1
|
Sun H, Li L, Yan J, Huang T. Prioritization of drug targets for thyroid cancer: a multi-omics Mendelian randomization study. Endocrine 2024; 86:732-743. [PMID: 38896366 DOI: 10.1007/s12020-024-03933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVES Recurrence or tumor metastasis and drug resistance remain the major challenge in the treatment of thyroid cancer. It is needed to identify novel drug targets for thyroid cancer. METHODS Summary data-based Mendelian randomization (SMR) and colocalization analysis were performed to evaluate the associations between gene methylation, expression, protein levels with thyroid cancer. We additionally performed protein-protein interaction (PPI) network, gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) analyses to further explore the potential roles of identified genes in thyroid cancer. RESULTS SDCCAG8 and VCAM1 genes were associated with risk of thyroid cancer with tier 1 evidence, while TCN2 gene was with tier 3 evidence. SDCCAG8 gene was associated with risk of papillary thyroid cancer with tier 1 evidence. At the level of circulating proteins, genetically predicted higher levels of SDCCAG8 (OR = 0.46, 95% CI 0.34-0.64) and VCAM1 (OR = 0.21, 95% CI 0.10-0.45) were inversely associated with thyroid cancer risk; higher level of TCN2 was associated with an increased risk of thyroid cancer (OR = 1.30, 95% CI 1.15-1.47); and the higher level of SDCCAG8 (OR = 0.40, 95% CI 0.28-0.58) was associated with a decreased risk of papillary thyroid cancer. The bioinformatics analysis showed that SDCCAG8, VCAM1 and TCN2 might play roles in immune-related pathways. CONCLUSION SDCCAG8, VCAM1 and TCN2 genes were associated with thyroid cancer risk with evidence at multi-omics levels. There were potential roles of SDCCAG8, VCAM1 and TCN2 in immune-related pathways. Our findings might improve the understanding of the pathogenesis of thyroid cancer and discovery of novel potential drug targets for this disease.
Collapse
Affiliation(s)
- Hong Sun
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Ling Li
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Jingchao Yan
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, China.
| | - Taomin Huang
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Jazwinska DE, Kulawiec DG, Zervantonakis IK. Cancer-mesothelial and cancer-macrophage interactions in the ovarian cancer microenvironment. Am J Physiol Cell Physiol 2023; 325:C721-C730. [PMID: 37545408 PMCID: PMC10635648 DOI: 10.1152/ajpcell.00461.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
The metastatic ovarian cancer microenvironment is characterized by an intricate interaction network between cancer cells and host cells. This complex heterotypic cancer-host cell crosstalk results in an environment that promotes cancer cell metastasis and treatment resistance, leading to poor patient prognosis and survival. In this review, we focus on two host cell types found in the ovarian cancer microenvironment: mesothelial cells and tumor-associated macrophages. Mesothelial cells make up the protective lining of organs in the abdominal cavity. Cancer cells attach and invade through the mesothelial monolayer to form metastatic lesions. Crosstalk between mesothelial and cancer cells can contribute to metastatic progression and chemotherapy resistance. Tumor-associated macrophages are the most abundant immune cell type in the ovarian cancer microenvironment with heterogeneous subpopulations exhibiting protumor or antitumor functions. Macrophage reprogramming toward a protumor or antitumor state can be influenced by chemotherapy and communication with cancer cells, resulting in cancer cell invasion and treatment resistance. A better understanding of cancer-mesothelial and cancer-macrophage crosstalk will uncover biomarkers of metastatic progression and therapeutic targets to restore chemotherapy sensitivity.
Collapse
Affiliation(s)
- Dorota E Jazwinska
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Diana G Kulawiec
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Ioannis K Zervantonakis
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
3
|
Song J, Sokoll LJ, Zhang Z, Chan DW. VCAM-1 complements CA-125 in detecting recurrent ovarian cancer. Clin Proteomics 2023; 20:25. [PMID: 37357306 PMCID: PMC10291808 DOI: 10.1186/s12014-023-09414-z] [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: 11/01/2022] [Accepted: 06/13/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Close to three-quarters of ovarian cancer cases are frequently diagnosed at an advanced stage, with more than 70% of them failing to respond to primary therapy and relapsing within 5 years. There is an urgent need to identify strategies for early detection of ovarian cancer recurrence, which may lead to earlier intervention and better outcomes. METHODS A customized magnetic bead-based 8-plex immunoassay was evaluated using a Bio-Plex 200 Suspension Array System. Target protein levels were analyzed in sera from 58 patients diagnosed with advanced ovarian cancer (including 34 primary and 24 recurrent tumors) and 46 healthy controls. The clinical performance of these biomarkers was evaluated individually and in combination for their ability to detect recurrent ovarian cancer. RESULTS An 8-plex immunoassay was evaluated with high analytical performance suitable for biomarker validation studies. Logistic regression modeling selected a two-marker panel of CA-125 and VCAM-1 that improved the performance of CA-125 alone in detecting recurrent ovarian cancer (AUC: 0.813 versus 0.700). At a fixed specificity of 83%, the two-marker panel significantly improved sensitivity in separating primary from recurrent tumors (70.8% versus 37.5%, P = 0.004), demonstrating that VCAM-1 was significantly complementary to CA-125 in detecting recurrent ovarian cancer. CONCLUSIONS A two-marker panel of CA-125 and VCAM-1 showed strong diagnostic performance and improvement over the use of CA-125 alone in detecting recurrent ovarian cancer. The experimental results warrant further clinical validation to determine their role in the early detection of recurrent ovarian cancer.
Collapse
Affiliation(s)
- Jin Song
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD, 21231, USA.
| | - Lori J Sokoll
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Zhen Zhang
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Daniel W Chan
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| |
Collapse
|
4
|
Zou M, Du Y, Liu R, Zheng Z, Xu J. Nanocarrier-delivered small interfering RNA for chemoresistant ovarian cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 12:e1648. [PMID: 33682310 DOI: 10.1002/wrna.1648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States. Because success in early screening is limited, and most patients with advanced disease develop resistance to multiple treatment modalities, the overall prognosis of ovarian cancer is poor. Despite the revolutionary role of surgery and chemotherapy in curing ovarian cancer, recurrence remains a major challenge in treatment. Thus, improving our understanding of the pathogenesis of ovarian cancer is essential for developing more effective treatments. In this review, we analyze the underlying molecular mechanisms leading to chemotherapy resistance. We discuss the clinical benefits and potential challenges of using nanocarrier-delivered small interfering RNA to treat chemotherapy-resistant ovarian cancer. We aim to elicit collaborative studies on nanocarrier-delivered small interfering RNA to improve the long-term survival rate and quality of life of patients with ovarian cancer. This article is categorized under: RNA Methods > RNA Nanotechnology Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Mingyuan Zou
- Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruizhen Liu
- The First People's Hospital of Wu'an, Wu'an, Hebei, China
| | - Zeliang Zheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Jakimovska M, Černe K, Verdenik I, Kobal B. High preoperative serum sVCAM-1 concentration as a predictor of early ovarian cancer recurrence. J Ovarian Res 2020; 13:107. [PMID: 32933568 PMCID: PMC7490865 DOI: 10.1186/s13048-020-00705-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 08/13/2020] [Indexed: 11/29/2022] Open
Affiliation(s)
- Marina Jakimovska
- Department of Obstetrics and Gyaecology, University Medical Centre, Ljubljana, Slovenia
| | - Katarina Černe
- Institute of Pharmacology and Experimental Toxicology, Faculty of medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ivan Verdenik
- Department of Obstetrics and Gyaecology, University Medical Centre, Ljubljana, Slovenia
| | - Borut Kobal
- Department of Obstetrics and Gyaecology, University Medical Centre, Ljubljana, Slovenia. .,Faculty of medicine, University of Ljubljana, Ljubljana, Slovenia. .,Department of Gynaecology and Obstetrics, Faculty of medicine, University of Ljubljana, Šlajmarjeva 3, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
6
|
Azizi M, Dianat-Moghadam H, Salehi R, Farshbaf M, Iyengar D, Sau S, Iyer AK, Valizadeh H, Mehrmohammadi M, Hamblin MR. Interactions Between Tumor Biology and Targeted Nanoplatforms for Imaging Applications. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910402. [PMID: 34093104 PMCID: PMC8174103 DOI: 10.1002/adfm.201910402] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Indexed: 05/04/2023]
Abstract
Although considerable efforts have been conducted to diagnose, improve, and treat cancer in the past few decades, existing therapeutic options are insufficient, as mortality and morbidity rates remain high. Perhaps the best hope for substantial improvement lies in early detection. Recent advances in nanotechnology are expected to increase the current understanding of tumor biology, and will allow nanomaterials to be used for targeting and imaging both in vitro and in vivo experimental models. Owing to their intrinsic physicochemical characteristics, nanostructures (NSs) are valuable tools that have received much attention in nanoimaging. Consequently, rationally designed NSs have been successfully employed in cancer imaging for targeting cancer-specific or cancer-associated molecules and pathways. This review categorizes imaging and targeting approaches according to cancer type, and also highlights some new safe approaches involving membrane-coated nanoparticles, tumor cell-derived extracellular vesicles, circulating tumor cells, cell-free DNAs, and cancer stem cells in the hope of developing more precise targeting and multifunctional nanotechnology-based imaging probes in the future.
Collapse
Affiliation(s)
- Mehdi Azizi
- Proteomics Research Centre, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| | - Hassan Dianat-Moghadam
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5165665621, Iran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 516615731, Iran
| | - Masoud Farshbaf
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 6581151656, Iran
| | - Disha Iyengar
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samaresh Sau
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Arun K Iyer
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Hadi Valizadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 516615731, Iran
| | | | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
7
|
Peptide-based nanosystems for vascular cell adhesion molecule-1 targeting: a real opportunity for therapeutic and diagnostic agents in inflammation associated disorders. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
8
|
VCAM-1 Upregulation Contributes to Insensitivity of Vemurafenib in BRAF-Mutant Thyroid Cancer. Transl Oncol 2020; 13:441-451. [PMID: 31911278 PMCID: PMC6948368 DOI: 10.1016/j.tranon.2019.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022] Open
Abstract
Vemurafenib, an inhibitor of mutant BRAF activity, is a promising anticancer agent for patients with BRAF-mutant metastatic melanoma. However, it is less effective in BRAF-mutant thyroid cancer, and the reason for this discrepancy is not yet fully elucidated. By RNA sequencing analysis, we identified vascular cell adhesion molecular-1 (VCAM-1) to be highly upregulated in both time- and dose-dependent manners during BRAF inhibition (BRAFi) in a BRAF-mutant papillary thyroid cancer cell line (BCPAP). Cell cytotoxicity and apoptosis assays showed that knockdown of the induced VCAM-1 in BCPAP cells augmented the antitumor effects of vemurafenib, with decreased IC50 values of 1.4 to 0.8 μM. Meanwhile, overexpression of VCAM-1 in a BRAF-mutant anaplastic thyroid cancer cell line (FRO) reduced the sensitivity to vemurafenib, with increased IC50 values of 1.9 to 5.8 μM. Further investigation showed that PI3K-Akt-mTOR pathway was activated during BRAFi. Co-treatment with Akt signaling inhibitor MK2206 decreased the induced expression of VCAM-1 during BRAFi. This combination further improved the efficacy of vemurafenib. Moreover, VCAM-1 promoted migration and invasion in thyroid cancer cells in vitro, which was also indicated in thyroid cancer patients. The present study is the first to demonstrate that VCAM-1 is upregulated in thyroid cancer cells treated with vemurafenib and contributes to vemurafenib resistance in BRAF-mutant thyroid cancer cells. Targeting the PI3K-Akt-mTOR pathway–mediated VCAM-1 response may be an alternative strategy to sensitize BRAF-mutant thyroid cancers to vemurafenib.
Collapse
|
9
|
Montemagno C, Dumas L, Cavaillès P, Ahmadi M, Bacot S, Debiossat M, Soubies A, Djaïleb L, Leenhardt J, Leiris ND, Dufies M, Pagès G, Hernot S, Devoogdt N, Perret P, Riou L, Fagret D, Ghezzi C, Broisat A. In Vivo Assessment of VCAM-1 Expression by SPECT/CT Imaging in Mice Models of Human Triple Negative Breast Cancer. Cancers (Basel) 2019; 11:cancers11071039. [PMID: 31340603 PMCID: PMC6678795 DOI: 10.3390/cancers11071039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 12/31/2022] Open
Abstract
Recent progress in breast cancer research has led to the identification of Vascular Cell Adhesion Molecule-1 (VCAM-1) as a key actor of metastatic colonization. VCAM-1 promotes lung-metastases and is associated with clinical early recurrence and poor outcome in triple negative breast cancer (TNBC). Our objective was to perform the in vivo imaging of VCAM-1 in mice models of TNBC. The Cancer Genomic Atlas (TCGA) database was analyzed to evaluate the prognostic role of VCAM-1 in TNBC. MDA-MB-231 (VCAM-1+) and control HCC70 (VCAM-1-) TNBC cells were subcutaneously xenografted in mice and VCAM-1 expression was assessed in vivo by single-photon emission computed tomography (SPECT) imaging using 99mTc-cAbVCAM1-5. Then, MDA-MB-231 cells were intravenously injected in mice and VCAM-1 expression in lung metastasis was assessed by SPECT imaging after 8 weeks. TCGA analysis showed that VCAM-1 is associated with a poor prognosis in TNBC patients. In subcutaneous tumor models, 99mTc-cAbVCAM1-5 uptake was 2-fold higher in MDA-MB-231 than in HCC70 (p < 0.01), and 4-fold higher than that of the irrelevant control (p < 0.01). Moreover, 99mTc-cAbVCAM1-5 uptake in MDA-MB-231 lung metastases was also higher than that of 99mTc-Ctl (p < 0.05). 99mTc-cAbVCAM1-5 is therefore a suitable tool to evaluate the role of VCAM-1 as a marker of tumor aggressiveness of TNBC.
Collapse
Affiliation(s)
- Christopher Montemagno
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Laurent Dumas
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
- Advanced Accelator Applications, 01630 Saint-Genis-Pouilly, France
| | - Pierre Cavaillès
- Natural Barriers and Infectiosity, Universite Grenoble Alpes, CNRS, CHU Grenoble Alpes, TIMC-IMAG, 38000 Grenoble, France
| | - Mitra Ahmadi
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Sandrine Bacot
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Marlène Debiossat
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Audrey Soubies
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Loic Djaïleb
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Julien Leenhardt
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Nicolas de Leiris
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Maeva Dufies
- Biomedical Department, Centre Scientifique de Monaco, 980000 Monaco, Monaco
| | - Gilles Pagès
- Biomedical Department, Centre Scientifique de Monaco, 980000 Monaco, Monaco
- Institute for Research on Cancer and Aging of Nice, Universite Cote d'Azur, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, 061489 Nice, France
| | - Sophie Hernot
- Laboratory of In Vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Laarbeeklan 103, B-1090 Brussels, Belgium
| | - Nick Devoogdt
- Laboratory of In Vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Laarbeeklan 103, B-1090 Brussels, Belgium
| | - Pascale Perret
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Laurent Riou
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Daniel Fagret
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Catherine Ghezzi
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France
| | - Alexis Broisat
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, Inserm, CHU Grenoble Alpes, LRB, 38000 Grenoble, France.
| |
Collapse
|
10
|
Molecular imaging to enlighten cancer immunotherapies and underlying involved processes. Cancer Treat Rev 2018; 70:232-244. [DOI: 10.1016/j.ctrv.2018.09.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 01/04/2023]
|
11
|
Huang D, Yuan W, Li H, Li S, Chen Z, Yang H. Identification of key pathways and biomarkers in sorafenib-resistant hepatocellular carcinoma using bioinformatics analysis. Exp Ther Med 2018; 16:1850-1858. [PMID: 30186410 PMCID: PMC6122189 DOI: 10.3892/etm.2018.6427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/26/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant types of cancer, with a high mortality rate. Sorafenib is the sole approved oral clinical therapy against advanced HCC. However, individual patients exhibit varying responses to sorafenib and the development of sorafenib resistance has been a new challenge for its clinical efficacy. The current study identified gene biomarkers and key pathways in sorafenib-resistant HCC using bioinformatics analysis. Gene dataset GSE73571 was obtained from the Gene Expression Omnibus (GEO) database, including four sorafenib-acquired resistant and three sorafenib-sensitive HCC phenotypes. Differentially expressed genes (DEGs) were identified using the web tool GEO2R. Functional and pathway enrichment of DEGs were analyzed using the Database for Annotation, Visualization and Integrated Discovery and the protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins and Cytoscape. A total of 1,319 DEGs were selected, which included 593 upregulated and 726 downregulated genes. Functional and pathway enrichment analysis revealed DEGs enriched in negative regulation of endopeptidase activity, cholesterol homeostasis, DNA replication and repair, coagulation cascades, insulin resistance, RNA transport, cell cycle and others. Eight hub genes, including kininogen 1, vascular cell adhesion molecule 1, apolipoprotein C3, alpha 2-HS glycoprotein, erb-b2 receptor tyrosine kinase 2, secreted protein acidic and cysteine rich, vitronectin and vimentin were identified from the PPI network. In conclusion, the present study identified DEGs and key genes in sorafenib-resistant HCC, which further the knowledge of potential mechanisms in the development of sorafenib resistance and may provide potential targets for early diagnosis and new treatments for sorafenib-resistant HCC.
Collapse
Affiliation(s)
- Danping Huang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Weiqu Yuan
- Acupuncture Department, The Fourth Clinical Medical College of Guangzhou University Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Hanmin Li
- Hepatopathy Institution, Affiliated Hospital Hubei University Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| | - Shaodong Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Zuanguang Chen
- Pharmaceutical Analysis Department, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Hongzhi Yang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
12
|
Evaluation of 99mTc-HYNIC-VCAM-1 scFv as a Potential Qualitative and Semiquantitative Probe Targeting Various Tumors. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:7832805. [PMID: 29853809 PMCID: PMC5960529 DOI: 10.1155/2018/7832805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/25/2018] [Indexed: 12/19/2022]
Abstract
Vascular cell adhesion molecule 1 (VCAM-1) is overexpressed in varieties of cancers. This study aimed to evaluate the application of a single chain variable fragment (scFv) of anti-VCAM-1 antibody labeled with 99mTc as a possible imaging agent in several tumors. VCAM-1 scFv was labeled with 99mTc using succinimidyl 6-hydrazinium nicotinate hydrochloride, and 99mTc-HYNIC-VCAM-1scFv was successfully synthesized with a high radiolabeling yield. VCAM-1 expression was evaluated in six cell lines by immunofluorescence staining. In vitro binding assays showed different binding affinities of 99mTc-HYNIC-VCAM-1scFv in different tumor cell lines, with high uptake in B16F10 melanoma and HT1080 fibrosarcoma cells, which was consistent with immunofluorescence staining results. In vivo SPECT planar imaging demonstrated that B16F10 and HT1080 tumors could be clearly visualized. Less intense uptake was observed in human SKOV3.ip ovarian tumor, and weak uptake was observed in human A375m melanoma, MDA-MB-231 breast cancer, and 786-O renal tumors. These findings were confirmed by biodistribution and immunofluorescence studies. High uptake by B16F10 tumors was inhibited by excess unlabeled VCAM-1scFv. 99mTc-HYNIC-VCAM-1scFv, which selectively binds to VCAM-1, can provide a qualitative and semiquantitative method for noninvasive evaluation of VCAM-1 expression by tumors.
Collapse
|
13
|
Zhang X, Liu C, Hu F, Zhang Y, Wang J, Gao Y, Jiang Y, Zhang Y, Lan X. PET Imaging of VCAM-1 Expression and Monitoring Therapy Response in Tumor with a 68Ga-Labeled Single Chain Variable Fragment. Mol Pharm 2018; 15:609-618. [PMID: 29308904 DOI: 10.1021/acs.molpharmaceut.7b00961] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) is a transmembrane glycoprotein closely related to tumorigenicity as well as tumor metastasis. It is also a well-known candidate for detecting tumors. LY2409881, an IKKβ inhibitor, could induce apoptosis of VCAM-1 positive cells. Our purpose is to prepare a novel tracer to evaluate its feasibility of detecting VCAM-1 expression and monitoring LY2409881 tumor curative effect. The tracer was prepared by conjugating the single chain variable fragment (scFv) of VCAM-1 and NOTA-NHS-ester and then labeled with 68Ga. 68Ga-NOTA-VCAM-1scFv was successfully prepared with high radiochemical yield. VCAM-1 overexpression and underexpression melanoma cell lines, B16F10 and A375m, were used in this study. The results of microPET/CT imaging in small animals indicated that the uptake of 68Ga-NOTA-VCAM-1scFv in B16F10 tumor was much higher than that of A375m, which was also confirmed by the biodistribution and autoradiography results. LY2409881 inhibits the growth of B16F10 melanoma in vivo by inducing dose- and time-dependent growth inhibition and apoptosis of the cells. The LY2409881 treated group and DMSO control group were established and imaged by microPET/CT. In the LY2409881 group, uptake of the tracer in tumor was decreased at the first week, and then gradually recovered to the initial level. In DMSO control, the uptake of the tracer remained at the same level during the whole time. The results suggested that LY2409881 inhibits the expression of VCAM-1 and suppresses tumor growth. 68Ga-NOTA-VCAM-1scFv, an easily synthesized probe, has a potential clinical application in the visual monitoring of IKKβ inhibitor intervention on VCAM-1 positive tumors.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Chunbao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Fan Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yingying Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University , Xi'an, 710032, China
| | - Yongheng Gao
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University , Xi'an, 710032, China
| | - Yaqun Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| |
Collapse
|
14
|
Falzone N, Ackerman NL, Rosales LDLF, Bernal MA, Liu X, Peeters SGJA, Soto MS, Corroyer-Dulmont A, Bernaudin M, Grimoin E, Touzani O, Sibson NR, Vallis KA. Dosimetric evaluation of radionuclides for VCAM-1-targeted radionuclide therapy of early brain metastases. Theranostics 2018; 8:292-303. [PMID: 29290808 PMCID: PMC5743475 DOI: 10.7150/thno.22217] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/02/2017] [Indexed: 11/13/2022] Open
Abstract
Brain metastases develop frequently in patients with breast cancer, and present a pressing therapeutic challenge. Expression of vascular cell adhesion molecule 1 (VCAM-1) is upregulated on brain endothelial cells during the early stages of metastasis and provides a target for the detection and treatment of early brain metastases. The aim of this study was to use a model of early brain metastasis to evaluate the efficacy of α-emitting radionuclides, 149Tb, 211At, 212Pb, 213Bi and 225Ac; β-emitting radionuclides, 90Y, 161Tb and 177Lu; and Auger electron (AE)-emitters 67Ga, 89Zr, 111In and 124I, for targeted radionuclide therapy (TRT). METHODS Histologic sections and two photon microscopy of mouse brain parenchyma were used to inform a cylindrical vessel geometry using the Geant4 general purpose Monte Carlo (MC) toolkit with the Geant4-DNA low energy physics models. Energy deposition was evaluated as a radial function and the resulting phase spaces were superimposed on a DNA model to estimate double-strand break (DSB) yields for representative β- and α-emitters, 177Lu and 212Pb. Relative biological effectiveness (RBE) values were determined by only evaluating DNA damage due to physical interactions. RESULTS 177Lu produced 2.69 ± 0.08 DSB per GbpGy, without significant variation from the lumen of the vessel to a radius of 100 µm. The DSB yield of 212Pb included two local maxima produced by the 6.1 MeV and 8.8 MeV α-emissions from decay products, 212Bi and 212Po, with yields of 7.64 ± 0.12 and 9.15 ± 0.24 per GbpGy, respectively. Given its higher DSB yield 212Pb may be more effective for short range targeting of early micrometastatic lesions than 177Lu. CONCLUSION MC simulation of a model of early brain metastases provides invaluable insight into the potential efficacy of α-, β- and AE-emitting radionuclides for TRT. 212Pb, which has the attributes of a theranostic radionuclide since it can be used for SPECT imaging, showed a favorable dose profile and RBE.
Collapse
Affiliation(s)
- Nadia Falzone
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Nicole L. Ackerman
- Department of Physics and Astronomy, Agnes Scott College, Decatur, GA, United States of America
| | | | - Mario A. Bernal
- Departamento de Física Aplicada, Instituto de Física "Gleb Wataghin", UNICAMP, Campinas, Brazil
| | - Xiaoxuan Liu
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sarah GJA Peeters
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Manuel Sarmiento Soto
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Aurélien Corroyer-Dulmont
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, Caen, France
| | - Elisa Grimoin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, Caen, France
| | - Omar Touzani
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, Caen, France
| | - Nicola R. Sibson
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Katherine A. Vallis
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Scalici JM, Arapovic S, Saks E, Atkins KA, Petroni G, Duska LR, Slack-Davis JK. Mesothelium expression of vascular cell adhesion molecule-1 (VCAM-1) is associated with an unfavorable prognosis in epithelial ovarian cancer (EOC). Cancer 2017; 123:977-984. [PMID: 28263384 PMCID: PMC5341143 DOI: 10.1002/cncr.30415] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/06/2016] [Accepted: 10/04/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Mesothelium vascular cell adhesion molecule-1 (VCAM-1) expression in the metastatic epithelial ovarian cancer (EOC) microenvironment is induced by tumor and mediates tumor cell invasion. VCAM-1 imaging suggests expression during treatment is an indicator of platinum resistance. Here, we assess the potential prognostic significance of mesothelium VCAM-1 expression and prospectively evaluate whether soluble VCAM-1 (sVCAM-1) is a surrogate for mesothelium expression. METHODS A retrospective review of EOC patients was performed to evaluate outcomes with mesothelium VCAM-1 expression determined by immunohistochemistry of peritoneum or omentum specimens. A prospective cohort of EOC patients was identified and followed through primary treatment. Serum for sVCAM-1 evaluation, which was performed via enzyme-linked immunosorbent assay, was collected before surgery or neoadjuvant chemotherapy and at each treatment cycle. Peritoneal specimens were obtained during debulking to assess mesothelial VCAM-1 expression. RESULTS A retrospective review identified 54 advanced-stage EOC patients. Patients expressing mesothelium VCAM-1 had shortened overall survival (44 vs 79 months, P = 0.035) and progression-free survival (18 vs 67 months, P = 0.010); the median time to platinum resistance was 36 months for VCAM-1-expressing patients and not yet determined for the VCAM-1-negative group. In our prospective observational cohort, 18 EOC patients completed primary treatment; 3 were negative for mesothelium VCAM-1 expression, and sVCAM-1 did not vary between groups. CONCLUSIONS Mesothelium VCAM-1 expression is negatively associated with progression-free and overall survival in EOC. This is especially compelling in light of previous data suggesting that persistent VCAM-1 expression during treatment is an indicator of platinum resistance. Our pilot study had insufficient cases to determine whether sVCAM-1 would substitute for mesothelium expression. Cancer 2017;123:977-84. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Jennifer M. Scalici
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA
| | - Sanja Arapovic
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA
| | - Erin Saks
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA
| | - Kristen A. Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
- Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Gina Petroni
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Linda R. Duska
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA
- Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Jill K. Slack-Davis
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Cancer Center, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
16
|
Calderon LE, Keeling JK, Rollins J, Black CA, Collins K, Arnold N, Vance DE, Ndinguri MW. Pt-Mal-LHRH, a Newly Synthesized Compound Attenuating Breast Cancer Tumor Growth and Metastasis by Targeting Overexpression of the LHRH Receptor. Bioconjug Chem 2016; 28:461-470. [PMID: 27997127 DOI: 10.1021/acs.bioconjchem.6b00610] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A new targeting chemotherapeutic agent, Pt-Mal-LHRH, was synthesized by linking activated cisplatin to luteinizing hormone releasing hormone (LHRH). The compound's efficacy and selectivity toward 4T1 breast cancer cells were evaluated. Carboplatin was selected as the comparative platinum complex, since the Pt-Mal-LHRH malonate linker chelates platinum in a similar manner to carboplatin. Breast cancer and normal cell viability were analyzed by an MTT assay comparing Pt-Mal-LHRH with carboplatin. Cells were also treated with either Pt-Mal-LHRH or carboplatin to evaluate platinum uptake by ICP-MS and cell migration using an in vitro scratch-migration assay. Tumor volume and metastasis were evaluated using an in vivo 4T1 mouse tumor model. Mice were administered Pt-Mal-LHRH (carboplatin molar equivalent dosage) through ip injection and compared to those treated with carboplatin (5 (mg/kg)/week), no treatment, and LHRH plus carboplatin (unbound) controls. An MTT assay showed a reduction in cell viability (p < 0.01) in 4T1 and MDA-MB-231 breast cancer cells treated with Pt-Mal-LHRH compared to carboplatin. Pt-Mal-LHRH was confirmed to be cytotoxic by flow cytometry using a propidium iodide stain. Pt-Mal-LHRH displayed a 20-fold increase in 4T1 cellular uptake compared to carboplatin. There was a decrease (p < 0.0001) in 4T1 cell viability compared to 3T3 normal fibroblast cells. Treatment with Pt-Mal-LHRH also resulted in a significant decrease in cell-migration compared to carboplatin. In vivo testing found a significant reduction in tumor volume (p < 0.05) and metastatic tumor colonization in the lungs with Pt-Mal-LHRH compared to carboplatin. There was a slight decrease in lung weight and no difference in liver weight between treatment groups. Together, our data indicate that Pt-Mal-LHRH is a more potent and selective chemotherapeutic agent than untargeted carboplatin.
Collapse
Affiliation(s)
- Lindsay E Calderon
- Department of Biology, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Jonathan K Keeling
- Department of Chemistry, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Joseph Rollins
- Department of Biology, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Carrie A Black
- Department of Chemistry, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Kendall Collins
- Department of Biology, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Nova Arnold
- Department of Biology, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Diane E Vance
- Department of Chemistry, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Margaret W Ndinguri
- Department of Chemistry, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| |
Collapse
|
17
|
Significance of TNF-α and the Adhesion Molecules: L-Selectin and VCAM-1 in Papillary Thyroid Carcinoma. J Thyroid Res 2016; 2016:8143695. [PMID: 26881177 PMCID: PMC4737049 DOI: 10.1155/2016/8143695] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/29/2015] [Indexed: 12/13/2022] Open
Abstract
Circulating levels of TNF-α and the adhesion molecules L-Selectin and VCAM-1 as well as their expression in the primary tumors of patients with benign thyroid diseases and papillary thyroid carcinoma (PTC) have been determined in this study. The serum levels of TNF-α, L-Selectin, and VCAM-1 were significantly higher in patients with both benign thyroid diseases and PTC as compared to the healthy individuals. However, the levels of only TNF-α and L-Selectin, and not VCAM-1, were significantly higher in patients with PTC in comparison to those observed in patients with benign thyroid diseases. Further the expression of TNF-α and L-Selectin was also significantly higher in the primary tumors of PTC patients, relative to the benign thyroid diseases. The expression of L-Selectin and VCAM-1 significantly correlated with aggressive tumor behavior. In PTC patients, the circulating TNF-α levels significantly positively correlated with the levels of L-Selectin, while TNF-α immunoreactivity was significantly associated with VCAM-1 expression. Serum TNF-α was found to be a significant prognosticator for OS in PTC patients. Overall the results signify that the interaction between TNF-α and the adhesion molecules may have a role in thyroid carcinogenesis and understanding this complexity may offer potential therapeutic targets for better management of thyroid cancer.
Collapse
|
18
|
De Souza R, Spence T, Huang H, Allen C. Preclinical imaging and translational animal models of cancer for accelerated clinical implementation of nanotechnologies and macromolecular agents. J Control Release 2015; 219:313-330. [PMID: 26409122 DOI: 10.1016/j.jconrel.2015.09.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/22/2015] [Accepted: 09/22/2015] [Indexed: 01/08/2023]
Abstract
The majority of animal models of cancer have performed poorly in terms of predicting clinical performance of new therapeutics, which are most often first evaluated in patients with advanced, metastatic disease. The development and use of metastatic models of cancer may enhance clinical translatability of preclinical studies focused on the development of nanotechnology-based drug delivery systems and macromolecular therapeutics, potentially accelerating their clinical implementation. It is recognized that the development and use of such models are not without challenge. Preclinical imaging tools offer a solution by allowing temporal and spatial characterization of metastatic lesions. This paper provides a review of imaging methods applicable for evaluation of novel therapeutics in clinically relevant models of advanced cancer. An overview of currently utilized models of oncology in small animals is followed by image-based development and characterization of visceral metastatic cancer models. Examples of imaging tools employed for metastatic lesion detection, evaluation of anti-tumor and anti-metastatic potential and biodistribution of novel therapies, as well as the co-development and/or use of imageable surrogates of response, are also discussed. While the focus is on development of macromolecular and nanotechnology-based therapeutics, examples with small molecules are included in some cases to illustrate concepts and approaches that can be applied in the assessment of nanotechnologies or macromolecules.
Collapse
Affiliation(s)
- Raquel De Souza
- Leslie Dan Faculty of Pharmacy, 144 College Street, Toronto, Ontario M5S 3M2, Canada.
| | - Tara Spence
- Leslie Dan Faculty of Pharmacy, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Huang Huang
- DLVR Therapeutics, 661 University Avenue, Toronto, Ontario M5G 0A3, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, 144 College Street, Toronto, Ontario M5S 3M2, Canada.
| |
Collapse
|
19
|
Bergamo A, Sava G. Linking the future of anticancer metal-complexes to the therapy of tumour metastases. Chem Soc Rev 2015; 44:8818-35. [DOI: 10.1039/c5cs00134j] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer chemotherapy is almost always applied to patients with one or more diagnosed metastases and is expected to impact these lesions, thus providing significant benefits to the patient.
Collapse
Affiliation(s)
| | - Gianni Sava
- Callerio Foundation Onlus
- 34127 Trieste
- Italy
- Department of Life Sciences
- University of Trieste
| |
Collapse
|
20
|
Hong H, Chen F, Zhang Y, Cai W. New radiotracers for imaging of vascular targets in angiogenesis-related diseases. Adv Drug Deliv Rev 2014; 76:2-20. [PMID: 25086372 DOI: 10.1016/j.addr.2014.07.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 07/14/2014] [Accepted: 07/22/2014] [Indexed: 01/03/2023]
Abstract
Tremendous advances over the last several decades in positron emission tomography (PET) and single photon emission computed tomography (SPECT) allow for targeted imaging of molecular and cellular events in the living systems. Angiogenesis, a multistep process regulated by the network of different angiogenic factors, has attracted world-wide interests, due to its pivotal role in the formation and progression of different diseases including cancer, cardiovascular diseases (CVD), and inflammation. In this review article, we will summarize the recent progress in PET or SPECT imaging of a wide variety of vascular targets in three major angiogenesis-related diseases: cancer, cardiovascular diseases, and inflammation. Faster drug development and patient stratification for a specific therapy will become possible with the facilitation of PET or SPECT imaging and it will be critical for the maximum benefit of patients.
Collapse
|
21
|
Soendergaard M, Newton-Northup JR, Deutscher SL. In vivo phage display selection of an ovarian cancer targeting peptide for SPECT/CT imaging. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2014; 4:561-570. [PMID: 25250205 PMCID: PMC4171842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/17/2014] [Indexed: 06/03/2023]
Abstract
The often fatal outcome of ovarian cancer (OC) is related to inadequate detection methods, which may be overcome by development of nuclear imaging agents. Cancer targeting peptides have been identified using in vivo bacteriophage (phage) display technology; however, the majority of these ligands target tumor vasculature. To overcome this problem, a two-tier phage display method was employed to select an ovarian cancer targeting peptide with good pharmacokinetic and imaging properties. A fUSE5 15-amino acid peptide library was screened against xenografted human OC SKOV-3 tumors in mice, which was followed by selection against enriched SKOV-3 cells. The selected peptide RSLWSDFYASASRGP (J18) was synthesized with a GSG-spacer and a 1,4,7,10-tetraazacyclodecane-1,4,7,10-tetraacetic acid (DOTA) chelator and radiolabeled with (111)In. SKOV-3 xenografted mice were used to evaluate the biodistribution and single photon emission computed tomography (SPECT) imaging capabilities of the radiolabeled peptide. Competitive binding experiments using (111)In-DOTA-GSG-J18 indicated that the peptide displayed a half maximal inhibitory concentration (IC50) value of 10.5 ± 1.1 μM. Biodistribution studies revealed that tumor uptake was 1.63 ± 0.68, 0.60 ± 0.32, 0.31 ± 0.12 and 0.10 ± 0.02% injected dose/g at 30 min, 1 h, 2 h and 4 h post-injection of (111)In-DOTA-GSG-J18, respectively. SPECT/CT imaging demonstrated good tumor uptake and minimal background binding. This study demonstrated successful utilization of a two-tier phage display selection process to identify an ovarian cancer avid peptide with excellent SPECT/CT imaging capabilities.
Collapse
Affiliation(s)
- Mette Soendergaard
- Department of Biochemistry, University of MissouriRm117 Schweitzer Hall, Columbia, MO 65211
| | | | - Susan L Deutscher
- Department of Biochemistry, University of MissouriRm117 Schweitzer Hall, Columbia, MO 65211
- Harry S. Truman Memorial Veterans Hospital, 1 Hospital DriveColumbia, MO 65201
| |
Collapse
|
22
|
Scalici JM, Harrer C, Allen A, Jazaeri A, Atkins KA, McLachlan KR, Slack-Davis JK. Inhibition of α4β1 integrin increases ovarian cancer response to carboplatin. Gynecol Oncol 2013; 132:455-61. [PMID: 24378876 DOI: 10.1016/j.ygyno.2013.12.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/18/2013] [Accepted: 12/20/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The inability to successfully treat women with ovarian cancer is due to the presence of metastatic disease at diagnosis and the development of platinum resistance. Ovarian cancer metastasizes throughout the peritoneal cavity by attaching to and invading through the mesothelium lining the peritoneum using a mechanism that involves α4β1 integrin and its ligand (vascular cell adhesion molecule) VCAM-1. Integrin α4β1 expression on tumor cells is known to confer protection from therapy in other cancers, notably multiple myeloma. We evaluated the role of α4β1 integrin in response to platinum-based therapy in a mouse model of peritoneal ovarian cancer metastasis by treatment with a humanized anti-α4β1 integrin function-blocking antibody. METHODS Integrin α4β1 expression on primary human ovarian cancer cells, fallopian tube and ovarian surface epithelia and fresh tumor was assessed by flow-cytometry. The therapeutic impact of anti-α4β1 treatment was assessed in murine models of platinum-resistant peritoneal disease and in vitro using the platinum resistant ovarian cancer cell lines. RESULTS Treatment of tumor-bearing mice with human-specific α4β1 integrin function-blocking antibodies, anti-VCAM-1 antibody or carboplatin alone had no effect on tumor burden compared to the IgG control group. However, the combined treatment of anti-α4β1 integrin or anti-VCAM-1 with carboplatin significantly reduced tumor burden. In vitro, the combination of carboplatin and anti-α4β1 integrin antibodies resulted in increased cell death and doubling time. CONCLUSIONS Our findings support a role for α4β1 integrin in regulating treatment response to carboplatin, implicating α4β1 integrin as a potential therapeutic target to influence platinum responsiveness in otherwise resistant disease.
Collapse
Affiliation(s)
- Jennifer M Scalici
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA
| | - Christine Harrer
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Anne Allen
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Amir Jazaeri
- Department of Obstetrics and Gynecology, University of Virginia, Charlottesville, VA, USA; The Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Kristen A Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA, USA; The Cancer Center, University of Virginia, Charlottesville, VA, USA
| | | | - Jill K Slack-Davis
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA; The Cancer Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|