1201
|
|
Tamborlin L, Sumere BR, de Souza MC, Pestana NF, Aguiar AC, Eberlin MN, Simabuco FM, Rostagno MA, Luchessi AD. Characterization of pomegranate peel extracts obtained using different solvents and their effects on cell cycle and apoptosis in leukemia cells. Food Sci Nutr 2020;8:5483-96. [PMID: 33133551 DOI: 10.1002/fsn3.1831] [Cited by in Crossref: 5] [Cited by in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Pomegranate (Punica granatum L.) has been used in traditional herbal medicine by several cultures as an anti‐inflammatory, antioxidant, antihyperglycemic, and for treatment and prevention of cancer and other diseases. Different parts of the fruit, extraction methods, and solvents can define the chemical profile of the obtained extracts and their biological activities. This study aimed to characterize the chemical profile of peel extracts collected using different extraction solvents and their biological effects on the cell cycle and apoptosis of THP‐1 leukemic cells. Aqueous extract presented the highest content of punicalagins (α pun = 562.26 ± 47.14 mg/L and β pun = 1,251.13 ± 22.21 mg/L) and the lowest content of ellagic acid (66.38 ± 0.21 mg/L), and it promoted a significant impairment of the cell cycle S phase. In fact, punicalagin‐enriched fraction, but not an ellagic acid‐enriched fraction, caused an S phase cell cycle arrest. All extracts increased the number of apoptotic cells. Punicalagin‐enriched fraction increased the percentage of cells with fragmented DNA, which was intensified by ellagic acid combination. The treatment combining punicalagin and ellagic acid fractions increased the apoptotic cleaved PARP1 protein and reduced the activation of the growth‐related mTOR pathway. Thus, these results evidence that solvent choice is critical for the phenolic compounds profile of pomegranate peel extracts and their biological activities.
Collapse
|
1202
|
|
Detti C, Dos Santos Nascimento LB, Brunetti C, Ferrini F, Gori A. Optimization of a Green Ultrasound-Assisted Extraction of Different Polyphenols from Pistacia lentiscus L. Leaves Using a Response Surface Methodology. Plants (Basel) 2020;9:E1482. [PMID: 33153159 DOI: 10.3390/plants9111482] [Cited by in Crossref: 4] [Cited by in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures]
Abstract
Pistacia lentiscus leaves are used in several applications, thanks to their polyphenolic abundance. Thiswork aimed to characterize the polyphenols and to optimize the extraction conditions to shorten the time, decrease the consumption of solvent, and to maximize the yield of different classes of phenolics, which have diverse industrial applications. The variables were optimized by applying a Box–Behnken design. Galloyl and myricetin derivatives were the most abundant compounds, and two new tetragalloyl derivatives were identified by LC-MS/MS. According to the models, the maximum yields of polyphenols (51.3 ± 1.8 mg g−1 DW) and tannins (40.2 ± 1.4 mg g−1 DW) were obtained using 0.12 L g−1 of 40% ethanol at 50 °C. The highest content of flavonoids (10.2 ± 0.8 mg g−1 DW) was obtained using 0.13 L g−1 of 50% ethanol at 50 °C, while 0.1 L g−1 of 30% ethanol at 30 °C resulted in higher amounts of myricitrin (2.6 ± 0.19 mg g−1 DW). Our optimized extraction decreased the ethanolic fraction by 25% and halved the time compared to other methods. These conditions can be applied differently to obtain P. lentiscus extracts richer in tannins or flavonoids, which might be employed for various purposes.
Collapse
|
1203
|
|
Russi S, Maresca V, Zoppoli P, Aieta M, Marino G, Sgambato A, Ignomirelli O, Ciuffi M, Notarangelo T, Basile A, Falco G, Laurino S. Effect of Feijoa Sellowiana Acetonic Extract on Proliferation Inhibition and Apoptosis Induction in Human Gastric Cancer Cells. Appl Sci (Basel) 2020;10:7756. [DOI: 10.3390/app10217756] [Cited by in Crossref: 2] [Cited by in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] Open
Abstract
Gastric cancer (GC) still represents a relevant health problem in the world for both incidence and mortality rates. Many studies underlined that natural products consumption could reduce GC risk, indicating flavonoids as responsible for the beneficial effects through the modulation of several biological processes, such as the inhibition of cancer antioxidant defense and induction of apoptosis. Since Feijoa sellowiana fruit is known to contain high amounts of flavonoids, among which is flavone, we evaluated the antiproliferative and proapoptotic effects of F. sellowiana acetonic extract on GC cell lines through MTS and Annexin-V FITC assays. Among three GC cell lines tested, SNU-1 results being sensitive to both the F. sellowiana acetonic extract and synthetic flavone, which was used as the reference treatment. Moreover, we evaluated their antioxidant effects, assessing the activity of the antioxidant enzymes supeoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) in polymorphonuclear cells. We found a significant increase of their activity after exposure to both F. sellowiana acetonic extract and flavone, supporting the idea that a diet that includes flavone-rich fruits could be of benefit for health. In addition to this antioxidant effect on normal cells, this study indicates, for the first time, an anticancer effect of F. sellowiana acetonic extract in GC cells.
Collapse
|
1204
|
|
Zarrilli G, Businello G, Dieci MV, Paccagnella S, Carraro V, Cappellesso R, Miglietta F, Griguolo G, Guarneri V, Lo Mele M, Fassan M. The Tumor Microenvironment of Primitive and Metastatic Breast Cancer: Implications for Novel Therapeutic Strategies. Int J Mol Sci 2020;21:E8102. [PMID: 33143050 DOI: 10.3390/ijms21218102] [Cited by in Crossref: 11] [Cited by in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Breast cancer evolves thanks to a dense and close interaction with the surrounding tumor microenvironment (TME). Fibroblasts, leukocytes, blood and lymphatic endothelial cells and extracellular matrix are the constituents of this entity, and they synergistically play a pivotal role in all of the stages of breast cancer development, from its onset to its metastatic spread. Moreover, it has been widely demonstrated that variations to the TME can correspond to prognosis variations. Breast cancer not only modulates the transformation of the environment within the mammary gland, but the same process is observed in metastases as well. In this minireview, we describe the features of TME within the primitive breast cancer, throughout its evolution and spread into the main metastatic sites.
Collapse
|
1205
|
|
Jardim DL, Goodman A, de Melo Gagliato D, Kurzrock R. The Challenges of Tumor Mutational Burden as an Immunotherapy Biomarker. Cancer Cell 2021;39:154-73. [PMID: 33125859 DOI: 10.1016/j.ccell.2020.10.001] [Cited by in Crossref: 141] [Cited by in RCA: 174] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
Tumor mutational burden (TMB) reflects cancer mutation quantity. Mutations are processed to neo-antigens and presented by major histocompatibility complex (MHC) proteins to T cells. To evade immune eradication, cancers exploit checkpoints that dampen T cell reactivity. Immune checkpoint inhibitors (ICIs) have transformed cancer treatment by enabling T cell reactivation; however, response biomarkers are required, as most patients do not benefit. Higher TMB results in more neo-antigens, increasing chances for T cell recognition, and clinically correlates with better ICI outcomes. Nevertheless, TMB is an imperfect response biomarker. A composite predictor that also includes critical variables, such as MHC and T cell receptor repertoire, is needed.
Collapse
|
1206
|
|
Ramirez-Valles EG, Rodríguez-Pulido A, Barraza-Salas M, Martínez-Velis I, Meneses-Morales I, Ayala-García VM, Alba-Fierro CA. A Quest for New Cancer Diagnosis, Prognosis and Prediction Biomarkers and Their Use in Biosensors Development. Technol Cancer Res Treat 2020;19:1533033820957033. [PMID: 33107395 DOI: 10.1177/1533033820957033] [Cited by in Crossref: 3] [Cited by in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Traditional techniques for cancer diagnosis, such as nuclear magnetic resonance, ultrasound and tissue analysis, require sophisticated devices and highly trained personnel, which are characterized by elevated operation costs. The use of biomarkers has emerged as an alternative for cancer diagnosis, prognosis and prediction because their measurement in tissues or fluids, such as blood, urine or saliva, is characterized by shorter processing times. However, the biomarkers used currently, and the techniques used for their measurement, including ELISA, western-blot, polymerase chain reaction (PCR) or immunohistochemistry, possess low sensitivity and specificity. Therefore, the search for new proteomic, genomic or immunological biomarkers and the development of new noninvasive, easier and cheaper techniques that meet the sensitivity and specificity criteria for the diagnosis, prognosis and prediction of this disease has become a relevant topic. The purpose of this review is to provide an overview about the search for new cancer biomarkers, including the strategies that must be followed to identify them, as well as presenting the latest advances in the development of biosensors that possess a high potential for cancer diagnosis, prognosis and prediction, mainly focusing on their relevance in lung, prostate and breast cancers.
Collapse
|
1207
|
|
Hossam N, Matboli M, Shehata HH, Aboelhussein MM, Hassan MK, Eissa S. Toll-like receptor immune modulatory role in personalized management of colorectal cancer, review of literature. Expert Review of Precision Medicine and Drug Development 2020;5:455-68. [DOI: 10.1080/23808993.2020.1816136] [Cited by in Crossref: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text]
|
1208
|
|
Saorin A, Di Gregorio E, Miolo G, Steffan A, Corona G. Emerging Role of Metabolomics in Ovarian Cancer Diagnosis. Metabolites 2020;10:E419. [PMID: 33086611 DOI: 10.3390/metabo10100419] [Cited by in Crossref: 13] [Cited by in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] Open
Abstract
Ovarian cancer is considered a silent killer due to the lack of clear symptoms and efficient diagnostic tools that often lead to late diagnoses. Over recent years, the impelling need for proficient biomarkers has led researchers to consider metabolomics, an emerging omics science that deals with analyses of the entire set of small-molecules (≤1.5 kDa) present in biological systems. Metabolomics profiles, as a mirror of tumor–host interactions, have been found to be useful for the analysis and identification of specific cancer phenotypes. Cancer may cause significant metabolic alterations to sustain its growth, and metabolomics may highlight this, making it possible to detect cancer in an early phase of development. In the last decade, metabolomics has been widely applied to identify different metabolic signatures to improve ovarian cancer diagnosis. The aim of this review is to update the current status of the metabolomics research for the discovery of new diagnostic metabolomic biomarkers for ovarian cancer. The most promising metabolic alterations are discussed in view of their potential biological implications, underlying the issues that limit their effective clinical translation into ovarian cancer diagnostic tools.
Collapse
|
1209
|
|
Emens LA, Esteva FJ, Beresford M, Saura C, De Laurentiis M, Kim SB, Im SA, Wang Y, Salgado R, Mani A, Shah J, Lambertini C, Liu H, de Haas SL, Patre M, Loi S. Trastuzumab emtansine plus atezolizumab versus trastuzumab emtansine plus placebo in previously treated, HER2-positive advanced breast cancer (KATE2): a phase 2, multicentre, randomised, double-blind trial. Lancet Oncol 2020;21:1283-95. [PMID: 33002436 DOI: 10.1016/S1470-2045(20)30465-4] [Cited by in Crossref: 84] [Cited by in RCA: 100] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
BACKGROUND HER2-positive metastatic breast cancer is incurable and new treatments are needed. Addition of atezolizumab to trastuzumab emtansine might potentiate anticancer immunity and enhance the HER2-targeted cytotoxic activity of trastuzumab emtansine. We aimed to test this combination in HER2-positive advanced breast cancer that had progressed after previous treatment with trastuzumab and a taxane. METHODS The KATE2 study is a randomised, double-blind, placebo-controlled, phase 2 study at 68 centres from nine countries across Asia, Australia, North America, and western Europe. Eligible patients were adults (aged ≥18 years) with an Eastern Cooperative Oncology Group performance status of 0 or 1 and centrally confirmed, measurable, HER2-positive advanced breast cancer previously treated with trastuzumab and a taxane. Patients were randomly assigned (2:1) either trastuzumab emtansine (3·6 mg/kg of bodyweight) plus atezolizumab (1200 mg) or trastuzumab emtansine plus placebo; all study drugs were administered by intravenous infusion every 3 weeks. Randomisation was done via an interactive voice and web response system using a permuted block scheme (block size of six) and was stratified by PD-L1 status, world region, and liver metastases. Patients, investigators, and study team members were masked to treatment allocation. The primary endpoint was investigator-assessed progression-free survival in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, NCT02924883, and the study has been completed. FINDINGS Between Sept 26, 2016, and Aug 7, 2017, 330 patients were screened for the study, of whom 202 were randomly allocated either atezolizumab (n=133) or placebo (n=69). At the recommendation of the independent data monitoring committee, treatment assignment was unmasked on Dec 11, 2017, due to futility and the numerically higher frequency of adverse events among patients assigned atezolizumab. This date was set as the clinical cutoff for the primary analysis. Median follow-up was 8·5 months (IQR 6·1-11·5) for patients assigned atezolizumab and 8·4 months (5·3-11·1) for those assigned placebo. Median progression-free survival was 8·2 months (95% CI 5·8-10·7) for patients assigned atezolizumab versus 6·8 months (4·0-11·1) for those assigned placebo (stratified hazard ratio 0·82, 95% CI 0·55-1·23; p=0·33). The most common grade 3 or worse adverse events were thrombocytopenia (17 [13%] among 132 patients who received atezolizumab vs three [4%] among 68 who received placebo), increased aspartate aminotransferase (11 [8%] vs two [3%]), anaemia (seven [5%] vs 0), neutropenia (six [5%] vs three [4%]), and increased alanine aminotransferase (six [5%] vs two [3%]). Serious adverse events occurred in 43 (33%) of 132 patients who received atezolizumab and 13 (19%) of 68 patients who received placebo. One patient who received atezolizumab died due to a treatment-related adverse event (haemophagocytic syndrome). INTERPRETATION Addition of atezolizumab to trastuzumab emtansine did not show a clinically meaningful improvement in progression-free survival and was associated with more adverse events. Further study of trastuzumab emtansine plus atezolizumab is warranted in a subpopulation of patients with PD-L1-positive, HER2-positive advanced breast cancer. FUNDING F Hoffman-La Roche.
Collapse
|
1210
|
|
Laugsand EA, Brenne SS, Skorpen F. DNA methylation markers detected in blood, stool, urine, and tissue in colorectal cancer: a systematic review of paired samples. Int J Colorectal Dis 2021;36:239-51. [PMID: 33030559 DOI: 10.1007/s00384-020-03757-x] [Cited by in Crossref: 14] [Cited by in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures]
Abstract
Purpose Methylated cell-free DNA in liquid biopsies are promising non-invasive biomarkers for colorectal cancer (CRC). Optimal markers would have high sensitivity and specificity for early detection of CRC and could be detected in more than one type of material from the patient. We systematically reviewed the literature on DNA methylation markers of colorectal cancer, detected in more than one type of material, regarding their potential as contributors to a panel for screening and follow-up of CRC. Methods The databases MEDLINE, Web of Science, and Embase were systematically searched. Data extraction and review was performed by two authors independently. Agreement between methylation status in tissue and other materials (blood/stool/urine) was analyzed using the McNemar test and Cohen’s kappa. Results From the 51 included studies, we identified seven single markers with sensitivity ≥ 75% and specificity ≥ 90% for CRC. We also identified one promising plasma panel and two stool panels. The correspondence of methylation status was evaluated as very good for four markers, but only marginal for most of the other markers investigated (12 of 21). Conclusion The included studies reported only some of the variables and markers of interest and included few patients. Hence, a meta-analysis was not possible at this point. Larger, prospective studies must be designed to study the discordant detection of markers in tissue and liquid biopsies. When reporting their findings, such studies should use a standardized format. Electronic supplementary material The online version of this article (10.1007/s00384-020-03757-x) contains supplementary material, which is available to authorized users.
Collapse
|
1211
|
|
Erber R, Hartmann A. Understanding PD-L1 Testing in Breast Cancer: A Practical Approach. Breast Care (Basel) 2020;15:481-90. [PMID: 33223991 DOI: 10.1159/000510812] [Cited by in Crossref: 14] [Cited by in RCA: 16] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] Open
Abstract
Background Immune checkpoint inhibitors (ICI) have changed therapy strategies for cancer patients tremendously. Some approved ICI acquire testing of PD-L1 expression on tumor and/or immune cells. However, since PD-L1 testing is a comprehensive issue with various assays, antibody clones, scoring methods, and cut-offs, we aimed to summarize the recommendations and technical and histopathological issues of diagnostic PD-L1 assessment with an emphasis on invasive breast cancer (IBC). Summary Besides other (pre)analytical considerations, selecting the most adequate PD-L1 immunohistochemical assay/antibody clone is important. In-house assay validation, prediagnostic training, and internal and external quality assurance should be implemented. The current most relevant PD-L1 assays and scores will be explained in this review. Moreover, recommendations for PD-L1 testing in IBC are outlined. Key Messages Atezolizumab plus nab-paclitaxel therapy is approved for adult patients with locally advanced or metastatic triple negative breast cancer (mTNBC), if the tumor-associated immune cells express PD-L1. - This PD-L1 immune cell positivity is defined as an immune cell (IC) score, which refers to the area occupied by PD-L1 positive immune cells (lymphocytes, dendritic cells, macrophages, and granulocytes) as a percentage of the whole tumor area. The cut-off is an IC score ≥1%. In the approval study for atezolizumab in mTNBC, IC score was assessed using the Ventana PD-L1 SP142 assay. Other assays or laboratory developed tests may be used depending on country-specific drug approvals. However, harmonization studies have to show whether other PD-L1 tests are reliable and of clinical value to predict the response of breast cancer patients to ICI.
Collapse
|
1212
|
|
Alami Merrouni I, Elachouri M. Anticancer medicinal plants used by Moroccan people: Ethnobotanical, preclinical, phytochemical and clinical evidence. J Ethnopharmacol 2021;266:113435. [PMID: 33022340 DOI: 10.1016/j.jep.2020.113435] [Cited by in Crossref: 21] [Cited by in RCA: 29] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cancer is a major health problem worldwide. Drugs' side effects and high cost of treatment remain the main limitations of conventional therapy. Nowadays, developing new therapeutic strategies is necessary. Therefore, medicinal plants can be used to promote novel, safe, and potent anticancer drugs through their natural compounds. AIM OF THE STUDY This review aims to provide scientific evidence related to the anticancer activities of medicinal plants used by Moroccan people as well as approving their efficiency as an alternative cancer therapy. METHODS An ethnopharmacological review approach was conducted by analyzing Moroccan published ethnobotanical surveys from 1991 to 2019 and consulting peer-reviewed articles worldwide to investigate the pharmacological, phytochemical, and clinical effects related to the anticancer activities. Plants with anticancer proprieties were classified into four groups: (a) plants only cited as anticancer, (b) plants pharmacologically investigated, (c) plants with bioactive compounds tested as anticancer, and (d) plants clinically investigated. RESULTS A total of 103 plant species belonging to 47 botanical families used by Moroccans to treat cancer have been recorded. Aristolochia fontanesii Boiss. & Reut, Marrubium vulgare L., and Allium sativum L. are the most referred species in Morocco. Medicinal plants used for cancer treatment were classified into four groups: 48 species were used traditionally as anticancer (group a), 41 species pharmacologically investigated for their anticancer activities (group b), 32 plants with bioactive compounds tested against cancer (group c), and eight plants were clinically investigated for their anticancer effects (group d). Out of 82 plants' extracts pharmacologically tested (from plants of group b), only 24 ones show a significant cytotoxic effect. A total of seventy-seven compounds are isolated from plants of group (c). However, only six ones were clinically evaluated, and most of them exhibit a beneficial effect on cancerous patients with few side effects. CONCLUSION Medicinal plants can be a promising candidate for alternative cancer therapy. Nevertheless, it is critical to increasing the clinical trials to confirm their beneficial effect on patients with cancer. Overall, this review can serve as a database for further studies.
Collapse
|
1213
|
|
Sadeghalvad M, Mohammadi-Motlagh HR, Rezaei N. Immune microenvironment in different molecular subtypes of ductal breast carcinoma. Breast Cancer Res Treat 2021;185:261-79. [PMID: 33011829 DOI: 10.1007/s10549-020-05954-2] [Cited by in Crossref: 13] [Cited by in RCA: 15] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
PURPOSE Ductal breast carcinoma as a heterogeneous disease has different molecular subtypes associated with clinical prognosis and patients' survival. The role of immune system as a consistent part of the tumor microenvironment (TME) has been documented in progression of ductal breast carcinoma. Here, we aimed to describe the important immune cells and the immune system-associated molecules in Ductal Carcinoma In situ (DCIS) and Invasive Ductal Carcinoma (IDC) with special emphasis on their associations with different molecular subtypes and patients' prognosis. RESULTS The immune cells have a dual role in breast cancer (BC) microenvironment depending on the molecular subtype or tumor grade. These cells with different frequencies are present in the TME of DCIS and IDC. The presence of regulatory cells including Tregs, MDSC, Th2, Th17, M2 macrophages, HLADR- T cells, and Tγδ cells is related to more immunosuppressive microenvironment, especially in ER- and TN subtypes. In contrast, NK cells, CTL, Th, and Tfh cells are associated to the anti-tumor activity. These cells are higher in ER+ BC, although in other subtypes such as TN or HER2+ are associated with a favorable prognosis. CONCLUSION Determining the specific immune response in each subtype could be helpful in estimating the possible behavior of the tumor cells in TME. It is important to realize that different frequencies of immune cells in BC environment likely determine the patients' prognosis and their survival in each subtype. Therefore, elucidation of the distinct immune players in TME would be helpful toward developing targeted therapies in each subtype.
Collapse
|
1214
|
|
Kaur S, Kumar A, Pandit K, Kaur S. Modulation of mutagenicity in Salmonella typhimurium and antioxidant properties and antiproliferative effects of fractions from Cassia fistula L. on human cervical HeLa and breast MCF-7 cancer cells. Environ Sci Pollut Res Int 2021;28:6619-34. [DOI: 10.1007/s11356-020-10771-7] [Cited by in Crossref: 6] [Cited by in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text]
|
1215
|
|
Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, Li X, Cao K, Deng H, He Y, Liao Q, Xiang B, Zhou M, Guo C, Zeng Z, Li G, Xiong W. The role of microenvironment in tumor angiogenesis. J Exp Clin Cancer Res. 2020;39:204. [PMID: 32993787 DOI: 10.1186/s13046-020-01709-5] [Cited by in Crossref: 111] [Cited by in RCA: 119] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures]
Abstract
Tumor angiogenesis is necessary for the continued survival and development of tumor cells, and plays an important role in their growth, invasion, and metastasis. The tumor microenvironment—composed of tumor cells, surrounding cells, and secreted cytokines—provides a conducive environment for the growth and survival of tumors. Different components of the tumor microenvironment can regulate tumor development. In this review, we have discussed the regulatory role of the microenvironment in tumor angiogenesis. High expression of angiogenic factors and inflammatory cytokines in the tumor microenvironment, as well as hypoxia, are presumed to be the reasons for poor therapeutic efficacy of current anti-angiogenic drugs. A combination of anti-angiogenic drugs and antitumor inflammatory drugs or hypoxia inhibitors might improve the therapeutic outcome.
Collapse
|
1216
|
|
Shklovskaya E, Rizos H. Spatial and Temporal Changes in PD-L1 Expression in Cancer: The Role of Genetic Drivers, Tumor Microenvironment and Resistance to Therapy. Int J Mol Sci 2020;21:E7139. [PMID: 32992658 DOI: 10.3390/ijms21197139] [Cited by in Crossref: 18] [Cited by in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Immunotherapies blocking immune inhibitory receptors programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) on T-cells have dramatically improved patient outcomes in a range of advanced cancers. However, the lack of response, and the development of resistance remain major obstacles to long-term improvements in patient outcomes. There is significant interest in the clinical use of biomarkers to improve patient selection, and the expression of PD-1 ligand 1 (PD-L1) is often reported as a potential biomarker of response. However, accumulating evidence suggests that the predictive value of PD-L1 expression in tumor biopsies is relatively low due, in part, to its complex biology. In this review, we discuss the biological consequences of PD-L1 expression by various cell types within the tumor microenvironment, and the complex mechanisms that regulate PD-L1 expression at the genomic, transcriptomic and proteomic levels.
Collapse
|
1217
|
|
El Bairi K, Trapani D, Petrillo A, Le Page C, Zbakh H, Daniele B, Belbaraka R, Curigliano G, Afqir S. Repurposing anticancer drugs for the management of COVID-19. Eur J Cancer 2020;141:40-61. [PMID: 33125946 DOI: 10.1016/j.ejca.2020.09.014] [Cited by in Crossref: 44] [Cited by in RCA: 46] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures]
Abstract
Since its outbreak in the last December, coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 has rapidly spread worldwide at a pandemic proportion and thus is regarded as a global public health emergency. The existing therapeutic options for COVID-19 beyond the intensive supportive care are limited, with an undefined or modest efficacy reported so far. Drug repurposing represents an enthusiastic mechanism to use approved drugs outside the scope of their original indication and accelerate the discovery of new therapeutic options. With the emergence of COVID-19, drug repurposing has been largely applied for early clinical testing. In this review, we discuss some repurposed anticancer drugs for the treatment of COVID-19, which are under investigation in clinical trials or proposed for the clinical testing.
Collapse
|
1218
|
|
Huynh M, Pambid MR, Jayanthan A, Dorr A, Los G, Dunn SE. The dawn of targeted therapies for triple negative breast cancer (TNBC): a snapshot of investigational drugs in phase I and II trials. Expert Opin Investig Drugs 2020;29:1199-208. [DOI: 10.1080/13543784.2020.1818067] [Cited by in Crossref: 9] [Cited by in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text]
|
1219
|
|
Bleker de Oliveira M, Koshkin V, Liu G, Krylov SN. Analytical Challenges in Development of Chemoresistance Predictors for Precision Oncology. Anal Chem 2020;92:12101-10. [PMID: 32790291 DOI: 10.1021/acs.analchem.0c02644] [Cited by in Crossref: 2] [Cited by in RCA: 3] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
Chemoresistance, i.e., tumor insensitivity to chemotherapy, shortens life expectancy of cancer patients. Despite the availability of new treatment options, initial systemic regimens for solid tumors are dominated by a set of standard chemotherapy drugs, and alternative therapies are used only when a patient has demonstrated chemoresistance clinically. Chemoresistance predictors use laboratory parameters measured on tissue samples to predict the patient's response to chemotherapy and help to avoid application of chemotherapy to chemoresistant patients. Despite thousands of publications on putative chemoresistance predictors, there are only about a dozen predictors that are sufficiently accurate for precision oncology. One of the major reasons for inaccuracy of predictors is inaccuracy of analytical methods utilized to measure their laboratory parameters: an inaccurate method leads to an inaccurate predictor. The goal of this study was to identify analytical challenges in chemoresistance-predictor development and suggest ways to overcome them. Here we describe principles of chemoresistance predictor development via correlating a clinical parameter, which manifests disease state, with a laboratory parameter. We further classify predictors based on the nature of laboratory parameters and analyze advantages and limitations of different predictors using the reliability of analytical methods utilized for measuring laboratory parameters as a criterion. Our eventual focus is on predictors with known mechanisms of reactions involved in drug resistance (drug extrusion, drug degradation, and DNA damage repair) and using rate constants of these reactions to establish accurate and robust laboratory parameters. Many aspects and conclusions of our analysis are applicable to all types of disease biomarkers built upon the correlation of clinical and laboratory parameters.
Collapse
|
1220
|
|
Herrington CS, Poulsom R, Coates PJ. Recent Advances in Pathology: the 2020 Annual Review Issue of The Journal of Pathology. J Pathol 2020;250:475-9. [PMID: 32346919 DOI: 10.1002/path.5425] [Cited by in Crossref: 4] [Cited by in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
This year's Annual Review Issue of The Journal of Pathology contains 18 invited reviews on current research areas in pathology. The subject areas reflect the broad range of topics covered by the journal and this year encompass the development and application of software in digital histopathology, implementation of biomarkers in pathology practice; genetics and epigenetics, and stromal influences in disease. The reviews are authored by experts in their field and provide comprehensive updates in the chosen areas, in which there has been considerable recent progress in our understanding of disease. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
|
1221
|
|
Osei E. A review of clinical and emerging biomarkers for breast cancers: towards precision medicine for patients. J Radiother Pract 2022;21:245-58. [DOI: 10.1017/s1460396920000746] [Cited by in Crossref: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
AbstractBackground:Breast cancer is the most commonly diagnosed malignancy among women and accounts for about 25% of all new cancer cases and 13% of all cancer deaths in Canadian women. It is a highly heterogeneous disease, encompassing multiple tumour entities, each characterised by distinct morphology, behaviour and clinical implications. Moreover, different breast tumour subtypes have different risk factors, clinical presentation, histopathological features, outcome and response to systemic therapies. Therefore, any strategies capable of the stratification of breast cancer by clinically relevant subtypes are an important requirement for personalised and targeted treatment. Therefore, in the advancement towards the concept of precision medicine that takes individual patient variability into account, several investigators have focused on the identification of effective clinical breast cancer biomarkers that interrogate key aberrant pathways potentially targetable with molecular targeted or immunological therapies.Methods and materials:This paper reports on a review of 11 current clinical and emerging biomarkers used in screening for early detection and diagnosis, to stratify patients by disease subtype, to identify patients’ risk for metastatic disease and subsequent relapse, to monitor patient response to specific treatment and to provide clinicians the possibility of prospectively identifying groups of patients who will benefit from a particular treatment.Conclusion:The future holds promising for the use of effective clinical breast cancer biomarkers for early detection and personalised patient-specific targeted treatment and increased patient survival. Breast cancer biomarkers can potentially assist in early-staged, non-invasive, sensitive and specific breast cancer detection and screening, provide clinically useful information for identification of patients with a greater likelihood of benefiting from the specific treatment, offer a better understanding of the metastatic process in cancer patients, predict disease and for patients with the established disease can assist define the nature of the disease, monitor the success of treatment and guide the clinical management of the disease.
Collapse
|
1222
|
|
Pang HF, Li XX, Zhao YH, Kang JK, Li JY, Tian W, Wang CM, Hou HX, Li DR. Confirming whether novel rhein derivative 4a induces paraptosis-like cell death by endoplasmic reticulum stress in ovarian cancer cells. Eur J Pharmacol 2020;886:173526. [PMID: 32890460 DOI: 10.1016/j.ejphar.2020.173526] [Cited by in Crossref: 5] [Cited by in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
Ovarian cancer is the leading cause of death among gynecologic cancer patients. Although platinum-based chemotherapy as a frontline treatment for ovarian cancer has been widely used in clinical settings, its clinical efficacy is not satisfactory due to the resistance of ovarian cancer cells to apoptosis. Therefore, it is of great significance to induce non-apoptotic programed cell death patterns, such as paraptosis, in ovarian cancer. In this study, we aimed to explore the potential anticancer mechanisms of novel rhein derivative 4a, which was modified with rhein as a lead compound. The results showed that a wide range of vacuoles from the endoplasmic reticulum and mitochondria appeared in ovarian SKOV3, SKOV3-PM4, and A2780 cells treated with derivative 4a, and the cell death caused by derivative 4a is a type of non-apoptotic and non-autophagic death, which is caused by expansion and damage of the endoplasmic reticulum or mitochondria, showing the characteristics of para-apoptotic death. Furthermore, derivative 4a stimulated the unfolded protein reaction of ovarian cancer cells by upregulating the expression of Bip78 and activating the PERK-eIF2α-ATF4 pathways. Notably, rhein derivative 4a-induced cell death was positively correlated with activation of p38, ERK, and JNK, and negatively correlated with Alix, a known protein that inhibits paraptosis. In addition, derivative 4a treatment also induced G2/M phase arrest in ovarian cancer cells. Taken together, our study reveals that derivative 4a induces paraptosis, and this finding can serve as a basis in developing a new strategy for the treatment of antiapoptotic ovarian cancer.
Collapse
|
1223
|
|
Lu T, Zhang L, Zhu W, Zhang Y, Zhang S, Wu B, Deng N. CRISPR/Cas9-Mediated OC-2 Editing Inhibits the Tumor Growth and Angiogenesis of Ovarian Cancer. Front Oncol 2020;10:1529. [PMID: 32984003 DOI: 10.3389/fonc.2020.01529] [Cited by in Crossref: 3] [Cited by in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Ovarian cancer is the leading cancer-related cause of death in women worldwide. It is of great relevance to understand the mechanism responsible for tumor progression and identify unique oncogenesis markers for a higher chance of preventing this malignant disease. The high-expression OC-2 gene has been shown to be a potential candidate for regulating oncogenesis and angiogenesis in ovarian cancer. Hence, we wished to investigate the impact of OC-2 gene on ovarian cancer aggressiveness. CRISPR/Cas9, a gene editing tool, allows for direct ablation of OC-2 at the genomic level, and we successfully generated OC-2 KO cell lines from SKOV3 and CAOV3 cells. In an apoptosis assay, OC-2 KO induced the apoptosis activation of tumor cells, with the up-regulation of Bax/Caspase-8 and the down-regulation of Bcl-2. Consequently, the proliferation, migration, and invasion of OC-2 KO cell lines were significantly inhibited. Assays of qRT-PCR and Western blotting showed that the expression levels of pro-angiogenic growth factors VEGFA, FGF2, HGF, and HIF-1α and the activation of Akt/ERK pathways were significantly down-regulated at the loss of OC-2. In the xenograft model, OC-2 KO potently suppressed the subcutaneous tumor growth, with the inhibition exceeding 56%. The down-regulation of CD31 and relevant pro-angiogenic growth factors were observed in OC-2 KO tumor tissues. Taken together, OC-2 depletion negatively regulated the ovarian cancer progression possibly by apoptosis activation and angiogenesis inhibition. This work revealed a pivotal regulator of apoptosis and angiogenesis networks in ovarian cancer, and we applied the CRISPR/Cas9 system to the transcription factor pathway for developing a broad-acting anti-tumor gene therapy.
Collapse
|
1224
|
|
Anisiewicz A, Pawlik A, Filip-Psurska B, Wietrzyk J. Differential Impact of Calcitriol and Its Analogs on Tumor Stroma in Young and Aged Ovariectomized Mice Bearing 4T1 Mammary Gland Cancer. Int J Mol Sci 2020;21:E6359. [PMID: 32887237 DOI: 10.3390/ijms21176359] [Cited by in Crossref: 6] [Cited by in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
(1) Background: Vitamin D compounds (VDC) are extensively studied in the field of anticancer properties, including breast cancer. Previously, we showed that calcitriol and its analogs (PRI-2191 and PRI-2205) stimulate metastasis in 4T1 murine mammary gland cancer models in young mice, whereas the reverse effect was observed in aged ovariectomized (OVX) mice; (2) Methods: We determined the phenotype of monocytes/macrophages using FACS and examined the expression of selected genes and proteins by Real-Time PCR and ELISA; (3) Results: Activities of VDC are accompanied by an increase in the percentage of Ly6Clow anti-inflammatory monocytes in the spleen of young and a decrease in aged OVX mice. Treatment of young mice with VDC resulted in an increase of CCL2 plasma and tumor concentration and Arg1 in tumor. In later stage of tumor progression the expression of genes related to metastasis in lung tissue was decreased or increased, in old OVX or young mice, respectively; (4) Conclusions: Pro- or anti-metastatic effects of calcitriol and its analogs in young or aged OVX mice, respectively, can be attributed to the differences in the effects of VDC on the tumor microenvironment, as a consequence of differences in the immunity status of young and aged mice.
Collapse
|
1225
|
|
Ebrahimibasabi E, Ebrahimi A, Momeni M, Amerian MR. Elevated expression of diosgenin-related genes and stimulation of the defense system in Trigonella foenum-graecum (Fenugreek) by cold plasma treatment. SCI HORTIC-AMSTERDAM 2020;271:109494. [DOI: 10.1016/j.scienta.2020.109494] [Cited by in Crossref: 8] [Cited by in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text]
|
1226
|
|
Díez-villanueva A, Sanz-pamplona R, Carreras-torres R, Moratalla-navarro F, Alonso M, Paré-brunet L, Aussó S, Guinó E, Solé X, Cordero D, Salazar R, Berdasco M, Peinado MA, Moreno V. DNA methylation events in transcription factors and gene expression changes in colon cancer. Epigenomics 2020;12:1593-610. [DOI: 10.2217/epi-2020-0029] [Cited by in Crossref: 9] [Cited by in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] Open
Abstract
Aim: Gain insight about the role of DNA methylation in the malignant growth of colon cancer. Patients & methods: Methylation and gene expression from 90 adjacent-tumor paired tissues and 48 healthy tissues were analyzed. Tumor genes whose change in expression was explained by changes in methylation were identified using linear models adjusted for tumor stromal content. Results: No differences in methylation were found between adjacent and healthy tissues, but clear differences were found between adjacent and tumor samples. We identified hypermethylated CpG islands located in promoter regions that drive differential gene expression of transcription factors and their target genes. Conclusion: Changes in methylation of a few genes provoke important changes in gene expression, by expanding the signal through transcription activation/repression.
Collapse
|
1227
|
|
Klepacka J, Najda A. Effect of commercial processing on polyphenols and antioxidant activity of buckwheat seeds. Int J Food Sci Technol 2021;56:661-70. [DOI: 10.1111/ijfs.14714] [Cited by in Crossref: 4] [Cited by in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text]
|
1228
|
|
Dhawi F, S El-Beltagi H, Aly E, Hamed AM. Antioxidant, Antibacterial Activities and Mineral Content of Buffalo Yoghurt Fortified with Fenugreek and Moringa oleifera Seed Flours. Foods 2020;9:E1157. [PMID: 32825777 DOI: 10.3390/foods9091157] [Cited by in Crossref: 7] [Cited by in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Recently, there is an increasing demand for functional yoghurts by consumer, especially those produced through the incorporation of food of plant origin or its bioactive components. The current research was devoted to formulating functional buffalo yoghurt through the addition of 0.1 and 0.2% of fenugreek (Trigonella foenum-graecum) seed flour (F1 and F2) and Moringa oleifera seed flour (M1 and M2). The effects of fortification were evaluated on physicochemical, total phenolic content (TPC), antioxidant activity (AOA), the viability of yoghurt starter, and sensory acceptability of yoghurts during cold storage. Moringa oleifera seed flour had higher contents of TPC (140.12 mg GAE/g) and AOA (31.30%) as compared to fenugreek seed flour (47.4 mg GAE/g and 19.1%, respectively). Values of TPC and AOA significantly increased in fortified yoghurts, and M2 treatment had the highest values of TPC (31.61, 27.29, and 25.69 mg GAE/g) and AOA (89.32, 83.5, and 80.35%) at 1, 7, and 14 days of storage, respectively. M2 showed significantly higher antibacterial activity against E. coli, S. aureus, L. monocytogenes, and Salmonella spp. and the zones of inhibition were 12.65, 13.14, 17.23 and 14.49 mm, respectively. On the other hand, control yoghurt showed the lowest antibacterial activity and the zones of inhibition were (4.12, 5.21, 8.55, and 8.39 mm against E. coli, S. aureus, L. monocytogenes, and Salmonella spp., respectively). Incorporation of 0.1% and 0.2% of moringa seed flour (M1 and M2) led to a higher content of Ca, P, K, and Fe and lower content of Mg and Zn as compared to F1 and F2, respectively. Thus, it could be concluded that fenugreek and Moringa oleifera seed flour can be exploited in the preparation of functional novel yoghurt.
Collapse
|
1229
|
|
Sharma S, Awad IE, Yadav A, Poirier RA. Molecular level investigation of curcumin self-assembly induced by trigonelline and nanoparticle formation. Appl Nanosci 2020;:1-12. [PMID: 32837805 DOI: 10.1007/s13204-020-01526-4] [Cited by in Crossref: 4] [Cited by in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures]
Abstract
Nanoparticle-facilitated drug delivery forms the core of medicine nowadays with the drug being delivered right at the target, reducing side effects and enhancing therapeutic value. Nanoparticles derived from natural compounds are further a point of focus being biocompatible and safe by and large. In this study, we have performed HF/6-31G calculations coupled with intermolecular interaction calculations and nanoscale molecular dynamics simulations to investigate self-assemblage in curcumin induced by trigonelline. Similar to recently reported self-assemblage in curcumin induced by sugar, trigonelline, a natural antidiabetic derived from fenugreek, can also induce auto-catalyzed self-assemblage in curcumin to form nanoparticles. It has been shown that these nanoparticles may be utilized for the delivery of drugs with severe side effects especially for diabetic patients with triple benefit of being antidiabetic, biocompatible and safe. As an example, carriage of antidiabetic drug pioglitazone and anticancer drug taxol have been depicted utilizing nanoparticles of curcumin and trigonelline. Twenty five taxol molecules could be comfortably carried in a 50 nm nanoparticle with an average overall root mean square deviation of 2.89 Å with reference to initial positions. For the first time, this study shows the possibility of developing antidiabetic nanoparticles with plethora of opportunities for diabetic patients. The study is expected to motivate experimental verification and has a long lasting impact in medicinal chemistry.
Collapse
|
1230
|
|
Famurewa AC, Ekeleme‐egedigwe CA, Onwe CS, Egedigwe UO, Okoro CO, Egedigwe UJ, Asogwa NT. Ginger juice prevents cisplatin‐induced oxidative stress, endocrine imbalance and NO/iNOS/NF‐κB signalling via modulating testicular redox‐inflammatory mechanism in rats. Andrologia 2020;52. [DOI: 10.1111/and.13786] [Cited by in Crossref: 8] [Cited by in RCA: 11] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text]
|
1231
|
|
Wang D, Huang J, Yeung AWK, Tzvetkov NT, Horbańczuk JO, Willschke H, Gai Z, Atanasov AG. The Significance of Natural Product Derivatives and Traditional Medicine for COVID-19. Processes (Basel) 2020;8:937. [DOI: 10.3390/pr8080937] [Cited by in Crossref: 14] [Cited by in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To date, there have been more than 10 million reported cases, more than 517,000 deaths in 215 countries, areas or territories. There is no effective antiviral medicine to prevent or treat COVID-19. Natural products and traditional medicine products with known safety profiles are a promising source for the discovery of new drug leads. There is increasing number of publications reporting the effect of natural products and traditional medicine products on COVID-19. In our review, we provide an overview of natural products and their derivatives or mimics, as well as traditional medicine products, which were reported to exhibit potential to inhibit SARS-CoV-2 infection in vitro, and to manage COVID-19 in vivo, or in clinical reports or trials. These natural products and traditional medicine products are categorized in several classes: (1) anti-malaria drugs including chloroquine and hydroxychloroquine, (2) antivirals including nucleoside analogs (remdesivir, favipiravir, β-D-N4-hydroxycytidine, ribavirin and among others), lopinavir/ritonavir and arbidol, (3) antibiotics including azithromycin, ivermectin and teicoplanin, (4) anti-protozoal drug, emetine, anti-cancer drug, homoharringtonine, and others, as well as (5) traditional medicine (Lian Hua Qing Wen Capsule, Shuang Huang Lian Oral Liquid, Qingfei Paidu Decoction and Scutellariae Radix). Randomized, double-blind and placebo-controlled large clinical trials are needed to provide solid evidence for the potential effective treatment. Currently, drug repurposing is a promising strategy to quickly find an effective treatment for COVID-19. In addition, carefully combined cocktails need to be examined for preventing a COVID-19 pandemic and the resulting global health concerns.
Collapse
|
1232
|
|
Barry Z, Park B, Corson TW. Pharmacological Potential of Small Molecules for Treating Corneal Neovascularization. Molecules 2020;25:E3468. [PMID: 32751576 DOI: 10.3390/molecules25153468] [Cited by in Crossref: 3] [Cited by in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Under healthy conditions, the cornea is an avascular structure which allows for transparency and optimal visual acuity. Its avascular nature is maintained by a balance of proangiogenic and antiangiogenic factors. An imbalance of these factors can result in abnormal blood vessel proliferation into the cornea. This corneal neovascularization (CoNV) can stem from a variety of insults including hypoxia and ocular surface inflammation caused by trauma, infection, chemical burns, and immunological diseases. CoNV threatens corneal transparency, resulting in permanent vision loss. Mainstay treatments of CoNV have partial efficacy and associated side effects, revealing the need for novel treatments. Numerous natural products and synthetic small molecules have shown potential in preclinical studies in vivo as antiangiogenic therapies for CoNV. Such small molecules include synthetic inhibitors of the vascular endothelial growth factor (VEGF) receptor and other tyrosine kinases, plus repurposed antimicrobials, as well as natural source-derived flavonoid and non-flavonoid phytochemicals, immunosuppressants, vitamins, and histone deacetylase inhibitors. They induce antiangiogenic and anti-inflammatory effects through inhibition of VEGF, NF-κB, and other growth factor receptor pathways. Here, we review the potential of small molecules, both synthetics and natural products, targeting these and other molecular mechanisms, as antiangiogenic agents in the treatment of CoNV.
Collapse
|
1233
|
|
El Bairi K, Afqir S, Amrani M. Is HE4 Superior over CA-125 in the Follow-up of Patients with Epithelial Ovarian Cancer? Curr Drug Targets 2020;21:1026-33. [PMID: 32334501 DOI: 10.2174/1389450121666200425211732] [Cited by in Crossref: 4] [Cited by in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
Notwithstanding important advances in the treatment of epithelial ovarian cancer (EOC),
this disease is still a leading cause of global high mortality from gynecological malignancies. Recurrence
in EOC is inevitable and it is responsible for poor survival rates. There is a critical need for novel
effective biomarkers with improved accuracy compared to the standard carbohydrate antigen-125
(CA-125) for follow-up. The human epididymis protein 4 (HE4) is used for early detection of EOC
(ROMA algorithm) as well as for predicting optimal cytoreduction after neoadjuvant chemotherapy
and survival outcomes. Notably, the emerging HE4 is a promising prognostic biomarker that has displayed
better accuracy in various recent studies for detecting recurrent disease. In this mini-review, we
discussed the potential of HE4 as an accurate predictor of EOC recurrence.
Collapse
|
1234
|
|
Cardona AF, Arrieta O, Ruiz-Patiño A, Sotelo C, Zamudio-Molano N, Zatarain-Barrón ZL, Ricaurte L, Raez L, Álvarez MPP, Barrón F, Rojas L, Rolfo C, Karachaliou N, Molina-Vila MA, Rosell R. Precision medicine and its implementation in patients with NTRK fusion genes: perspective from developing countries. Ther Adv Respir Dis 2020;14:1753466620938553. [PMID: 32643553 DOI: 10.1177/1753466620938553] [Cited by in Crossref: 5] [Cited by in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Precision oncology is the field that places emphasis on the diagnosis and treatment of tumors that harbor specific genomic alterations susceptible to inhibition or modulation. Although most alterations are only present in a minority of patients, a substantial effect on survival can be observed in this subgroup. Mass genome sequencing has led to the identification of a specific driver in the translocations of the tropomyosin receptor kinase family (NTRK) in a subset of rare tumors both in children and in adults, and to the development and investigation of Larotrectinib. This medication was granted approval by the US Food and Drug Administration for NTRK-positive tumors, regardless of histology or age group, as such, larotrectinib was the first in its kind to be approved under the premise that molecular pattern is more important than histology in terms of therapeutic approach. It yielded significant results in disease control with good tolerability across a wide range of diseases including rare pediatric tumors, salivary gland tumors, gliomas, soft-tissue sarcomas, and thyroid carcinomas. In addition, and by taking different approaches in clinical trial design and conducting allocation based on biomarkers, the effects of target therapies can be isolated and quantified. Moreover, and considering developing nations and resource-limited settings, precision oncology could offer a tool to reduce cancer-related disability and hospital costs. In addition, developing nations also present patients with rare tumors that lack a chance of treatment, outside of clinical trials. This, in turn, offers the possibility for international collaboration, and contributes to employment, education, and health service provisions. The reviews of this paper are available via the supplemental material section.
Collapse
|
1235
|
|
Cervena K, Siskova A, Buchler T, Vodicka P, Vymetalkova V. Methylation-Based Therapies for Colorectal Cancer. Cells 2020;9:E1540. [PMID: 32599894 DOI: 10.3390/cells9061540] [Cited by in Crossref: 14] [Cited by in RCA: 10] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Colorectal carcinogenesis (CRC) is caused by the gradual long-term accumulation of both genetic and epigenetic changes. Recently, epigenetic alterations have been included in the classification of the CRC molecular subtype, and this points out their prognostic impact. As epigenetic modifications are reversible, they may represent relevant therapeutic targets. DNA methylation, catalyzed by DNA methyltransferases (DNMTs), regulates gene expression. For many years, the deregulation of DNA methylation has been considered to play a substantial part in CRC etiology and evolution. Despite considerable advances in CRC treatment, patient therapy response persists as limited, and their profit from systemic therapies are often hampered by the introduction of chemoresistance. In addition, inter-individual changes in therapy response in CRC patients can arise from their specific (epi)genetic compositions. In this review article, we summarize the options of CRC treatment based on DNA methylation status for their predictive value. This review also includes the therapy outcomes based on the patient’s methylation status in CRC patients. In addition, the current challenge of research is to develop therapeutic inhibitors of DNMT. Based on the essential role of DNA methylation in CRC development, the application of DNMT inhibitors was recently proposed for the treatment of CRC patients, especially in patients with DNA hypermethylation.
Collapse
|
1236
|
|
Garcia-Lazaro RS, Lamdan H, Caligiuri LG, Lorenzo N, Berengeno AL, Ortega HH, Alonso DF, Farina HG. In vitro and in vivo antitumor activity of Yerba Mate extract in colon cancer models. J Food Sci 2020;85:2186-97. [PMID: 32567699 DOI: 10.1111/1750-3841.15169] [Cited by in Crossref: 12] [Cited by in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
Yerba Mate (Ilex paraguariensis St. Hill. Aquifoliaceae) is a native South American tree and has a large amount of bioactive compounds. Colorectal cancer (CRC) is one of the so-called westernized diseases and is the third most common cancer in both men and women. Efficient strategies for the treatment of CRC are extensively being explored including dietary intervention. The objective of our research was to evaluate the effects of Yerba Mate extract on cell proliferation, invasive capacity of tumor cells, and angiogenesis. For this, in vitro and in vivo experimentation was carried out using CRC models. The extract was generated by aqueous extraction and prepared according to traditional American procedure of preparing mate infusion. In vitro results showed that the Yerba Mate extract inhibits CT26 and COLO 205 cell proliferation with IC50 values of 0.25 and 0.46 mg/mL, respectively. We demonstrated by TUNEL assay that one of the mechanisms by which Yerba Mate extract decreases cell proliferation is by induction of apoptosis. In a murine syngeneic tumor model, oral administration of Yerba Mate extract in a dose of 1.6 g/kg/day significantly inhibited angiogenesis and tumor growth without affecting biological parameters or body weight. Our findings suggest that Yerba Mate may be a promising agent for the treatment of colon cancer and could be used as an herbal medicine or functional food ingredient. PRACTICAL APPLICATION: Considering the chemical composition and presence of phenolic compounds with their free-radical scavenging activities and bioactivities against colon cancer cells, Yerba Mate can be a promising candidate as healthy food sources in human nutrition, and also be considered a natural source of potential antitumor agents. Taking into account the economic importance of Yerba Mate in Argentina, this vegetable would have a greater commercial value as a functional food.
Collapse
|
1237
|
|
Wu T, Yue R, He M, Xu C. Effect of Fenugreek on vasomotor symptoms in menopausal women: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2020;99:e20526. [PMID: 32502006 DOI: 10.1097/MD.0000000000020526] [Cited by in Crossref: 1] [Cited by in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Background: Vasomotor symptoms (hot flashes or night sweats) are closely related to the impaired quality of life in menopausal women. Fenugreek is the ripe seed of Trigonella foenum graecum Linn. In China, this plant is used to relieve menopausal symptoms in women. Although recent studies have shown that fenugreek may have a good effect on the menopausal symptoms, there is no meta-analysis to systematically evaluate its efficacy in improving menopausal vasomotor symptoms. Methods: Randomized controlled trials that met the inclusion criteria will be retrieved in 5 English online databases and 4 Chinese online databases. The primary outcomes are changes in frequency and intensity of vasomotor symptoms that measured by validated scales. The secondary outcomes will include quality of life, blood hormone parameters, blood biochemical parameters, and adverse events. Heterogeneity of data will be assessed by I2 and Cochrane Q statistics. Sensitivity analysis and subgroup analysis will be performed to explore the sources of heterogeneity. Egger test and Begg test will be used to assess the publication bias. Finally, we will evaluate the quality of evidence by the GRADE approach. All the data statistics will be performed using the STATA 15.0 software. Results: All the results of will be published in a peer-reviewed journal. Conclusions: This meta-analysis will systematically evaluate the efficacy and safety of fenugreek in the treatment of menopausal vasomotor symptoms. OSF registration number: 10.17605/OSF.IO/3BCY8.
Collapse
|
1238
|
|
Michel LL, von Au A, Mavratzas A, Smetanay K, Schütz F, Schneeweiss A. Immune Checkpoint Blockade in Patients with Triple-Negative Breast Cancer. Target Oncol 2020;15:415-28. [DOI: 10.1007/s11523-020-00730-0] [Cited by in Crossref: 13] [Cited by in RCA: 16] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text]
|
1239
|
|
El Bairi K, Jabi R, Trapani D, Boutallaka H, Ouled Amar Bencheikh B, Bouziane M, Amrani M, Afqir S, Maleb A. Can the microbiota predict response to systemic cancer therapy, surgical outcomes, and survival? The answer is in the gut. Expert Rev Clin Pharmacol 2020;13:403-21. [PMID: 32308061 DOI: 10.1080/17512433.2020.1758063] [Cited by in Crossref: 3] [Cited by in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
INTRODUCTION The gut microbiota seems to play a key role in tumorigenesis, across various hallmarks of cancer. Recent evidence suggests its potential use as a biomarker predicting drug response and adding prognostic information, generally in the context of immuno-oncology. AREAS COVERED In this review, we focus on the modulating effects of gut microbiota dysbiosis on various anticancer molecules used in practice, including cytotoxic and immune-modulating agents, primarily immune-checkpoint inhibitors (ICI). Pubmed/Medline-based literature search was conducted to find potential original studies that discuss gut microbiota as a prognostic and predictive biomarker for cancer therapy. We also looked at the US ClinicalTrials.gov website to find additional studies particularly ongoing human clinical trials. EXPERT COMMENTARY Sequencing of stool-derived materials and tissue samples from cancer patients and animal models has shown a significant enrichment of various bacteria such as Fusobacterium nucleatum and Bacteroides fragilis were associated with resistant disease and poorer outcomes. Gut microbiota was also found to be associated with surgical outcomes and seems to play a significant role in anastomotic leak (ATL) after surgery mainly by collagen breakdown. However, this research field is just at the beginning and the current findings are not yet ready to change clinical practice.
Collapse
|
1240
|
|
Sugie T, Sato E, Miyashita M, Yamaguchi R, Sakatani T, Kozuka Y, Moritani S, Suzuki E, Kakimi K, Mikami Y, Moriya T. Multispectral quantitative immunohistochemical analysis of tumor-infiltrating lymphocytes in relation to programmed death-ligand 1 expression in triple-negative breast cancer. Breast Cancer 2020;27:519-26. [PMID: 32447649 DOI: 10.1007/s12282-020-01110-2] [Cited by in Crossref: 5] [Cited by in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) expression on immune cells (ICs) is a predictive marker for PD-L1 checkpoint blockade in patients with triple-negative breast cancer (TNBC). However, the level of PD-L1 expression and the percentage of cells that are PD-L1+ are continuous variables not dichotomous variables for tumor-infiltrating lymphocytes (TILs) and other cells. METHODS Multiplexed immunohistochemistry was applied to 31 archived surgical specimens from untreated TNBC patients. TIL levels were visually scored, and CD8+ T cells and PD-L1+ ICs were quantified using an automated multispectral imaging system. PD-L1 expression was assessed within a multiplexed context (CD8 combined spectral composite). RESULTS The mean value of stromal TILs (i.e., the percentage of the stromal area with a dese mononuclear infiltrate) was 20%. The frequency of patients with PD-L1-positive tumor cells (TC) and ICs was 38.7% and 32.2%, respectively, with a significant association between them. TIL levels were correlated with CD8+ T cell infiltration in the stroma (Spearman r = 0.795, p < 0.0001). PD-L1 expression on IC was significantly associated with TIL levels (Spearman r = 0.790, p < 0.001) and infiltration of CD8+ T cells (Spearman r = 0.683, p < 0.0001). CONCLUSIONS The level of PD-L1 on IC was correlated with the level of PD-L1 on TC as well as TIL levels and infiltration of CD8+ T cells. These results suggest that high PD-L1 on IC may reflect T cell-inflamed tumors with the amount of TILs present, including the CD8+ T cells required for anti-tumor responses.
Collapse
|
1241
|
|
Asemani S, Montazeri V, Foroutan-Ghaznavi M, Pirouzpanah SS, Baradaran B, Jafari S, Barzegar A, Shanehbandi D, Asadi N, Pirouzpanah S. Dietary patterns and relative expression levels of PPAR-γ, VEGF-A and HIF-1α genes in benign breast diseases: case-control and consecutive case-series designs. Br J Nutr 2020;124:832-43. [PMID: 32406342 DOI: 10.1017/S0007114520001737] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text]
Abstract
We aimed to study dietary patterns in association with the relative expression levels of PPAR-γ, vascular endothelial growth factor-A (VEGF-A) and hypoxia-inducible factor-1α (HIF-1α) in women with benign breast disease (BBD). The study design was combinative, included a case-series and case-control compartments. Initially, eligible BBD patients (n 77, aged 19-52 years old) were recruited at Nour-Nejat hospital, Tabriz, Iran (2012-2014). A hospital-based group of healthy controls was matched for age (n 231, aged 20-63 years old) and sex. Dietary data were collected using a valid 136-item FFQ. Principal component analysis generated two main components (Kaiser-Meyer-Olkin = 0·684), including a Healthy pattern (whole bread, fruits, vegetables, vegetable oils, legumes, spices, seafood, low-fat meat, skinless poultry, low-fat dairy products, nuts and seeds) and a Western pattern (starchy foods, high-fat meat and poultry, high-fat dairy products, hydrogenated fat, fast food, salt and sweets). High adherence to the Western pattern increased the risk of BBD (ORadj 5·59; 95 % CI 2·06, 15·10; P < 0·01), whereas high intake of the Healthy pattern was associated with a 74 % lower risk of BBD (95 % CI 0·08, 0·81; P < 0·05). In the BBD population, the Western pattern was correlated with over-expression of HIF-1α (radj 0·309, P < 0·05). There were inverse correlations between the Healthy pattern and expressions of PPAR-γ (radj -0·338, P < 0·05), HIF-1α (radj -0·340, P < 0·05) and VEGF-A (radj -0·286, P < 0·05). In conclusion, new findings suggested that the Healthy pattern was associated inversely with the risk of BBD, and this could be correlated with down-regulation of PPAR-γ, VEGF-A and HIF-1α genes, which might hold promise to preclude BBD of malignant pathological transformation.
Collapse
|
1242
|
|
Abbink K, Zusterzeel PLM, Geurts-Moespot A, van der Steen R, Span PN, Sweep FCGJ. Prognostic significance of VEGF and components of the plasminogen activator system in endometrial cancer. J Cancer Res Clin Oncol 2020;146:1725-35. [PMID: 32394054 DOI: 10.1007/s00432-020-03225-7] [Cited by in Crossref: 4] [Cited by in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures]
Abstract
Objective The plasminogen activator system (PAS) and vascular endothelial growth factor (VEGF) are important in the carcinogenesis and play a key role in cancer invasion and mediating metastasis of carcinomas. The aim of the study was to evaluate the correlation of serum levels of VEGF and components of the PAS with clinicopathological risk factors and outcome in patients with endometrial cancer (EC). Methods Preoperative blood was collected from 173 patients treated for EC between 1999 and 2009. Serum concentrations of VEGF, urokinase plasminogen activator (uPA) tissue plasminogen activator (tPA), plasminogen activator inhibitor type-1 (PAI-1) and -2 (PAI-2) were assessed by enzyme-linked immunosorbent assays (ELISA). Results Serum levels of VEGF and components of the PAS were significantly associated with stage of the disease, tumor histology, tumor grade, myometrial invasion (MI), presence of lymphovascular space invasion (LVSI) and lymph node metastases (LNM). Preoperative serum levels of PAI-1 and -2 and tPA were higher in patients who experienced a recurrence than in patients who remained disease free (p < 0.01). PAI-1 and -2 and tPA were significantly independent prognostic factors for DFS with a HR of 3.85 (95% CI 1.84–8.07), 3.90 (95% CI 1.75–8.66) and 2.53 (95% CI 1.16–5.55), respectively. PAI-1 and tPA turned out to be independent prognostic factors for OS, with a HR of 2.09 (95% CI 1.08–4.05) and 2.16 (95% CI 1.06–4.44), respectively. Conclusion Serum levels of VEGF and components of the PAS at primary diagnosis were associated with well-known clinicopathological risk factors such as; FIGO stage, tumor histology, tumor grade, MI, LVSI and LNM. High concentrations of PAI-1 and-2 and tPA are independent factors for poor prognosis in patients with endometrial cancer.
Collapse
|
1243
|
|
Kos Z, Roblin E, Kim RS, Michiels S, Gallas BD, Chen W, van de Vijver KK, Goel S, Adams S, Demaria S, Viale G, Nielsen TO, Badve SS, Symmans WF, Sotiriou C, Rimm DL, Hewitt S, Denkert C, Loibl S, Luen SJ, Bartlett JMS, Savas P, Pruneri G, Dillon DA, Cheang MCU, Tutt A, Hall JA, Kok M, Horlings HM, Madabhushi A, van der Laak J, Ciompi F, Laenkholm AV, Bellolio E, Gruosso T, Fox SB, Araya JC, Floris G, Hudeček J, Voorwerk L, Beck AH, Kerner J, Larsimont D, Declercq S, Van den Eynden G, Pusztai L, Ehinger A, Yang W, AbdulJabbar K, Yuan Y, Singh R, Hiley C, Bakir MA, Lazar AJ, Naber S, Wienert S, Castillo M, Curigliano G, Dieci MV, André F, Swanton C, Reis-Filho J, Sparano J, Balslev E, Chen IC, Stovgaard EIS, Pogue-Geile K, Blenman KRM, Penault-Llorca F, Schnitt S, Lakhani SR, Vincent-Salomon A, Rojo F, Braybrooke JP, Hanna MG, Soler-Monsó MT, Bethmann D, Castaneda CA, Willard-Gallo K, Sharma A, Lien HC, Fineberg S, Thagaard J, Comerma L, Gonzalez-Ericsson P, Brogi E, Loi S, Saltz J, Klaushen F, Cooper L, Amgad M, Moore DA, Salgado R; International Immuno-Oncology Biomarker Working Group. Pitfalls in assessing stromal tumor infiltrating lymphocytes (sTILs) in breast cancer. NPJ Breast Cancer 2020;6:17. [PMID: 32411819 DOI: 10.1038/s41523-020-0156-0] [Cited by in Crossref: 66] [Cited by in RCA: 68] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Stromal tumor-infiltrating lymphocytes (sTILs) are important prognostic and predictive biomarkers in triple-negative (TNBC) and HER2-positive breast cancer. Incorporating sTILs into clinical practice necessitates reproducible assessment. Previously developed standardized scoring guidelines have been widely embraced by the clinical and research communities. We evaluated sources of variability in sTIL assessment by pathologists in three previous sTIL ring studies. We identify common challenges and evaluate impact of discrepancies on outcome estimates in early TNBC using a newly-developed prognostic tool. Discordant sTIL assessment is driven by heterogeneity in lymphocyte distribution. Additional factors include: technical slide-related issues; scoring outside the tumor boundary; tumors with minimal assessable stroma; including lymphocytes associated with other structures; and including other inflammatory cells. Small variations in sTIL assessment modestly alter risk estimation in early TNBC but have the potential to affect treatment selection if cutpoints are employed. Scoring and averaging multiple areas, as well as use of reference images, improve consistency of sTIL evaluation. Moreover, to assist in avoiding the pitfalls identified in this analysis, we developed an educational resource available at www.tilsinbreastcancer.org/pitfalls.
Collapse
|
1244
|
|
Hudeček J, Voorwerk L, van Seijen M, Nederlof I, de Maaker M, van den Berg J, van de Vijver KK, Sikorska K, Adams S, Demaria S, Viale G, Nielsen TO, Badve SS, Michiels S, Symmans WF, Sotiriou C, Rimm DL, Hewitt SM, Denkert C, Loibl S, Loi S, Bartlett JMS, Pruneri G, Dillon DA, Cheang MCU, Tutt A, Hall JA, Kos Z, Salgado R, Kok M, Horlings HM; International Immuno-Oncology Biomarker Working Group. Application of a risk-management framework for integration of stromal tumor-infiltrating lymphocytes in clinical trials. NPJ Breast Cancer 2020;6:15. [PMID: 32436923 DOI: 10.1038/s41523-020-0155-1] [Cited by in Crossref: 9] [Cited by in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Stromal tumor-infiltrating lymphocytes (sTILs) are a potential predictive biomarker for immunotherapy response in metastatic triple-negative breast cancer (TNBC). To incorporate sTILs into clinical trials and diagnostics, reliable assessment is essential. In this review, we propose a new concept, namely the implementation of a risk-management framework that enables the use of sTILs as a stratification factor in clinical trials. We present the design of a biomarker risk-mitigation workflow that can be applied to any biomarker incorporation in clinical trials. We demonstrate the implementation of this concept using sTILs as an integral biomarker in a single-center phase II immunotherapy trial for metastatic TNBC (TONIC trial, NCT02499367), using this workflow to mitigate risks of suboptimal inclusion of sTILs in this specific trial. In this review, we demonstrate that a web-based scoring platform can mitigate potential risk factors when including sTILs in clinical trials, and we argue that this framework can be applied for any future biomarker-driven clinical trial setting.
Collapse
|
1245
|
|
Bouyahya A, Belmehdi O, Benjouad A, Ameziane El Hassani R, Amzazi S, Dakka N, Bakri Y. Pharmacological properties and mechanism insights of Moroccan anticancer medicinal plants: What are the next steps? Ind Crops Prod 2020;147:112198. [DOI: 10.1016/j.indcrop.2020.112198] [Cited by in Crossref: 22] [Cited by in RCA: 25] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text]
|
1246
|
|
Kralj Cigić I, Rupnik S, Rijavec T, Poklar Ulrih N, Cigić B. Accumulation of Agmatine, Spermidine, and Spermine in Sprouts and Microgreens of Alfalfa, Fenugreek, Lentil, and Daikon Radish. Foods 2020;9:E547. [PMID: 32369919 DOI: 10.3390/foods9050547] [Cited by in Crossref: 9] [Cited by in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Sprouts and microgreens are a rich source of various bioactive compounds. Seeds of lentil, fenugreek, alfalfa, and daikon radish seeds were germinated and the contents of the polyamines agmatine (AGM), putrescine (PUT), cadaverine (CAD), spermidine (SPD), and spermine (SPM) in ungerminated seeds, sprouts, and microgreens were determined. In general, sprouting led to the accumulation of the total polyamine content. The highest levels of AGM (5392 mg/kg) were found in alfalfa microgreens, PUT (1079 mg/kg) and CAD (3563 mg/kg) in fenugreek sprouts, SPD (579 mg/kg) in lentil microgreens, and SPM (922 mg/kg) in fenugreek microgreens. A large increase in CAD content was observed in all three legume sprouts. Conversely, the nutritionally beneficial polyamines AGM, SPD, and SPM were accumulated in microgreens, while their contents of CAD were significantly lower. In contrast, daikon radish sprouts exhibited a nutritionally better profile of polyamines than the microgreens. Freezing and thawing of legume sprouts resulted in significant degradation of CAD, PUT, and AGM by endogenous diamine oxidases. The enzymatic potential of fenugreek sprouts can be used to degrade exogenous PUT, CAD, and tyramine at pH values above 5.
Collapse
|
1247
|
|
Moura DS, Sanchez-Bustos P, Fernandez-Serra A, Lopez-Alvarez M, Mondaza-Hernandez JL, Blanco-Alcaina E, Gavilan-Naranjo A, Martinez-Delgado P, Lacerenza S, Santos-Fernandez P, Carrasco-Garcia I, Hidalgo-Rios S, Gutierrez A, Ramos R, Hindi N, Taron M, Lopez-Guerrero JA, Martin-Broto J. CUL4A, ERCC5, and ERCC1 as Predictive Factors for Trabectedin Efficacy in Advanced Soft Tissue Sarcomas (STS): A Spanish Group for Sarcoma Research (GEIS) Study. Cancers (Basel) 2020;12:E1128. [PMID: 32365979 DOI: 10.3390/cancers12051128] [Cited by in Crossref: 6] [Cited by in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
A translational study was designed to analyze the expression of nucleotide excision repair (NER) and homologous recombination (HR) genes as potential predictive biomarkers for trabectedin in soft-tissue sarcoma (STS). This study is part of a randomized phase II trial comparing trabectedin plus doxorubicin versus doxorubicin in advanced STS. Gene expression levels were evaluated by qRT-PCR, while CUL4A protein levels were quantified by immunohistochemistry. Expression levels were correlated with patients’ progression-free survival (PFS) and overall survival (OS). Gene expression was also evaluated in cell lines and correlated with trabectedin sensitivity. In doxorubicin arm and in the whole series, which includes samples from both arms, no significant differences in terms of PFS were observed amongst the analyzed genes. In the group treated with trabectedin plus doxorubicin, the median of PFS was significantly longer in cases with CUL4A, ERCC1, or ERCC5 overexpression, while BRCA1 expression did not correlated with PFS. Gene expression had no prognostic influence in OS. CUL4A protein levels correlated with worse PFS in doxorubicin arm and in the whole series. In cell lines, only overexpression of ERCC1 was significantly correlated with trabectedin sensitivity. In conclusion, CUL4A, ERCC5, and mainly ERCC1 acted as predictive factors for trabectedin efficacy in advanced STS.
Collapse
|
1248
|
|
Vitelli-Storelli F, Zamora-Ros R, Molina AJ, Fernández-Villa T, Castelló A, Barrio JP, Amiano P, Ardanaz E, Obón-Santacana M, Gómez-Acebo I, Fernández-Tardón G, Molina-Barceló A, Alguacil J, Marcos-Gragera R, Ruiz-Moreno E, Pedraza M, Gil L, Guevara M, Castaño-Vinyals G, Dierssen-Sotos T, Kogevinas M, Aragonés N, Martín V. Association between Polyphenol Intake and Breast Cancer Risk by Menopausal and Hormone Receptor Status. Nutrients 2020;12:E994. [PMID: 32260135 DOI: 10.3390/nu12040994] [Cited by in Crossref: 2] [Cited by in RCA: 3] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
There is limited evidence of phenolic compounds acting as protective agents on several cancer types, including breast cancer (BC). Nevertheless, some polyphenol classes have not been investigated and there is a lack of studies assessing the effect on menopausal status and hormone receptor status as influenced by these compounds. The objective of this study is to evaluate the association between the intake of all polyphenol classes in relation to the BC risk by menopausal and hormone receptor status. We used data from a population-based multi-case-control study (MCC-Spain) including 1472 BC cases and 1577 controls from 12 different regions of Spain. The odds ratios (ORs) with 95% CI were calculated using logistic regression of mixed effects by quartiles and log2 of polyphenol intakes (adjusted for the residual method) of overall BC, menopausal and receptor status. No associations were found between total intake of polyphenols and BC risk. However, inverse associations were found between stilbenes and all BC risk (ORQ4 vs. Q1: 0.70, 95%CI: 0.56–0.89, Ptrend = 0.001), the consumption of hydroxybenzaldehydes (ORQ4 vs. Q1: 0.75, 95%CI: 0.59–0.93, Ptrend = 0.012) and hydroxycoumarins (ORQ4 vs. Q1: 0.73, 95%CI: 0.57–0.93; Ptrend = 0.005) were also inversely associated. The intake of stilbenes, hydroxybenzaldehydes and hydroxycoumarins can contribute to BC reduction risk on all menopausal and receptor statuses.
Collapse
|
1249
|
|
Sama IE, Woolley RJ, Nauta JF, Romaine SPR, Tromp J, Ter Maaten JM, van der Meer P, Lam CSP, Samani NJ, Ng LL, Metra M, Dickstein K, Anker SD, Zannad F, Lang CC, Cleland JGF, van Veldhuisen DJ, Hillege HL, Voors AA. A network analysis to identify pathophysiological pathways distinguishing ischaemic from non-ischaemic heart failure. Eur J Heart Fail 2020;22:821-33. [PMID: 32243695 DOI: 10.1002/ejhf.1811] [Cited by in Crossref: 14] [Cited by in RCA: 15] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Aims Heart failure (HF) is frequently caused by an ischaemic event (e.g. myocardial infarction) but might also be caused by a primary disease of the myocardium (cardiomyopathy). In order to identify targeted therapies specific for either ischaemic or non‐ischaemic HF, it is important to better understand differences in underlying molecular mechanisms. Methods and results We performed a biological physical protein–protein interaction network analysis to identify pathophysiological pathways distinguishing ischaemic from non‐ischaemic HF. First, differentially expressed plasma protein biomarkers were identified in 1160 patients enrolled in the BIOSTAT‐CHF study, 715 of whom had ischaemic HF and 445 had non‐ischaemic HF. Second, we constructed an enriched physical protein–protein interaction network, followed by a pathway over‐representation analysis. Finally, we identified key network proteins. Data were validated in an independent HF cohort comprised of 765 ischaemic and 100 non‐ischaemic HF patients. We found 21/92 proteins to be up‐regulated and 2/92 down‐regulated in ischaemic relative to non‐ischaemic HF patients. An enriched network of 18 proteins that were specific for ischaemic heart disease yielded six pathways, which are related to inflammation, endothelial dysfunction superoxide production, coagulation, and atherosclerosis. We identified five key network proteins: acid phosphatase 5, epidermal growth factor receptor, insulin‐like growth factor binding protein‐1, plasminogen activator urokinase receptor, and secreted phosphoprotein 1. Similar results were observed in the independent validation cohort. Conclusions Pathophysiological pathways distinguishing patients with ischaemic HF from those with non‐ischaemic HF were related to inflammation, endothelial dysfunction superoxide production, coagulation, and atherosclerosis. The five key pathway proteins identified are potential treatment targets specifically for patients with ischaemic
HF.
Collapse
|
1250
|
|
Farooq M, Abutaha N, Mahboob S, Baabbad A, Almoutiri ND, Wadaan MAAM. Investigating the antiangiogenic potential of Rumex vesicarius (humeidh), anticancer activity in cancer cell lines and assessment of developmental toxicity in zebrafish embryos. Saudi J Biol Sci 2020;27:611-22. [PMID: 32210679 DOI: 10.1016/j.sjbs.2019.11.042] [Cited by in Crossref: 6] [Cited by in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] Open
Abstract
Recent trends in anticancer therapy is to use therapeutic agents which not only kill the cancer cell, but are less toxic to surrounding normal cells/tissue. One approach is to cut the nutrient supply to growing tumor cells, by blocking the formation of new blood vessels around the tumor. As the phytochemicals and botanical crude extracts have proven their efficacy as natural antiangiogenic agents with minimum toxicities, there is need to explore varieties of medicinal plants for novel antiangiogenic compounds. Rumex vesicarius L. (Humeidh), is an annual herbal plant with proven medicinal values. The antiangiogenic potential, and developmental toxicity of humeidh in experimental animal models has never been studied before. The crude extracts were prepared from the roots, stems, leaves and flowers of Rumex vesicarius L. in methanol, chloroform, ethyl acetate and n-hexane. The developmental toxicity screening in zebrafish embryos, has revealed that Rumex vesicarius was not toxic to zebrafish embryos. The chloroform stem extract showed significant level of antiangiogenic activity in zebrafish angiogenic assay on a dose dependent manner. Thirty five (35) bioactive compounds were identified by gas chromatography mass spectrophotometry (GC–MS) analysis in the stem extract of Rumex vesicarius. Propanoic acid, 2-[(trimethylsilyl)oxy]-, trimethylsilyl ester, Butane, 1,2,3-tris(trimethylsiloxy), and Butanedioic acid, bis(trimethylsilyl) ester were identified as major compound present in the stem of R. vasicarius. The anticancer activity of roots, stem, leaves and flowers crude extract was evaluated in human breast cancer (MCF7), human colon carcinoma (Lovo, and Caco-2), human hepatocellular carcinoma (HepG2) cell lines. Most of the crude extracts did not show significant level of cytotoxicity in tested cancer cells line, except, chloroform extract of stem which exhibited strong anticancer activity in all tested cancer cells with IC50 values in micro molar range. Based on these results, it is recommended that formulation prepared from R. vesicarius can further be tested in clinical trials in order to explore its therapeutic potential as an effective and safe natural anticancer product.
Collapse
|