1
|
Lee Y, Fang Y, Kuila S, Imoukhuede PI. Cross-family interactions of vascular endothelial growth factors and platelet-derived growth factors on the endothelial cell surface: A computational model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640640. [PMID: 40093087 PMCID: PMC11908192 DOI: 10.1101/2025.02.27.640640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Angiogenesis, the formation of new vessels from existing vessels, is mediated by vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF). Despite discoveries supporting the cross-family interactions between VEGF and PDGF families, sharing the binding partners between them makes it challenging to identify growth factors that predominantly affect angiogenesis. Systems biology offers promises to untangle this complexity. Thus, in this study, we developed a mass-action kinetics-based computational model for cross-family interactions between VEGFs (VEGF-A, VEGF-B, and PlGF) and PDGFs (PDGF-AA, PDGF-AB, and PDGF-BB) with their receptors (VEGFR1, VEGFR2, NRP1, PDGFRα, and PDGFRβ). The model, parametrized with our literature mining and surface resonance plasmon assays, was validated by comparing the concentration of VEGFR1 complexes with a previously constructed angiogenesis model. The model predictions include five outcomes: 1) the percentage of free or bound ligands and 2) receptors, 3) the concentration of free ligands, 4) the percentage of ligands occupying each receptor, and 5) the concentration of ligands that is bound to each receptor. We found that at equimolar ligand concentrations (1 nM), PlGF and VEGF-A were the main binding partners of VEGFR1 and VEGFR2, respectively. Varying the density of receptors resulted in the following five outcomes: 1) Increasing VEGFR1 density depletes the free PlGF concentration, 2) increasing VEGFR2 density decreases PDGF:PDGFRα complexes, 3) increased NRP1 density generates a biphasic concentration of the free PlGF, 4) increased PDGFRα density increases PDGFs:PDGFRα binding, and 5) increasing PDGFRβ density increases VEGF-A:PDGFRβ. Our model offers a reproducible, fundamental framework for exploring cross-family interactions that can be extended to the tissue level or intracellular molecular level. Also, our model may help develop therapeutic strategies in pathological angiogenesis by identifying the dominant complex in the cell signaling. Author summary New blood vessel formation from existing ones is essential for growth, healing, and reproduction. However, when this process is disrupted-either too much or too little-it can contribute to diseases such as cancer and peripheral arterial disease. Two key families of proteins, vascular endothelial growth factors (VEGFs) and platelet-derived growth factors (PDGFs), regulate this process. Traditionally, scientists believed that VEGFs only bind to VEGF receptors and PDGFs to PDGF receptors. However, recent findings show that these proteins can interact with each other's receptors, making it more challenging to understand and control blood vessel formation. To clarify these complex interactions, we combined computer modeling with biological data to map out which proteins bind to which receptors and to what extent. Our findings show that when VEGFs and PDGFs are present in equal amounts, VEGFs are the primary binding partners for VEGF receptors. We also explored how changes in receptor levels affect these interactions in disease-like conditions. This work provides a foundational computational model for studying cross-family interactions, which can be expanded to investigate tissue-level effects and processes inside cells. Ultimately, our model may help develop better treatments for diseases linked to abnormal blood vessel growth by identifying key protein-receptor interactions.
Collapse
|
2
|
Johri S, Bi K, Titchen BM, Fu J, Conway J, Crowdis JP, Vokes NI, Fan Z, Fong L, Park J, Liu D, He MX, Van Allen EM. Dissecting tumor cell programs through group biology estimation in clinical single-cell transcriptomics. Nat Commun 2025; 16:2090. [PMID: 40025015 PMCID: PMC11873288 DOI: 10.1038/s41467-025-57377-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
With the growth of clinical cancer single-cell RNA sequencing studies, robust differential expression methods for case/control analyses (e.g., treatment responders vs. non-responders) using gene signatures are pivotal to nominate hypotheses for further investigation. However, many commonly used methods produce a large number of false positives, do not adequately represent the patient-specific hierarchical structure of clinical single-cell RNA sequencing data, or account for sample-driven confounders. Here, we present a nonparametric statistical method, BEANIE, for differential expression of gene signatures between clinically relevant groups that addresses these issues. We demonstrate its use in simulated and real-world clinical datasets in breast cancer, lung cancer and melanoma. BEANIE outperforms existing methods in specificity while maintaining sensitivity, as demonstrated in simulations. Overall, BEANIE provides a methodological strategy to inform biological insights into unique and shared differentially expressed gene signatures across different tumor states, with utility in single-study, meta-analysis, and cross-validation across cell types.
Collapse
Affiliation(s)
- Shreya Johri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Breanna M Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Graduate Program in Biological and Biomedical Sciences, Boston, MA, USA
| | - Jingxin Fu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jake Conway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Graduate Program in Bioinformatics and Integrative Genomics, Boston, MA, USA
| | - Jett P Crowdis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Natalie I Vokes
- Department of Thoracic and Head and Neck Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Zenghua Fan
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
3
|
Basheer I, Wang H, Li G, Jehan S, Raza A, Du C, Ullah N, Li D, Sui G. β-caryophyllene sensitizes hepatocellular carcinoma cells to chemotherapeutics and inhibits cell malignancy through targeting MAPK signaling pathway. Front Pharmacol 2024; 15:1492670. [PMID: 39734415 PMCID: PMC11671526 DOI: 10.3389/fphar.2024.1492670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Background β-caryophyllene (BCP) is a naturally occurring bicyclic sesquiterpene extracted from various plants, and widely used as a medicinal agent for various diseases. During hepatocellular carcinoma (HCC) development, cancer cells generally exhibit increased cell proliferation due to mutations or aberrant expression of key regulatory genes. The current study determines the cytotoxic effects of BCP alone or in combination with doxorubicin (DOX) and cisplatin (DDP) on HCC cells, and elucidates the underlying mechanism of BCP to exert its anticancer activities. Materials and methods HepG2, SMMC-7721 HCC cells, and HL-7702 normal liver cells were treated with BCP, DOX, and DDP individually or combinatorially. Cell proliferation assay, flow cytometric assay, and Western blot were employed to evaluate the cytotoxic effects of these treatments. Transwell assays were used to examine BCP's effects on HCC cell migration and invasion. RNA-seq analysis was used to determine BCP's primary target genes in HepG2 cells. Integrative analysis of differentially expressed genes (DEGs) of RNA-seq data with an HCC TCGA dataset identified BCP-targeted genes that were verified by RT-qPCR analysis. Ectopic gene expression, cell viability, and colony formation assay were performed to validate the primary targets of BCP. Results BCP selectively inhibited HCC cell proliferation while exhibited relatively low toxicity in normal liver cells; however, DOX and DDP showed higher toxicity in normal cells than that in HCC cells. In combinatorial treatments, BCP synergistically enhanced cytotoxicity of DOX and DDP in HCC cells but this effect was markedly reduced in HL-7702 cells. BCP treatment reduced migration and invasion of HCC cells. Furthermore, RNA-seq analyses of BCP-treated HepG2 cells identified 433 protein-coding DEGs. Integrative analyses revealed five BCP-targeted DEGs regulating the MAPK signaling pathway. Among these five genes, three displayed a significantly positive correlation of their expression with the overall survival of HCC patients. As a primary target, PGF was significantly downregulated by BCP treatment, and its exogenous expression desensitized HCC cells to BCP-mediated inhibition. Discussion BCP inhibits malignant properties of HCC and synergistically sensitizes the anticancer activity of DOX and DDP. In HCC cells, BCP primarily targets the PGF gene and MAPK signaling pathway.
Collapse
Affiliation(s)
- Irum Basheer
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Hai Wang
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Guangyue Li
- Intelligent Biomedical Labs, Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Shah Jehan
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ali Raza
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Chentao Du
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Najeeb Ullah
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Dangdang Li
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Guangchao Sui
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| |
Collapse
|
4
|
Malekan M, Haass NK, Rokni GR, Gholizadeh N, Ebrahimzadeh MA, Kazeminejad A. VEGF/VEGFR axis and its signaling in melanoma: Current knowledge toward therapeutic targeting agents and future perspectives. Life Sci 2024; 345:122563. [PMID: 38508233 DOI: 10.1016/j.lfs.2024.122563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Melanoma is responsible for most skin cancer-associated deaths globally. The progression of melanoma is influenced by a number of pathogenic processes. Understanding the VEGF/VEGFR axis, which includes VEGF-A, PlGF, VEGF-B, VEGF-C, and VEGF-D and their receptors, VEGFR-1, VEGFR-2, and VEGFR-3, is of great importance in melanoma due to its crucial role in angiogenesis. This axis generates multifactorial and complex cellular signaling, engaging the MAPK/ERK, PI3K/AKT, PKC, PLC-γ, and FAK signaling pathways. Melanoma cell growth and proliferation, migration and metastasis, survival, and acquired resistance to therapy are influenced by this axis. The VEGF/VEGFR axis was extensively examined for their potential as diagnostic/prognostic biomarkers in melanoma patients and results showed that VEGF overexpression can be associated with unfavorable prognosis, higher level of tumor invasion and poor response to therapy. MicroRNAs linking to the VEGF/VEGFR axis were identified and, in this review, divided into two categories according to their functions, some of them promote melanoma angiogenesis (promotive group) and some restrict melanoma angiogenesis (protective group). In addition, the approach of treating melanoma by targeting the VEGF/VEGFR axis has garnered significant interest among researchers. These agents can be divided into two main groups: anti-VEGF and VEGFR inhibitors. These therapeutic options may be a prominent step along with the modern targeting and immune therapies for better coverage of pathological processes leading to melanoma progression and therapy resistance.
Collapse
Affiliation(s)
- Mohammad Malekan
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | | | - Ghasem Rahmatpour Rokni
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Armaghan Kazeminejad
- Department of Dermatology, Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences,Sari, Iran
| |
Collapse
|
5
|
Mäenpää N, Tiainen L, Hämäläinen M, Luukkaala T, Tanner M, Lahdenperä O, Vihinen P, Karihtala P, Kellokumpu-Lehtinen PL, Moilanen E, Jukkola A. Neuropilin-1 and placental growth factor as prognostic factors in metastatic breast cancer. BMC Cancer 2024; 24:331. [PMID: 38468231 DOI: 10.1186/s12885-024-12070-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/28/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Angiogenesis is crucial for tumor development, progression, and metastasizing. The most important regulator of angiogenesis is the vascular endothelial growth factor (VEGF) family, which is involved in multiple pathways in tumor microenvironment. The objective of this study was to investigate the prognostic value of the VEGF family in patients treated for metastatic breast cancer. The emphasis was on neuropilin-1 (NRP-1) and placental growth factor (PlGF). MATERIALS AND METHODS An analysis of eight members of the VEGF family was performed using baseline plasma samples of 65 patients treated for metastatic HER2 negative breast cancer in a phase II first-line bevacizumab plus chemotherapy trial. The patients were divided into two groups, high or low, according to the median for each VEGF family member. Progression-free survival (PFS) and overall survival (OS) were determined for each VEGF family member. RESULTS The patients with low plasma levels of NRP-1 and PlGF had a longer OS than those with high plasma levels [multivariable adjusted hazard ratios (HRs) 2.54 (95% confidence interval (CI) 1.11-5.82, p = 0.02) and 3.11 (95% CI 1.30-7.47, p = 0.01), respectively]. The patients with low levels of both NRP-1 and PlGF had a remarkably long OS with HR of 6.24, (95% CI 1.97-19.76, p = 0.002). In addition, high baseline NRP-1 level was associated with a significantly shorter PFS [multivariable adjusted HR 2.90 (95% CI 1.02-8.28, p = 0.04)] than that in the low-level group, and a high baseline vascular endothelial growth factor receptor-2 level was associated with a longer PFS [multivariable adjusted HR 0.43 (95% CI 0.19-0.98, p = 0.04)]. CONCLUSION Especially NRP-1 and PlGF have prognostic potential in metastatic breast cancer patients treated with a bevacizumab-taxane combination. Patients with low plasma levels of NRP-1 or PlGF have longer OS than patients with high levels. Patients with both low NRP-1 and PlGF levels appear to have excellent long-term survival. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT00979641, registration date 18/09/2009. The regional Ethics Committee: R08142M, registration date 18/11/2008.
Collapse
Affiliation(s)
- Niina Mäenpää
- Faculty of Medicine and Health Technology, Tampere University, FI-33014, Tampere, Finland.
- Department of Oncology, Tays Cancer Centre, Tampere University Hospital, FICAN Mid, Teiskontie 35, FI-33521, Tampere, Finland.
| | - Leena Tiainen
- Faculty of Medicine and Health Technology, Tampere University, FI-33014, Tampere, Finland
- Department of Oncology, Tays Cancer Centre, Tampere University Hospital, FICAN Mid, Teiskontie 35, FI-33521, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere University, 33014, Tampere, P.O. Box 100, Finland
| | - Tiina Luukkaala
- Research, Development and Innovation Center, Tampere University Hospital, Teiskontie 35, FI-33521, Tampere, Finland
- Health Sciences, Faculty of Social Sciences, Tampere University, FI-33521, Tampere, P.O. Box 2000, Finland
| | - Minna Tanner
- Department of Oncology, Tays Cancer Centre, Tampere University Hospital, FICAN Mid, Teiskontie 35, FI-33521, Tampere, Finland
| | - Outi Lahdenperä
- FICAN West Cancer Centre, Turku University Hospital, 20521, Turku, P.O. Box 52, Finland
| | - Pia Vihinen
- FICAN West Cancer Centre, Turku University Hospital, 20521, Turku, P.O. Box 52, Finland
| | - Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Centre, University of Helsinki, FI-00029, Helsinki, P.O. Box 180, Finland
| | - Pirkko-Liisa Kellokumpu-Lehtinen
- Faculty of Medicine and Health Technology, Tampere University, FI-33014, Tampere, Finland
- Research, Development and Innovation Center, Tampere University Hospital, Teiskontie 35, FI-33521, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere University, 33014, Tampere, P.O. Box 100, Finland
| | - Arja Jukkola
- Faculty of Medicine and Health Technology, Tampere University, FI-33014, Tampere, Finland
- Department of Oncology, Tays Cancer Centre, Tampere University Hospital, FICAN Mid, Teiskontie 35, FI-33521, Tampere, Finland
| |
Collapse
|
6
|
Yao M, Chen L, Peng P, Zhong Z. Women with preeclampsia may have reduced risk of breast cancer: a meta-analysis of cohort studies with 7.8 million participants. Hypertens Pregnancy 2023; 42:2265482. [PMID: 37847190 DOI: 10.1080/10641955.2023.2265482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/21/2023] [Indexed: 10/18/2023]
Abstract
This review pooled data from the literature to examine the association between preeclampsia (PE) and subsequent risk of breast cancer in women. Cohort studies published in the databases of PubMed, Embase, Scopus, and Web of Science up to 18 July 2023 were searched. Adjusted data were pooled to obtain the risk ratio (RR). Eleven studies with 15 cohorts and a cumulative sample size of 7,838,693 women were included. Meta-analysis of all studies demonstrated a reduced risk of breast cancer in women with PE as compared to those without PE (RR: 0.89 95% CI: 0.83, 0.95 p < 0.001 I2 = 50%). Follow-up ranged from 8 to 29.2 years. Results did not change during sensitivity analysis. Outcomes varied on subgroup analysis based on location, study type, data extraction method, incidence of breast cancer, and follow-up. To conclude, women with PE may have a reduced risk of breast cancer later in life. However, the risk reduction is minimal and may not have much clinical significance. The evidence is also limited by high inter-study heterogeneity and lack of adjustment of all possible confounders.
Collapse
Affiliation(s)
- Min Yao
- Department of Breast Surgery, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Lijie Chen
- Department of Breast Surgery, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Puchao Peng
- Department of Breast Surgery, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Zhiwei Zhong
- Department of Breast Surgery, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| |
Collapse
|
7
|
Brogowska KK, Zajkowska M, Mroczko B. Vascular Endothelial Growth Factor Ligands and Receptors in Breast Cancer. J Clin Med 2023; 12:jcm12062412. [PMID: 36983412 PMCID: PMC10056253 DOI: 10.3390/jcm12062412] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy responsible for the largest number of deaths in women worldwide. The risk of developing BC is predisposed by many factors such as age, presence of genetic mutations or body weight. The diagnosis is mostly made relatively late, which is why patients are exposed to radical surgical treatments, long-term chemotherapy and lower survival rates. There are no sufficiently sensitive and specific screening tests; therefore, researchers are still looking for new diagnostic biomarkers that would indicate the appearance of neoplastic changes in the initial stage of neoplasm. The VEGF family of proteins (VEGF-A, VEGF-B, VEGF-C, VEGF-D, EG-VEGF, PlGF) and their receptors are significant factors in the pathogenesis of BC. They play a significant role in the process of angiogenesis and lymphangiogenesis in both physiological and pathological conditions. The usefulness of these proteins as potential diagnostic biomarkers has been initially proven. Moreover, the blockage of VEGF-related pathways seems to be a valid therapeutic target. Recent studies have tried to describe novel strategies, including targeting pericytes, use of miRNAs and extracellular tumor-associated vesicles, immunotherapeutic drugs and nanotechnology. This indicates their possible contribution to the formation of breast cancer and their usefulness as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | - Monika Zajkowska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
8
|
Valipour A, Namdar Ahmad Abad H, Vatanchian M, Arezumand R. Designing and Developing Enzyme-Linked Immunosorbent Assay Sandwich Kit for Measuring Placental Growth Factor Concentration. Monoclon Antib Immunodiagn Immunother 2023; 42:3-8. [DOI: https:/doi.org/10.1089/mab.2022.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Affiliation(s)
- Arezoo Valipour
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hasan Namdar Ahmad Abad
- Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehran Vatanchian
- Department of Anatomical Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Roghaye Arezumand
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Department of Natural Products and Medicinal Plants Research Center, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|
9
|
Valipour A, Namdar Ahmad Abad H, Vatanchian M, Arezumand R. Designing and Developing Enzyme-Linked Immunosorbent Assay Sandwich Kit for Measuring Placental Growth Factor Concentration. Monoclon Antib Immunodiagn Immunother 2023; 42:3-8. [PMID: 36779992 DOI: 10.1089/mab.2022.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023] Open
Abstract
Placental growth factor (PlGF) is an angiogenic factor belonging to vascular endothelial growth factor family. This factor is mainly expressed in the placenta and have important role in blood supply to embryonic tissues and fetal. According to accumulated data after 10th week of gestational age the expression of PlGF is increased. The peak of this factor is seen in the 30th week of pregnancy. The abnormal expression of PlGF have been seen in some diseases such as preeclampsia, eclampsia, cancer, and atherosclerotic lesions. Preeclampsia is a pregnancy complication characterized by high blood pressure and signs of damage to another organ system, most often the liver and kidneys. As noted the level of PlGF decreased in preeclampsia is, therefore, timely and accurate measurement of this factor could help in diagnosing preeclampsia. In this study, we worked on development of sandwich enzyme-linked immunosorbent assay (ELISA) kit for measurement of PlGF, to this end, bivalent single-domain monoclonal antibody with high affinity binding was used as detection antibody and rabbit polyclonal antibody with strong signal to PlGF was used as capture antibody. Both types of antibodies were produced in the laboratory. Therefore, this study showed that the designed kit can measure PlGF up to 7.5 pg/mL. Intra-assay accuracy was <10% and interassay accuracy was <15%. The ELISA sandwich kit had the appropriate sensitivity and accuracy in measuring human PlGF.
Collapse
Affiliation(s)
- Arezoo Valipour
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hasan Namdar Ahmad Abad
- Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehran Vatanchian
- Department of Anatomical Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Roghaye Arezumand
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.,Department of Natural Products and Medicinal Plants Research Center, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|
10
|
Azzarito G, Kurmann L, Leeners B, Dubey RK. Micro-RNA193a-3p Inhibits Breast Cancer Cell Driven Growth of Vascular Endothelial Cells by Altering Secretome and Inhibiting Mitogenesis: Transcriptomic and Functional Evidence. Cells 2022; 11:cells11192967. [PMID: 36230929 PMCID: PMC9562882 DOI: 10.3390/cells11192967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/12/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer (BC) cell secretome in the tumor microenvironment (TME) facilitates neo-angiogenesis by promoting vascular endothelial cell (VEC) growth. Drugs that block BC cell growth or angiogenesis can restrict tumor growth and are of clinical relevance. Molecules that can target both BC cell and VEC growth as well as BC secretome may be more effective in treating BC. Since small non-coding microRNAs (miRs) regulate cell growth and miR193a-3p has onco-suppressor activity, we investigated whether miR193a-3p inhibits MCF-7-driven growth (proliferation, migration, capillary formation, signal transduction) of VECs. Using BC cells and VECs grown in monolayers or 3D spheroids and gene microarrays, we demonstrate that: pro-growth effects of MCF-7 and MDA-MB231 conditioned medium (CM) are lost in CM collected from MCF-7/MDA-MB231 cells pre-transfected with miR193a-3p (miR193a-CM). Moreover, miR193a-CM inhibited MAPK and Akt phosphorylation in VECs. In microarray gene expression studies, miR193a-CM upregulated 553 genes and downregulated 543 genes in VECs. Transcriptomic and pathway enrichment analysis of differentially regulated genes revealed downregulation of interferon-associated genes and pathways that induce angiogenesis and BC/tumor growth. An angiogenesis proteome array confirmed the downregulation of 20 pro-angiogenesis proteins by miR193a-CM in VECs. Additionally, in MCF-7 cells and VECs, estradiol (E2) downregulated miR193a-3p expression and induced growth. Ectopic expression of miR193a-3p abrogated the growth stimulatory effects of estradiol E2 and serum in MCF-7 cells and VECs, as well as in MCF-7 and MCF-7+VEC 3D spheroids. Immunostaining of MCF-7+VEC spheroid sections with ki67 showed miR193a-3p inhibits cell proliferation. Taken together, our findings provide first evidence that miR193a-3p abrogates MCF-7-driven growth of VECs by altering MCF-7 secretome and downregulating pro-growth interferon signals and proangiogenic proteins. Additionally, miR193a-3p inhibits serum and E2-induced growth of MCF-7, VECs, and MCF-7+VEC spheroids. In conclusion, miRNA193a-3p can potentially target/inhibit BC tumor angiogenesis via a dual mechanism: (1) altering proangiogenic BC secretome/TME and (2) inhibiting VEC growth. It may represent a therapeutic molecule to target breast tumor growth.
Collapse
Affiliation(s)
- Giovanna Azzarito
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland
| | - Lisa Kurmann
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland
| | - Brigitte Leeners
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland
| | - Raghvendra K. Dubey
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Correspondence:
| |
Collapse
|
11
|
Characterization of the Expression of Angiogenic Factors in Cutaneous Squamous Cell Carcinoma of Domestic Cats. Vet Sci 2022; 9:vetsci9070375. [PMID: 35878392 PMCID: PMC9351683 DOI: 10.3390/vetsci9070375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is a common malignant skin cancer with a significant impact on health, and it is important to determine the degree of reliance of CSCC on angiogenesis for growth and metastasis. Major regulators of angiogenesis are the vascular endothelial growth factor (VEGF) family and their associated receptors. Alternative pre-mRNA splicing produces multiple isoforms of VEGF-A and PLGF with distinct biological properties. Several studies highlight the function of VEGF-A in CSCC, but there are no studies of the different isoforms of VEGF-A and PLGF for this neoplasm. We characterized the expression of three isoforms of VEGF-A, two isoforms of PLGF, and their receptors in cat CSCC biopsies compared to normal haired skin (NHS). Although our results revealed no significant changes in transcript levels of panVEGF-A or their isoforms, the mRNA levels of PLGF I and the receptors Flt-1 and KDR were downregulated in CSCC compared to NHS. Differences were observed in ligand:receptor mRNA expression ratio, with the expression of VEGF-A relative to its receptor KDR higher in CSCC, which is consistent with our hypothesis and prior human SCC studies. Immunolocalization in tissue showed increased expression of all measured factors and receptors in tumor cells compared to NHS and surrounding vasculature. We conclude that the factors measured may play a pivotal role in CSCC growth, although further studies are needed to clarify the role of angiogenic factors in feline CSCC.
Collapse
|
12
|
Zhen Z, Shen Z, Hu Y, Sun P. Screening and identification of angiogenesis-related genes as potential novel prognostic biomarkers of hepatocellular carcinoma through bioinformatics analysis. Aging (Albany NY) 2021; 13:17707-17733. [PMID: 34252885 PMCID: PMC8312452 DOI: 10.18632/aging.203260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 06/23/2021] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality, which makes the prognostic prediction challenging. Angiogenesis appears to be of critical importance in the progression and metastasis of HCC. Some of the angiogenesis-related genes promote this process, while other anti-angiogenesis genes suppress tumor growth and metastasis. Therefore, the comprehensive prognostic value of multiple angiogenesis-related genes in HCC needs to be further clarified. In this study, the mRNA expression profile of HCC patients and the corresponding clinical data were acquired from multiple public databases. Univariate Cox regression analysis was utilized to screen out differentially expressed angiogenesis-related genes with prognostic value. A multigene signature was established with the least absolute shrinkage and selection operator Cox regression in the Cancer Genome Atlas cohort, and validated through an independent cohort. The results suggested that a total of 16 differentially expressed genes (DEGs) were associated with overall survival (OS) and a 7-gene signature was constructed. The risk score of each patient was calculated using this signature, the median value of which was used to divide these patients into a high-risk group and a low-risk group. Compared with the low-risk group, the patients in the high-risk group had a poor prognosis. The risk score was an independent predictor for OS through multivariate Cox regression analysis. Then, unsupervised learning was used to verify the validity of this 7-gene signature. A nomogram by further integrating clinical information and the prognostic signature was utilized to predict prognostic risk and individual OS. Functional enrichment analyses demonstrated that these DEGs were enriched in the pathways of cell proliferation and mitosis, and the immune cell infiltration was significantly different between the two risk groups. In summary, a novel angiogenesis-related genes signature could be used to predict the prognosis of HCC and for targeted therapy.
Collapse
Affiliation(s)
- Zili Zhen
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai 201508, China.,Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai 201508, China.,Department of Surgery, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhemin Shen
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai 201508, China.,Department of Surgery, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yanmei Hu
- Department of Paediatrics, the Second Hospital of Jilin University, Changchun 130041, Jilin, China
| | - Peilong Sun
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai 201508, China.,Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai 201508, China
| |
Collapse
|
13
|
Persson E, Gregersson P, Gustafsson A, Fitzpatrick P, Rhost S, Ståhlberg A, Landberg G. Patient-derived scaffolds influence secretion profiles in cancer cells mirroring clinical features and breast cancer subtypes. Cell Commun Signal 2021; 19:66. [PMID: 34090457 PMCID: PMC8178857 DOI: 10.1186/s12964-021-00746-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/27/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Breast cancer is a common malignancy with varying clinical behaviors and for the more aggressive subtypes, novel and more efficient therapeutic approaches are needed. Qualities of the tumor microenvironment as well as cancer cell secretion have independently been associated with malignant clinical behaviors and a better understanding of the interplay between these two features could potentially reveal novel targetable key events linked to cancer progression. METHODS A newly developed human derived in vivo-like growth system, consisting of decellularized patient-derived scaffolds (PDSs) recellularized with standardized breast cancer cell lines (MCF7 and MDA-MB-231), were used to analyze how 63 individual patient specific microenvironments influenced secretion determined by proximity extension assays including 184 proteins and how these relate to clinical outcome. RESULTS The secretome from cancer cells in PDS cultures varied distinctly from cells grown as standard monolayers and besides a general increase in secretion from PDS cultures, several secreted proteins were only detectable in PDSs. Monolayer cells treated with conditioned media from PDS cultures, further showed increased mammosphere formation demonstrating a cancer stem cell activating function of the PDS culture induced secretion. The detailed secretomic profiles from MCF7s growing on 57 individual PDSs differed markedly but unsupervised clustering generated three separate groups having similar secretion profiles that significantly correlated to different clinical behaviors. The secretomic profile that associated with cancer relapse and high grade breast cancer showed induced secretion of the proteins IL-6, CCL2 and PAI-1, all linked to cancer stem cell activation, metastasis and priming of the pre-metastatic niche. Cancer promoting pathways such as "Suppress tumor immunity" and "Vascular and tissue remodeling" was also linked to this more malignant secretion cluster. CONCLUSION PDSs repopulated with cancer cells can be used to assess how cancer secretion is effected by specific and varying microenvironments. More malignant secretion patterns induced by specific patient based cancer microenvironments could further be identified pinpointing novel therapeutic opportunities targeting micro environmentally induced cancer progression via secretion of potent cytokines. Video abstract.
Collapse
Affiliation(s)
- Emma Persson
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1G, 41390, Gothenburg, Sweden
| | - Pernilla Gregersson
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1G, 41390, Gothenburg, Sweden
| | - Anna Gustafsson
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1G, 41390, Gothenburg, Sweden
| | - Paul Fitzpatrick
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1G, 41390, Gothenburg, Sweden
| | - Sara Rhost
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1G, 41390, Gothenburg, Sweden
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1G, 41390, Gothenburg, Sweden.,Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, 41390, Gothenburg, Sweden.,Department of Clinical Genetics and Genomics, Sahlgrenska University Hostpital, Region Västra Götaland, 41390, Gothenburg, Sweden
| | - Göran Landberg
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1G, 41390, Gothenburg, Sweden.
| |
Collapse
|
14
|
Oliveira MDMS, Salgado CDM, Viana LR, Gomes-Marcondes MCC. Pregnancy and Cancer: Cellular Biology and Mechanisms Affecting the Placenta. Cancers (Basel) 2021; 13:1667. [PMID: 33916290 PMCID: PMC8037654 DOI: 10.3390/cancers13071667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer during pregnancy is rarely studied due to its low incidence (1:1000). However, as a result of different sociocultural and economic changes, women are postponing pregnancy, so the number of pregnant women with cancer has been increasing in recent years. The importance of studying cancer during pregnancy is not only based on maternal and foetal prognosis, but also on the evolutionary mechanisms of the cell biology of trophoblasts and neoplastic cells, which point out similarities between and suggest new fields for the study of cancer. Moreover, the magnitude of how cancer factors can affect trophoblastic cells, and vice versa, in altering the foetus's nutrition and health is still a subject to be understood. In this context, the objective of this narrative review was to show that some researchers point out the importance of supplementing branched-chain amino acids, especially leucine, in experimental models of pregnancy associated with women with cancer. A leucine-rich diet may be an interesting strategy to preserve physiological placenta metabolism for protecting the mother and foetus from the harmful effects of cancer during pregnancy.
Collapse
Affiliation(s)
| | | | - Lais Rosa Viana
- Nutrition and Cancer Laboratory, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Sao Paulo 13083-862, Brazil; (M.d.M.S.O.); (C.d.M.S.)
| | - Maria Cristina Cintra Gomes-Marcondes
- Nutrition and Cancer Laboratory, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Sao Paulo 13083-862, Brazil; (M.d.M.S.O.); (C.d.M.S.)
| |
Collapse
|
15
|
Cechova M, Chocholaty M, Babjuk M, Zima T, Havlova K, Koldova M, Schmidt M, Kalousova M. Diagnostic and prognostic value of placental growth factor serum concentration in clear cell renal cell carcinoma. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2021; 165:375-379. [PMID: 33612835 DOI: 10.5507/bp.2021.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 01/08/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND AND AIM Placental Growth Factor (PlGF) plays a crucial role in angiogenesis and was identified as a potential prognostic biomarker in various types of cancer. Therefore, we evaluated the diagnostic accuracy and prognostic value of PlGF serum concentration in patients with clear cell renal cell carcinoma (ccRCC). PATIENTS AND METHODS A total of 49 patients subjected to partial or radical nephrectomy for ccRCC [localized without relapse (lccRCC; n=31), localized with later relapse (rccRCC; n=8), primary metastatic cancer (mccRCC; n=10); median of follow-up 4.4 years] were enrolled in a prospective study to assess the significance of PlGF serum concentration. PlGF was measured prior to surgery and 3 months postoperatively. Our control group consisted of 38 healthy subjects. RESULTS PlGF serum concentration was significantly higher in ccRCC compared to controls (P=0.002). The cut-off value of PlGF concentration for the risk of ccRCC was determined at 12.71 pg/mL (AUC=0.729; P=0.0001). Prior to surgery, among ccRCC subgroups, significantly higher PlGF concentration was detected in mccRCC compared to lccRCC (P=0.002). Postoperatively, we observed a tendency to higher PlGF serum concentration in rccRCC compared to lccRCC subgroup, however without significance (P=0.17). The cut-off value for the risk of relapse was 11.41 pg/mL (AUC=0.792; P=0.0003). In subjects with localized ccRCC with PlGF concentration below 11.41 pg/mL 3-years cancer specific survival was 93% compared to 61% in subject with concentration above the cut-off value (P=0.018). CONCLUSION Based on our findings, PlGF serum concentration seems to be a useful biomarker in diagnostics and prediction of prognosis in ccRCC.
Collapse
Affiliation(s)
| | | | - Marek Babjuk
- Department of Urology, University Hospital Motol and 2
| | - Tomas Zima
- Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1
| | - Klara Havlova
- Department of Urology, University Hospital Motol and 2
| | | | - Marek Schmidt
- Department of Urology, University Hospital Motol and 2
| | - Marta Kalousova
- Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1
| |
Collapse
|
16
|
Modulating the Crosstalk between the Tumor and the Microenvironment Using SiRNA: A Flexible Strategy for Breast Cancer Treatment. Cancers (Basel) 2020; 12:cancers12123744. [PMID: 33322132 PMCID: PMC7763441 DOI: 10.3390/cancers12123744] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/27/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary With this review we aimed to collect the most relevant scientific findings regarding siRNA therapeutic tools against breast cancer microenvironment. Remarkably, breast cancer treatments have been redirected towards the tumor microenvironment components, mainly involved in patients’ relapse and pharmacological resistance. Therefore, siRNAs represent a promising strategy to jeopardize the tumor microenvironment interplay thanks to their non-toxic and specific effects. Abstract Tumorigenesis is a complex and multistep process in which sequential mutations in oncogenes and tumor-suppressor genes result in enhanced proliferation and apoptosis escape. Over the past decades, several studies have provided evidence that tumors are more than merely a mass of malignant cancer cells, with the tumor microenvironment (TME) also contributing to cancer progression. For this reason, the focus of cancer research in recent years has shifted from the malignant cancer cell itself to the TME and its interactions. Since the TME actively participates in tumor progression, therapeutic strategies targeting it have created great interest. In this context, much attention has been paid to the potential application of small interfering RNA (siRNA), a class of non-coding RNA that has the ability to downregulate the expression of target genes in a sequence-specific way. This is paving the way for a novel therapeutic approach for the treatment of several diseases, including cancer. In this review, we describe recent efforts in developing siRNA therapeutics for the treatment of breast cancer, with particular emphasis on TME regulation. We focus on studies that adapt siRNA design to reprogram/re-educate the TME and eradicate the interplay between cancer cells and TME.
Collapse
|
17
|
Occurrence of Fibrotic Tumor Vessels in Grade I Meningiomas Is Strongly Associated with Vessel Density, Expression of VEGF, PlGF, IGFBP-3 and Tumor Recurrence. Cancers (Basel) 2020; 12:cancers12103075. [PMID: 33096816 PMCID: PMC7593950 DOI: 10.3390/cancers12103075] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is a key feature during oncogenesis and remains a potential target of antiangiogenic therapy. While commonly described in high-grade lesions, vascularization and its correlation with prognosis in grade I meningiomas is largely unexplored. In the histological classification, not only the number but also the composition of blood vessels seems to be important. Therefore, tumor vessel density and fibrosis were correlated with clinical and imaging variables and prognosis in 295 patients with intracranial grade I meningioma. Expression of pro-angiogenic proteins within the meningiomas was investigated by proteome analyses and further validated by immunohistochemical staining. Fibrotic tumor vessels (FTV) were detected in 48% of all tumors and strongly correlated with vessel density, but not with the histopathological tumor subtype. Occurrence of FTV was correlated with a 2-fold increased risk of recurrence in both univariate and multivariate analyses. Explorative proteome analyses revealed upregulation of VEGF (vascular endothelial growth factor), PlGF (placental growth factor), and IGFBP-3 (insulin-like growth factor-binding protein-3) in tumors displaying FTV. Immunohistochemical analyses confirmed strong correlations between tumor vessel fibrosis and expression of VEGF, PlGF, and IGFBP-3. Presence of FTV was strongly associated with disruption of the arachnoid layer on preoperative MRI in univariate and multivariate analyses. In summary, the occurrence of fibrotic tumor vessels in grade I meningiomas is strongly associated with vessel density, disruption of the arachnoid layer, expression of VEGF, PlGF, IGFBP-3 and tumor recurrence.
Collapse
|
18
|
Wang F, Zhang L, Zhang F, Wang J, Wang Y, Man D. First trimester serum PIGF is associated with placenta accreta. Placenta 2020; 101:39-44. [PMID: 32916477 DOI: 10.1016/j.placenta.2020.08.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/09/2020] [Accepted: 08/31/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES In this retrospective study, we investigated whether first trimester serum placental growth factor (PIGF) differed amongst pregnancies with placenta previa-accreta and non-adherent placenta previa and healthy pregnancies. METHODS In 1 January 2017-30 September 2019, a total of 177 pregnant females were included in the study, as follows: 35 cases of placenta previa-accreta, 30 cases of non-adherent placenta previa, and 112 cases of age and BMI-matched, healthy pregnant controls. PIGF multiples of the median (MoM) were acquired from laboratory data files. The predictor of placenta accreta was analyzed by using multiple logistic regression analysis. RESULTS PIGF MoM of placenta previa-accreta group was significantly higher than those of the non-adherent placenta previa group and control group (p = 0.0098 < 0.01, p = 0.0002 < 0.01). Serum PIGF was found to be significantly positively associated with placenta accreta after adjusted gestational week at time of blood sampling, BMI, and age (OR: 4.83; 95% CI: 1.91-12.24;p = 0.0009 < 0.01). In addition, previous cesarean section history (OR: 2.75; 95% CI: 1.23-6.17; p = 0.014 < 0.05) and smoking (OR: 9.17; 95% CI: 1.69-49.62; p = 0.010 < 0.05) were also significantly associated with placenta accreta. CONCLUSION Increased first trimester serum PIGF was significantly positively associated with placenta accreta, suggesting that the potential role of PIGF in identifying pregnancies at high risk for placenta accreta. Previous cesarean section history and smoking may be the risk factors for accreta in placenta previa patients.
Collapse
Affiliation(s)
- Fengge Wang
- Department of Obstetric, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Lihua Zhang
- Department of Obstetric, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Fanyong Zhang
- Department of Obstetric, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Jishui Wang
- Department of Obstetric, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yangping Wang
- Department of Obstetric, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Dongmei Man
- Department of Obstetric, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
19
|
Di Paolo V, Colletti M, Ferruzzi V, Russo I, Galardi A, Alessi I, Milano GM, Di Giannatale A. Circulating Biomarkers for Tumor Angiogenesis: Where Are We? Curr Med Chem 2020; 27:2361-2380. [PMID: 30129403 DOI: 10.2174/0929867325666180821151409] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/10/2018] [Accepted: 07/17/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND In recent years, several anti-angiogenic drugs have been developed and their addition to standard treatment has been associated with clinical benefits. However, the response to anti-angiogenic therapy is characterized by considerable variability. In this context, the development of dynamic non-invasive biomarkers would be helpful to elucidate the emergence of anti-angiogenic resistance as well as to correctly address the treatment. OBJECTIVES The purpose of this review is to describe current reports on circulating diagnostic and prognostic biomarkers related to angiogenesis. We further discuss how this non-invasive strategy could improve the monitoring of tumor treatment and help clinical strategy. RESULTS We discuss the latest evidence in the literature regarding circulating anti-angiogenic markers. Besides growth factor proteins, different circulating miRNAs could exert a pro- or anti-angiogenic activity so as to represent suitable candidates for a non-invasive strategy. Recent reports indicate that tumor-derived exosomes, which are small membrane vesicles abundant in biological fluids, also have an impact on vascular remodeling. CONCLUSION Numerous circulating biomarkers related to angiogenesis have been recently identified. Their use will allow identifying patients who are more likely to benefit from a specific anti-angiogenic treatment, as well as detecting those who will develop resistance and/or adverse effects. Nonetheless, further studies are required to elucidate the role of these biomarkers in clinical settings.
Collapse
Affiliation(s)
- Virginia Di Paolo
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Piazza di Sant'Onofrio, 4-00165 Rome, Italy
| | - Marta Colletti
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Piazza di Sant'Onofrio, 4-00165 Rome, Italy
| | - Valentina Ferruzzi
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Piazza di Sant'Onofrio, 4-00165 Rome, Italy
| | - Ida Russo
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Piazza di Sant'Onofrio, 4-00165 Rome, Italy
| | - Angela Galardi
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Piazza di Sant'Onofrio, 4-00165 Rome, Italy
| | - Iside Alessi
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Piazza di Sant'Onofrio, 4-00165 Rome, Italy
| | - Giuseppe Maria Milano
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Piazza di Sant'Onofrio, 4-00165 Rome, Italy
| | - Angela Di Giannatale
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Piazza di Sant'Onofrio, 4-00165 Rome, Italy
| |
Collapse
|
20
|
Fares J, Kanojia D, Rashidi A, Ulasov I, Lesniak MS. Genes that Mediate Metastasis across the Blood-Brain Barrier. Trends Cancer 2020; 6:660-676. [PMID: 32417182 DOI: 10.1016/j.trecan.2020.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/15/2020] [Accepted: 04/22/2020] [Indexed: 12/21/2022]
Abstract
Brain metastasis is an important cause of mortality in patients with cancer and represents the majority of all intracranial tumors. A key step during the metastatic journey of the cancer cell to the brain is the invasion through the blood-brain barrier (BBB). Nevertheless, the molecular mechanisms that govern this process remain unknown. The BBB has been blamed for limiting the access of therapeutic drugs to the brain, which provides a safe haven for cancer cells in the brain and confers poor prognosis for the patient. Here, we explore the genes that control the transmigration of metastatic cancer cells across the BBB, offering new targets for the development of gene and cell therapies against brain metastases.
Collapse
Affiliation(s)
- Jawad Fares
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
21
|
Van Cutsem E, Paccard C, Chiron M, Tabernero J. Impact of Prior Bevacizumab Treatment on VEGF-A and PlGF Levels and Outcome Following Second-Line Aflibercept Treatment: Biomarker Post Hoc Analysis of the VELOUR Trial. Clin Cancer Res 2019; 26:717-725. [PMID: 31727675 DOI: 10.1158/1078-0432.ccr-19-1985] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/07/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Aflibercept is a targeted anti-VEGF therapy used to treat patients with metastatic colorectal cancer (mCRC) following progression on oxaliplatin-based regimens. This post hoc study evaluated the effect of prior bevacizumab treatment and growth factor levels on patient outcomes associated with aflibercept in the VELOUR phase III trial. EXPERIMENTAL DESIGN Baseline biomarker plasma concentrations were measured using a bead-based multiplex assay. Patients were grouped according to prior bevacizumab treatment, second-line treatment, and serum biomarker concentrations, and analyzed for overall survival (OS) and progression-free survival (PFS). RESULTS Plasma samples were available for 553 patients (placebo n = 265; aflibercept n = 288), of which 169 had received prior bevacizumab. Nine biomarkers implicated in angiogenesis or bevacizumab resistance correlated with prior bevacizumab therapy. VEGF-A and placental growth factor (PlGF) were the most significantly increased in patients who had received prior bevacizumab compared with those who had not received prior bevacizumab. In the placebo group, patients with high VEGF-A (>144 pg/mL) levels at baseline had worse OS and PFS compared with patients with lower levels at baseline (9.6 vs. 12.9 months). This was also seen in patients who received placebo and had high baseline PlGF (>8 pg/mL; 9.7 vs. 11.7 months). In the aflibercept group, prolonged OS and PFS were observed regardless of baseline VEGF-A or PlGF levels. CONCLUSIONS High VEGF-A and PlGF serum levels may underlie development of resistance to bevacizumab in patients with mCRC. Aflibercept retains its activity regardless of baseline VEGF-A and PlGF levels and may be an effective second-line treatment for patients with bevacizumab-induced resistance.
Collapse
Affiliation(s)
| | | | | | - Josep Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), IOB-Quiron, UVic, Barcelona, Spain
| |
Collapse
|
22
|
Ma Q, Gu JT, Wang B, Feng J, Yang L, Kang XW, Duan P, Sun X, Liu PJ, Wang JC. PlGF signaling and macrophage repolarization contribute to the anti-neoplastic effect of metformin. Eur J Pharmacol 2019; 863:172696. [PMID: 31562866 DOI: 10.1016/j.ejphar.2019.172696] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/05/2023]
Abstract
Placental growth factor (PlGF) related signaling pathway has been shown to have close relationship with the progression of cancers. Metformin has been reported to have an inhibitory effect on PlGF expression in a breast cancer model. However, little is known about whether the anti-neoplastic activity of metformin is contributed by its inhibitory effect on PlGF expression. Protein, mRNA and secretion levels of PlGF were tested and the proliferation of cancer cells was determined. After treatment of metformin, BALB/c mice bearing 4T1 tumors were sacrificed and immunohistochemistry staining of the tumor sections was obtained. Baseline expression of autocrine PlGF varied between different breast cancer cell lines, while the expression of vascular endothelial growth factor receptor-1 (VEGFR-1) was comparable between cell lines. Other clinical data showed that the expression of PlGF other than VEGFR-1 had a prognostic value for patients with breast cancers. Metformin significantly decreased the secretion and mRNA levels of PlGF, which greatly contributed to its inhibitory effect on the proliferation of breast cancer cells with high P1GF expression. The unresponsiveness of tumor cells with low PlGF expression to genetic silencing was reversed by the supplementation of exogenous PlGF. Systemic metformin administration apparently inhibited the in vivo growth of 4T1 carcinoma, which was accompanied by the repolarization of macrophages from M2 to M1. These findings indicated that both autocrine and paracrine PlGF signaling and macrophage repolarization are involved in the progression of breast cancer, which could be targeted by metformin.
Collapse
Affiliation(s)
- Qiang Ma
- Department of peripheral vascular diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Jing-Tao Gu
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Bo Wang
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Jun Feng
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Lin Yang
- Department of psychiatry, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Xiao-Wei Kang
- Education Administration Office, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Peng Duan
- Emergency Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Xin Sun
- Department of Thoracic Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Pei-Jun Liu
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| | - Ji-Chang Wang
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China; Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, PR China.
| |
Collapse
|
23
|
Wei SC, Tsao PN, Wang YT, Lin BR, Wu DC, Tsai WS, Chen JS, Wong JM. Using serum placenta growth factor could improve the sensitivity of colorectal cancer screening in fecal occult blood negative population: A multicenter with independent cohort validation study. Cancer Med 2019; 8:3583-3591. [PMID: 31063258 PMCID: PMC6601572 DOI: 10.1002/cam4.2216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/08/2019] [Accepted: 04/14/2019] [Indexed: 12/13/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common cancers worldwide. Screening for CRC using the fecal occult blood test (FOBT) is feasible and useful for decreasing disease‐related mortality; however, its sensitivity and compliance are unsatisfactory. Methods This study examined the efficacy of using serum placenta growth factor (PlGF) for a novel CRC screening strategy. To investigate a potential novel screening tool for CRC, we compared the sensitivity, specificity, positive predictive value, and negative predictive value of the FOBT, serum PlGF, and their combination through an examination of two independent cohorts and validation using the second cohort. All the patients and control group received the colonoscopy and FOBT, the colonoscopy was used as the gold standard for the result. Results Serum PlGF levels were significantly increased in CRC patients (16.8 ± 11.4 pg/mL) compared with controls (12.0 ± 11.2 pg/mL). The predictive model that used the serum PlGF level alone was as effective as the FOBT (AUC: 0.60 vs 0.68, P = 0.891), and it had significantly higher sensitivity than the FOBT (0.81 vs 0.39). In addition, we found serum PlGF level has a good value for predicting CRC patients in those FOBT negative populations. Finally, combining serum PlGF level and the FOBT improved the predictive power and demonstrated satisfactory sensitivity (0.71) and specificity (0.71). This result was confirmed and validated in the second independent cohort. Furthermore, no matter the stages (early/advanced) and the location (distal/proximal) of CRC, the efficacy of serum PlGF and the combined model remained quite stable. Conclusion Serum PlGF level is a potential alternative screening tool for CRC, especially for those who are reluctant to stool‐based screening methods and who were tested as negative FOBT. In addition, combining serum PlGF level and the FOBT could increase the power of CRC screening.
Collapse
Affiliation(s)
- Shu-Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Po-Nien Tsao
- Department of Pediatrics, National Taiwan University Children's Hospital and National Taiwan University, Taipei, Taiwan.,The Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Wang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Been-Ren Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wen-Sy Tsai
- Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jinn-Shiun Chen
- Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Jau-Min Wong
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| |
Collapse
|
24
|
Al-Zeheimi N, Naik A, Bakheit CS, Al Riyami M, Al Ajarrah A, Al Badi S, Al Baimani K, Malik K, Al Habsi Z, Al Moundhri MS, Adham SA. Neoadjuvant Chemotherapy Alters Neuropilin-1, PlGF, and SNAI1 Expression Levels and Predicts Breast Cancer Patients Response. Front Oncol 2019; 9:323. [PMID: 31106153 PMCID: PMC6494932 DOI: 10.3389/fonc.2019.00323] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/10/2019] [Indexed: 12/19/2022] Open
Abstract
Circulating proteins hold a potential benefit as biomarkers for precision medicine. Previously, we showed that systemic levels of neuropilin-1 (NRP-1) and its associated molecules correlated with poor-prognosis breast cancer. To further identify the role of NRP-1 and its interacting molecules in correspondence with patients' response to neoadjuvant chemotherapy (NAC), we conducted a comparative study on blood and tissue samples collected from a cohort of locally advanced breast cancer patients, before and after neoadjuvant chemotherapy (NAC). From a panel of tested proteins and genes, we found that the levels of plasma NRP-1, placenta growth factor (PlGF) and immune cell expression of the transcription factor SNAI1 before and after NAC were significantly different. Paired t-test analysis of 22 locally advanced breast cancer patients showed that plasma NRP-1 levels were increased significantly (p = 0.018) post-NAC in patients with pathological partial response (pPR). Kaplan–Meier analysis indicated that patients who received NAC cycles and their excised tumors remained with high levels of NRP-1 had a lower overall survival compared with patients whose tissue NRP-1 decreased post-NAC (log-rank p = 0.049). In vitro validation of the former result showed an increase in the secreted and cellular NRP-1 levels in resistant MDA-MB-231 cells to the most common NAC regimen Adriyamicin/cyclophosphamide+Paclitaxel (AC+PAC). In addition, NRP-1 knockdown in MDA-MB-231 cells sensitized the cells to AC and more profoundly to PAC treatment and the cells sensitivity was proportional to the expressed levels of NRP-1. Unlike NRP-1, circulating PlGF was significantly increased (p = 0.014) in patients with a pathological complete response (pCR). SNAI1 expression in immune cells showed a significant increase (p = 0.018) in patients with pCR, consistent with its posited protective role. We conclude that increased plasma and tissue NRP-1 post-NAC correlate with pPR and shorter overall survival, respectively. These observations support the need to consider anti-NRP-1 as a potential targeted therapy for breast cancer patients who are identified with high NRP-1 levels. Meanwhile, the increase in both PlGF and SNAI1 in pCR patients potentially suggests their antitumorigenic role in breast cancer that paves the way for further mechanistic investigation to validate their role as potential predictive markers for pCR in breast cancer.
Collapse
Affiliation(s)
- Noura Al-Zeheimi
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Adviti Naik
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Charles Saki Bakheit
- Department of Mathematics and Statistics, Sultan Qaboos University, Muscat, Oman
| | - Marwa Al Riyami
- Department of Pathology, College of Medicine, Sultan Qaboos University, Muscat, Oman
| | - Adil Al Ajarrah
- Department of Surgery, Sultan Qaboos University Hospital, Muscat, Oman
| | - Suaad Al Badi
- Department of Pathology, College of Medicine, Sultan Qaboos University, Muscat, Oman
| | - Khalid Al Baimani
- Medical Oncology Unit, Department of Medicine, College of Medicine, Sultan Qaboos University Hospital, Muscat, Oman
| | - Kamran Malik
- Department of Surgery, Wrexham Maelor Hospital, Wrexham, United Kingdom
| | - Zamzam Al Habsi
- Department of Surgery, Sultan Qaboos University Hospital, Muscat, Oman
| | - Mansour S Al Moundhri
- Medical Oncology Unit, Department of Medicine, College of Medicine, Sultan Qaboos University Hospital, Muscat, Oman
| | - Sirin A Adham
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
25
|
Theile D, Cho WC. Pharmacodynamic monitoring using biomarkers to individualize pharmacotherapy. Biomark Med 2019; 13:393-408. [DOI: 10.2217/bmm-2018-0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Drug doses are often titrated upon their clinical effects (e.g., blood pressure). Unfortunately, for many drugs there is no direct, clinical read-out to estimate dose adequateness. Alternatively, drug dosing is based on the maximum tolerated dose approach or therapeutic drug monitoring. However, the concentration-response curves may be flattened or bell-shaped as suggested for some ‘biologicals’. Together, these aspects raise the question why drug dosing is not individualized by pharmacodynamic monitoring. Evaluating the effects of drugs at their pharmacological target or meaningful biomarkers might indicate nonresponders, objectively quantify the maximum molecular effect and thus restrict overdose and underdosing. This review outlines the theory and biological or technical prerequisites for biomarker-based pharmacodynamic monitoring, and highlights selected examples from different fields of clinical medicine.
Collapse
Affiliation(s)
- Dirk Theile
- Department of Clinical Pharmacology & Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg 69120, Germany
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Kowloon, Hong Kong
| |
Collapse
|
26
|
Herrera-Martínez AD, Hofland LJ, Gálvez Moreno MA, Castaño JP, de Herder WW, Feelders RA. Neuroendocrine neoplasms: current and potential diagnostic, predictive and prognostic markers. Endocr Relat Cancer 2019; 26:R157-R179. [PMID: 30615596 DOI: 10.1530/erc-18-0354] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/03/2019] [Indexed: 12/13/2022]
Abstract
Some biomarkers for functioning and non-functioning neuroendocrine neoplasms (NENs) are currently available. Despite their application in clinical practice, results should be interpreted cautiously. Considering the variable sensitivity and specificity of these parameters, there is an unmet need for novel biomarkers to improve diagnosis and predict patient outcome. Nowadays, several new biomarkers are being evaluated and may become future tools for the management of NENs. These biomarkers include (1) peptides and growth factors; (2) DNA and RNA markers based on genomics analysis, for example, the so-called NET test, which has been developed for analyzing gene transcripts in circulating blood; (3) circulating tumor/endothelial/progenitor cells or cell-free tumor DNA, which represent minimally invasive methods that would provide additional information for monitoring treatment response and (4) improved imaging techniques with novel radiolabeled somatostatin analogs or peptides. Below we summarize some future directions in the development of novel diagnostic and predictive/prognostic biomarkers in NENs. This review is focused on circulating and selected tissue markers.
Collapse
Affiliation(s)
- Aura D Herrera-Martínez
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC); Reina Sofia University Hospital, Córdoba, Spain
| | - Leo J Hofland
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - María A Gálvez Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC); Reina Sofia University Hospital, Córdoba, Spain
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC); Reina Sofia University Hospital, Córdoba, Spain
| | - Wouter W de Herder
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Richard A Feelders
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
27
|
Cuny T, de Herder W, Barlier A, Hofland LJ. Role of the tumor microenvironment in digestive neuroendocrine tumors. Endocr Relat Cancer 2018; 25:R519-R544. [PMID: 30306777 DOI: 10.1530/erc-18-0025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) represent a group of heterogeneous tumors whose incidence increased over the past few years. Around half of patients already present with metastatic disease at the initial diagnosis. Despite extensive efforts, cytotoxic and targeted therapies have provided only limited efficacy for patients with metastatic GEP-NETs, mainly due to the development of a certain state of resistance. One factor contributing to both the failure of systemic therapies and the emergence of an aggressive tumor phenotype may be the tumor microenvironment (TME), comprising dynamic and adaptative assortment of extracellular matrix components and non-neoplastic cells, which surround the tumor niche. Accumulating evidence shows that the TME can simultaneously support both tumor growth and metastasis and contribute to a certain state of resistance to treatment. In this review, we summarize the current knowledge of the TME of GEP-NETs and discuss the current therapeutic agents that target GEP-NETs and those that could be of interest in the (near) future.
Collapse
Affiliation(s)
- Thomas Cuny
- Division Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Marseille, France
- Department of Endocrinology, Assistance Publique - Hôpitaux de Marseille (AP-HM), Hôpital de la Conception, Centre de Référence des Maladies Rares Hypophysaires HYPO, Marseille, France
| | - Wouter de Herder
- Division Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Anne Barlier
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Marseille, France
- Department of Endocrinology, Assistance Publique - Hôpitaux de Marseille (AP-HM), Hôpital de la Conception, Centre de Référence des Maladies Rares Hypophysaires HYPO, Marseille, France
| | - Leo J Hofland
- Division Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
28
|
Serum angiogenesis profile in gestational trophoblastic neoplasm using multiplex immunoassay. Life Sci 2018; 211:25-30. [PMID: 30195618 DOI: 10.1016/j.lfs.2018.08.070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 11/23/2022]
Abstract
AIMS Gestational trophoblastic neoplasms (GTN) exemplify a rare, mostly curable but highly aggressive disease. It is often associated with a rapid formation of distant metastases and most likely with an intense neoangiogenesis processes. The aim of the study was to analyze markers in serum of patients with GTN before chemotherapy compared to healthy pregnant women. MAIN METHODS In this study sixteen protein angiogenesis markers were evaluated in serum of 21 patients with GTN before chemotherapy and compared with healthy pregnant women. Markers were measured using BioPlex Pro Human Cancer Biomarker Panel 1 immunoassay. t-Tests and receiver operating characteristic curves were used for statistical analysis. KEY FINDINGS Receiver operator curve analysis identified six proteins (sTIE-2, osteopontin, sIL-6α, sVEGFR-2, sEGFR, PECAM-1) which had sufficient sensitivity and specificity (AUC > 0,70) to distinguish GTN patients before the treatment from pregnant controls. The levels of three proteins (sTIE-2, osteopontin and sIL-6α) were altered in GTN patients before the treatment as compared to healthy controls (p = 0,0112; p = 0,0442; p = 0,0488, respectively) and thereby may serve as potential disease markers. SIGNIFICANCE Serum concentration of proteins related to angiogenesis changes in the course of GTN and may appear useful in the diagnostic process of this disease.
Collapse
|
29
|
Song Y, Tang C, Yin C. Combination antitumor immunotherapy with VEGF and PIGF siRNA via systemic delivery of multi-functionalized nanoparticles to tumor-associated macrophages and breast cancer cells. Biomaterials 2018; 185:117-132. [PMID: 30241030 DOI: 10.1016/j.biomaterials.2018.09.017] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 09/06/2018] [Accepted: 09/10/2018] [Indexed: 12/14/2022]
Abstract
Given that vascular endothelial growth factor (VEGF) and placental growth factor (PIGF), over-expressed in breast cancer cells and M2-like tumor-associated macrophages (M2-TAMs) within tumor microenvironment (TME), work synergistically and independently in mediating tumor progression and immunosuppression, combinatorial immune-based approaches targeting them are expected to be a potent therapeutic modality for patients. Here, polyethylene glycol (PEG) and mannose doubly modified trimethyl chitosan (PEG = MT) along with citraconic anhydride grafted poly (allylamine hydrochloride) (PC)-based nanoparticles (NPs) (PEG = MT/PC NPs) with dual pH-responsiveness were developed to deliver VEGF siRNA (siVEGF)/PIGF siRNA (siPIGF) to both M2-TAMs and breast cancer cells for antitumor immunotherapy. With prolonged blood circulation and intelligent pH-sensitivity, PEG = MT/PC NPs were highly accumulated in tumor tissues and then internalized in M2-TAMs and breast cancer cells via mannose-mediated active targeting and passive targeting, respectively. With the charge-reversal of PC, PEG = MT/PC NPs presented effective endosomal/lysosomal escape and intracellular siRNA release, resulting in efficient gene silencing. Due to the synergism between siVEGF and siPIGF in anti-proliferation of tumor cells and reversal of the TME from pro-oncogenic to anti-tumoral, PEG = MT/PC/siVEGF/siPIGF NPs (PEG = MT/PC/siV-P NPs) exerted robust suppression of breast tumor growth and lung metastasis. This combination strategy may provide a promising alternative for breast cancer therapy.
Collapse
Affiliation(s)
- Yudong Song
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai, 200438, China
| |
Collapse
|
30
|
Li H, Jin Y, Hu Y, Jiang L, Liu F, Zhang Y, Hao Y, Chen S, Wu X, Liu Y. The PLGF/c-MYC/miR-19a axis promotes metastasis and stemness in gallbladder cancer. Cancer Sci 2018; 109:1532-1544. [PMID: 29575299 PMCID: PMC5980328 DOI: 10.1111/cas.13585] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/14/2018] [Accepted: 03/17/2018] [Indexed: 12/13/2022] Open
Abstract
Gallbladder cancer (GBC) is the most common malignant tumor of the biliary tract system. Epithelial-mesenchymal transition (EMT) plays a vital role in the process of tumor metastasis. Mesenchymal-like cells can serve as a source of cancer stem cells, which can confer the EMT phenotype. Placental growth factor (PLGF) belongs to the vascular endothelial growth factor family and plays a vital role in cancer. However, the underlying molecular mechanisms about the influence of PLGF on EMT in GBC remain unknown. Here we show that PLGF expression levels were higher in GBC tissues than in normal adjacent tissues and were associated with poor prognosis in GBC patients. Exogenous PLGF enhanced the migration, invasion, and tumorsphere formation of GBC cells. Conversely, knockdown of PLGF decreased the aggressive phenotype of GBC cells. Mechanistically, exogenous PLGF upregulated microRNA-19a (miR-19a) expression through the activation of c-MYC. Moreover, Spearman's correlation analysis showed a positive pairwise correlation among PLGF, c-MYC, and miR-19a expression in GBC tissues. Taken together, these results suggest that PLGF promotes EMT and tumorsphere formation through inducing miR-19a expression by upregulating c-MYC. Thus, PLGF could be a promising molecular therapeutic target for GBC.
Collapse
Affiliation(s)
- Huaifeng Li
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yunpeng Jin
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yunping Hu
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lin Jiang
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fatao Liu
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yijian Zhang
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yajuan Hao
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shili Chen
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiangsong Wu
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yingbin Liu
- Department of General Surgery and Laboratory of General SurgeryXinhua HospitalAffiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Biliary Tract DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
31
|
Santalahti K, Havulinna A, Maksimow M, Zeller T, Blankenberg S, Vehtari A, Joensuu H, Jalkanen S, Salomaa V, Salmi M. Plasma levels of hepatocyte growth factor and placental growth factor predict mortality in a general population: a prospective cohort study. J Intern Med 2017; 282:340-352. [PMID: 28682476 DOI: 10.1111/joim.12648] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Circulating levels of growth factors involved in leucocyte production and angiogenesis could be indicative of underlying aberrations of tissue homeostasis and therefore be utilized as predictors of risk for all-cause cardiovascular disease (CVD) or cancer mortality. METHODS Baseline plasma levels of a range of growth factors were measured in two cohorts of the population-based FINRISK study (1997 Discovery cohort, N = 8444, aged 25-74; 2002 Replication cohort, N = 2951, aged 51-74 years) using a multiplexed bead array methodology and ELISA. Participants were followed up by linking them to registry data. RESULTS In the Discovery cohort (653 deaths; 216 CVD-related, 231 cancer-related), fully adjusted Cox proportional hazard regression models showed that increased plasma hepatocyte growth factor (HGF) and placental growth factor (PlGF) were associated with higher risk of 10-year mortality (HR, 1.29 [95% confidence interval (CI), 1.18-1.41] and HR, 1.23 [95% CI, 1.14-1.32], respectively). In the Replication cohort (259 deaths; 83 CVD-related, 90 cancer-related), baseline HGF levels also predicted all-cause mortality (HR, 1.2 [95% CI, 1.08-1.32]; PlGF data not available). By including HGF levels in a CVD mortality model, 9% of all CVD deaths were correctly reclassified in the Discovery cohort (categorical net reclassification improvement [NRI] for events, P = 4.0 × 10-4 ). Moreover, adding HGF to all-cause and CVD mortality models resulted in an overall clinical NRI of 0.10-0.18 in the Discovery cohort and meta-analyses (P < 0.05 for all tests). CONCLUSION Blood levels of HGF and PlGF may serve as new biomarkers for predicting increased risk of death in the general population.
Collapse
Affiliation(s)
- K Santalahti
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - A Havulinna
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - M Maksimow
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - T Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK eV), Partner Site Hamburg/Lübeck/Kiel/Hamburg, Germany
| | - S Blankenberg
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK eV), Partner Site Hamburg/Lübeck/Kiel/Hamburg, Germany
| | - A Vehtari
- Department of Biomedical Engineering and Computational Science, Aalto University, Espoo, Finland
| | - H Joensuu
- Department of Oncology, Helsinki University Hospital, Helsinki, Finland.,University of Helsinki, Helsinki, Finland
| | - S Jalkanen
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - V Salomaa
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - M Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| |
Collapse
|
32
|
Lassig AAD, Joseph AM, Lindgren BR, Yueh B. Association of Oral Cavity and Oropharyngeal Cancer Biomarkers in Surgical Drain Fluid With Patient Outcomes. JAMA Otolaryngol Head Neck Surg 2017; 143:670-678. [PMID: 28418447 DOI: 10.1001/jamaoto.2016.3595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Importance Survival rates for head and neck cancer have been relatively stable for several decades. Individualized prognostic indicators are needed to identify patients at risk for poorer outcomes. Objective To determine whether biomarker levels in surgical drain fluid of patients with head and neck cancer are associated with poor cancer outcomes. Design, Setting, and Participants This prospective cohort study enrolled patients with squamous cell carcinoma (SCC) of the oral cavity and oropharynx who required surgical treatment from April 1, 2011, to February 1, 2016, at a tertiary or academic care center. Twenty patients, including 14 with stage IV disease, had complete specimen collection. Differences in cytokine and MMP levels by disease outcomes were evaluated. Interventions Patients underwent surgical treatment with drain placement as dictated by the standard of care. Drain fluid samples were collected every 8 hours postoperatively until drains were removed because of clinical criteria. Levels of cytokines and matrix metalloproteinases (MMPs) were measured using electrochemiluminescent, patterned array, multiplex technology. Main Outcomes and Measures The primary clinical outcome measures were survival outcome and recurrence. The biomarkers measured included the cytokines basic fibroblastic growth factor, vascular endothelial growth factor isoform A, soluble fms-like tyrosine kinase-1 (sFlt-1), and placental growth factor (PIGF) and MMP-1, MMP-3, and MMP-9. Other clinical and pathologic cancer characteristics were recorded. Results In this cohort of 20 patients with SCC (15 men and 5 women; mean [SD] age, 63.5 [9.9] years), a significant association with recurrence was found for levels of MMP-1 (relative difference between groups, 2.78; 95% CI, 1.23-6.29), MMP-3 (relative difference between groups, 5.29; 95% CI, 2.14-13.05), and sFlt-1 (relative difference between groups, 3.75; 95% CI, 1.84-7.65). No biomarkers were associated with disease outcome. Vascular endothelial growth factor isoform A was associated with nodal metastasis (relative difference between groups, 1.98; 95% CI, 1.12-3.51), and basic fibroblastic growth factor was associated with lymphovascular invasion (relative difference between groups, 1.74; 95% CI, 1.02-2.97). Conclusions and Relevance In this pilot sample of patients with SCC of the oral cavity and oropharynx, MMP-1, MMP-3, and sFlt-1 levels in wound fluid were associated with poor clinical cancer outcomes in the form of recurrence. This finding is consistent with the literature of tumor microenvironment in saliva, serum, and tumor tissue biomarkers. To our knowledge, this report is the first of such findings in surgical drain fluid, an easily accessible means of cytokine measurement. Measurement of these biomarkers in surgical fluid potentially represents a novel means of assessing cancer prognosis in this population.
Collapse
Affiliation(s)
- Amy Anne D Lassig
- Division of Head and Neck Surgery, Department of Otolaryngology-Head and Neck Surgery, University of Minnesota, Minneapolis2Department of Otolaryngology-Head and Neck Surgery, Hennepin County Medical Center, Minneapolis, Minnesota
| | - Anne M Joseph
- Division of General Internal Medicine, Department of Medicine, University of Minnesota, Minneapolis
| | - Bruce R Lindgren
- Division of Biostatistics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Bevan Yueh
- Division of Head and Neck Surgery, Department of Otolaryngology-Head and Neck Surgery, University of Minnesota, Minneapolis
| |
Collapse
|
33
|
Fei Y, Guo P, Wang F, Li H, Lei Y, Li W, Xun X, Lu F. Identification of miRNA-mRNA crosstalk in laryngeal squamous cell carcinoma. Mol Med Rep 2017; 16:4179-4186. [DOI: 10.3892/mmr.2017.7123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 06/06/2017] [Indexed: 11/06/2022] Open
|
34
|
Neuropilin-1 Associated Molecules in the Blood Distinguish Poor Prognosis Breast Cancer: A Cross-Sectional Study. Sci Rep 2017; 7:3301. [PMID: 28607365 PMCID: PMC5468252 DOI: 10.1038/s41598-017-03280-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/28/2017] [Indexed: 12/12/2022] Open
Abstract
Circulating plasma and peripheral blood mononuclear (PBMCs) cells provide an informative snapshot of the systemic physiological state. Moreover, they provide a non-invasively accessible compartment to identify biomarkers for personalized medicine in advanced breast cancer. The role of Neuropilin-1 (NRP-1) and its interacting molecules in breast tumor tissue was correlated with cancer progression; however, the clinical impact of their systemic levels was not extensively evaluated. In this cross-sectional study, we found that circulating and tumor tissue expression of NRP-1 and circulating placental growth factor (PlGF) increase in advanced nodal and metastatic breast cancer compared with locally advanced disease. Tumor tissue expression of NRP-1 and PlGF is also upregulated in triple negative breast cancer (TNBC) compared to other subtypes. Conversely, in PBMCs, NRP-1 and its interacting molecules SEMA4A and SNAI1 are significantly downregulated in breast cancer patients compared to healthy controls, indicating a protective role. Moreover, we report differential PBMC expression profiles that correlate inversely with disease stage (SEMA4A, SNAI1, PLXNA1 and VEGFR3) and can differentiate between the TNBC and non-TNBC tumor subtypes (VEGFR3 and PLXNA1). This work supports the importance of NRP-1-associated molecules in circulation to characterize poor prognosis breast cancer and emphasizes on their role as favorable drug targets.
Collapse
|
35
|
Falk RT, Staff AC, Bradwin G, Karumanchi SA, Troisi R. A prospective study of angiogenic markers and postmenopausal breast cancer risk in the prostate, lung, colorectal, and ovarian cancer screening trial. Cancer Causes Control 2016; 27:1009-17. [PMID: 27357932 PMCID: PMC4958123 DOI: 10.1007/s10552-016-0779-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/11/2016] [Indexed: 11/12/2022]
Abstract
PURPOSE Pro-angiogenic factors are positively associated with breast tumor staging and poorer prognosis, but their role in the etiology of breast cancer has not been assessed. METHODS We measured serum levels of the pro-angiogenic vascular endothelial growth factor A (VEGF), and placental growth factor (PlGF) and anti-angiogenic soluble fms-like tyrosine kinase-1 (sFlt-1) in 352 incident breast cancer cases [mean age at diagnosis 67 (range 55-83)] and 352 non-cases in the prostate, lung, colorectal, and ovarian screening trial (women enrolled 1993-2001, followed through 2005) matched on age and date of enrollment. Cases were followed on average 4.2 years from blood draw to diagnosis, range 3.9-12.8 years; 53 % were estrogen receptor positive/progesterone receptor positive (ER+/PR+), and 13 % were ER-/PR-. Quartile-specific hazard ratios (HR) and 95 % confidence intervals (CI) were estimated using weighted Cox proportional hazards regression models adjusted for known breast cancer risk factors. An ordinal variable for the angiogenic markers was used to test for trend in the HR. RESULTS Comparing the highest to lowest quartile, multivariable HR were 0.90 for VEGF (95 % CI 0.33-2.43, p trend = 0.88), 1.38 for sFlt-1 (95 % CI 0.63-3.04, p trend = 0.63), and 0.62 for PlGF (95 % CI 0.19-2.00, p trend = 0.73). Risk patterns were not altered when all angiogenic markers were included in the model simultaneously, or by restricting analyses to invasive breast cancers, to cases diagnosed two or more years after blood collection or to ER+ tumors. CONCLUSIONS There was no evidence of an increased breast cancer risk associated with circulating levels of pro-angiogenic markers VEGF and PlGF or a reduced risk with circulating levels of anti-angiogenic marker sFlt-1.
Collapse
Affiliation(s)
- Roni T. Falk
- />Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, 9609 Medical Center Drive, Bethesda, MD 20852 USA
| | - Annetine Cathrine Staff
- />Women and Children’s Division, Department of Gynecology and Obstetrics, Oslo University Hospital, Ullevål, 0424 Oslo, Norway
| | - Gary Bradwin
- />Clinical and Epidemiologic Research Laboratory, Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA USA
| | - S. Ananth Karumanchi
- />Deparments of Medicine, Obstetrics and Gynecology, Center for Vascular Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Rebecca Troisi
- />Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20852 USA
| |
Collapse
|
36
|
Zucchini C, De Sanctis P, Facchini C, Di Donato N, Montanari G, Bertoldo V, Farina A, Curti A, Seracchioli R. Performance of Circulating Placental Growth Factor as A Screening Marker for Diagnosis of Ovarian Endometriosis: A Pilot Study. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2015; 9:483-9. [PMID: 26985335 PMCID: PMC4793168 DOI: 10.22074/ijfs.2015.4606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/19/2014] [Indexed: 12/02/2022]
Abstract
Background The aim of this study is to compare the circulating placental growth
factor (PlGF) concentration in women with and without endometrioma to verify the
performance of this marker to diagnose the disease. Materials and Methods In this case-control study, thirteen women with histological diagnosis of ovarian endometriosis were compared with women without endometriosis disease.
PlGF plasma levels of endometriotic patients and controls were investigated using a fluorescence immunoassay technique. Results PlGF showed a direct correlation with body mass index (BMI) only in the
control group (P=0.013). After adjustment for BMI values, PlGF median value in
endometriosis group (14.7 pg/mL) resulted higher than in control group (13.8 pg/
mL, P=0.004). Conclusion PlGF is a promising peripheral blood marker that can discriminate between
patients with and without ovarian endometriosis.
Collapse
Affiliation(s)
- Cinzia Zucchini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Paola De Sanctis
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Chiara Facchini
- Pelvic Endoscopy and Minimally Invasive Gynaecologic Surgery, St. Orsola Malpighi University Hospital, Bologna, Italy
| | - Nadine Di Donato
- Pelvic Endoscopy and Minimally Invasive Gynaecologic Surgery, St. Orsola Malpighi University Hospital, Bologna, Italy
| | - Giulia Montanari
- Pelvic Endoscopy and Minimally Invasive Gynaecologic Surgery, St. Orsola Malpighi University Hospital, Bologna, Italy
| | - Valentina Bertoldo
- Pelvic Endoscopy and Minimally Invasive Gynaecologic Surgery, St. Orsola Malpighi University Hospital, Bologna, Italy
| | - Antonio Farina
- Department of Medicine and Surgery DIMEC, Division of Prenatal Medicine, St. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Alessandra Curti
- Department of Medicine and Surgery DIMEC, Division of Prenatal Medicine, St. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Renato Seracchioli
- Pelvic Endoscopy and Minimally Invasive Gynaecologic Surgery, St. Orsola Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
37
|
Lan X, Sun W, Zhang P, He L, Dong W, Wang Z, Liu S, Zhang H. Downregulation of long noncoding RNA NONHSAT037832 in papillary thyroid carcinoma and its clinical significance. Tumour Biol 2015; 37:6117-23. [PMID: 26611646 DOI: 10.1007/s13277-015-4461-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNA (lncRNA) is a kind of RNA that is longer than 200 nucleotides with limited or no protein-coding potential. Studies have proved that lncRNAs play important regulatory roles in gene expression and contribute to oncogenesis and cancer metastasis. However, the expression level of lncRNAs and their clinicopathologic significance in papillary thyroid carcinoma (PTC) have not been well studied. In this study, we investigated the expression level of a novel lncRNA NONHSAT037832 in PTC and paired noncancerous thyroid tissues as well as some cell lines by quantitative real-time polymerase chain reaction. The association between the expression level of NONHSAT037832 and clinicopathologic characteristics of patients with PTC was further analyzed. Three receiver operating characteristic curves (ROCs) were established to evaluate the diagnostic value of NONHSAT037832. The results suggested that the expression level of NONHSAT037832 was significantly decreased in PTC compared with paired noncancerous tissues (P < 0.01). And, NONHSAT037832 was also significantly downregulated in two PTC cell lines (K1 and IHH-4) compared to normal thyroid follicular epithelial cell line Nthy-ori 3-1 (P < 0.01). Downregulated NONHSAT037832 was significantly associated with lymph node metastasis (P = 0.015) and tumor size (P = 0.032). The ROCs revealed that NONHSAT037832 had a high diagnostic value for differentiating between PTC and noncancerous diseases as well as identifying PTC with lymph node metastasis and larger tumors (≥3 cm). The area under curve was up to 0.897 (95%CI = 0.852-0.942, P = 0.000), 0.641 (95%CI = 0.519-0.762, P = 0.033), and 0.702 (95%CI = 0.567-0.827, P = 0.008), respectively. This study indicated that NONHSAT037832 might serve as a potential biomarker of PTC.
Collapse
Affiliation(s)
- Xiabin Lan
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Wei Sun
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Ping Zhang
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Liang He
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Wenwu Dong
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Zhihong Wang
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Siming Liu
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| |
Collapse
|
38
|
Engelmann JC, Amann T, Ott-Rötzer B, Nützel M, Reinders Y, Reinders J, Thasler WE, Kristl T, Teufel A, Huber CG, Oefner PJ, Spang R, Hellerbrand C. Causal Modeling of Cancer-Stromal Communication Identifies PAPPA as a Novel Stroma-Secreted Factor Activating NFκB Signaling in Hepatocellular Carcinoma. PLoS Comput Biol 2015; 11:e1004293. [PMID: 26020769 PMCID: PMC4447342 DOI: 10.1371/journal.pcbi.1004293] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 04/17/2015] [Indexed: 01/26/2023] Open
Abstract
Inter-cellular communication with stromal cells is vital for cancer cells. Molecules involved in the communication are potential drug targets. To identify them systematically, we applied a systems level analysis that combined reverse network engineering with causal effect estimation. Using only observational transcriptome profiles we searched for paracrine factors sending messages from activated hepatic stellate cells (HSC) to hepatocellular carcinoma (HCC) cells. We condensed these messages to predict ten proteins that, acting in concert, cause the majority of the gene expression changes observed in HCC cells. Among the 10 paracrine factors were both known and unknown cancer promoting stromal factors, the former including Placental Growth Factor (PGF) and Periostin (POSTN), while Pregnancy-Associated Plasma Protein A (PAPPA) was among the latter. Further support for the predicted effect of PAPPA on HCC cells came from both in vitro studies that showed PAPPA to contribute to the activation of NFκB signaling, and clinical data, which linked higher expression levels of PAPPA to advanced stage HCC. In summary, this study demonstrates the potential of causal modeling in combination with a condensation step borrowed from gene set analysis [Model-based Gene Set Analysis (MGSA)] in the identification of stromal signaling molecules influencing the cancer phenotype. All living cells rely on communication with other cells to ensure their function and survival. Molecular signals are sent among cells of the same cell type and from cells of one cell type to another. In cancer, not only the cancer cells themselves are responsible for the malignancy, but also stromal (non-cancerous) cells and the molecular signals they send to cancer cells are important factors that determine the severity and outcome of the disease. Therefore, the identification of stromal signals and their influence on cancer cells is important for the development of novel treatment strategies. With a computational systems biology model of stroma-cancer cell communication, we have compiled a set of ten proteins secreted by stromal cells that shape the cancer phenotype. Most importantly, our causal analysis uncovered Pregnancy-Associated Plasma Protein A (PAPPA) as a novel paracrine inducer of the pro-tumorigenic NFκB signaling pathway. In liver cancer patients, higher levels of PAPPA protein indicate a more progressed tumor stage, confirming its clinical relevance.
Collapse
Affiliation(s)
- Julia C. Engelmann
- Department of Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
- * E-mail: (JCE); (RS); (CH)
| | - Thomas Amann
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Birgitta Ott-Rötzer
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Margit Nützel
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Yvonne Reinders
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Jörg Reinders
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Wolfgang E. Thasler
- Biobank under the authority of Human Tissue and Cell Research (HTCR) and Center for Liver Cell Research, Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Theresa Kristl
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Salzburg, Austria
| | - Andreas Teufel
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Christian G. Huber
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Salzburg, Austria
| | - Peter J. Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Rainer Spang
- Department of Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
- * E-mail: (JCE); (RS); (CH)
| | - Claus Hellerbrand
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
- * E-mail: (JCE); (RS); (CH)
| |
Collapse
|
39
|
Tebbe C, Chhina J, Dar SA, Sarigiannis K, Giri S, Munkarah AR, Rattan R. Metformin limits the adipocyte tumor-promoting effect on ovarian cancer. Oncotarget 2015; 5:4746-64. [PMID: 24970804 PMCID: PMC4148096 DOI: 10.18632/oncotarget.2012] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Omental adipocytes promote ovarian cancer by secretion of adipokines, cytokines and growth factors, and acting as fuel depots. We investigated if metformin modulates the ovarian cancer promoting effects of adipocytes. Effect of conditioned media obtained from differentiated mouse 3T3L1 preadipoctes on the proliferation and migration of a mouse ovarian surface epithelium cancer cell line (ID8) was estimated. Conditioned media from differentiated adipocytes increased the proliferation and migration of ID8 cells, which was attenuated by metformin. Metformin inhibited adipogenesis by inhibition of key adipogenesis regulating transcription factors (CEBPα, CEBPß, and SREBP1), and induced AMPK. A targeted Cancer Pathway Finder RT-PCR (real-time polymerase chain reaction) based gene array revealed 20 up-regulated and 2 down-regulated genes in ID8 cells exposed to adipocyte conditioned media, which were altered by metformin. Adipocyte conditioned media also induced bio-energetic changes in the ID8 cells by pushing them into a highly metabolically active state; these effects were reversed by metformin. Collectively, metformin treatment inhibited the adipocyte mediated ovarian cancer cell proliferation, migration, expression of cancer associated genes and bio-energetic changes. Suggesting, that metformin could be a therapeutic option for ovarian cancer at an early stage, as it not only targets ovarian cancer, but also modulates the environmental milieu.
Collapse
Affiliation(s)
- Calvin Tebbe
- Division of Gynecology Oncology, Department of Women's Health, Obstetrics and Gynecology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Jasdeep Chhina
- Division of Gynecology Oncology, Department of Women's Health, Obstetrics and Gynecology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Sajad A Dar
- Division of Gynecology Oncology, Department of Women's Health, Obstetrics and Gynecology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Kalli Sarigiannis
- Division of Gynecology Oncology, Department of Women's Health, Obstetrics and Gynecology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Adnan R Munkarah
- Division of Gynecology Oncology, Department of Women's Health, Obstetrics and Gynecology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women's Health, Obstetrics and Gynecology, Henry Ford Hospital, Detroit, MI 48202, USA
| |
Collapse
|
40
|
Preeclampsia and subsequent breast cancer risk. Cancer Causes Control 2015; 26:955-6. [DOI: 10.1007/s10552-015-0577-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 04/06/2015] [Indexed: 10/23/2022]
|
41
|
Yajima I, Kumasaka MY, Ohnuma S, Ohgami N, Naito H, Shekhar HU, Omata Y, Kato M. Arsenite-mediated promotion of anchorage-independent growth of HaCaT cells through placental growth factor. J Invest Dermatol 2015; 135:1147-1156. [PMID: 25493652 DOI: 10.1038/jid.2014.514] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 11/09/2014] [Accepted: 11/25/2014] [Indexed: 12/19/2022]
Abstract
Various cancers including skin cancer are increasing in 45 million people exposed to arsenic above the World Health Organization's guideline value of 10 μg l(-1). However, there is limited information on key molecules regulating arsenic-mediated carcinogenesis. Our fieldwork in Bangladesh demonstrated that levels of placental growth factor (PlGF) in urine samples from residents of cancer-prone areas with arsenic-polluted drinking water were higher than those in urine samples from residents of an area that was not polluted with arsenic. Our experimental study in human nontumorigenic HaCaT skin keratinocytes showed that arsenite promoted anchorage-independent growth with increased expression and secretion of PlGF, a ligand of vascular endothelial growth factor receptor1 (VEGFR1), and increased VEGFR1/mitogen-activated protein kinase/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) activities. The arsenite-mediated promotion of anchorage-independent growth was strongly inhibited by PlGF depletion with decreased activities of the PlGF/VEGFR1/MEK/ERK pathway. Moreover, arsenite proteasome-dependently degrades metal-regulatory transcription factor-1 (MTF-1) protein, resulting in a decreased amount of MTF-1 protein binding to the PlGF promoter. MTF-1 negatively controlled PlGF transcription in HaCaT cells, resulting in increased PlGF transcription. These results suggest that arsenite-mediated MTF-1 degradation enhances the activity of PlGF/VEGFR1/MEK/ERK signaling, resulting in promotion of the malignant transformation of keratinocytes. Thus, this study proposed a molecular mechanism for arsenite-mediated development of skin cancer.
Collapse
Affiliation(s)
- Ichiro Yajima
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan; Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan; Voluntary Body for International Health Care in Universities, Nagoya, Japan
| | - Mayuko Y Kumasaka
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan; Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan; Voluntary Body for International Health Care in Universities, Nagoya, Japan
| | - Shoko Ohnuma
- Voluntary Body for International Health Care in Universities, Nagoya, Japan
| | - Nobutaka Ohgami
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan; Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan; Voluntary Body for International Health Care in Universities, Nagoya, Japan
| | - Hisao Naito
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hossain U Shekhar
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka-1000, Bangladesh
| | - Yasuhiro Omata
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan; Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan; Voluntary Body for International Health Care in Universities, Nagoya, Japan; Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka-1000, Bangladesh.
| |
Collapse
|
42
|
Rasmussen LG, Lykke JA, Staff AC. Angiogenic biomarkers in pregnancy: defining maternal and fetal health. Acta Obstet Gynecol Scand 2015; 94:820-32. [DOI: 10.1111/aogs.12629] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 03/04/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Lene G. Rasmussen
- Department of Obstetrics and Gynecology; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; University of Copenhagen; Copenhagen Denmark
| | - Jacob A. Lykke
- Faculty of Medicine; University of Copenhagen; Copenhagen Denmark
- Department of Obstetrics and Gynecology; Hvidovre University Hospital; Copenhagen Denmark
- Departement of Obstetrics; Rigshospitalet Copenhagen University Hospital; Copenhagen Denmark
| | - Anne C. Staff
- Department of Obstetrics and Gynecology; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; University of Oslo; Oslo Norway
| |
Collapse
|
43
|
Joosten SC, Hamming L, Soetekouw PM, Aarts MJ, Veeck J, van Engeland M, Tjan-Heijnen VC. Resistance to sunitinib in renal cell carcinoma: From molecular mechanisms to predictive markers and future perspectives. Biochim Biophys Acta Rev Cancer 2014; 1855:1-16. [PMID: 25446042 DOI: 10.1016/j.bbcan.2014.11.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/15/2022]
Abstract
The introduction of agents that inhibit tumor angiogenesis by targeting vascular endothelial growth factor (VEGF) signaling has made a significant impact on the survival of patients with metastasized renal cell carcinoma (RCC). Sunitinib, a tyrosine kinase inhibitor of the VEGF receptor, has become the mainstay of treatment for these patients. Although treatment with sunitinib substantially improved patient outcome, the initial success is overshadowed by the occurrence of resistance. The mechanisms of resistance are poorly understood. Insight into the molecular mechanisms of resistance will help to better understand the biology of RCC and can ultimately aid the development of more effective therapies for patients with this infaust disease. In this review we comprehensively discuss molecular mechanisms of resistance to sunitinib and the involved biological processes, summarize potential biomarkers that predict response and resistance to treatment with sunitinib, and elaborate on future perspectives in the treatment of metastasized RCC.
Collapse
Affiliation(s)
- S C Joosten
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - L Hamming
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - P M Soetekouw
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - M J Aarts
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - J Veeck
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands; Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - M van Engeland
- Dept. of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - V C Tjan-Heijnen
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| |
Collapse
|
44
|
Lack of evidence for PlGF mediating the tumor resistance after anti-angiogenic therapy in malignant gliomas. J Neurooncol 2014; 121:269-78. [DOI: 10.1007/s11060-014-1647-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 10/26/2014] [Indexed: 10/25/2022]
|
45
|
|
46
|
The role of angiogenic factors in endometrial cancer. MENOPAUSE REVIEW 2014; 13:122-6. [PMID: 26327841 PMCID: PMC4520350 DOI: 10.5114/pm.2014.42714] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/04/2014] [Accepted: 02/17/2014] [Indexed: 01/09/2023]
Abstract
Endometrial cancer is the most common malignancy within the female reproductive system (37.7%). The incidence increases with age. Frequently this type of cancer is diagnosed in peri- and post-menopausal women. 60-70% of cancers occur in women over 60 years of age, and less than 5% in women below 40 years of age. Angiogenesis is a process of formation of new microvessels from existing capillaries. There are four different mechanisms of new vessel growth: sprouting, intussusception, vessel elongation and incorporation of endothelial progenitor cells into new microvessels. Angiogenesis plays important roles in growth of endometrial cancers. This process is controlled by many angiogenic factors, for example vascular endothelial growth factor (VEGF). VEGF is the most powerful and most specific endothelial cell growth factor. It plays a crucial role in the initiation of physiological and pathological angiogenesis, lymphangiogenesis, and vasculogenesis. The VEGF family consists of VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, VEGF-F and PLGF (placental growth factor). The effects of VEGF are mediated through binding to the two specific and homologous receptors VEGFR-1 (FLT-1) and VEGFR-2 (KDR). Placental growth factor (PLGF) belongs to the VEGF family and it is also a very important growth factor. So far four isoforms of PLGF have been identified: PLGF-1 (PLGF131), PLGF-2 (PLGF152), PLGF-3 (PLGF203) and PLGF-4 (PLGF224).
Collapse
|
47
|
d'Audigier C, Gautier B, Yon A, Alili JM, Guérin CL, Evrard SM, Godier A, Haviari S, Reille-Serroussi M, Huguenot F, Dizier B, Inguimbert N, Borgel D, Bièche I, Boisson-Vidal C, Roncal C, Carmeliet P, Vidal M, Gaussem P, Smadja DM. Targeting VEGFR1 on endothelial progenitors modulates their differentiation potential. Angiogenesis 2014; 17:603-16. [PMID: 24419917 DOI: 10.1007/s10456-013-9413-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 12/26/2013] [Indexed: 01/16/2023]
Abstract
OBJECTIVES We studied whether plasma levels of angiogenic factors VEGF and placental growth factor (PlGF) in coronary artery disease patients or undergoing cardiac surgery are modified, and whether those factors modulate endothelial progenitor's angiogenic potential. METHODS AND RESULTS A total of 143 patients' plasmas from two different studies were analyzed (30 coronary artery disease patients, 30 patients with stable angina, coupled with 30 age and sex-matched controls; 53 patients underwent cardiac surgery). Among factors screened, only PlGF was found significantly increased in these pathological populations. PlGF-1 and PlGF-2 were then tested on human endothelial-colony-forming cells (ECFCs). We found that PlGF-1 and PlGF-2 induce VEGFR1 phosphorylation and potentiate ECFCs tubulogenesis in vitro. ECFCs VEGFR1 was further inhibited using a specific small interfering RNA (siRNA) and the chemical compound 4321. We then observed that the VEGFR1-siRNA and the compound 4321 decrease ECFCs tubulogenesis potential in vitro. Finally, we tested the compound 4321 in the preclinical Matrigel(®)-plug model with C57Bl/6J mice as well as in the murine hindlimb ischemia model. We found that 4321 inhibited the plug vascularization, attested by the hemoglobin content and the VE-Cadherin expression level and that 4321 inhibited the post-ischemic revascularization. CONCLUSION PlGF plasma levels were found increased in cardiovascular patients. Disrupting PlGF/VEGFR1 pathway could modulate ECFC-induced tubulogenesis, the cell type responsible for newly formed vessels in vivo.
Collapse
|
48
|
Abstract
The function of vascular endothelial growth factor (VEGF) in cancer is not limited to angiogenesis and vascular permeability. VEGF-mediated signalling occurs in tumour cells, and this signalling contributes to key aspects of tumorigenesis, including the function of cancer stem cells and tumour initiation. In addition to VEGF receptor tyrosine kinases, the neuropilins are crucial for mediating the effects of VEGF on tumour cells, primarily because of their ability to regulate the function and the trafficking of growth factor receptors and integrins. This has important implications for our understanding of tumour biology and for the development of more effective therapeutic approaches.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Cancer Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | | |
Collapse
|
49
|
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: 1.7] [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
|
50
|
Lieu CH, Tran H, Jiang ZQ, Mao M, Overman MJ, Lin E, Eng C, Morris J, Ellis L, Heymach JV, Kopetz S. The association of alternate VEGF ligands with resistance to anti-VEGF therapy in metastatic colorectal cancer. PLoS One 2013; 8:e77117. [PMID: 24143206 PMCID: PMC3797099 DOI: 10.1371/journal.pone.0077117] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 08/29/2013] [Indexed: 12/15/2022] Open
Abstract
Background Circulating angiogenic factors are altered in patients with mCRC receiving bevacizumab. Evaluation of alterations in levels of VEGF ligands may provide insights into possible resistance mechanisms. Methods PlGF, VEGF-A, VEGF-C, and VEGF-D were measured from two cohorts of patients. Sequential plasma samples were obtained from a discovery cohort of 42 patients treated with chemotherapy and bevacizumab. A validation cohort included plasma samples from a cross-sectional of 403 patients prior to chemotherapy, or after progression on a regimen with or without bevacizumab. Results In the discovery cohort, VEGF-C was increased prior to progression and at progression (+49% and +95%, respectively, p<0.01), consistent with previously reported elevations in PlGF. Levels of VEGF-D were increased (+23%) at progression (p=0.05). In the validation cohort, samples obtained from patients after progression on a regimen with bevacizumab had higher levels of PlGF and VEGF-D (+43% and +6%, p=0.02, p=0.01, respectively) compared to untreated patients, but failed to validate the increase in VEGF-C seen in the first cohort. Patients who progressed on chemotherapy with bevacizumab had significantly elevated levels of PlGF (+88%) but not VEGF-C and VEGF-D compared to patients treated with chemotherapy alone. Elevations of PlGF and VEGF-D appeared transient and returned to baseline with a half-life of 6 weeks. Conclusions Increases in PlGF and VEGF-D were observed after progression on chemotherapy with bevacizumab. These changes appear to be reversible after discontinuing therapy. These ligands are associated with resistance to bevacizumab-containing chemotherapy in mCRC, but causation remains to be established.
Collapse
Affiliation(s)
- Christopher H. Lieu
- Division of Medical Oncology, University of Colorado, Denver, Colorado, United States of America
- * E-mail:
| | - Hai Tran
- Department of Thoracic/Head & Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Zhi-Qin Jiang
- Department of Gastrointestinal Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Muling Mao
- Department of Gastrointestinal Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - E. Lin
- Department of Gastrointestinal Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Cathy Eng
- Department of Gastrointestinal Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jeffrey Morris
- Department of Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Lee Ellis
- Department of Surgical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - John V. Heymach
- Department of Thoracic/Head & Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|