Wang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Yong Wu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Fei Wang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Susu Liu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Shijie Zhai
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Xiao Zhang
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Haoyang Zhao
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Meiling Hu
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina
| | - Wenda Gu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Yanwei Yang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Sanlong Wang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety EvaluationNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Chunnan Liang
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Jianjun Lyu
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Tiangong Lu
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Institute for Biological Product ControlNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and AntibodySinocelltech LtdBeijingChina,Beijing Key Laboratory of Monoclonal Antibody Research and DevelopmentSino Biological Inc.BeijingChina,Cell Culture Engineering CenterChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Changfa Fan
- Division of Animal Model Research, National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal ResourcesNational Institutes for Food and Drug Control (NIFDC)BeijingChina
| |
Collapse
|
3
|
Savino A, De Marzo N, Provero P, Poli V. Meta-Analysis of Microdissected Breast Tumors Reveals Genes Regulated in the Stroma but Hidden in Bulk Analysis. Cancers (Basel) 2021; 13:3371. [PMID: 34282769 PMCID: PMC8268805 DOI: 10.3390/cancers13133371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023] Open
Abstract
Transcriptome data provide a valuable resource for the study of cancer molecular mechanisms, but technical biases, sample heterogeneity, and small sample sizes result in poorly reproducible lists of regulated genes. Additionally, the presence of multiple cellular components contributing to cancer development complicates the interpretation of bulk transcriptomic profiles. To address these issues, we collected 48 microarray datasets derived from laser capture microdissected stroma or epithelium in breast tumors and performed a meta-analysis identifying robust lists of differentially expressed genes. This was used to create a database with carefully harmonized metadata that we make freely available to the research community. As predicted, combining the results of multiple datasets improved statistical power. Moreover, the separate analysis of stroma and epithelium allowed the identification of genes with different contributions in each compartment, which would not be detected by bulk analysis due to their distinct regulation in the two compartments. Our method can be profitably used to help in the discovery of biomarkers and the identification of functionally relevant genes in both the stroma and the epithelium. This database was made to be readily accessible through a user-friendly web interface.
Collapse
Affiliation(s)
- Aurora Savino
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy;
| | - Niccolò De Marzo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy;
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Corso Massimo D’Azeglio 52, 10126 Turin, Italy;
- Center for Omics Sciences, Ospedale San Raffaele IRCCS, Via Olgettina 60, 20132 Milan, Italy
| | - Valeria Poli
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy;
| |
Collapse
|
4
|
Kabak YB, Sozmen M, Devrim AK, Sudagidan M, Yildirim F, Guvenc T, Yarim M, Gulbahar YM, Ahmed I, Karaca E, Inal S. Expression levels of angiogenic growth factors in feline squamous cell carcinoma. Acta Vet Hung 2020; 68:37-48. [PMID: 32384073 DOI: 10.1556/004.2020.00005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 11/13/2019] [Indexed: 01/16/2023]
Abstract
Squamous cell carcinoma (SCC) is the most common malignant neoplasm of the skin in cats. Tumour angiogenesis is the pivotal event for tumour progression and metastasis. We assessed protein and gene expression of angiogenic growth factors including bFGF, VEGF-C, TGF-β, PDGF-A, PDGF-C and PDGFR-α that possibly contribute to the angiogenic phenotype of feline SCC (FSCC) and could, therefore, be a good target in the treatment of SCC. In the present study, a total of 27 FSCC cases were investigated. Tumour cases were histopathologically classified as well differentiated (10/27), moderately differentiated (5/27), and poorly differentiated (12/27). The expression levels of the growth factors were detected using immunohistochemistry and assessed semi-quantitatively. Growth factor expression levels were evaluated at different locations: in the oral region, in areas exposed to solar UV radiation including the ears, eyelids and nasal planum, and other miscellaneous locations. Our findings have revealed that FSCC arising from different anatomical sites of the body and showing differences in aggressiveness, metastasis, and prognosis may be angiogenesis dependent, and angiogenic key regulators could play a role in the development of FSCC.
Collapse
Affiliation(s)
- Yonca B. Kabak
- 1Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, Kurupelit, 55200 Atakum, Samsun, Turkey
| | - Mahmut Sozmen
- 1Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, Kurupelit, 55200 Atakum, Samsun, Turkey
| | - Alparslan K. Devrim
- 2Department of Biochemistry, Faculty of Veterinary Medicine, Kırıkkale University, Kırıkkale, Turkey
| | - Mert Sudagidan
- 3Konya Food and Agriculture University, Kit-Argem, Konya, Turkey
| | - Funda Yildirim
- 4Department of Pathology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Tolga Guvenc
- 1Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, Kurupelit, 55200 Atakum, Samsun, Turkey
| | - Murat Yarim
- 1Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, Kurupelit, 55200 Atakum, Samsun, Turkey
| | - Yavuz M. Gulbahar
- 1Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, Kurupelit, 55200 Atakum, Samsun, Turkey
| | - Ishtiaq Ahmed
- 5Department of Pathology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Efe Karaca
- 1Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, Kurupelit, 55200 Atakum, Samsun, Turkey
| | - Sinem Inal
- 1Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, Kurupelit, 55200 Atakum, Samsun, Turkey
| |
Collapse
|
5
|
Lu RM, Chiu CY, Liu IJ, Chang YL, Liu YJ, Wu HC. Novel human Ab against vascular endothelial growth factor receptor 2 shows therapeutic potential for leukemia and prostate cancer. Cancer Sci 2019; 110:3773-3787. [PMID: 31578782 PMCID: PMC6890446 DOI: 10.1111/cas.14208] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) is highly expressed in tumor‐associated endothelial cells, where it modulates tumor‐promoting angiogenesis, and it is also found on the surface of tumor cells. Currently, there are no Ab therapeutics targeting VEGFR2 approved for the treatment of prostate cancer or leukemia. Therefore, development of novel efficacious anti‐VEGFR2 Abs will benefit cancer patients. We used the Institute of Cellular and Organismic Biology human Ab library and affinity maturation to develop a fully human Ab, anti‐VEGFR2‐AF, which shows excellent VEGFR2 binding activity. Anti‐VEGFR2‐AF bound Ig‐like domain 3 of VEGFR2 extracellular region to disrupt the interaction between VEGF‐A and VEGFR2, neutralizing downstream signaling of the receptor. Moreover, anti‐VEGFR2‐AF inhibited capillary structure formation and exerted Ab‐dependent cell‐mediated cytotoxicity and complement‐dependent cytotoxicity in vitro. We found that VEGFR2 is expressed in PC‐3 human prostate cancer cell line and associated with malignancy and metastasis of human prostate cancer. In a PC‐3 xenograft mouse model, treatment with anti‐VEGFR2‐AF repressed tumor growth and angiogenesis as effectively and safely as US FDA‐approved anti‐VEGFR2 therapeutic, ramucirumab. We also report for the first time that addition of anti‐VEGFR2 Ab can enhance the efficacy of docetaxel in the treatment of a prostate cancer mouse model. In HL‐60 human leukemia‐xenografted mice, anti‐VEGFR2‐AF showed better efficacy than ramucirumab with prolonged survival and reduced metastasis of leukemia cells to ovaries and lymph nodes. Our findings suggest that anti‐VEGFR2‐AF has strong potential as a cancer therapy that could directly target VEGFR2‐expressing tumor cells in addition to its anti‐angiogenic action.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chiung-Yi Chiu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - I-Ju Liu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yu-Ling Chang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yaw-Jen Liu
- Research and Development Center, United Biopharma Inc., Hsinshu, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
6
|
Mei Y, Liu H. IL-37: An anti-inflammatory cytokine with antitumor functions. Cancer Rep (Hoboken) 2018; 2:e1151. [PMID: 32935478 DOI: 10.1002/cnr2.1151] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND IL-37 is a newly identified IL-1 family cytokine. Unlike other members in IL-1 family, IL-37 has been demonstrated to be an anti-inflammatory cytokine in many inflammatory and autoimmune diseases. IL-37 is regarded as a dual-function cytokine as both the extracellular and intracellular IL-37 are biologically functional. Extracellular IL-37 can bind to IL-18Rα and IL-1R8 to form a triple complex, regulating the downstream STAT3 and PTEN signaling. Intracellular IL-37 can interact with Smad3, translocate into nucleus, and regulate downstream target gene expressions. Recently, the role of IL-37 in tumor development has been extensively studied. RECENT FINDINGS IL-37 has been found to play an antitumor role in various types of tumors, such as non-small cell lung cancer, hepatocellular carcinoma, and renal cell carcinoma. Many mechanism studies have been carried out to elaborate the possible effects of IL-37 on tumor growth, immune responses, and tumor angiogenesis. More importantly, the function of IL-37 may be dependent on its concentration and receptor expression. It can form dimers at high concentrations to be inactivated, thus inhibiting its anti-inflammatory function. We focused on the role of IL-37 in various tumor types and provided the hypothesis regarding the underlying mechanisms. CONCLUSION IL-37 may affect tumor development through multiple mechanisms: (1) IL-37 directly influences tumor cell viability; (2) IL-37 regulates the immune response to promote the antitumor immunity; and (3) IL-37 suppresses tumor angiogenesis in the tumor microenvironment. Future studies are warranted to further investigate the mechanisms of these multifaceted functions of IL-37 in animal models and cancer patients.
Collapse
Affiliation(s)
- Yu Mei
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore
| |
Collapse
|
7
|
Martini T, Heinkele J, Mayr R, Weis CA, Wezel F, Wahby S, Eckstein M, Schnöller T, Breyer J, Wirtz R, Ritter M, Bolenz C, Erben P. Predictive value of lymphangiogenesis and proliferation markers on mRNA level in urothelial carcinoma of the bladder after radical cystectomy. Urol Oncol 2018; 36:530.e19-530.e27. [PMID: 30446441 DOI: 10.1016/j.urolonc.2018.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/10/2018] [Accepted: 09/03/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To evaluate the mRNA expression of lymphangiogenesis and proliferation markers and to examine its association with histopathological characteristics and clinical outcome in patients with urothelial carcinoma of the bladder (UCB) after radical cystectomy (RC). PATIENTS AND METHODS Gene expression analysis of the vascular endothelial growth -C and -D (VEGF-C/-D), its receptor VEGF receptor-3 (VEGFR-3), MKI67, and RACGAP1 was performed in 108 patients after radical cystectomy and their correlation with clinical-pathological parameters was investigated. Uni- and multivariate regression analyses were used to identify predictors for cancer-specific survival (CSS), recurrence-free survival (RFS) and overall survival (OS) after RC. RESULTS The expression of RACGAP1 and VEGFR-3 showed an association with a higher pT stage (P = 0.049; P = 0.009). MKI67 showed an association with a high-grade urothelial carcinoma of the bladder (P = 0.021). VEGFR-3 expression was significantly associated with the presence of lymphovascular invasion (LVI) (P = 0.016) and lymph node metastases (pN+) (P = 0.028). With the univariate analysis, overexpression of VEGFR-3 (P = 0.029) and the clinical-pathological parameters pT stage (P < 0.0001), pN+ (P = 0.0004), LVI (P < 0.0001) and female gender (P = 0.021) were significantly associated with a reduced CSS. Multivariate analysis identified a higher pT stage (P = 0.017) and LVI (P = 0.008) as independent predictors for reduced CSS. Independent predictors for reduced OS were a higher pT stage (P = 0.0007) and LVI (P = 0.0021), while overexpression of VEGF-D was associated with better OS (P < 0.0001). CONCLUSIONS The mRNA expression of the investigated markers showed associations with common histopathological parameters. Increased expression of VEGF-D is independently associated with better overall survival.
Collapse
Affiliation(s)
| | - Jakob Heinkele
- Department of Urology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Roman Mayr
- Department of Urology, University of Regensburg, Caritas St. Josef Medical Center, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Felix Wezel
- Department of Urology, University of Ulm, Ulm, Germany
| | - Sarah Wahby
- Department of Urology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Markus Eckstein
- Institute of Pathology, University of Erlangen-Nüremberg, Erlangen, Germany
| | | | - Johannes Breyer
- Department of Urology, University of Regensburg, Caritas St. Josef Medical Center, Germany
| | - Ralph Wirtz
- STRATIFYER Molecular Pathology GmbH, Cologne, Germany; Institute of Pathology, The St. Elisabeth Hospital Köln-Hohenlind, Cologne, Germany
| | - Manuel Ritter
- Department of Urology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Philipp Erben
- Department of Urology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
8
|
Chen B, Liu J, Wang X, Shen Q, Li C, Dai C. Co-expression of PDGF-B and VEGFR-3 strongly correlates with poor prognosis in hepatocellular carcinoma patients after hepatectomy. Clin Res Hepatol Gastroenterol 2018; 42:126-133. [PMID: 29273278 DOI: 10.1016/j.clinre.2016.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 10/19/2016] [Accepted: 11/08/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND The ability to evaluate the prognosis of hepatocellular carcinoma (HCC) patients following hepatectomy with biological markers is of great importance. METHODS In this study, we collected samples from 90 patients with HCC after hepatectomy. Immunohistochemistry was used to detect the expression of PDGF-B and VEGFR-3 in these HCC samples. RESULTS According to the immunohistochemical results, PDGF-B and VEGFR-3 staining were significantly associated with clinical features. Additionally, a significant association between high PDGF-B and VEGFR-3 levels and shorter overall survival was noted, when PDGF-B and VEGFR-3 co-expression been analyzed. CONCLUSION These results suggest that the correlative expression level of PDGF-B and VEGFR-3 has strong value in the prognosis of HCC patients following hepatectomy.
Collapse
Affiliation(s)
- Bin Chen
- Department of General Surgery, The affiliated Ningbo Traditional Chinese Medical Hospital of Zhejiang Chinese Medical University, 315010 Ningbo, Zhejiang province, P.R. China
| | - Jinghua Liu
- Department of Hepatobiliary Surgery, Linyi people's hospital, 27th of East Jiefang Road, 276000 Linyi, Shandong province, P.R. China
| | - Xionghua Wang
- Department of General Surgery, The affiliated Ningbo Traditional Chinese Medical Hospital of Zhejiang Chinese Medical University, 315010 Ningbo, Zhejiang province, P.R. China
| | - Qian Shen
- Department of General Surgery, The affiliated Ningbo Traditional Chinese Medical Hospital of Zhejiang Chinese Medical University, 315010 Ningbo, Zhejiang province, P.R. China
| | - Chao Li
- Department of General Surgery, The affiliated Ningbo Traditional Chinese Medical Hospital of Zhejiang Chinese Medical University, 315010 Ningbo, Zhejiang province, P.R. China
| | - Chunshan Dai
- Department of General Surgery, The affiliated Ningbo Traditional Chinese Medical Hospital of Zhejiang Chinese Medical University, 315010 Ningbo, Zhejiang province, P.R. China.
| |
Collapse
|
9
|
Kiselev Y, Andersen S, Johannessen C, Fjukstad B, Standahl Olsen K, Stenvold H, Al-Saad S, Donnem T, Richardsen E, Bremnes RM, Rasmussen Busund LT. Transcription factor PAX6 as a novel prognostic factor and putative tumour suppressor in non-small cell lung cancer. Sci Rep 2018; 8:5059. [PMID: 29568088 PMCID: PMC5864921 DOI: 10.1038/s41598-018-23417-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 03/09/2018] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths. Novel predictive biomarkers are needed to improve treatment selection and more accurate prognostication. PAX6 is a transcription factor with a proposed tumour suppressor function. Immunohistochemical staining was performed on tissue microarrays from 335 non-small cell lung cancer (NSCLC) patients for PAX6. Multivariate analyses of clinico-pathological variables and disease-specific survival (DSS) was carried out, and phenotypic changes of two NSCLC cell lines with knockdown of PAX6 were characterized. While PAX6 expression was only associated with a trend of better disease-specific survival (DSS) (p = 0.10), the pN+ subgroup (N = 103) showed significant correlation between high PAX6 expression and longer DSS (p = 0.022). Median survival for pN + patients with high PAX6 expression was 127.4 months, versus 22.9 months for patients with low PAX6 expression. In NCI-H661 cells, knockdown of PAX6 strongly activated serum-stimulated migration. In NCI-H460 cells, PAX6 knockdown activated anchorage-independent growth. We did not observe any significant effect of PAX6 on proliferation in either of cell lines. Our findings strongly support the proposition of PAX6 as a valid and positive prognostic marker in NSCLC in node-positive patients. There is a need for further studies, which should provide mechanistical explanation for the role of PAX6 in NSCLC.
Collapse
Affiliation(s)
- Yury Kiselev
- Department of Life Sciences and Health, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway. .,Department of Pharmacy, UiT The Arctic University of Norway, Tromso, Norway. .,Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway.
| | - Sigve Andersen
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.,Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Charles Johannessen
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Bjørn Fjukstad
- Department of Computer Science, Faculty of Science and Technology, UiT The Arctic University of Norway, Tromso, Norway
| | - Karina Standahl Olsen
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromso, Norway
| | - Helge Stenvold
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Samer Al-Saad
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway.,Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Tom Donnem
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.,Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Elin Richardsen
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway.,Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Roy M Bremnes
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.,Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Lill-Tove Rasmussen Busund
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway.,Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| |
Collapse
|
10
|
Kowalczuk O, Laudanski J, Laudanski W, Niklinska WE, Kozlowski M, Niklinski J. Lymphatics-associated genes are downregulated at transcription level in non-small cell lung cancer. Oncol Lett 2018; 15:6752-6762. [PMID: 29849784 DOI: 10.3892/ol.2018.8159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 10/17/2017] [Indexed: 12/30/2022] Open
Abstract
The present study aimed to verify a possibility of ongoing lymphangiogenesis in non-small cell lung cancer (NSCLC) via examination of mRNA levels of a number of lymphangiogenesis-associated genes in tumors. It was hypothesized that transcriptional activation of these genes would occur in tumors that stimulate new lymphatic vessel formation. The study was performed on 140 pairs of fresh-frozen surgical specimens of cancer and unaffected lung tissues derived from NSCLC stage I-IIIA patients. mRNA levels were evaluated with the reverse transcription-quantitative polymerase chain reaction method and expressed as fold change differences between the tumor and normal tissues. Possible associations between expression and patient clinicopathological characteristics and survival were analyzed. In the NSCLC tissue samples, vascular endothelial growth factor (VEGF) C, VEGFD, VEGFR3, VEGFR2, VEGFR1, lymphatic vessel endothelial hyaluronan receptor 1, integrin subunit α 9, FOX2, neuropilin 2, fibroblast growth factor 2 genes were significantly downregulated (P<0.001 for all) compared with matched normal lung tissues, whereas mRNA levels for VEGFA, spleen associated tyrosine kinase, podoplanin, and prospero homeobox 1 genes were similar in both tissues. Neither lymph node status, nor disease pathological stage influenced expression, whereas more profound suppression of gene activities appeared to occur in squamous cell carcinomas compared with adenocarcinomas. The VEGFR1 mRNA expression level was significantly connected with patient survival in the univariate analysis, and was an independent prognostic factor for overall survival in the multivariate Cox's proportional hazards model (HR 2.103; 95% confidence interval: 1.005-4.401; P=0.049). The results support a hypothesis of absence of new lymphatic vessel formation inside growing NSCLC tumor mass, however do not exclude a possibility of lymphangiogenesis in narrow marginal tumor parts.
Collapse
Affiliation(s)
- Oksana Kowalczuk
- Department of Clinical Molecular Biology, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Jerzy Laudanski
- Department of Thoracic Surgery, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Wojciech Laudanski
- Department of Thoracic Surgery, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Wieslawa Ewa Niklinska
- Department of Histology and Embryology, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Miroslaw Kozlowski
- Department of Thoracic Surgery, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Jacek Niklinski
- Department of Clinical Molecular Biology, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
11
|
Sasahira T, Nishiguchi Y, Kurihara-Shimomura M, Nakashima C, Kuniyasu H, Kirita T. NIPA-like domain containing 1 is a novel tumor-promoting factor in oral squamous cell carcinoma. J Cancer Res Clin Oncol 2018; 144:875-882. [PMID: 29464350 DOI: 10.1007/s00432-018-2612-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/16/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE In our previous global gene expression analysis, we identified NIPA-like domain containing 1 (NIPAL1), which encodes a magnesium transporter, as one of the most overexpressed genes in recurrent oral squamous cell carcinoma (OSCC). Although has been NIPAL1 linked with gout pathogenesis, little is known about its expression and function in human malignancies. METHODS In this study, we examined NIPAL1 expression in 192 cases of OSCC by immunohistochemistry and performed a functional analysis of human OSCC cells. RESULTS NIPAL1 immunostaining was observed in 39 of 192 OSCC patients (20.3%). NIPAL1 expression correlated significantly with cancer cell intravsation (P = 0.0062), as well as with poorer disease-free survival in a Kaplan-Meier analysis (P < 0.0001). Moreover, a multivariate Cox proportional hazards model analysis revealed that NIPAL1 expression was an independent predictor of disease-free survival in OSCC (P < 0.0001). In a functional analysis, NIPAL1 regulated the growth and adhesion of OSCC tumor cells and endothelial cells. CONCLUSIONS Our findings suggest that NIPAL1 might be a novel factor promoting OSCC tumorigenesis, as well as a useful molecular marker of OSCC.
Collapse
Affiliation(s)
- Tomonori Sasahira
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan.
| | - Yukiko Nishiguchi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| | - Miyako Kurihara-Shimomura
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan.,Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Chie Nakashima
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara, 634-8521, Japan
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| |
Collapse
|
12
|
Kilvaer TK, Rakaee M, Hellevik T, Østman A, Strell C, Bremnes RM, Busund LT, Dønnem T, Martinez-Zubiaurre I. Tissue analyses reveal a potential immune-adjuvant function of FAP-1 positive fibroblasts in non-small cell lung cancer. PLoS One 2018; 13:e0192157. [PMID: 29415055 PMCID: PMC5802915 DOI: 10.1371/journal.pone.0192157] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/17/2018] [Indexed: 01/08/2023] Open
Abstract
Objectives Selective targeting of cancer-associated fibroblasts (CAFs) has been proposed to synergize with immune-checkpoint inhibitors. While the roles of CAFs in cancer development are well described, their immune-regulatory properties remain incompletely understood. This study investigates correlations between CAF and immune-markers in tumor stroma from non-small cell lung cancer (NSCLC) patients, and examines whether a combination of CAF and immune cell scores impact patient prognosis. Methods Tumor specimens from 536 primary operable stage I-III NSCLC patients were organized in tissue microarrays. Expression of protein-markers was evaluated by immunohistochemistry. Results Fibroblast and stromal markers PDGFRα, PDGFRβ, FAP-1 and vimentin showed weak correlations while αSMA, and Masson’s trichrome did not correlate with any of the investigated markers. Hierarchical clustering indicated the existence of different CAF-subsets. No relevant correlations were found between any CAF-marker and the immune-markers CD3, CD4, CD8, CD20, CD68, CD1a, CD56, FoxP3 and CD45RO. High density of fibroblast-activation protein positive mesenchymal cells (CAFFAP) was associated with better prognosis in tumors with high infiltration of CD8 and CD3 T-lymphocytes. Conclusions The presented data suggest that CAFs, irrespective of identity, have low influence on the degree of tumor infiltration by inflammatory- and/or immune-cells. However, CAFFAP may exert immuno-adjuvant roles in NSCLC, and targeting CAFs should be cautiously considered.
Collapse
Affiliation(s)
- Thomas Karsten Kilvaer
- Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
- Department of Clinical Medicine, UiT The Artic University of Norway, Tromsø, Norway
| | - Mehrdad Rakaee
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Turid Hellevik
- Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
| | - Arne Østman
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Carina Strell
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Roy M. Bremnes
- Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
- Department of Clinical Medicine, UiT The Artic University of Norway, Tromsø, Norway
| | - Lill-Tove Busund
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromsø, Norway
| | - Tom Dønnem
- Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
- Department of Clinical Medicine, UiT The Artic University of Norway, Tromsø, Norway
| | | |
Collapse
|
13
|
Hald SM, Rakaee M, Martinez I, Richardsen E, Al-Saad S, Paulsen EE, Blix ES, Kilvaer T, Andersen S, Busund LT, Bremnes RM, Donnem T. LAG-3 in Non-Small-cell Lung Cancer: Expression in Primary Tumors and Metastatic Lymph Nodes Is Associated With Improved Survival. Clin Lung Cancer 2017; 19:249-259.e2. [PMID: 29396238 DOI: 10.1016/j.cllc.2017.12.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/22/2017] [Accepted: 12/01/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Lymphocyte activation gene-3 (LAG-3) is an immune checkpoint receptor and a putative therapeutic target in non-small-cell lung cancer (NSCLC). We explored the prognostic effect of LAG-3+ tumor-infiltrating lymphocytes (TILs) in primary tumors and metastatic lymph nodes in NSCLC and its potential for inclusion in an immunoscore, supplementing the TNM classification. MATERIALS AND METHODS Primary tumor tissue from 553 stage I-IIIB NSCLC patients and 143 corresponding metastatic lymph nodes were collected. The expression of LAG-3 was evaluated by immunohistochemistry on tissue microarrays. RESULTS On univariate analysis, LAG-3+ TILs in the intraepithelial and stromal compartments of primary tumors and in the intraepithelial and extraepithelial compartments of metastatic lymph nodes were associated with improved disease-specific survival (DSS). On multivariate analysis, stromal LAG-3+ TILs were a significant independent predictor of improved DSS (hazard ratio [HR], 0.59; 95% confidence interval [CI], 0.43-0.82; P = .002). Stromal LAG-3+ TILs did not have prognostic impact across all pathologic stages. In the metastatic lymph nodes, intraepithelial (HR, 0.61; 95% CI, 0.38-0.99; P = .049) and extraepithelial (HR, 0.54; 95% CI, 0.29-0.70; P < .001) LAG-3+ TILs were independently associated with favorable DSS. CONCLUSION LAG-3+ TILs are an independent positive prognostic factor in stage I-IIIB NSCLC. LAG-3 in metastatic lymph nodes is a candidate marker for an immunoscore in NSCLC.
Collapse
Affiliation(s)
- Sigurd M Hald
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.
| | - Mehrdad Rakaee
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Inigo Martinez
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Elin Richardsen
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway; Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Samer Al-Saad
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway; Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Erna-Elise Paulsen
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway; Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Egil Støre Blix
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway; Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Thomas Kilvaer
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway; Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Sigve Andersen
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway; Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Lill-Tove Busund
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway; Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Roy M Bremnes
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway; Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Tom Donnem
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway; Department of Oncology, University Hospital of North Norway, Tromso, Norway
| |
Collapse
|
14
|
Al-Saad S, Richardsen E, Kilvaer TK, Donnem T, Andersen S, Khanehkenari M, Bremnes RM, Busund LT. The impact of MET, IGF-1, IGF1R expression and EGFR mutations on survival of patients with non-small-cell lung cancer. PLoS One 2017; 12:e0181527. [PMID: 28742836 PMCID: PMC5526580 DOI: 10.1371/journal.pone.0181527] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/03/2017] [Indexed: 01/10/2023] Open
Abstract
Introduction To compare the efficacy of silver in situ hybridization (SISH) and immunohistochemistry (IHC) in detecting MET and IGF1R alterations and to investigate their prevalence and prognostic significance. A possible correlation between MET receptor expression, MET gene alterations and the two most frequent occurring EGFR gene mutations was also investigated. Materials and methods Stage I to IIIA tumors from 326 patients with NSCLC were immunohistochemically tested for protein expression of MET and IGF-1. Their cytoplasmic expression was compared with the gene copy number of the MET and IGF1Rgenes by SISH in paraffin-embedded, formalin-fixed material. Correlations were made with the immunohistochemical expression of two frequent EGFR mutations and clinicopathological variables. Univariate and multivariate survival analyses was used to evaluate the prognostic efficacy of the tested markers. Results In univariate analyses, high cytoplasmic MET expression showed a significant negative prognostic effect in adenocarcinoma patients (p = 0.026). MET gene to chromosome 7 ratio was a significant positive prognostic marker (p = 0.005), probably only due to the highly negative prognostic significance of chromosome 7 polysomy (p = 0.002). High IGF1R gene copy number was a negative prognostic marker for all NSCLC patients (p = 0.037). In the multivariate analysis, polysomy of chromosome 7 in tumor cells correlated significantly and independently with a poor prognosis (p = 0.011). In patients with adenocarcinoma, a high cytoplasmic MET expression was an independent negative prognostic marker (p = 0.013). In males a high IGF1R gene copy number to chromosome 15 count ratio was significantly and independently correlated to a poor prognosis (p = 0.018). Conclusion MET protein expression provides superior prognostic information compared with SISH. Polysomy of chromosome 7 is an independent negative prognostic factor in NSCLC patients. This finding has an important implication while examining genes located on chromosome 7 by means of SISH. High IGF1R gene copy number to chromosome 15 count ratio is an independent predictor of inferior survival in male patients with primary NSCLC.
Collapse
Affiliation(s)
- Samer Al-Saad
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
- * E-mail:
| | - Elin Richardsen
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
| | - Thomas K. Kilvaer
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Tom Donnem
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Sigve Andersen
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Mehrdad Khanehkenari
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Roy M. Bremnes
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- Department of Oncology, University Hospital of Northern Norway, Tromso, Norway
| | - Lill-Tove Busund
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of Northern Norway, Tromso, Norway
| |
Collapse
|
15
|
Wang Y, Wang H, Pan T, Li L, Li J, Yang H. STIM1 silencing inhibits the migration and invasion of A549 cells. Mol Med Rep 2017; 16:3283-3289. [PMID: 28713917 PMCID: PMC5547937 DOI: 10.3892/mmr.2017.7010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 05/12/2017] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to explore the effects of stromal interaction molecule 1 (STIM1) knockdown on the migration, invasion and metastasis of A549 cells in vitro and in vivo. Western blotting and immunohistochemistry were used to detect protein expression levels. Wound healing and Transwell invasion assays were used to assess the migratory and invasive abilities of A549 cells transfected with STIM1-specific short hairpin (sh)RNA (shSTIM1). In addition, a tail vein metastatic assay was performed. The results demonstrated that the frequency of STIM1 high-expression was significantly increased in metastatic lung cancer tissues (72.2%) compared with in non-metastatic lung cancer tissues (33.0%). STIM1 knockdown inhibited A549 cell migration and invasion in vitro and tumor metastasis in vivo. The protein expression levels of Snail1, Vimentin, matrix metalloproteinase (MMP) 2 and MMP9 were markedly decreased in A549-shSTIM1 compared with in A549 cells transfected with control shRNA (shcon). In addition, the protein expression levels of E-cadherin were markedly increased in A549-shSTIM1 cells compared with in A549-shcon cells. These results suggested that STIM1 knockdown may inhibit the migration and invasion of A549 cells in vitro, and metastasis in vivo.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Haiyu Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Teng Pan
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Li Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Jiangmin Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
16
|
Paulsen EE, Kilvaer TK, Rakaee M, Richardsen E, Hald SM, Andersen S, Busund LT, Bremnes RM, Donnem T. CTLA-4 expression in the non-small cell lung cancer patient tumor microenvironment: diverging prognostic impact in primary tumors and lymph node metastases. Cancer Immunol Immunother 2017; 66:1449-1461. [PMID: 28707078 PMCID: PMC5645427 DOI: 10.1007/s00262-017-2039-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 07/07/2017] [Indexed: 12/11/2022]
Abstract
The immune checkpoint receptor CTLA-4 plays a crucial part in negatively regulating T cell activation and maintaining self-tolerance. It is frequently overexpressed in a variety of malignancies, yet its prognostic impact in non-small cell lung cancer (NSCLC) remains unclear. We constructed tissue microarrays from tumor tissue samples and evaluated the immunohistochemical expression of CTLA-4 in 536 patients with primary resected stage I-IIIA NSCLC. Expression of CTLA-4 was analyzed in tumor and stromal primary tumor tissue and in locoregional metastatic lymph nodes. CTLA-4 expression in neither tumor epithelial cells (T-CTLA-4) nor stromal cells (S-CTLA-4) of primary tumors was significantly associated with disease-specific survival (DSS) in all patients. However, high S-CTLA-4 expression independently predicted significantly improved DSS in the squamous cell carcinoma subgroup (HR 0.62, 95% CI 0.41-0.93, P = 0.021). In contrast, there was an independent negative prognostic impact of T-CTLA-4 expression in metastatic lymph nodes (HR 1.65, 95% CI 1.03-2.65, P = 0.039). Our results indicate that the expression of CTLA-4 has diverging prognostic impacts in metastatic NSCLC lymph nodes versus primary tumors. The presented results highlight important differences in the tumor microenvironments of primary and metastatic NSCLC tissues, and have potential to guide treatment and clinical sampling strategies.
Collapse
Affiliation(s)
- Erna-Elise Paulsen
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway.
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway.
- Translational Cancer Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9038, Tromso, Norway.
| | - Thomas K Kilvaer
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Mehrdad Rakaee
- Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Elin Richardsen
- Department of Clinical Pathology, University Hospital of North Norway, Mailbox 46, 9038, Tromso, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Sigurd M Hald
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Sigve Andersen
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Lill-Tove Busund
- Department of Clinical Pathology, University Hospital of North Norway, Mailbox 46, 9038, Tromso, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Roy M Bremnes
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| | - Tom Donnem
- Department of Oncology, University Hospital of North Norway, Mailbox 13, 9038, Tromso, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050, Langnes, 9037, Tromso, Norway
| |
Collapse
|
17
|
Dieu-Nosjean MC, Giraldo NA, Kaplon H, Germain C, Fridman WH, Sautès-Fridman C. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev 2016; 271:260-75. [PMID: 27088920 DOI: 10.1111/imr.12405] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The characterization of the microenvironment of human tumors led to the description of tertiary lymphoid structures (TLS) characterized by mature dendritic cells in a T-cell zone adjacent to B-cell follicle including a germinal center. TLS represent sites of lymphoid neogenesis that develop in most solid cancers. Analysis of the current literature shows that the TLS presence is associated with a favorable clinical outcome for cancer patients, regardless of the approach used to quantify TLS and the stage of the disease. Using several approaches that combine immunohistochemistry, gene expression assays, and flow cytometry on large series of lung tumors, our work demonstrated that TLS are important sites for the initiation and/or maintenance of the local and systemic T- and B-cell responses against tumors. Surrounded by high endothelial venules, they represent a privileged area for the recruitment of lymphocytes into tumors and generation of central-memory T and B cells that circulate and limit cancer progression. TLS can be considered as a novel biomarker to stratify the overall survival risk of untreated cancer patients and as a marker of efficient immunotherapies. The induction and manipulation of cancer-associated TLS using drug agonists and/or biotherapies should open new avenues to treat cancer patients.
Collapse
Affiliation(s)
- Marie-Caroline Dieu-Nosjean
- INSERM, UMR_S 1138, Cordeliers Research Center, Team 13 Cancer, Immune Control and Escape, Paris, France.,Sorbonne Paris Cité, UMR_S 1138, Cordeliers Research Center, University Paris Descartes, Paris, France.,Sorbonne Universités, UMR_S 1138, Cordeliers Research Center, UPMC University Paris 06, Paris, France
| | - Nicolas A Giraldo
- INSERM, UMR_S 1138, Cordeliers Research Center, Team 13 Cancer, Immune Control and Escape, Paris, France.,Sorbonne Paris Cité, UMR_S 1138, Cordeliers Research Center, University Paris Descartes, Paris, France.,Sorbonne Universités, UMR_S 1138, Cordeliers Research Center, UPMC University Paris 06, Paris, France
| | - Hélène Kaplon
- INSERM, UMR_S 1138, Cordeliers Research Center, Team 13 Cancer, Immune Control and Escape, Paris, France.,Sorbonne Paris Cité, UMR_S 1138, Cordeliers Research Center, University Paris Descartes, Paris, France.,Sorbonne Universités, UMR_S 1138, Cordeliers Research Center, UPMC University Paris 06, Paris, France
| | - Claire Germain
- INSERM, UMR_S 1138, Cordeliers Research Center, Team 13 Cancer, Immune Control and Escape, Paris, France.,Sorbonne Paris Cité, UMR_S 1138, Cordeliers Research Center, University Paris Descartes, Paris, France.,Sorbonne Universités, UMR_S 1138, Cordeliers Research Center, UPMC University Paris 06, Paris, France
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team 13 Cancer, Immune Control and Escape, Paris, France.,Sorbonne Paris Cité, UMR_S 1138, Cordeliers Research Center, University Paris Descartes, Paris, France.,Sorbonne Universités, UMR_S 1138, Cordeliers Research Center, UPMC University Paris 06, Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team 13 Cancer, Immune Control and Escape, Paris, France.,Sorbonne Paris Cité, UMR_S 1138, Cordeliers Research Center, University Paris Descartes, Paris, France.,Sorbonne Universités, UMR_S 1138, Cordeliers Research Center, UPMC University Paris 06, Paris, France
| |
Collapse
|
18
|
Paulsen EE, Kilvaer TK, Khanehkenari MR, Al-Saad S, Hald SM, Andersen S, Richardsen E, Ness N, Busund LT, Bremnes RM, Donnem T. Assessing PDL-1 and PD-1 in Non-Small Cell Lung Cancer: A Novel Immunoscore Approach. Clin Lung Cancer 2016; 18:220-233.e8. [PMID: 27816392 DOI: 10.1016/j.cllc.2016.09.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) or its ligand, PD-L1, have gained momentum in the treatment of non-small cell lung cancer (NSCLC). However, their prognostic significance remains controversial. The present study evaluated the expression of PD-L1 and PD-1 and their potential role in an Immunoscore, supplementing the TNM classification of NSCLC. MATERIALS AND METHODS Tissue microarrays constructed from tumor tissue samples from 2 cohorts of a total of 536 patients (University Hospital of North Norway, n = 285; Nordland Hospital, n = 251) with primary resected stage I to IIIA NSCLC. PD-L1 and PD-1 were evaluated by immunohistochemistry in the primary tumor and metastatic lymph node tissue. RESULTS In univariate analysis, a high density of PD-L1+ immune cells in the stromal compartment (S-PD-L1) and PD-1+ intraepithelial tumor infiltrating lymphocytes (T-PD-1) was associated with favorable disease-specific survival (DSS; S-PD-L1, P = .004; T-PD-1, P = .012), both limited to the squamous cell carcinoma histologic subgroup (S-PD-L1, P = .002; T-PD-1, P = .034). A combined low S-PD-L1 and T-PD-1 was associated with poor survival in all patients (DSS: hazard ratio [HR], 1.81; 95% confidence interval [CI], 1.37-2.40; P < .001) at both centers and for all pathologic stages. In multivariate analysis, S-PD-L1 and T-PD-1 were independent positive prognostic factors, and combined low scores remained an independent prognosticator for poor survival (DSS: HR, 1.72; 95% CI, 1.29-2.28; P < .001; disease-free survival, P = .001; overall survival, P = .005). CONCLUSION Our study identified S-PD-L1 and T-PD-1 as independent positive prognostic factors for NSCLC patients. Their combination added significant prognostic impact within each pathologic stage and hence are feasible to include in a TNM Immunoscore.
Collapse
Affiliation(s)
- Erna-Elise Paulsen
- Department of Oncology, University Hospital of North Norway, Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.
| | - Thomas K Kilvaer
- Department of Oncology, University Hospital of North Norway, Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | | | - Samer Al-Saad
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Sigurd M Hald
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Sigve Andersen
- Department of Oncology, University Hospital of North Norway, Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Elin Richardsen
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway; Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Nora Ness
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Lill-Tove Busund
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway; Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Roy M Bremnes
- Department of Oncology, University Hospital of North Norway, Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Tom Donnem
- Department of Oncology, University Hospital of North Norway, Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| |
Collapse
|
19
|
Paulsen EE, Kilvaer T, Khanehkenari MR, Maurseth RJ, Al-Saad S, Hald SM, Al-Shibli K, Andersen S, Richardsen E, Busund LT, Bremnes R, Donnem T. CD45RO(+) Memory T Lymphocytes--a Candidate Marker for TNM-Immunoscore in Squamous Non-Small Cell Lung Cancer. Neoplasia 2016; 17:839-48. [PMID: 26678911 PMCID: PMC4681889 DOI: 10.1016/j.neo.2015.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 12/04/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are vital in limiting cancer progression and may supplement the TNM classification. CD45RO+ memory TILs show major prognostic impact in various malignancies but have not been extensively explored in non–small cell lung cancer (NSCLC). In this study, we aimed to evaluate their potential in a NSCLC TNM-Immunoscore. Tissue microarrays were constructed from tumor tissue samples from two cohorts including in total 536 patients (University Hospital of North Norway, n = 285; Nordland Hospital, n = 251) with primary resected stage I to IIIA NSCLC. The density of CD45RO+ and CD8+ TILs in tumor epithelial and stromal compartments of the tumors was evaluated by immunohistochemistry. In univariate analyses, intraepithelial CD45RO+ TIL density (T-CD45RO) was a significant prognostic factor for disease-specific survival (P = .007), limited to the squamous cell carcinoma (SCC) histology subgroup (P < .001), where it was significant in both cohorts (University Hospital of North Norway, P = .003; Nordland Hospital, P = .022). Combining T-CD45RO and stromal CD8+ TIL density (S-CD8) increased the prognostic impact in SCC (P < .001) and showed a significant impact within all pathological stages (I, P = .025; II, P < .001; III, P = .001). In the multivariate analysis, T-CD45RO was an independent positive prognostic factor for SCC (hazard ratio 2.65, 95% confidence interval 1.64-4.28, P < .001), and in combination with S-CD8, the prognostic impact increased vastly (high + high versus low + low: hazard ratio 6.50, 95% confidence interval 3.54-11.91, P < .001). In conclusion, T-CD45RO was an independent prognostic factor for SCC NSCLC. When combined with S-CD8, the prognostic impact increased and was significant within each pathological stage. We propose CD45RO as a candidate marker for TNM-Immunoscore in SCC NSCLC.
Collapse
Affiliation(s)
- Erna-Elise Paulsen
- Department of Oncology, University Hospital of North Norway, Mailbox 100, N-9038 Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050 Langnes, N-9037 Tromso, Norway.
| | - Thomas Kilvaer
- Department of Oncology, University Hospital of North Norway, Mailbox 100, N-9038 Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050 Langnes, N-9037 Tromso, Norway.
| | - Mehrdad Rakaee Khanehkenari
- Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050 Langnes, N-9037 Tromso, Norway.
| | | | - Samer Al-Saad
- Department of Clinical Pathology, University Hospital of North Norway, Mailbox 100, N-9038 Tromso, Norway.
| | - Sigurd M Hald
- Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050 Langnes, N-9037 Tromso, Norway.
| | - Khalid Al-Shibli
- Department of Pathology, Nordland Hospital, Mailbox 1480, 8092 Bodoe, Norway.
| | - Sigve Andersen
- Department of Oncology, University Hospital of North Norway, Mailbox 100, N-9038 Tromso, Norway.
| | - Elin Richardsen
- Department of Clinical Pathology, University Hospital of North Norway, Mailbox 100, N-9038 Tromso, Norway; Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050 Langnes, N-9037 Tromso, Norway.
| | - Lill-Tove Busund
- Department of Clinical Pathology, University Hospital of North Norway, Mailbox 100, N-9038 Tromso, Norway; Department of Medical Biology, UiT The Arctic University of Norway, Mailbox 6050 Langnes, N-9037 Tromso, Norway.
| | - Roy Bremnes
- Department of Oncology, University Hospital of North Norway, Mailbox 100, N-9038 Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050 Langnes, N-9037 Tromso, Norway.
| | - Tom Donnem
- Department of Oncology, University Hospital of North Norway, Mailbox 100, N-9038 Tromso, Norway; Department of Clinical Medicine, UiT The Arctic University of Norway, Mailbox 6050 Langnes, N-9037 Tromso, Norway.
| |
Collapse
|
20
|
Skjefstad K, Grindstad T, Khanehkenari MR, Richardsen E, Donnem T, Kilvaer T, Andersen S, Bremnes RM, Busund LT, Al-Saad S. Prognostic relevance of estrogen receptor α, β and aromatase expression in non-small cell lung cancer. Steroids 2016; 113:5-13. [PMID: 27234503 DOI: 10.1016/j.steroids.2016.05.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/16/2016] [Accepted: 05/20/2016] [Indexed: 02/07/2023]
Abstract
Sex steroids and their receptors are important in the fetal development of normal lung tissue. In addition emerging evidence reveals their significance in lung cancer pathogenesis. This encourages the exploitation of hormone receptors as treatment targets in lung cancer, as it has been successfully used in breast cancer. This study investigates the prognostic impact of estrogen receptor (ER) α and β and the aromatase (AR) enzyme in non-small cell lung cancer (NSCLC) patients. Tumor tissue from 335 NSCLC patients was collected and tissue microarrays (TMAs) were constructed. Immunohistochemical analyses were performed to evaluate the expression of ERα, ERβ and AR in the cytoplasme and nuclei of cells in the tumor epithelial and stromal compartment. By use of survival statistics we investigated the markers impact on disease-specific survival (DSS). Nuclear ERβ expression in tumor epithelial cells in female patients (HR 3.03; 95% CI 1.39-6.61) and tumor cell AR expression in all patients (HR 1.55; 95% CI 1.08-2.23) were significant negative prognostic markers of disease-specific survival in our cohort. High ERβ expression correlates with worse outcome in female patients. Further, patients with high AR expression had an unfavorable prognostic outcome compared with patients expressing low AR levels. These results emphasize the importance of sex steroids role in NSCLC, and, as anti-hormonal drugs are widely available, could lead to the development of novel palliative or even adjuvant treatment strategies in this patient population.
Collapse
Affiliation(s)
- Kaja Skjefstad
- Department of Medical Biology, UiT - The Arctic University of Norway, 9037 Tromso, Norway.
| | - Thea Grindstad
- Department of Medical Biology, UiT - The Arctic University of Norway, 9037 Tromso, Norway
| | | | - Elin Richardsen
- Department of Medical Biology, UiT - The Arctic University of Norway, 9037 Tromso, Norway; Department of Clinical Pathology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Tom Donnem
- Department of Clinical Medicine, UiT - The Arctic University of Norway, 9037 Tromso, Norway; Department of Oncology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Thomas Kilvaer
- Department of Clinical Pathology, University Hospital of North Norway, 9037 Tromso, Norway; Department of Oncology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Sigve Andersen
- Department of Clinical Medicine, UiT - The Arctic University of Norway, 9037 Tromso, Norway; Department of Oncology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Roy M Bremnes
- Department of Clinical Medicine, UiT - The Arctic University of Norway, 9037 Tromso, Norway; Department of Oncology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Lill-Tove Busund
- Department of Medical Biology, UiT - The Arctic University of Norway, 9037 Tromso, Norway; Department of Clinical Pathology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Samer Al-Saad
- Department of Medical Biology, UiT - The Arctic University of Norway, 9037 Tromso, Norway; Department of Clinical Pathology, University Hospital of North Norway, 9037 Tromso, Norway
| |
Collapse
|
21
|
Tao L, Wang S, Zhao Y, Wang AY, Zhang L, Ruan JS, Fan FT, Liu YP, Li Y, Yue ZQ, Qian WH, Chen WX, Lu Y. Pleiotropic effects of herbs characterized with blood-activating and stasis-resolving functions on angiogenesis. Chin J Integr Med 2016; 22:795-800. [PMID: 27358204 DOI: 10.1007/s11655-015-2405-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Indexed: 11/25/2022]
Abstract
Accumulative evidences have underpinned the nature candidates from Chinese medicine (CM), particularly CM served as blood activating and stasis resolving (BASR, Huoxue Huayu in Chinese) by targeting tumor-associated angiogenesis. However, recent experiment research on the therapeutic angiogenesis by BASR-CM attracts wide attention and discussion. This opinion review focused on the underlying link between two indications and anticipated that (1) BASR-CM might emphasize on a balanced multi-cytokines network interaction; (2) BASR-CM might address on the nature of diseases prior to differently affecting physiological and pathological angiogenesis; (3) BASR-CM might mainly act on perivascular cells, either promotes arteriogenesis by increasing arteriogenic factors in ischemic diseases, or simultaneously keep a quiescent vasculature to impede angiogenesis in tumor context.
Collapse
Affiliation(s)
- Li Tao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Sheng Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ai-Yun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lei Zhang
- Department of Pharmacy, Provincial Hospital Affiliated to Anhui Medical University, Hefei, 230001, China
| | - Jun-Shan Ruan
- Fujian Provincial Hospital, Clinical College of Fujian Medical University, Fuzhou, 350001, China
| | - Fang-Tian Fan
- Department of Pharmacology, Hanlin College, Nanjing University of Chinese Medicine, Taizhou Jiangsu Province, 225300, China
| | - Yu-Ping Liu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yao Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhi-Qiang Yue
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wen-Hui Qian
- Department of Pharmaceutics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Wen-Xing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
22
|
Zheng CL, Qiu C, Shen MX, Qu X, Zhang TH, Zhang JH, Du JJ. Prognostic impact of elevation of vascular endothelial growth factor family expression in patients with non-small cell lung cancer: an updated meta-analysis. Asian Pac J Cancer Prev 2016; 16:1881-95. [PMID: 25773840 DOI: 10.7314/apjcp.2015.16.5.1881] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The vascular endothelial growth factor family has been implicated in tumorigenesis and metastasis. The prognostic value of each vascular endothelial growth factor family member, particular VEGF/ VEGFR co-expression, in patients with non-small lung cancer remains controversial. MATERIALS AND METHODS Relevant literature was identified by searching PubMed, EMBASE and Web of Science. Studies evaluating expression of VEGFs and/or VEGFRs by immunohistochemistry or ELISA in lung cancer tissue were eligible for inclusion. Hazard ratios (HRs) and 95% confidence intervals (CIs) from individual study were pooled by using a fixed- or random-effect model, heterogeneity and publication bias analyses were also performed. RESULTS 74 studies covering 7,631 patients were included in the meta-analysis. Regarding pro-angiogenesis factors, the expression of VEGFA (HR=1.633, 95%CI: 1.490-1.791) and VEGFR1 (HR=1.924, 95%CI: 1.220-3.034) was associated separately with poor survival. Especially, VEGFA over-expression was an independent prognostic factor in adenocarcinoma (ADC) (HR=1.775, 95%CI: 1.384-2.275) and SCC (HR=2.919, 95%CI: 2.060-4.137). Co-expression of VEGFA/VEGFR2 (HR=2.011, 95%CI: 1.405-2.876) was also significantly associated with worse survival. For lymphangiogenesis factors, the expression of VEGFC (HR=1.611, 95%CI: 1.407-1.844) predicted a poor prognosis. Co-expression of VEGFC/VEGFR3 (HR=2.436, 95%CI: 1.468-4.043) emerged as a preferable prognostic marker. CONCLUSIONS The expression of VEGFA (particularly in SCC and early stage NSCLC), VEGFC, VEGFR1 indicates separately an unfavorable prognosis in patients with NSCLC. Co-expression VEGFA/ VEGFR2 is comparable with VEGFC/VEGFR3, both featuring sufficient discrimination value as preferable as prognostic biologic markers.
Collapse
Affiliation(s)
- Chun-Long Zheng
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, China E-mail :
| | | | | | | | | | | | | |
Collapse
|
23
|
Kilvaer TK, Paulsen EE, Hald SM, Wilsgaard T, Bremnes RM, Busund LT, Donnem T. Lymphangiogenic Markers and Their Impact on Nodal Metastasis and Survival in Non-Small Cell Lung Cancer--A Structured Review with Meta-Analysis. PLoS One 2015; 10:e0132481. [PMID: 26305218 PMCID: PMC4549062 DOI: 10.1371/journal.pone.0132481] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/15/2015] [Indexed: 12/17/2022] Open
Abstract
Background In non-small cell lung cancer (NSCLC), nodal metastasis is an adverse prognostic factor. Several mediating factors have been implied in the development of nodal metastases and investigated for predictive and prognostic properties in NSCLC. However, study results differ. In this structured review and meta-analysis we explore the published literature on commonly recognized pathways for molecular regulation of lymphatic metastasis in NSCLC. Methods A structured PubMed search was conducted for papers reporting on the expression of known markers of lymhangiogenesis in NSCLC patients. Papers of sufficient quality, presenting survival and/or correlation data were included. Results High levels of vascular endothelial growth factor C (VEGF-C, HR 1.57 95% CI 1.34–1.84) and high lymphatic vascular density (LVD, HR 1.84 95% CI 1.18–2.87) were significant prognostic markers of poor survival and high expression of VEGF-C, vascular endothelial growth factor receptor 3 (VEGFR3) and LVD was associated with lymph node metastasis in NSCLC. Conclusion Lymphangiogenic markers are prognosticators of survival and correlate with lymph node metastasis in NSCLC. Their exact role and clinical implications should be further elucidated.
Collapse
Affiliation(s)
- Thomas K. Kilvaer
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- * E-mail:
| | - Erna-Elise Paulsen
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Sigurd M. Hald
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Tom Wilsgaard
- Department of Community Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Roy M. Bremnes
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Lill-Tove Busund
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Tom Donnem
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| |
Collapse
|
24
|
Kilvaer TK, Khanehkenari MR, Hellevik T, Al-Saad S, Paulsen EE, Bremnes RM, Busund LT, Donnem T, Martinez IZ. Cancer Associated Fibroblasts in Stage I-IIIA NSCLC: Prognostic Impact and Their Correlations with Tumor Molecular Markers. PLoS One 2015; 10:e0134965. [PMID: 26252379 PMCID: PMC4529239 DOI: 10.1371/journal.pone.0134965] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/15/2015] [Indexed: 01/31/2023] Open
Abstract
Background Cancer Associated Fibroblasts (CAFs) are thought to regulate tumor growth and metastasis. Fibroblast Activating Protein 1 (FAP-1) is a marker for fibroblast activation and by many recognized as the main marker of CAFs. Alpha Smooth Muscle Actin (α-SMA) is a general myofibroblast marker, and can be used to identify CAFs. This study investigates the prognostic impact of FAP-1 and α-SMA in non-small cell lung cancer (NSCLC) patients and correlates their expression to 105 proteins investigated in the same cohort. Methods Tumor specimens from 536 NSCLC patients were obtained and tissue micro-arrays were constructed. Immunohistochemistry was used to evaluate the expression of FAP-1 and α-SMA and explore their impact on survival and association with other tumor molecular markers in NSCLC patients. Results High expression of FAP-1, but not α-SMA, in squamous cell carcinoma (SCC, P = 0.043, HR = 0.63 95% CI 0.40–0.99) was significantly associated with increased disease-specific survival. FAP-1 and α-SMA were not significantly correlated to each other. Analyses of FAP-1 and α-SMA associated with other tumor-related proteins revealed histotype-specific correlation patterns. Conclusion The presence of FAP-1 expressing CAFs is an indicator of positive outcome for NSCLC-SCC patients. In addition, correlation analyses suggest FAP-1 and α-SMA to label different subsets of fibroblasts and their associations with other tumor-related proteins diverge according to histological subtype.
Collapse
Affiliation(s)
- Thomas K. Kilvaer
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
- * E-mail:
| | | | - Turid Hellevik
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Samer Al-Saad
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Erna-Elise Paulsen
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Roy M. Bremnes
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Lill-Tove Busund
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | - Tom Donnem
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Inigo Z. Martinez
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| |
Collapse
|
25
|
Zhou L, Yu L, Wu S, Feng Z, Song W, Gong X. Clinicopathological significance of KAI1 expression and epithelial-mesenchymal transition in non-small cell lung cancer. World J Surg Oncol 2015; 13:234. [PMID: 26231404 PMCID: PMC4522085 DOI: 10.1186/s12957-015-0657-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/20/2015] [Indexed: 01/31/2023] Open
Abstract
Background KAI1 and epithelial-mesenchymal transition (EMT) is related to both angiogenesis and lymphangiogenesis and is an important target in new cancer treatment strategies. We aimed to investigate the KAI1 and marker of EMT expression and correlation with lymph node metastasis (LNM) and explore their prognostic impact in non-small cell lung cancer (NSCLC). Methods Tumor tissue specimens from 312 resected patients with stage I–IIIA NSCLC were obtained. Immunohistochemistry was used to assess the expression of the molecular markers KAI1, E-cadherin (E-cad), vimentin, CD34, and D2-40. Results There were 153 N0 and 159 N+ patients. Tumor cell expression of KAI1and the marker of EMT, lymphatic vessel density (LVD), and microvessel density (MVD) were related to LNM. In multivariate analyses, the ages of patients, high tumor cell KAI1 expression, EMT, and the scores of MVD were independent factor of prognosis. Conclusions Tumor cell KAI1 expression, EMT, LVD, and MVD correlate with LNM. Thus, the detection of KAI1, expression of markers of EMT, and the scores of MVD may be used as a potential indicator of NSCLC prognosis.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Pathology, the First Hospital Affiliated of Bengbu Medical College, Bengbu Medical College, No. 287 Changhuai Ave, Longzihu, Bengbu, Anhui Province, 233003, China.
| | - Lan Yu
- Department of Pathology, the First Hospital Affiliated of Bengbu Medical College, Bengbu Medical College, No. 287 Changhuai Ave, Longzihu, Bengbu, Anhui Province, 233003, China.
| | - Shiwu Wu
- Department of Pathology, the First Hospital Affiliated of Bengbu Medical College, Bengbu Medical College, No. 287 Changhuai Ave, Longzihu, Bengbu, Anhui Province, 233003, China.
| | - Zhenzhong Feng
- Department of Pathology, the First Hospital Affiliated of Bengbu Medical College, Bengbu Medical College, No. 287 Changhuai Ave, Longzihu, Bengbu, Anhui Province, 233003, China.
| | - Wenqing Song
- Department of Pathology, the First Hospital Affiliated of Bengbu Medical College, Bengbu Medical College, No. 287 Changhuai Ave, Longzihu, Bengbu, Anhui Province, 233003, China.
| | - Xiaomeng Gong
- Department of Pathology, the First Hospital Affiliated of Bengbu Medical College, Bengbu Medical College, No. 287 Changhuai Ave, Longzihu, Bengbu, Anhui Province, 233003, China.
| |
Collapse
|
26
|
Devery AM, Wadekar R, Bokobza SM, Weber AM, Jiang Y, Ryan AJ. Vascular endothelial growth factor directly stimulates tumour cell proliferation in non-small cell lung cancer. Int J Oncol 2015; 47:849-56. [PMID: 26179332 PMCID: PMC4532222 DOI: 10.3892/ijo.2015.3082] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/30/2015] [Indexed: 01/29/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a key stimulator of physiological and pathological angiogenesis. VEGF signals primarily through VEGF receptor 2 (VEGFR2), a receptor tyrosine kinase whose expression is found predominantly on endothelial cells. The purpose of this study was to determine the role of VEGFR2 expression in NSCLC cells. NSCLC cells and tissue sections were stained for VEGFR2 expression by immunohistochemistry (IHC). Immunoblotting and ELISA were used to determine the activation and inhibition of VEGFR2 and its downstream signalling pathways. Five-day proliferation assays were carried out in the presence or absence of VEGF. IHC analysis of NSCLC demonstrated tumour cell VEGFR2 expression in 20% of samples. Immunoblot analysis showed expression of VEGFR2 protein in 3/8 NSCLC cell lines that correlated with VEGFR2 mRNA expression levels. VEGF-dependent VEGFR2 activation was apparent in NSCLC cells, and was associated with increased tumor cell proliferation. Cediranib treatment or siRNA against VEGFR2 inhibited VEGF-dependent increases in cell proliferation. Inhibition of VEGFR2 tyrosine kinase activity using cediranib was more effective than inhibition of AKT (MK2206) or MEK (AZD6244) for overcoming VEGFR2-driven cell proliferation. VEGF treatment did not affect cell survival following treatment with radiation, cisplatin, docetaxel or gemcitabine. Our data suggest that a subset of NSCLC tumour cells express functional VEGFR2 which can act to promote VEGF-dependent tumour cell growth. In this tumour subset, therapies targeting VEGFR2 signalling, such as cediranib, have the potential to inhibit both tumour cell proliferation and angiogenesis.
Collapse
Affiliation(s)
- Aoife M Devery
- CRUK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, UK
| | - Rekha Wadekar
- CRUK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, UK
| | - Sivan M Bokobza
- CRUK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, UK
| | - Anika M Weber
- CRUK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, UK
| | - Yanyan Jiang
- CRUK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, UK
| | - Anderson J Ryan
- CRUK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
27
|
Pøhl M, Olsen KE, Holst R, Donnem T, Busund LT, Bremnes RM, Al-Saad S, Andersen S, Richardsen E, Ditzel HJ, Hansen O. Keratin 34betaE12/keratin7 expression is a prognostic factor of cancer-specific and overall survival in patients with early stage non-small cell lung cancer. Acta Oncol 2015; 55:167-77. [PMID: 26057535 DOI: 10.3109/0284186x.2015.1049291] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Carcinomas and their metastases often retain the keratin patterns of their epithelial origin, and are therefore useful as lineage-specific markers in diagnostic pathology. Recently, it has become clear that intermediate filaments composed by keratins play a role in modulation of cell proliferation, migration, and possibly cancer invasion, factors impacting prognosis in early stage non-small cell lung cancer (NSCLC). MATERIAL AND METHODS Tumor tissue from a retrospective Danish cohort of 177 patients with completely resected NSCLC, stage I-IIIA tumors, were analyzed for keratin 7 (K7) and keratin 34βE12 expression by immunohistochemistry and validated in a comparable independent Norwegian cohort of 276 stage I-IIIA NSCLC patients. RESULTS Based on keratin 34βE12/K7 expression, three subgroups with significantly different median cancer-specific survival rates were identified (34βE12+/K7+, 168 months vs. 34βE12+/K7+, 73 months vs. 34βE12-/K7+, 30 months; p = 0.0004). In multivariate analysis, stage II-IIIA (HR 2.9), 34βE12+/K7+ (HR 1.90) and 34βE12-/K7+ (HR 3.7), were prognostic factors of poor cancer-specific survival (CSS) (p < 0.001). Validation in the Norwegian cohort confirmed that stage II-IIIA (HR 2.3), 34βE12+/K7+ (HR 1.6), and 34βE12-/K7+ (HR 2.0) were prognostic factors of poor CSS (p < 0.05). Multivariate Cox proportional-hazard analysis demonstrated that 34βE12+/K7 + and 34βE12+/K7 + status was significantly associated with poor overall survival (p < 0.05). CONCLUSION Keratin 34βE12/K7 expression is a prognostic parameter in resected early stage NSCLC that allows identification of high-risk NSCLC patients with poor cancer-specific and overall survival.
Collapse
Affiliation(s)
- Mette Pøhl
- a Department of Oncology , Odense University Hospital , Odense , Denmark
- b Institute of Clinical Research, University of Southern Denmark , Odense , Denmark
- f Department of Oncology , Rigshospitalet , Copenhagen , Denmark
| | - Karen Ege Olsen
- b Institute of Clinical Research, University of Southern Denmark , Odense , Denmark
- c Department of Pathology , Odense University Hospital , Odense , Denmark
| | - Rene Holst
- d Department of Statistics , University of Southern Denmark , Odense , Denmark
| | - Tom Donnem
- g Institute of Clinical Medicine, University of Tromso , Tromso , Norway
- h Department of Oncology , University Hospital of North Norway , Tromso , Norway
| | - Lill-Tove Busund
- i Institute of Medical Biology, University of Tromso , Tromso , Norway
- j Department of Clinical Pathology , University Hospital of North Norway , Tromso , Norway
| | - Roy M Bremnes
- g Institute of Clinical Medicine, University of Tromso , Tromso , Norway
- h Department of Oncology , University Hospital of North Norway , Tromso , Norway
| | - Samer Al-Saad
- i Institute of Medical Biology, University of Tromso , Tromso , Norway
- j Department of Clinical Pathology , University Hospital of North Norway , Tromso , Norway
| | - Sigve Andersen
- g Institute of Clinical Medicine, University of Tromso , Tromso , Norway
- h Department of Oncology , University Hospital of North Norway , Tromso , Norway
| | - Elin Richardsen
- i Institute of Medical Biology, University of Tromso , Tromso , Norway
- j Department of Clinical Pathology , University Hospital of North Norway , Tromso , Norway
| | - Henrik J Ditzel
- a Department of Oncology , Odense University Hospital , Odense , Denmark
- e Department of Cancer and Inflammation Research , Institute of Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Olfred Hansen
- a Department of Oncology , Odense University Hospital , Odense , Denmark
- b Institute of Clinical Research, University of Southern Denmark , Odense , Denmark
| |
Collapse
|
28
|
Skjefstad K, Richardsen E, Donnem T, Andersen S, Kiselev Y, Grindstad T, Hald SM, Al-Shibli K, Bremnes RM, Busund LT, Al-Saad S. The prognostic role of progesterone receptor expression in non-small cell lung cancer patients: Gender-related impacts and correlation with disease-specific survival. Steroids 2015; 98:29-36. [PMID: 25668612 DOI: 10.1016/j.steroids.2015.01.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/21/2015] [Accepted: 01/22/2015] [Indexed: 01/05/2023]
Abstract
PURPOSE Progesterone has been shown to impact the development of hormone-sensitive cancers, such as breast and ovarian cancers. Emerging evidence has revealed a possible role of progesterone in the tumorigenesis of other cancers, including lung cancer. Herein, we aimed to elucidate the prevalence and prognostic significance of progesterone receptor (PR) expression in non-small cell lung cancer (NSCLC) tissue. EXPERIMENTAL Tumor tissue samples were collected from our patient cohort consisting of 335 NSCLC patients with stage I-IIIA disease. Tissue microarrays (TMAs) were constructed, and immunohistochemical (IHC) analyses were performed to evaluate the PR expression in the tumor epithelial and stromal compartments. RESULTS In a univariate analysis, positive PR expression in the stromal tumor compartment (P=0.005) was significantly and independently associated with a favorable outcome for both genders. Furthermore, positive PR expression in tumor epithelial cells (P=0.003) correlated with a poor prognosis for female patients. In a multivariate analysis, positive PR expression in the tumor stroma (P=0.007) was an independent prognostic factor for improved disease-specific survival (DSS). Positive PR expression in tumor epithelial cells emerged as an independent prognostic factor in female patients (P=0.001) for poor DSS. CONCLUSIONS We show that PR expression in tumor-surrounding stromal cells is associated with improved DSS for both male and female patients. Additionally, we reveal that positive PR expression in tumor epithelial cells is an independent, unfavorable prognosticator for DSS in female patients, making PR expression a potential marker for prognostic stratification in NSCLC.
Collapse
Affiliation(s)
- Kaja Skjefstad
- Department of Medical Biology, University of Tromso, 9037 Tromso, Norway.
| | - Elin Richardsen
- Department of Medical Biology, University of Tromso, 9037 Tromso, Norway; Department of Clinical Pathology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Tom Donnem
- Department of Clinical Medicine, University of Tromso, 9037 Tromso, Norway; Department of Oncology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Sigve Andersen
- Department of Clinical Medicine, University of Tromso, 9037 Tromso, Norway; Department of Oncology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Yury Kiselev
- Department of Medical Biology, University of Tromso, 9037 Tromso, Norway; Department of Pharmacy, University of Tromso, 9037 Tromso, Norway
| | - Thea Grindstad
- Department of Medical Biology, University of Tromso, 9037 Tromso, Norway
| | - Sigurd M Hald
- Department of Clinical Medicine, University of Tromso, 9037 Tromso, Norway
| | - Khalid Al-Shibli
- Department of Medical Biology, University of Tromso, 9037 Tromso, Norway; Department of Pathology, Nordland Central Hospital, 8005 Bodo, Norway
| | - Roy M Bremnes
- Department of Clinical Medicine, University of Tromso, 9037 Tromso, Norway; Department of Oncology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Lill-Tove Busund
- Department of Medical Biology, University of Tromso, 9037 Tromso, Norway; Department of Clinical Pathology, University Hospital of North Norway, 9037 Tromso, Norway
| | - Samer Al-Saad
- Department of Medical Biology, University of Tromso, 9037 Tromso, Norway; Department of Clinical Pathology, University Hospital of North Norway, 9037 Tromso, Norway
| |
Collapse
|
29
|
Pomme G, Augustin F, Fiegl M, Droeser RA, Sterlacci W, Tzankov A. Detailed assessment of microvasculature markers in non-small cell lung cancer reveals potentially clinically relevant characteristics. Virchows Arch 2015; 467:55-66. [DOI: 10.1007/s00428-015-1767-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/11/2015] [Accepted: 03/20/2015] [Indexed: 11/28/2022]
|
30
|
Donnem T, Hald SM, Paulsen EE, Richardsen E, Al-Saad S, Kilvaer TK, Brustugun OT, Helland A, Lund-Iversen M, Poehl M, Olsen KE, Ditzel HJ, Hansen O, Al-Shibli K, Kiselev Y, Sandanger TM, Andersen S, Pezzella F, Bremnes RM, Busund LT. Stromal CD8+ T-cell Density—A Promising Supplement to TNM Staging in Non-Small Cell Lung Cancer. Clin Cancer Res 2015; 21:2635-43. [PMID: 25680376 DOI: 10.1158/1078-0432.ccr-14-1905] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 02/01/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunoscore is a prognostic tool defined to quantify in situ immune cell infiltrates, which appears to be superior to the tumor-node-metastasis (TNM) classification in colorectal cancer. In non-small cell lung cancer (NSCLC), no immunoscore has been established, but in situ tumor immunology is recognized as highly important. We have previously evaluated the prognostic impact of several immunological markers in NSCLC, yielding the density of stromal CD8(+) tumor-infiltrating lymphocytes (TIL) as the most promising candidate. Hence, we validate the impact of stromal CD8(+) TIL density as an immunoscore in NSCLC. EXPERIMENTAL DESIGN The prognostic impact of stromal CD8(+) TILs was evaluated in four different cohorts from Norway and Denmark consisting of 797 stage I-IIIA NSCLC patients. The Tromso cohort (n = 155) was used as training set, and the results were further validated in the cohorts from Bodo (n = 169), Oslo (n = 295), and Denmark (n = 178). Tissue microarrays and clinical routine CD8 staining were used for all cohorts. RESULTS Stromal CD8(+) TIL density was an independent prognostic factor in the total material (n = 797) regardless of the endpoint: disease-free survival (P < 0.001), disease-specific survival (P < 0.001), or overall survival (P < 0.001). Subgroup analyses revealed significant prognostic impact of stromal CD8(+) TIL density within each pathologic stage (pStage). In multivariate analysis, stromal CD8(+) TIL density and pStage were independent prognostic variables. CONCLUSIONS Stromal CD8(+) TIL density has independent prognostic impact in resected NSCLC, adds prognostic impact within each pStage, and is a good candidate marker for establishing a TNM-Immunoscore.
Collapse
Affiliation(s)
- Tom Donnem
- Department of Oncology, University Hospital of North Norway, Tromso, Norway. Institute of Clinical Medicine, The Arctic University of Norway, Tromso, Norway.
| | - Sigurd M Hald
- Institute of Clinical Medicine, The Arctic University of Norway, Tromso, Norway
| | - Erna-Elise Paulsen
- Department of Oncology, University Hospital of North Norway, Tromso, Norway. Institute of Clinical Medicine, The Arctic University of Norway, Tromso, Norway
| | - Elin Richardsen
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway. Institute of Medical Biology, The Arctic University of Norway, Tromso, Norway
| | - Samer Al-Saad
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway. Institute of Medical Biology, The Arctic University of Norway, Tromso, Norway
| | - Thomas K Kilvaer
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Odd Terje Brustugun
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway. Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Aslaug Helland
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway. Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Mette Poehl
- Department of Oncology, Rigshospitalet, Copenhagen, Denmark. Department of Oncology, Odense University Hospital, Odense, Denmark. Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Karen Ege Olsen
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark. Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Henrik J Ditzel
- Department of Oncology, Odense University Hospital, Odense, Denmark. Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Olfred Hansen
- Department of Oncology, Odense University Hospital, Odense, Denmark. Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Yury Kiselev
- Institute of Medical Biology, The Arctic University of Norway, Tromso, Norway. Department of Pharmacy, The Arctic University of Tromso, Tromso, Norway
| | - Torkjel M Sandanger
- Department of Community Medicine, The Artic University of Tromso, Tromso, Norway
| | - Sigve Andersen
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
| | - Francesco Pezzella
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Roy M Bremnes
- Department of Oncology, University Hospital of North Norway, Tromso, Norway. Institute of Clinical Medicine, The Arctic University of Norway, Tromso, Norway
| | - Lill-Tove Busund
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway. Institute of Medical Biology, The Arctic University of Norway, Tromso, Norway
| |
Collapse
|
31
|
Lønvik K, Sørbye SW, Nilsen MN, Paulssen RH. Prognostic value of the MicroRNA regulators Dicer and Drosha in non-small-cell lung cancer: co-expression of Drosha and miR-126 predicts poor survival. BMC Clin Pathol 2014; 14:45. [PMID: 25525410 PMCID: PMC4269969 DOI: 10.1186/1472-6890-14-45] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/03/2014] [Indexed: 12/26/2022] Open
Abstract
Background Dicer and Drosha are important enzymes for processing microRNAs. Recent studies have exhibited possible links between expression of different miRNAs, levels of miRNA processing enzymes, and cancer prognosis. We have investigated the prognostic impact of Dicer and Drosha and their correlation with miR-126 expression in a large cohort of non-small cell lung cancer (NSCLC) patients. We aimed to find patient groups within the cohort that might have an advantage of receiving adjunctive therapies. Methods Dicer expression in the cytoplasm and Drosha expression in the nucleus were evaluated by manual immunohistochemistry of tissue microarrays (TMAs), including tumor tissue samples from 335 patients with resected stages I to IIIA NSCLC. In addition, in situ hybridizations of TMAs for visualization of miR-126 were performed. Kaplan–Meier analysis was performed, and the log-rank test via SPSS v.22 was used for estimating significance levels. Results In patients with normal performance status (ECOG = 0, n = 197), high Dicer expression entailed a significantly better prognosis than low Dicer expression (P = 0.024). Dicer had no significant prognostic value in patients with reduced performance status (ECOG = 1–2, n = 138). High Drosha expression was significantly correlated with high levels of the microRNA 126 (miR-126) (P = 0.004). Drosha/miR-126 co-expression had a significant negative impact on the disease-specific survival (DSS) rate (P < 0.001). Multivariate analyses revealed that the interaction Dicer*Histology (P = 0.049) and Drosha/miR-126 co-expression (P = 0.033) were independent prognostic factors. Conclusions In NSCLC patients with normal performance status, Dicer is a positive prognostic factor. The importance of Drosha as a prognostic factor in our material seems to be related to miR-126 and possibly other microRNAs. Electronic supplementary material The online version of this article (doi:10.1186/1472-6890-14-45) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kenneth Lønvik
- Department of Clinical Pathology, University Hospital of Northern Norway, N-9038 Tromsø, Norway ; Department of Medical Biology, Tromsø, Norway
| | - Sveinung W Sørbye
- Department of Clinical Pathology, University Hospital of Northern Norway, N-9038 Tromsø, Norway
| | | | - Ruth H Paulssen
- Department of Clinical Medicine, UiT - The Arctic University of Norway, N-9037 Tromsø, Norway
| |
Collapse
|
32
|
Karim NA, Bui H, Pathrose P, Starnes S, Patil N, Shehata M, Mostafa A, Rao M, Zarzour A, Anderson M. The use of pharmacogenomics for selection of therapy in non-small-cell lung cancer. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2014; 8:139-44. [PMID: 25520568 PMCID: PMC4259862 DOI: 10.4137/cmo.s18369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/21/2014] [Accepted: 09/24/2014] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Performance status (PS) is the only known clinical predictor of outcome in patients with advanced non-small-cell lung cancer (NSCLC), although pharmacogenomic markers may also correlate with outcome. The aim of our study was to correlate clinical and pharmacogenomic measures with overall survival. METHODS This was an IRB approved, retrospective study in which the medical records of 50 patients with advanced NSCLC from 1998–2008 were reviewed, and gender, race, PS, and chemotherapy regimens were documented. Stromal expression of pharmacogenomic markers (VEGFR, ERCC1, 14-3-3σ, pAKT, and PTEN) was measured. Clinical factors and pharmacogenomics markers were compared to overall survival using a Cox proportional hazards model. RESULTS Forty patients received platinum-based therapy. Median age was 65 years. Improved PS, female gender, and gemcitabine therapy were significantly associated with longer overall survival (P = 0.004, P = 0.04, and P = 0.003, respectively). Age was not associated with survival. Caucasians had better overall survival in comparison to African Americans with median survival of 14.8 months versus 10.4 months (P = 0.1). Patients treated with platinum-based therapy had better survival of 15 months versus 8 months for non-platinum based therapy (P = 0.01). There was no significant association between any of the pharmacogenomics markers and overall survival other than in patients treated with platinum, in whom ERCC1 negativity was strongly associated with longer survival (P = 0.007). CONCLUSION ERCC1 negativity with platinum therapy, gemcitabine therapy, good PS, and female gender all correlated with improved overall survival in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Nagla A Karim
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Hai Bui
- Department of Pathology, Cincinnati VA Medical Center, Cincinnati, OH, USA
| | - Peterson Pathrose
- Divison of Thoracic Surgery, Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Sandra Starnes
- Divison of Thoracic Surgery, Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Ninad Patil
- Department of Pathology, Mount Sinai Medical Center, New York, NY, USA
| | - Mahmoud Shehata
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Ahmed Mostafa
- Division of Pulmonary, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Mb Rao
- Division of Environmental Health, University of Cincinnati, Cincinnati, OH, USA
| | - Ahmad Zarzour
- Internal Medicine Department, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Marshall Anderson
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
33
|
Monocarboxylate transporters 1-4 in NSCLC: MCT1 is an independent prognostic marker for survival. PLoS One 2014; 9:e105038. [PMID: 25225794 PMCID: PMC4165596 DOI: 10.1371/journal.pone.0105038] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/19/2014] [Indexed: 01/07/2023] Open
Abstract
Introduction Monocarboxylate transporters (MCTs) 1–4 are lactate transporters crucial for cancers cells adaption to upregulated glycolysis. Herein, we aimed to explore their prognostic impact on disease-specific survival (DSS) in both cancer and tumor stromal cells in NSCLC. Methods Tissue micro arrays (TMAs) were constructed, representing both cancer and stromal tumor tissue from 335 unselected patients diagnosed with stage I–IIIA NSCLC. Immunohistochemistry was used to evaluate the expression of MCT1-4. Results In univariate analyses; ↓MCT1 (P = 0.021) and ↑MCT4 (P = 0.027) expression in cancer cells, and ↑MCT1 (P = 0.003), ↓MCT2 (P = 0.006), ↓MCT3 (P = 0.020) expression in stromal cells correlated significantly with a poor DSS. In multivariate analyses; ↓MCT1 expression in cancer cells (HR: 1.9, CI 95%: 1.3–2.8, P = 0.001), ↓MCT2 (HR: 2.4, CI 95%: 1.5–3.9, P<0.001), ↓MCT3 (HR: 1.9, CI 95%: 1.1–3.5, P = 0.031) and ↑MCT1 expression in stromal cells (HR: 1.7, CI 95%: 1.1–2.7, P = 0.016) were significant independent poor prognostic markers for DSS. Conclusions We provide novel information of MCT1 as a candidate marker for prognostic stratification in NSCLC. Interestingly, MCT1 shows diverging, independent prognostic impact in the cancer cell and stromal cell compartments.
Collapse
|
34
|
Thromboxane synthase expression and correlation with VEGF and angiogenesis in non-small cell lung cancer. Biochim Biophys Acta Mol Basis Dis 2014; 1842:747-55. [DOI: 10.1016/j.bbadis.2014.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 12/20/2013] [Accepted: 01/22/2014] [Indexed: 10/25/2022]
|
35
|
Sasahira T, Ueda N, Yamamoto K, Kurihara M, Matsushima S, Bhawal UK, Kirita T, Kuniyasu H. Prox1 and FOXC2 act as regulators of lymphangiogenesis and angiogenesis in oral squamous cell carcinoma. PLoS One 2014; 9:e92534. [PMID: 24647631 PMCID: PMC3960274 DOI: 10.1371/journal.pone.0092534] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/24/2014] [Indexed: 12/22/2022] Open
Abstract
Prospero homeobox 1 (Prox1) and forkhead box (FOX) C2 regulate angiogenesis and/or lymphangiogenesis. However, the detailed role and function of Prox1 and FOXC2 in cancer remains controversial. In the present study, we examined the expression of Prox1 and FOXC2 proteins in specimens from 163 cases with oral squamous cell carcinoma (OSCC). Furthermore, the role of Prox1 and FOXC2 in cancer cell growth and invasion was evaluated in cultured OSCC cells. Prox1 expression was significantly associated with local progression of the tumor (P = 0.0023), clinical stage (P<0.0001), lymphovessel density (LVD) (P<0.0001), nodal metastasis (P<0.0001), and worse prognosis (P<0.0001). Immunoreactivity of FOXC2 was strongly correlated with microvessel density (MVD) (P<0.0001) and poor prognosis (P = 0.0076). In vitro analysis demonstrated that Prox1 regulates cell growth, proliferation, invasion, and lymphangiogenesis by activating vascular endothelial growth factor (VEGF)-C expression. Furthermore, FOXC2 enhanced the expression level of Prox1 and promoted angiogenesis by enhancement of VEGF-A expression. Our results suggested that Prox1 and FOXC2 play key roles in OSCC progression and that further studies focusing on these proteins may yield useful insights for diagnosis and therapy of OSCC.
Collapse
Affiliation(s)
- Tomonori Sasahira
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Nobuhiro Ueda
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Kazuhiko Yamamoto
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Miyako Kurihara
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Sayako Matsushima
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Ujjal K. Bhawal
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
- * E-mail:
| |
Collapse
|
36
|
Stenvold H, Donnem T, Andersen S, Al-Saad S, Busund LT, Bremnes RM. Stage and tissue-specific prognostic impact of miR-182 in NSCLC. BMC Cancer 2014; 14:138. [PMID: 24575749 PMCID: PMC3996062 DOI: 10.1186/1471-2407-14-138] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 02/12/2014] [Indexed: 11/10/2022] Open
Abstract
Background MicroRNA (miR)-182 is frequently upregulated in cancers, has generally been viewed as an oncogene and is possibly connected to angiogenesis. We aimed to explore what impact miR-182 has in non-small cell lung cancer (NSCLC), and more explicitly its correlation with angiogenic markers. Methods From 335 unselected stage I to IIIA NSCLC carcinomas, duplicate tumor and tumor-associated stromal cores were collected in tissue microarray blocks (TMAs). In situ hybridization (ISH) was used to detect the expression of miR-182 in tumor cells, and immunohistochemistry (IHC) was used to detect the expression of angiogenesis related protein markers. Results In univariate analyses, high tumor cell expression of miR-182 was a positive prognostic factor for patients with squamous cell carcinoma (SCC, P = 0.042) and stage II patients (P = 0.003). Also in the multivariate analysis, high tumor cell miR-182 expression was associated with a good prognosis in the same groups (SCC: HR 0.57, CI 95% 0.33-0.99, P = 0.048; stage II: HR 0.50, CI 95% 0.28-0.90, P = 0.020). We found significant correlations between miR-182 and the angiogenesis related markers FGF2, HIF2α and MMP-7. Conclusion In patients with SCC and in stage II patients, high tumor cell miR-182 expression is an independent positive prognostic factor.
Collapse
Affiliation(s)
- Helge Stenvold
- Institute of Clinical Medicine, University of Tromso, Tromso, Norway.
| | | | | | | | | | | |
Collapse
|
37
|
Stenvold H, Donnem T, Andersen S, Al-Saad S, Valkov A, Pedersen MI, Busund LT, Bremnes RM. High tumor cell expression of microRNA-21 in node positive non-small cell lung cancer predicts a favorable clinical outcome. BMC Clin Pathol 2014; 14:9. [PMID: 24524655 PMCID: PMC3931486 DOI: 10.1186/1472-6890-14-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 01/27/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND MicroRNA (miR)-21 has been revealed as an oncogene in cancer development, and is one of the miRNAs closely connected to angiogenesis. We aimed to explore the impact of miR-21 expression in both tumor and stromal compartments of non-small cell lung cancer (NSCLC), and correlations between miR-21 and angiogenic protein markers. METHODS From 335 unselected stage I to IIIA NSCLC carcinomas, duplicate tumor and tumor-associated stromal cores were collected in tissue microarrays (TMAs). In situ hybridization (ISH) was used to detect the expression of miR-21 separately in tumor cells and stromal cells of the tumor, and immunohistochemistry (IHC) was used to detect the expression of the protein markers protein kinase B (Akt), phosphatidylinositol-3-kinase (PI3K), hypoxia induced factor 1 (HIF1α) and vascular endothelial growth factor-A (VEGF-A). RESULTS In univariate analyses, high tumor cell expression of miR-21 in patients with lymph node metastasis was a positive prognostic factor (P = 0.024). High stromal miR-21 expression had a negative prognostic impact (P = 0.022). In the multivariate analysis, low tumor mir-21 expression in node positive patients was an independent adverse prognostic factor (HR 2.03, CI 95% 1.09-3.78, P = 0.027). CONCLUSIONS In patients with lymph node metastasis, miR-21 expression in tumor cells is an independent positive prognostic factor. High stromal miR-21 expression is a negative prognostic factor.
Collapse
Affiliation(s)
- Helge Stenvold
- Institute of Clinical Medicine, University of Tromso, Tromso, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Kim HS, Lee HE, Yang HK, Kim WH. High lactate dehydrogenase 5 expression correlates with high tumoral and stromal vascular endothelial growth factor expression in gastric cancer. Pathobiology 2013; 81:78-85. [PMID: 24401755 DOI: 10.1159/000357017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 11/05/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Lactate dehydrogenase 5 (LDH5) is a major lactate dehydrogenase isoenzyme catalyzing the transformation of pyruvate to lactate to provide anaerobic energy. Vascular endothelial growth factor (VEGF) is expressed in both tumor and stromal cells in gastric cancer. Our aim was to study the prognostic effect of LDH5, and tumoral and stromal expression of the angiogenic factor VEGF in gastric cancer, and the intercorrelation of tissue expression of both factors. METHODS Tissue microarray analysis of 382 consecutive gastric cancer resection specimens was used for immunohistochemistry of LDH5 and VEGF, and expression of LDH5, tumoral VEGF, and stromal VEGF was categorized into low and high groups. RESULTS High expression was observed for LDH5 in 57.9% (219/378), tumoral VEGF in 35.7% (136/381), and stromal VEGF in 58.5% (223/381) of the specimens. Regarding high expression of LDH5 and VEGF, significant associations with intestinal type, advanced gastric cancer, lymph node metastasis, higher TNM stage, and upper-third location were noted. Positive intercorrelations occurred among the expression of LDH5 and VEGF. Results of survival analyses revealed a significant association of high expression of LDH5 and VEGF with lower survival (overall and disease-free survival). Five-year survival rates were significantly lower in tumors with high LDH5 and tumoral VEGF expression in diffuse- or mixed-type cancers and high expression of stromal VEGF in intestinal-type cancer. CONCLUSION The results of our study showed that high LDH5 and VEGF expression in both tumor and stroma was a prognostic factor for patients with gastric cancers, especially diffuse- or mixed-type cancers. Therefore, LDH5 expression may play a role in the regulation of tumoral and stromal VEGF expression in gastric cancer. Our results suggest the potential use of LDH5 expression as a biomarker for response to VEGF-targeted therapy.
Collapse
Affiliation(s)
- Hee Sung Kim
- Department of Pathology, Chung Ang University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
39
|
Holzer TR, Fulford AD, Nedderman DM, Umberger TS, Hozak RR, Joshi A, Melemed SA, Benjamin LE, Plowman GD, Schade AE, Ackermann BL, Konrad RJ, Nasir A. Tumor cell expression of vascular endothelial growth factor receptor 2 is an adverse prognostic factor in patients with squamous cell carcinoma of the lung. PLoS One 2013; 8:e80292. [PMID: 24244672 PMCID: PMC3828187 DOI: 10.1371/journal.pone.0080292] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/02/2013] [Indexed: 11/19/2022] Open
Abstract
A robust immunohistochemical (IHC) assay for VEGFR2 was developed to investigate its utility for patient tailoring in clinical trials. The sensitivity, specificity, and selectivity of the IHC assay were established by siRNA knockdown, immunoblotting, mass spectrometry, and pre-absorption experiments. Characterization of the assay included screening a panel of multiple human cancer tissues and an independent cohort of non-small cell lung carcinoma (NSCLC, n = 118) characterized by TTF-1, p63, CK5/6, and CK7 IHC. VEGFR2 immunoreactivity was interpreted qualitatively (VEGFR2 positive/negative) in blood vessels and by semi-quantitative evaluation using H-scores in tumor cells (0-300). Associations were determined among combinations of VEGFR2 expression in blood vessels and tumor cells, and clinico-pathologic characteristics (age, sex, race, histologic subtype, disease stage) and overall survival using Kaplan-Meier analyses and appropriate statistical models. VEGFR2 expression both in blood vessels and in tumor cells in carcinomas of the lung, cervix, larynx, breast, and others was demonstrated. In the validation cohort, 99/118 (83.9%) NSCLC tissues expressed VEGFR2 in the blood vessels and 46/118 (39.0%) showed high tumor cell positivity (H-score ≥10). Vascular and tumor cell expression were inversely correlated (p = 0.0175). High tumor cell expression of VEGFR2 was associated with a 3.7-fold reduction in median overall survival in lung squamous-cell carcinoma (SCC, n = 25, p = 0.0134). The inverse correlation between vascular and tumor cell expression of VEGFR2 and the adverse prognosis associated with high VEGFR2 expression in immunohistochemically characterized pulmonary SCC are new findings with potential therapeutic implications. The robustness of this novel IHC assay will support further evaluation of its utility for patient tailoring in clinical trials of antiangiogenic agents.
Collapse
Affiliation(s)
- Timothy R. Holzer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Angie D. Fulford
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Drew M. Nedderman
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Tara S. Umberger
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Rebecca R. Hozak
- Oncology Statistics-Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Adarsh Joshi
- Oncology Statistics-Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Symantha A. Melemed
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Laura E. Benjamin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Gregory D. Plowman
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Andrew E. Schade
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Bradley L. Ackermann
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Robert J. Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Aejaz Nasir
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
40
|
Positive prognostic impact of miR-210 in non-small cell lung cancer. Lung Cancer 2013; 83:272-8. [PMID: 24305009 DOI: 10.1016/j.lungcan.2013.11.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 10/10/2013] [Accepted: 11/06/2013] [Indexed: 12/20/2022]
Abstract
OBJECTIVES miR-210 is an important regulator of the cellular response to hypoxia. Therefore, we aimed to explore the prognostic significance of miR-210 in non-small cell lung cancer (NSCLC) patients with stage I-IIIA disease. MATERIALS AND METHODS In addition to clinicopathological and demograpic information, tumor tissues were collected and tissue micro arrays (TMAs) were constructed from 335 patients with stage I-IIIA NSCLC. Expression of miR-210 in cancer cells and stromal cells of the tumor was assessed by in situ hybridization. RESULTS In univariate analyses, high cancer cell (p=0.039) and high stromal cell expression (p=0.008) of miR-210 were both significantly associated with an improved disease-spesific survival (DSS). High co-expression of miR-210 in cancer and stromal cells was also a positive prognostic factor for DSS (p=0.010). In multivariate analysis, miR-210 in stromal cells (p=0.011), and miR-210 co-expressed in cancer and stromal cells was an independent prognosticator for DSS (p=0.011). CONCLUSIONS We show that miR-210 in stromal cells, and co-expressed in cancer cells and stromal cells mediates an independent prognostic impact. It is a candidate marker for prognostic stratification in NSCLC.
Collapse
|
41
|
Sanmartín E, Sirera R, Usó M, Blasco A, Gallach S, Figueroa S, Martínez N, Hernando C, Honguero A, Martorell M, Guijarro R, Rosell R, Jantus-Lewintre E, Camps C. A gene signature combining the tissue expression of three angiogenic factors is a prognostic marker in early-stage non-small cell lung cancer. Ann Surg Oncol 2013; 21:612-20. [PMID: 24145997 DOI: 10.1245/s10434-013-3330-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Angiogenesis and lymphangiogenesis are key mechanisms for tumor growth and dissemination. They are mainly regulated by the vascular endothelial growth factor (VEGF) family of ligands and receptors. The aim of this study was to analyze relative expression levels of angiogenic markers in resectable non-small cell lung cancer patients in order to asses a prognostic signature that could improve characterization of patients with worse clinical outcomes. METHODS RNA was obtained from tumor and normal lung specimens from 175 patients. Quantitative polymerase chain reaction was performed to analyze the relative expression of HIF1A, PlGF, VEGFA, VEGFA165b, VEGFB, VEGFC, VEGFD, VEGFR1, VEGFR2, VEGFR3, NRP1 and NRP2. RESULTS Univariate analysis showed that tumor size and ECOG-PS are prognostic factors for time to progression (TTP) and overall survival (OS). This analysis in the case of angiogenic factors also revealed that PlGF, VEGFA, VEGFB and VEGFD distinguish patients with different outcomes. Taking into account the complex interplay between the different ligands of the VEGF family and to more precisely predict the outcome of the patients, we considered a new analysis combining several VEGF ligands. In order to find independent prognostic variables, we performed a multivariate Cox analysis, which showed that the subgroup of patients with higher relative expression of VEGFA plus lower VEGFB and VEGFD presented the poorest outcome for both TTP and OS. CONCLUSIONS The relative expression of these three genes can be considered as an angiogenic gene signature whose applicability for the selection of candidates for targeted therapies needs to be further validated.
Collapse
Affiliation(s)
- Elena Sanmartín
- Molecular Oncology Laboratory, Fundación Investigación, Hospital General Universitario de Valencia, Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Goc J, Fridman WH, Sautès-Fridman C, Dieu-Nosjean MC. Characteristics of tertiary lymphoid structures in primary cancers. Oncoimmunology 2013; 2:e26836. [PMID: 24498556 PMCID: PMC3912008 DOI: 10.4161/onci.26836] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 10/15/2013] [Accepted: 10/16/2013] [Indexed: 12/14/2022] Open
Abstract
Tumors are sustained by complex networks of interactions between malignant cells, stromal cells and tumor-infiltrating immune cells. These networks differ from patient to patient in terms of nature, composition and organization as well as with regard to the precise localization of tumor-infiltrating cells. Of note, the heterogeneity of the immunological component of the tumor microenvironment, as opposed to its mere abundance, has been shown to influence disease outcome. However, a key question remains: where does the activation of tumor-specific T cells take place? The recently described, tumor-associated lymph node-like entities termed “tertiary lymphoid structures” exhibit a structural organization that is reminiscent of secondary lymphoid organs, and thus may imprint the local immune contexture. Here, we discuss how cancer-associated tertiary lymphoid structures impact on the tumor micro-architecture, immune microenvironment, and ultimately, patient survival.
Collapse
Affiliation(s)
- Jérémy Goc
- The Laboratory of Immune Microenvironment and Tumors; INSERM U872; Cordeliers Research Center; Paris, France ; University Pierre and Marie Curie; UMRS872; Paris, France ; University Paris Descartes; UMRS872; Paris, France
| | - Wolf-Herman Fridman
- The Laboratory of Immune Microenvironment and Tumors; INSERM U872; Cordeliers Research Center; Paris, France ; University Pierre and Marie Curie; UMRS872; Paris, France ; University Paris Descartes; UMRS872; Paris, France
| | - Catherine Sautès-Fridman
- The Laboratory of Immune Microenvironment and Tumors; INSERM U872; Cordeliers Research Center; Paris, France ; University Pierre and Marie Curie; UMRS872; Paris, France ; University Paris Descartes; UMRS872; Paris, France
| | - Marie-Caroline Dieu-Nosjean
- The Laboratory of Immune Microenvironment and Tumors; INSERM U872; Cordeliers Research Center; Paris, France ; University Pierre and Marie Curie; UMRS872; Paris, France ; University Paris Descartes; UMRS872; Paris, France
| |
Collapse
|
43
|
Jiang H, Shao W, Zhao W. VEGF-C in non-small cell lung cancer: meta-analysis. Clin Chim Acta 2013; 427:94-9. [PMID: 24144865 DOI: 10.1016/j.cca.2013.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 09/30/2013] [Accepted: 10/03/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND We sought to clarify the prognostic value of vascular endothelial growth factor C (VEGF-C) in survival of patients with non-small cell lung cancer (NSCLC). METHODS We performed a meta-analysis of relevant literature to aggregate the available survival results, using studies published in English until May 2013. Eligible studies dealt with VEGF-C assessment in NSCLC patients on primary lesions and reported survival data according to VEGF-C expression. RESULTS We aggregated 16 trials, comprising 1988 patients, in this meta-analysis. The overall combined hazard ratio (HR) was 1.65 (95% confidence interval (CI): 1.37-1.98) and was calculated using a random-effects model. It associated high VEGF-C expression with poor survival in all NSCLC patients, including those with stage I NSCLC and high VEGF-C expression (HR: 2.00; 95% CI: 1.22-3.28). However, VEGF-C expression did not significantly correlate with survival in patients with lung adenocarcinoma (ADC) (HR: 1.48; 95% CI: 1.01-2.18). CONCLUSION Our meta-analysis shows that VEGF-C expression is associated with poor prognosis for NSCLC patients, including patients with stage I NSCLC. However, VEGF-C expression is not significantly correlated with survival for patients with lung ADC.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Geriatric Oncology, The Second Affiliated Hospital, Southeast University, 1-1 Zhongfu Street, Nanjing, Jiangsu 210003, PR China.
| | | | | |
Collapse
|
44
|
Yao G, He P, Chen L, Hu X, Gu F, Ye C. MT1-MMP in breast cancer: induction of VEGF-C correlates with metastasis and poor prognosis. Cancer Cell Int 2013; 13:98. [PMID: 24119788 PMCID: PMC3852241 DOI: 10.1186/1475-2867-13-98] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 10/08/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Recent evidence suggests that vascular endothelial growth factor-C (VEGF-C)- dependent tumour production promotes lymphangiogenesis, while membrane-type matrix 1 metalloproteinase (MT1-MMP) is involved in the critical steps leading to carcinogenesis. However, the role of MT1-MMP in lymphangiogenesis and lymphatic metastasis remains poorly understood. In the present study, we investigated the relationship between MT1-MMP and VEGF-C in human breast cancer and correlated MT1-MMP and VEGF-C expression with lymphangiogenesis and prognosis. METHODS MT1-MMP and VEGF-C levels were compared in five breast carcinoma cell lines. We used a membrane invasion assay to assess the effect of MT1-MMP and VEGF-C expression, as well as anti-MT1-MMP and VEGF-C antibodies, on cancer cell invasion. We further assessed MT1-MMP and VEGF-C immunoreactivity and lymph vessels in a cohort of human breast cancer specimens (n = 106) and associated MT1-MMP and VEGF-C expression with clinicopathological parameters, such as lymphatic vessel density (LVD), and patient prognosis. RESULTS MT1-MMP and VEGF-C expression differed among the five breast cancer cell lines and MT1-MMP and VEGF-C expression were correlated with tumour cell invasion. VEGF-C mRNA expression levels and invasive activity of MDA-MB-231 cells was inhibited by an anti-MT1-MMP antibody in a concentration-dependent manner. A significant correlation was found between the expression of MT1-MMP and VEGF-C in breast cancer patient samples and elevated MT1-MMP and VEGF-C expression was associated with higher LVD, lymph node metastasis, cancer stage, and a decline in overall survival rates. CONCLUSIONS Our data demonstrate that MT1-MMP expression is closely correlated with VEGF-C expression, and that MT1-MMP promotes lymphangiogenesis by up-regulating VEGF-C expression in human breast cancer. Thus, elevated MT1-MMP may serve as a significant prognostic factor in breast cancer.
Collapse
Affiliation(s)
- Guangyu Yao
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Ping He
- Department of Pathology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, Guangdong Province, China
| | - Lujia Chen
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Xiaolei Hu
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Fan Gu
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Changsheng Ye
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
45
|
Hein M, Graver S. Tumor cell response to bevacizumab single agent therapy in vitro. Cancer Cell Int 2013; 13:94. [PMID: 24059699 PMCID: PMC3849065 DOI: 10.1186/1475-2867-13-94] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 09/05/2013] [Indexed: 11/17/2022] Open
Abstract
Background Angiogenesis represents a highly multi-factorial and multi-cellular complex (patho-) physiologic event involving endothelial cells, tumor cells in malignant conditions, as well as bone marrow derived cells and stromal cells. One main driver is vascular endothelial growth factor (VEGFA), which is known to interact with endothelial cells as a survival and mitogenic signal. The role of VEGFA on tumor cells and /or tumor stromal cell interaction is less clear. Condition specific (e.g. hypoxia) or tumor specific expression of VEGFA, VEGF receptors and co-receptors on tumor cells has been reported, in addition to the expression on the endothelium. This suggests a potential paracrine/autocrine loop that could affect changes specific to tumor cells. Methods We used the monoclonal antibody against VEGFA, bevacizumab, in various in vitro experiments using cell lines derived from different tumor entities (non small cell lung cancer (NSCLC), colorectal cancer (CRC), breast cancer (BC) and renal cell carcinoma (RCC)) in order to determine if potential VEGFA signaling could be blocked in tumor cells. The experiments were done under hypoxia, a major inducer of VEGFA and angiogenesis, in an attempt to mimic the physiological tumor condition. Known VEGFA induced endothelial biological responses such as proliferation, migration, survival and gene expression changes were evaluated. Results Our study was able to demonstrate expression of VEGF receptors on tumor cells as well as hypoxia regulated angiogenic gene expression. In addition, there was a cell line specific effect in tumor cells by VEGFA blockade with bevacizumab in terms of proliferation; however overall, there was a limited measurable consequence of bevacizumab therapy detected by migration and survival. Conclusion The present study showed in a variety of in vitro experiments with several tumor cell lines from different tumor origins, that by blocking VEGFA with bevacizumab, there was a limited autocrine or cell-autonomous function of VEGFA signaling in tumor cells, when evaluating VEGFA induced downstream outputs known in endothelial cells.
Collapse
Affiliation(s)
- Melanie Hein
- Physiological Chemistry, Biocenter, University of Wuerzburg, Wuerzburg, Germany.
| | | |
Collapse
|
46
|
CD4/CD8 co-expression shows independent prognostic impact in resected non-small cell lung cancer patients treated with adjuvant radiotherapy. Lung Cancer 2013; 80:209-15. [DOI: 10.1016/j.lungcan.2012.12.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/28/2012] [Accepted: 12/03/2012] [Indexed: 11/20/2022]
|
47
|
Associations between hepatocyte growth factor, c-Met, and basic fibroblast growth factor and survival in endometrial cancer patients. Br J Cancer 2012; 106:2004-9. [PMID: 22617129 PMCID: PMC3388566 DOI: 10.1038/bjc.2012.200] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background: Hepatocyte growth factor (HGF), c-Met, and basic fibroblast growth factor (bFGF) are molecular markers that contribute to angiogenesis and proliferation in numerous cancers. We assessed the prognostic significance of these factors in tumour and stroma of endometrial cancer (EC) patients (n=211). Methods: Immunohistochemistry (IHC) was used to detect tumour and stromal protein expression of the biomarkers. Associations between expression and clinicopathological factors were assessed using Chi-square tests. Kaplan–Meier curves, log-rank tests, and Cox regression were used to summarise associations between biomarker expression and overall survival (OS) and recurrence-free survival (RFS). Results: Tumour bFGF was significantly associated with high-grade endometrioid and clear cell histology (P<0.001), advanced stage (P=0.008), positive lymph-node involvement (P=0.002), poor OS (log-rank test, P=0.009), and poor RFS (P<0.001). In multivariable analyses, cases with HGF-positive, stromal bFGF-positive tumours had a lower risk of death compared with cases with HGF-positive, stromal bFGF-negative tumours (hazard ratio (HR): 0.14, 95% CI: 0.03, 0.60). Cases with HGF-positive, bFGF-positive tumours had a higher risk of recurrence compared with cases with negative expression of both markers (HR: 9.88, 95% CI: 2.63, 37.16). Conclusion: These IHC data show that tumour and stromal bFGF expression have opposite associations with survival outcomes in EC patients. If confirmed in larger studies, tumour-derived bFGF could be an attractive target in EC therapy.
Collapse
|
48
|
E-cadherin as a predictive marker of brain metastasis in non-small-cell lung cancer, and its regulation by pioglitazone in a preclinical model. J Neurooncol 2012; 109:219-27. [PMID: 22576972 DOI: 10.1007/s11060-012-0890-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 04/23/2012] [Indexed: 01/06/2023]
Abstract
It remains unclear whether patients with non-small-cell lung cancer (NSCLC) develop brain metastasis during or after standard therapy. We attempted to identify biological markers that predict brain metastasis, and investigated how to modulate expression of such markers. A case-control study of patients who were newly diagnosed with NSCLC and who had developed brain metastasis during follow-up was conducted between 2004 and 2009. These patients were compared with a control group of patients who had NSCLC but no evidence of brain metastasis. Immunohistochemical analysis of expression of Ki-67, p53, Bcl-2, Bax, vascular endothelial growth factor, epidermal growth factor receptor, caspase-3, and E-cadherin was conducted. The methylation status of the genes for O(6)-methylguanine-DNA-methyltransferase, tissue inhibitor of matrix metalloproteinase (TIMP)-2, TIMP-3, and death-associated protein-kinase was also determined, by use of a methylation-specific polymerase chain reaction. A significantly increased risk of developing brain metastasis was associated with the presence of primary tumors with low E-cadherin expression in patients with NSCLC. We also investigated the effects of pioglitazone, a peroxisome proliferator-activated receptor γ-activating drug, in tumor-bearing mouse models. We found that E-cadherin expression was proportional to pioglitazone exposure time. Interestingly, pioglitazone pretreatment before cancer cell inoculation prevented loss of E-cadherin expression and reduced expression of MMP9 and fibronectin, compared with the control group. E-cadherin expression could be a predictor of brain metastasis in patients with NSCLC. Preventive treatment with pioglitazone may be useful for modulating E-cadherin expression.
Collapse
|
49
|
Sorbye SW, Kilvaer TK, Valkov A, Donnem T, Smeland E, Al-Shibli K, Bremnes RM, Busund LT. Prognostic impact of CD57, CD68, M-CSF, CSF-1R, Ki67 and TGF-beta in soft tissue sarcomas. BMC Clin Pathol 2012; 12:7. [PMID: 22554285 PMCID: PMC3408340 DOI: 10.1186/1472-6890-12-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 05/03/2012] [Indexed: 01/22/2023] Open
Abstract
Background Prognostic markers in curable STS may have the potential to guide therapy after surgical resection. The purpose of this study was to clarify the prognostic impact of the presence of cells and growth factors belonging to the innate immune system in soft tissue sarcomas (STS). The significance of macrophages (CD68), their growth factor macrophage colony-stimulating factor (M-CSF), its receptor colony-stimulating factor-1 receptor (CSF-1R), natural killer cells (CD57) and the general immunomodulating molecule (TGF-beta) are all controversial in STS. Herein, these markers are evaluated and compared to the cell proliferation marker Ki67. Methods Tissue microarrays from 249 patients with non-gastrointestinal (non-GIST) STS were constructed from duplicate cores of viable and representative neoplastic tumor areas and duplicate cores of peritumoral capsule. Immunohistochemistry was used to evaluate the expression of CD68, M-CSF, CSF-1R, CD57, TGF-beta and Ki67 in tumor and peritumoral capsule. Results In univariate analyses increased expression of M-CSF (P = 0.034), Ki67 (P < 0.001) and TGF-beta (P = 0.003) in tumor correlated with shorter disease-specific survival (DSS). Increased expression of CD68 in tumor correlated significantly with malignancy grade (P = 0.016), but not DSS (P = 0.270). Increased expression of Ki67 in peritumoral capsule tended to correlate with a shorter DSS (P = 0.057). In multivariate analyses, co-expression of M-CSF and TGF-beta (P = 0.022) in tumor and high expression of Ki67 (P = 0.019) in peritumoral capsule were independent negative prognostic factors for DSS. Conclusions Increased co-expression of M-CSF and TGF-beta in tumor in patients with STS, and increased expression of Ki67 in peritumoral capsule were independent negative prognostic factors for DSS.
Collapse
Affiliation(s)
- Sveinung W Sorbye
- Dept of Clinical Pathology, University Hospital of North Norway, Tromso, 9038, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
SUN ZG, WANG Z, ZHANG M. Correlation between vascular endothelial growth factor C expression and prognosis in patients with esophageal squamous cell carcinomas after Ivor-Lewis esophagectomy. Asia Pac J Clin Oncol 2012; 8:e68-76. [DOI: 10.1111/j.1743-7563.2011.01514.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|