1
|
Liu M, Wang Y, Wang C, Li P, Qiu J, Yang N, Sun M, Han L. A Microfluidic 3D-Tumor-Spheroid Model for the Evaluation of Targeted Therapies from Angiogenesis-Related Cytokines at the Single Spheroid Level. Adv Healthc Mater 2024; 13:e2402321. [PMID: 39126126 DOI: 10.1002/adhm.202402321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Indexed: 08/12/2024]
Abstract
Angiogenesis is a key player in drug resistance to targeted therapies for breast cancer. The average expression of angiogenesis-related cytokines is widely associated with the treatments of target therapies for a population of cells or spheroids, overlooking the distinct responses for individuals. In this work, a highly integrated microfluidic platform is developed for the generation of monodisperse multicellular tumor spheroids (MTSs), drug treatments, and the measurement of cytokines for individual MTSs in a single chip. The platform allows the correlation evaluation between cytokine secretion and drug treatment at the level of individual spheroids. For validation, quantities of six representative proangiogenic cytokines are tested against treatments with four model drugs at varying times and concentrations. By applying a linear regression model, significant correlations are established between cytokine secretion and the treated drug concentration for individual spheroids. The proposed platform provides a high-throughput method for the investigation of the molecular mechanism of the cytokine response to targeted therapies and paves the way for future drug screening using predictive regression models at the single-spheroid level.
Collapse
Affiliation(s)
- Mengqi Liu
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Yihe Wang
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Chao Wang
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Ping Li
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Jiaoyan Qiu
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Ningkai Yang
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Mingyuan Sun
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Tsingdao, 266237, China
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, 250100, P. R. China
| |
Collapse
|
2
|
Rodríguez-Candela Mateos M, Carpintero-Fernández P, Freijanes PS, Mosquera J, Nebril BA, Mayán MD. Insights into the role of connexins and specialized intercellular communication pathways in breast cancer: Mechanisms and applications. Biochim Biophys Acta Rev Cancer 2024; 1879:189173. [PMID: 39154967 DOI: 10.1016/j.bbcan.2024.189173] [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: 05/27/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Gap junctions, membrane-based channels comprised of connexin proteins (Cxs), facilitate direct communication among neighbouring cells and between cells and the extracellular space through their hemichannels. The normal human breast expresses various Cxs family proteins, such as Cx43, Cx30, Cx32, Cx46, and Cx26, crucial for proper tissue development and function. These proteins play a significant role in breast cancer development, progression, and therapy response. In primary tumours, there is often a reduction and cytoplasmic mislocalization of Cx43 and Cx26, while metastatic lesions show an upregulation of these and other Cxs. Although existing research predominantly supports the tumour-suppressing role of Cxs in primary carcinomas through channel-dependent and independent functions, controversies persist regarding their involvement in the metastatic process. This review aims to provide an updated perspective on Cxs in human breast cancer, with a specific focus on intrinsic subtypes due to the heterogeneous nature of this disease. Additionally, the manuscript will explore the role of Cxs in immune interactions and novel forms of intercellular communication, such as tunneling nanotubes and extracellular vesicles, within the breast tumour context and tumour microenvironment. Recent findings suggest that Cxs hold potential as therapeutic targets for mitigating metastasis and drug resistance. Furthermore, they may serve as novel biomarkers for cancer prognosis, offering promising avenues for future research and clinical applications.
Collapse
Affiliation(s)
- Marina Rodríguez-Candela Mateos
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain
| | - Paula Carpintero-Fernández
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; CellCOM Research Group, Center for Research in Nanomaterials and Biomedicine (CINBIO), Universidade de Vigo, Edificio Olimpia Valencia, Campus Universitario Lagoas Marcosende, 36310 Vigo, Spain; Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS, Spain
| | - Paz Santiago Freijanes
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Anatomic Pathology Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - Joaquin Mosquera
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Surgery Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - Benigno Acea Nebril
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Surgery Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - María D Mayán
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; CellCOM Research Group, Center for Research in Nanomaterials and Biomedicine (CINBIO), Universidade de Vigo, Edificio Olimpia Valencia, Campus Universitario Lagoas Marcosende, 36310 Vigo, Spain; Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS, Spain.
| |
Collapse
|
3
|
Wang Z, Wang Z, Deng L, Wu X, Liang Y, Wei P. Basic Fibroblast Growth Factor Accumulation in Culture Medium Masks the Direct Antitumor Effect of Anti-VEGF Agent Bevacizumab. DOKL BIOCHEM BIOPHYS 2024; 517:285-290. [PMID: 39002014 DOI: 10.1134/s1607672924600283] [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: 03/20/2024] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 07/15/2024]
Abstract
The direct antitumor effect of bevacizumab (BEV) has long been debated. Evidence of the direct antitumor activities of drugs are mainly obtained from in vitro experiments, which are greatly affected by experimental conditions. In this study, we evaluated the effect of BEV-containing medium renewal on the results of in vitro cytotoxicity experiments in A549 and U251 cancer cells. We observed starkly different results between the experiments with and without BEV-containing medium renewal. Specifically, BEV inhibited the tumor cell growth in the timely replacement with a BEV-containing medium but promoted tumor cell growth without medium renewal. Meanwhile, compared with the control, a significant basic fibroblast growth factor (bFGF) accumulation in the supernatant was observed in the group without medium renewal but none in that with replaced medium. Furthermore, bFGF neutralization partially reversed the pro-proliferative effect of BEV in the medium non-renewed group, while exogenous bFGF attenuated the tumor cell growth inhibition of BEV in the medium-renewed group. Our data explain the controversy over the direct antitumor effect of BEV in different studies from the perspective of the compensatory autocrine cytokines in tumor cells.
Collapse
Affiliation(s)
- Zhiyong Wang
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Ziyi Wang
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Liyan Deng
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xiaolan Wu
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Yanfang Liang
- Department of Pathology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Pei Wei
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China.
| |
Collapse
|
4
|
Abdallah R, Shaito AA, Badran A, Baydoun S, Sobeh M, Ouchari W, Sahri N, Eid AH, Mesmar JE, Baydoun E. Fractionation and phytochemical composition of an ethanolic extract of Ziziphus nummularia leaves: antioxidant and anticancerous properties in human triple negative breast cancer cells. Front Pharmacol 2024; 15:1331843. [PMID: 38405665 PMCID: PMC10885810 DOI: 10.3389/fphar.2024.1331843] [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: 11/01/2023] [Accepted: 01/09/2024] [Indexed: 02/27/2024] Open
Abstract
Natural products have long been utilized in traditional medicine as remedies to improve health and treat illnesses, and have had a key role in modern drug discovery. Recently, there has been a revived interest in the search for bioactives from natural sources as alternative or complementary modalities to synthetic medicines; especially for cancer treatment, which incidence and mortality rates are on the rise worldwide. Ziziphus nummularia has been widely used in traditional medicine for the treatment of various diseases. Its traditional uses and numerous ethnopharmacological properties may be attributed to its richness in bioactive metabolites. However, its phytochemical composition or chemopreventive effects against the aggressive triple-negative breast cancer (TNBC) are still poorly explored. Here, phytochemical composition of an ethanolic extract of Z. nummularia leaves (ZNE) and its chromatographically isolated fractions was identified both qualitatively by spectrophotometric assays and analytically by HPLC-PDA-MS/MS. The anti-proliferative effects of ZNE were tested in several cancer cell lines, but we focused on its anti-TNBC effects since they were not explored yet. The anti-cancerous potential of ZNE and its fractions was tested in vitro in MDA-MB-231, a TNBC cell line. Results showed that ZNE and its Fraction 6 (F6) reduced the viability of MDA-MB-231 cells. F6 decreased MDA-MB-231 viability more than crude ZNE or its other fractions. ZNE and F6 are rich in phytochemicals and HPLC-PDA-MS/MS analysis identified several metabolites that were previously reported to have anti-cancerous effects. Both ZNE and F6 showed potent antioxidant capacity in the DPPH assay, but promoted reactive oxygen species (ROS) production in MDA-MB-231 cells; an effect which was blunted by the antioxidant N-acetyl cysteine (NAC). NAC also blunted ZNE- and F6-induced reduction in TNBC cell viability. We also demonstrated that ZNE and F6 induced an arrest of the cell cycle, and triggered apoptosis- and autophagy-mediated cell death. ZNE and F6 inhibited metastasis-related cellular processes by modifying cell migration, invasion, and adhesion. Taken together, our findings reveal that Z. nummularia is rich in phytochemicals that can attenuate the malignant phenotype of TNBC and may offer innovative avenues for the discovery of new drug leads for treatment of TNBC and other cancers.
Collapse
Affiliation(s)
- Rola Abdallah
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, and College of Medicine, Qatar University, Doha, Qatar
| | - Adnan Badran
- Department of Nutrition, University of Petra, Amman, Jordan
| | - Serine Baydoun
- Breast Imaging Section, Imaging Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Mansour Sobeh
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Wafae Ouchari
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Nihad Sahri
- Agrobiosciences Program, College for Agriculture and Environmental Science, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
5
|
Shaito AA, Omairi I, Al-Thani N, Seglab F, Ad-Darwish E, Kobeissy F, Nasreddine S. Determination of Medicago orbicularis Antioxidant, Antihemolytic, and Anti-Cancerous Activities and Its Augmentation of Cisplatin-Induced Cytotoxicity in A549 Lung Cancer Cells. PLANTS (BASEL, SWITZERLAND) 2024; 13:442. [PMID: 38337975 PMCID: PMC10857225 DOI: 10.3390/plants13030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 02/12/2024]
Abstract
The anti-lung cancer properties of the plant Medicago orbicularis have not been explored yet. Therefore, we identified its phytochemical composition and investigated the antioxidant, anti-hemolytic, and anti-cancerous properties of extracts of this plant in A549 human lung adenocarcinoma cells. The results show that all parts of M. orbicularis (stems, leaves, and fruits) exhibit remarkable hemolytic activities and modest antioxidant capacity. In addition, all extracts showed a dose-dependent anti-cancerous cytotoxic activity against A549 cells, with fruit extracts being the most potent. This cytotoxic effect could be related, at least partly, to the induction of apoptosis, where M. orbicularis fruit extracts reduced the ratio of anti-apoptotic BCL-2/pro-apoptotic BAX, thereby promoting cellular death. Furthermore, the use of M. orbicularis, in combination with a conventional chemotherapeutic agent, cisplatin, was assessed. Indeed, the combination of cisplatin and M. orbicularis fruit extracts was more cytotoxic and induced more aggregation of A549 cells than either treatment alone. GC-MS analysis and total polyphenol and flavonoid content determination indicated that M. orbicularis is rich in compounds that have anti-cancerous effects. We propose M. orbicularis as a potential source of anti-cancerous agents to manage the progression of lung cancer and its resistance to therapy.
Collapse
Affiliation(s)
- Abdullah A. Shaito
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
- College of Medicine and Department of Biomedical Sciences at College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Islam Omairi
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut 1105, Lebanon
- Biology Department, Faculty of Sciences-Section I, Group of Anti-Cancer Therapeutic Approaches (ATAC), Laboratory Rammal Rammal, Lebanese University, Beirut 1102, Lebanon
| | - Najlaa Al-Thani
- Research and Development Department, Barzan Holdings, Doha P.O. Box 7178, Qatar
| | - Fatiha Seglab
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Esraa Ad-Darwish
- Biotechnology in Forensic Science Program, Faculty of Health Sciences, American University of Science and Technology, Beirut 1100, Lebanon
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), More-House School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310, USA
| | - Salam Nasreddine
- Biology Department, Faculty of Sciences-Section I, Group of Anti-Cancer Therapeutic Approaches (ATAC), Laboratory Rammal Rammal, Lebanese University, Beirut 1102, Lebanon
- Doctoral School of Science and Technology, Research Platform for Environmental Science (PRASE), Lebanese University, Beirut 1102, Lebanon
| |
Collapse
|
6
|
Romanzi A, Milosa F, Marcelli G, Critelli RM, Lasagni S, Gigante I, Dituri F, Schepis F, Cadamuro M, Giannelli G, Fabris L, Villa E. Angiopoietin-2 and the Vascular Endothelial Growth Factor Promote Migration and Invasion in Hepatocellular Carcinoma- and Intrahepatic Cholangiocarcinoma-Derived Spheroids. Biomedicines 2023; 12:87. [PMID: 38255193 PMCID: PMC10813100 DOI: 10.3390/biomedicines12010087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Aggressive hepatocellular carcinoma (HCC) overexpressing Angiopoietin-2 (ANG-2) (a protein linked with angiogenesis, proliferation, and epithelial-mesenchymal transition (EMT)), shares 95% of up-regulated genes and a similar poor prognosis with the proliferative subgroup of intrahepatic cholangiocarcinoma (iCCA). We analyzed the pro-invasive effect of ANG-2 and its regulator vascular endothelial growth factor (VEGF) on HCC and CCA spheroids to uncover posUsible common ways of response. Four cell lines were used: Hep3B and HepG2 (HCC), HuCC-T1 (iCCA), and EGI-1 (extrahepatic CCA). We treated the spheroids with recombinant human (rh) ANG-2 and/or VEGF and then observed the changes at the baseline, after 24 h, and again after 48 h. Proangiogenic stimuli increased migration and invasion capability in HCC- and iCCA-derived spheroids and were associated with a modification in EMT phenotypic markers (a decrease in E-cadherin and an increase in N-cadherin and Vimentin), especially at the migration front. Inhibitors targeting ANG-2 (Trebananib) and the VEGF (Bevacizumab) effectively blocked the migration ability of spheroids that had been stimulated with rh-ANG-2 and rh-VEGF. Overall, our findings highlight the critical role played by ANG-2 and the VEGF in enhancing the ability of HCC- and iCCA-derived spheroids to migrate and invade, which are key processes in cancer progression.
Collapse
Affiliation(s)
- Adriana Romanzi
- Department of Biomedical, Metabolic and Neural Sciences, Clinical and Experimental Medicine Program, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.R.); (S.L.)
- Chimomo Department, Gastroenterology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.M.); (G.M.); (R.M.C.); (F.S.)
| | - Fabiola Milosa
- Chimomo Department, Gastroenterology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.M.); (G.M.); (R.M.C.); (F.S.)
| | - Gemma Marcelli
- Chimomo Department, Gastroenterology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.M.); (G.M.); (R.M.C.); (F.S.)
| | - Rosina Maria Critelli
- Chimomo Department, Gastroenterology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.M.); (G.M.); (R.M.C.); (F.S.)
| | - Simone Lasagni
- Department of Biomedical, Metabolic and Neural Sciences, Clinical and Experimental Medicine Program, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.R.); (S.L.)
- Chimomo Department, Gastroenterology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.M.); (G.M.); (R.M.C.); (F.S.)
| | - Isabella Gigante
- National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, 70013 Castellana Grotte, Italy; (I.G.); (F.D.); (G.G.)
| | - Francesco Dituri
- National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, 70013 Castellana Grotte, Italy; (I.G.); (F.D.); (G.G.)
| | - Filippo Schepis
- Chimomo Department, Gastroenterology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.M.); (G.M.); (R.M.C.); (F.S.)
| | - Massimiliano Cadamuro
- Department of Molecular Medicine, School of Medicine, University of Padua, 35121 Padua, Italy; (M.C.); (L.F.)
| | - Gianluigi Giannelli
- National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, 70013 Castellana Grotte, Italy; (I.G.); (F.D.); (G.G.)
| | - Luca Fabris
- Department of Molecular Medicine, School of Medicine, University of Padua, 35121 Padua, Italy; (M.C.); (L.F.)
| | - Erica Villa
- Chimomo Department, Gastroenterology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.M.); (G.M.); (R.M.C.); (F.S.)
| |
Collapse
|
7
|
Gershoni A, Barayev E, Daood RH, Yogev M, Gal-Or O, Reitblat O, Tsessler M, Schaap Fogler M, Tuuminen R, Ehrlich R. Anatomical and Functional Outcomes with Prompt versus Delayed Initiation of Anti-VEGF in Exudative Age-Related Macular Degeneration. J Clin Med 2023; 13:111. [PMID: 38202118 PMCID: PMC10779608 DOI: 10.3390/jcm13010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
PURPOSE To investigate the correlation between time from diagnosis of treatment-naïve exudative age-related macular degeneration (AMD) to the introduction of anti-VEGF treatment and anatomical and functional outcomes. DESIGN Retrospective cohort study. METHODS Included were treatment-naïve exudative AMD patients who presented to a single tertiary medical center between 2012 and 2018. All patients were treated within the first 30 days of their diagnosis with three monthly intravitreal injections of bevacizumab. Patients were divided into three groups: group 1 (prompt anti-VEGF) were injected with bevacizumab within ten days, group 2 (intermediate anti-VEGF) within 11-20 days, and group 3 (delayed anti-VEGF) within 21-30 days from diagnosis. Baseline characteristics and clinical outcomes were compared up to two years from treatment. RESULTS 146 eyes of 146 patients were included. Sixty-eight patients were in the prompt anti-VEGF group, 31 in the intermediate anti-VEGF group, and 47 in the delayed anti-VEGF group. Following the induction phase of three intravitreal bevacizumab injections, the mean central subfield macular thickness (328.0 ± 115.4 µm vs. 364.6 ± 127.2 µm vs. 337.7 ± 150.1 µm, p = 0.432) and the best-corrected visual acuity (0.47 ± 0.38 vs. 0.59 ± 0.48 vs. 0.47 ± 0.44 logMAR units, p = 0.458) were comparable between the prompt, intermediate and delayed anti-VEGF groups. Anatomical and functional outcomes, treatment burden, number of relapses and eyes with second-line anti-VEGF therapy were comparable between the groups at both 1-year and 2-year timepoints. CONCLUSIONS Our real-world evidence data emphasize that even if anti-VEGF induction cannot be initiated promptly within ten days from diagnosis of naïve exudative AMD, the visual and anatomical prognosis of the patients may not worsen if the treatment is started within one month of diagnosis.
Collapse
Affiliation(s)
- Assaf Gershoni
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Edward Barayev
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Rabeea H. Daood
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
| | - Maureen Yogev
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
| | - Orly Gal-Or
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Olga Reitblat
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Maria Tsessler
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
| | - Michal Schaap Fogler
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Raimo Tuuminen
- Helsinki Retina Research Group, University of Helsinki, 00170 Helsinki, Finland
- Department of Ophthalmology, Kymenlaakso Central Hospital, Kotkantie 41, 48210 Kotka, Finland
| | - Rita Ehrlich
- Ophthalmology Division, Rabin Medical Center, Petach Tikva 49414, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
8
|
Ünal Ç, Sağlam S. Metronomic Temozolomide (mTMZ) and Bevacizumab-The Safe and Effective Frontier for Treating Metastatic Neuroendocrine Tumors (NETs): A Single-Center Experience. Cancers (Basel) 2023; 15:5688. [PMID: 38067391 PMCID: PMC10705735 DOI: 10.3390/cancers15235688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/18/2023] [Accepted: 11/29/2023] [Indexed: 10/16/2024] Open
Abstract
Addressing the persistent challenges in treating metastatic neuroendocrine tumors (NETs) demands ongoing refinement and innovation in therapeutic strategies. This study investigates the potential advantages of combining metronomic temozolomide (mTMZ) with bevacizumab for patients diagnosed with metastatic NETs, particularly focusing on those with a Ki-67 index under 55%. Data from 30 patients were analyzed, using key performance indicators such as progression-free survival (PFS), overall survival (OS), and response rates to therapy, to gauge the treatment's efficacy. The results were encouraging: the median PFS recorded was 16.3 months, and the OS was 25.9 months. The disease control rate (DCR) reached an impressive 86.7%, and the objective response rate (ORR) stood at 63.3%. The treatment regimen was well-tolerated, with no reported instances of grade 4 toxicities. Such a safety profile indicates that this regimen may be particularly advantageous for older, fragile patients who might struggle with conventional dosage levels. These initial findings suggest that the mTMZ and bevacizumab combination could potentially rival the conventional temozolomide-capecitabine therapy in managing metastatic NETs. We aimed to meticulously assess the efficacy of the mTMZ and bevacizumab combination in treating metastatic NETs. Given the initial promising results, a more conclusive understanding of its efficacy will require further research through larger, multicenter prospective clinical trials.
Collapse
Affiliation(s)
- Çağlar Ünal
- Division of Medical Oncology, Department of Internal Medicine, Kartal Dr. Lütfi Kırdar City Hospital, İstanbul 34870, Turkey
| | - Sezer Sağlam
- Division of Medical Oncology, Department of Internal Medicine, Demiroglu Bilim University, İstanbul 34870, Turkey;
| |
Collapse
|
9
|
Cehofski LJ, Kruse A, Mæng MO, Kjaergaard B, Grauslund J, Honoré B, Vorum H. Proteome Analysis of Bevacizumab Intervention in Experimental Central Retinal Vein Occlusion. J Pers Med 2023; 13:1580. [PMID: 38003895 PMCID: PMC10672637 DOI: 10.3390/jpm13111580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/17/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Bevacizumab is a frequently used inhibitor of vascular endothelial growth factor (VEGF) in the management of macular edema in central retinal vein occlusion (CRVO). Studying retinal protein changes in bevacizumab intervention may provide insights into mechanisms of action. In nine Danish Landrace pigs, experimental CRVO was induced in both eyes with argon laser. The right eyes received an intravitreal injection of 0.05 mL bevacizumab (n = 9), while the left control eyes received 0.05 mL saline water (NaCl). Retinal samples were collected 15 days after induced CRVO. Label-free quantification nano-liquid chromatography-tandem mass spectrometry identified 59 proteins that were regulated following bevacizumab treatment. Following bevacizumab intervention, altered levels of bevacizumab components, including the Ig gamma-1 chain C region and the Ig kappa chain C region, were observed. Changes in other significantly regulated proteins ranged between 0.58-1.73, including for the NADH-ubiquinone oxidoreductase chain (fold change = 1.73), protein-transport protein Sec24B (fold change = 1.71), glycerol kinase (fold change = 1.61), guanine-nucleotide-binding protein G(T) subunit-gamma-T1 (fold change = 0.67), and prefoldin subunit 6 (fold change = 0.58). A high retinal concentration of bevacizumab was achieved within 15 days. Changes in the additional proteins were limited, suggesting a narrow mechanism of action.
Collapse
Affiliation(s)
- Lasse Jørgensen Cehofski
- Department of Ophthalmology, Odense University Hospital, 5000 Odense, Denmark;
- Biomedical Research Laboratory, Aalborg University Hospital, 9000 Aalborg, Denmark;
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Anders Kruse
- Department of Ophthalmology, Aalborg University Hospital, 9000 Aalborg, Denmark; (A.K.); (M.O.M.)
| | - Mads Odgaard Mæng
- Department of Ophthalmology, Aalborg University Hospital, 9000 Aalborg, Denmark; (A.K.); (M.O.M.)
| | - Benedict Kjaergaard
- Biomedical Research Laboratory, Aalborg University Hospital, 9000 Aalborg, Denmark;
| | - Jakob Grauslund
- Department of Ophthalmology, Odense University Hospital, 5000 Odense, Denmark;
| | - Bent Honoré
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark;
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| | - Henrik Vorum
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark;
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| |
Collapse
|
10
|
Huang X, Huang Y, Li P. How do serum lipid levels change and influence progression-free survival in epithelial ovarian cancer patients receiving bevacizumab treatment? Front Oncol 2023; 13:1168996. [PMID: 37064140 PMCID: PMC10090393 DOI: 10.3389/fonc.2023.1168996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundThis study aimed to investigate how serum lipid levels affect epithelial ovarian cancer (EOC) patients receiving bevacizumab treatment and to develop a model for predicting the patients’ prognosis.MethodsA total of 139 EOC patients receiving bevacizumab treatment were involved in this study. Statistical analysis was used to compare the median and average values of serum lipid level variables between the baseline and final follow-up. Additionally, a method based on machine learning was proposed to identify independent risk factors for estimating progression-free survival (PFS) in EOC patients receiving bevacizumab treatment. A PFS nomogram dividing the patients into low- and high-risk categories was created based on these independent prognostic variables. Finally, Kaplan–Meier curves and log-rank tests were utilized to perform survival analysis.ResultsAmong EOC patients involved in this study, statistical analysis of serum lipid level variables revealed a substantial increase in total cholesterol, triglycerides, apolipoprotein A1, and free fatty acids, and a significant decrease in apolipoprotein B from baseline to final follow-up. Our method identified FIGO stage, combined chemotherapy regimen, activated partial thromboplastin time, globulin, direct bilirubin, free fatty acids, blood urea nitrogen, high-density lipoprotein cholesterol, and triglycerides as risk factors. These risk factors were then included in our nomogram as independent predictors for EOC patients. PFS was substantially different between the low-risk group (total score < 298) and the high-risk group (total score ≥ 298) according to Kaplan–Meier curves (P < 0.05).ConclusionSerum lipid levels changed variously in EOC patients receiving bevacizumab treatment. A prediction model for PFS of EOC patients receiving bevacizumab treatment was constructed, and it can be beneficial in determining the prognosis, selecting a treatment plan, and monitoring these patients’ long-term care.
Collapse
Affiliation(s)
- Xiaoyu Huang
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Yong Huang
- Department of Medical Oncology, The Second People’s Hospital of Hefei, Hefei, China
| | - Ping Li
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Ping Li,
| |
Collapse
|
11
|
El-Hajjar L, Saliba J, Karam M, Shaito A, El Hajj H, El-Sabban M. Ubiquitin-Related Modifier 1 (URM-1) Modulates Cx43 in Breast Cancer Cell Lines. Int J Mol Sci 2023; 24:ijms24032958. [PMID: 36769280 PMCID: PMC9917400 DOI: 10.3390/ijms24032958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 02/05/2023] Open
Abstract
Gap-junction-forming connexins are exquisitely regulated by post-translational modifications (PTMs). In particular, the PTM of connexin 43 (Cx43), a tumor suppressor protein, regulates its turnover and activity. Here, we investigated the interaction of Cx43 with the ubiquitin-related modifier 1 (URM-1) protein and its impact on tumor progression in two breast cancer cell lines, highly metastatic triple-negative MDA-MB-231 and luminal breast cancer MCF-7 cell lines. To evaluate the subsequent modulation of Cx43 levels, URM-1 was downregulated in these cells. The transcriptional levels of epithelial-to-mesenchymal transition (EMT) markers and the metastatic phenotype were assessed. We demonstrated that Cx43 co-localizes and interacts with URM-1, and URMylated Cx43 was accentuated upon cellular stress. The significant upregulation of small ubiquitin-like modifier-1 (SUMO-1) was also observed. In cells with downregulated URM-1, Cx43 expression significantly decreased, and SUMOylation by SUMO-1 was affected. The concomitant expression of EMT markers increased, leading to increased proliferation, migration, and invasion potential. Inversely, the upregulation of URM-1 increased Cx43 expression and reversed EMT-induced processes, underpinning a role for this PTM in the observed phenotypes. This study proposes that the URMylation of Cx43 in breast cancer cells regulates its tumor suppression properties and contributes to breast cancer cell malignancy.
Collapse
Affiliation(s)
- Layal El-Hajjar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Jessica Saliba
- Department of Biology, Faculty of Sciences, Lebanese University, Beirut P.O. Box 90656, Lebanon
- Department of Public Health, Faculty of Health Sciences, University of Balamand, Beirut P.O. Box 100, Lebanon
| | - Mario Karam
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Abdullah Shaito
- Biomedical Research Center, Qatar University Doha, Doha P.O. Box 2713, Qatar
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Correspondence: ; Tel.: +961-(1)-350000 (ext. 4765)
| |
Collapse
|
12
|
Quiros-Gonzalez I, Tomaszewski MR, Golinska MA, Brown E, Ansel-Bollepalli L, Hacker L, Couturier DL, Sainz RM, Bohndiek SE. Photoacoustic Tomography Detects Response and Resistance to Bevacizumab in Breast Cancer Mouse Models. Cancer Res 2022; 82:1658-1668. [PMID: 35404400 PMCID: PMC9359720 DOI: 10.1158/0008-5472.can-21-0626] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 08/27/2021] [Accepted: 02/18/2022] [Indexed: 01/07/2023]
Abstract
Angiogenesis is an established prognostic factor in advanced breast cancer, yet response to antiangiogenic therapies in this disease remains highly variable. Noninvasive imaging biomarkers could help identify patients that will benefit from antiangiogenic therapy and provide an ideal tool for longitudinal monitoring, enabling dosing regimens to be altered with real-time feedback. Photoacoustic tomography (PAT) is an emerging imaging modality that provides a direct readout of tumor hemoglobin concentration and oxygenation. We hypothesized that PAT could be used in the longitudinal setting to provide an early indication of response or resistance to antiangiogenic therapy. To test this hypothesis, PAT was performed over time in estrogen receptor-positive and estrogen receptor-negative breast cancer xenograft mouse models undergoing treatment with the antiangiogenic bevacizumab as a single agent. The cohort of treated tumors, which were mostly resistant to the treatment, contained a subset that demonstrated a clear survival benefit. At endpoint, the PAT data from the responding subset showed significantly lower oxygenation and higher hemoglobin content compared with both resistant and control tumors. Longitudinal analysis revealed that tumor oxygenation diverged significantly in the responding subset, identifying early treatment response and the evolution of different vascular phenotypes between the subsets. Responding tumors were characterized by a more angiogenic phenotype when analyzed with IHC, displaying higher vessel density, yet poorer vascular maturity and elevated hypoxia. Taken together, our findings indicate that PAT shows promise in providing an early indication of response or resistance to antiangiogenic therapy. SIGNIFICANCE Photoacoustic assessment of tumor oxygenation is a noninvasive early indicator of response to bevacizumab therapy, clearly distinguishing between control, responding, and resistant tumors within just a few weeks of treatment.
Collapse
Affiliation(s)
- Isabel Quiros-Gonzalez
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Michal R. Tomaszewski
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Monika A. Golinska
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Emma Brown
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Laura Ansel-Bollepalli
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Lina Hacker
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Dominique-Laurent Couturier
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Rosa M. Sainz
- Cell Morphology and Biology Department, IUOPA and ISPA, Universidad de Oviedo, Oviedo, Spain
| | - Sarah E. Bohndiek
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| |
Collapse
|
13
|
Oikawa M, Yaegashi D, Yokokawa T, Misaka T, Sato T, Kaneshiro T, Kobayashi A, Yoshihisa A, Nakazato K, Ishida T, Takeishi Y. D-Dimer Is a Predictive Factor of Cancer Therapeutics-Related Cardiac Dysfunction in Patients Treated With Cardiotoxic Chemotherapy. Front Cardiovasc Med 2022; 8:807754. [PMID: 35127869 PMCID: PMC8813859 DOI: 10.3389/fcvm.2021.807754] [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: 11/02/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022] Open
Abstract
Background D-dimer is a sensitive biomarker for cancer-associated thrombosis, but little is known about its significance on cancer therapeutics-related cardiac dysfunction (CTRCD). Methods Consecutive 169 patients planned for cardiotoxic chemotherapy were enrolled and followed up for 12 months. All patients underwent echocardiography and blood test at baseline and at 3-, 6-, and 12 months. Results The patients were divided into two groups based on the level of D-dimer (>1.65 μg/ml or ≦ 1.65 μg/ml) at baseline before chemotherapy: high D-dimer group (n = 37) and low D-dimer group (n = 132). Left ventricular ejection fraction (LVEF) decreased at 3- and 6 months after chemotherapy in high D-dimer group [baseline, 65.2% (62.8–71.4%); 3 months, 62.9% (59.0–67.7%); 6 months, 63.1% (60.0–67.1%); 12 months, 63.3% (58.8–66.0%), p = 0.03], but no change was observed in low D-dimer group. The occurrence of CTRCD within the 12-month follow-up period was higher in the high D-dimer group than in the low D-dimer group (16.2 vs. 4.5%, p = 0.0146). Multivariable logistic regression analysis revealed that high D-dimer level at baseline was an independent predictor of the development of CTRCD [odds ratio 3.93, 95% CI (1.00–15.82), p = 0.047]. Conclusion We should pay more attention to elevated D-dimer levels not only as a sign of cancer-associated thrombosis but also the future occurrence of CTRCD.
Collapse
|
14
|
Widodo SS, Hutchinson RA, Fang Y, Mangiola S, Neeson PJ, Darcy PK, Barrow AD, Hovens CM, Dinevska M, Stylli SS, Mantamadiotis T. Toward precision immunotherapy using multiplex immunohistochemistry and in silico methods to define the tumor immune microenvironment. Cancer Immunol Immunother 2021; 70:1811-1820. [PMID: 33389014 PMCID: PMC10991574 DOI: 10.1007/s00262-020-02801-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022]
Abstract
Recent developments in cancer immunotherapy promise better outcomes for cancer patients, although clinical trials for difficult to treat cancers such as malignant brain cancer present special challenges, showing little response to first generation immunotherapies. Reasons for differences in immunotherapy response in some cancer types are likely due to the nature of tumor microenvironment, which harbors multiple cell types which interact with tumor cells to establish immunosuppression. The cell types which appear to hold the key in regulating tumor immunosuppression are the tumor-infiltrating immune cells. The current standard treatment for difficult to treat cancer, including the most malignant brain cancer, glioblastoma, continues to offer a bleak outlook for patients. Immune-profiling and correlation with pathological and clinical data will lead to a deeper understanding of the tumor immune microenvironment and contribute toward the selection, optimization and development of novel precision immunotherapies. Here, we review the current understanding of the tumor microenvironmental landscape in glioblastoma with a focus on next-generation technologies including multiplex immunofluorescence and computational approaches to map the brain tumor microenvironment to decipher the role of the immune system in this lethal malignancy.
Collapse
Affiliation(s)
- Samuel S Widodo
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | - Ryan A Hutchinson
- Department of Clinical Pathology, The University of Melbourne, Melbourne, Australia
- Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, Australia
| | - Yitong Fang
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | - Stefano Mangiola
- Bioinformatics Division, The Walter and Eliza Hall Institute, Parkville, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Alexander D Barrow
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | | | - Marija Dinevska
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, Australia
| | - Theo Mantamadiotis
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia.
- Department of Surgery, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
15
|
Tryptanthrin exerts anti-breast cancer effects both in vitro and in vivo through modulating the inflammatory tumor microenvironment. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2021; 71:245-266. [PMID: 33151167 DOI: 10.2478/acph-2021-0020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/21/2020] [Indexed: 01/19/2023]
Abstract
Tryptanthrin is an indole quinazoline alkaloid from the indigo-bearing plants, such as Isatis indigotica Fort. Typically, this natural compound shows a variety of pharmacological activities such as antitumor, antibacterial, anti-inflammatory and antioxidant effects. This study was conducted to assess the antitumor activity of tryptanthrin in breast cancer models both in vitro and in vivo, and to explore the important role of the inflammatory tumor microenvironment (TME) in the antitumor effects of tryptanthrin. Human breast adenocarcinoma MCF-7 cells were used to assess the antitumor effect of tryptanthrin in vitro. MTT assay and colony formation assay were carried out to monitor the antiproliferative effect of tryptanthrin (1.56~50.0 μmol L-1) on inhibiting the proliferation and colony formation of MCF-7 cells, respectively. The migration and invasion of MCF-7 cells were evaluated by wound healing assay and Transwell chamber assay, respectively. Moreover, the 4T1 murine breast cancer model was established to examine the pharmacological activity of tryptanthrin, and three groups with different doses of tryptanthrin (25, 50 and 100 mg kg-1) were set in study. Additionally, tumor volumes and organ coefficients were measured and calculated. After two weeks of tryptanthrin treatment, samples from serum, tumor tissue and different organs from tumor-bearing mice were collected, and the enzyme-linked immunosorbent assay (ELISA) was performed to assess the regulation of inflammatory molecules in mouse serum. Additionally, pathological examinations of tumor tissues and organs from mice were evaluated through hematoxylin and eosin (H&E) staining. The expression of inflammatory proteins in tumor tissues was measured by immunohistochemistry (IHC) and Western blotting. Tryptanthrin inhibited the proliferation, migration and invasion of MCF-7 cells, up-regulated the protein level of E-cadherin, and down-regulated those of MMP-2 and Snail, as suggested by the MCF-7 cell experiment. According to the results from in vivo experiment, tryptanthrin was effective in inhibiting tumor growth, and it showed favorable safety without inducing the fluctuations of body mass and organ coefficient (p > 0.05). In addition, tryptanthrin also suppressed the expression levels of NOS1, COX-2 and NF-κB in mouse tumor tissues, and regulated those of IL-2, IL-10 and TNF-α in the serum of tumor cells-transplanted mice. Tryptanthrin exerted its anti-breast cancer activities through modulating the inflammatory TME both in vitro and in vivo.
Collapse
|
16
|
Kingnate C, Charoenkwan K, Kumfu S, Apaijai N, Jaiwongkam T, Khunamornpong S, Chattipakorn N, Chattipakorn SC. Platinum-based chemotherapy and bevacizumab instigate the destruction of human ovarian cancers via different signaling pathways. Biochem Pharmacol 2021; 188:114587. [PMID: 33932471 DOI: 10.1016/j.bcp.2021.114587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/16/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
The standard chemotherapy regimens of ovarian cancer are platinum-based chemotherapy (carboplatin and paclitaxel) and bevacizumab (BEV). However, the effects of BEV alone or combined with carboplatin and paclitaxel on mitochondrial dynamics, mitochondrial function, mitophagy, apoptosis, inflammation and vascular endothelial growth factor (VEGF) in human ovarian cancer mitochondria and cells have not yet been investigated. Therefore, we aimed to test the hypothesis that 1) platinum-based chemotherapy and BEV equally damage isolated mitochondria from human ovarian cancers, and ovarian cancer cells through inducing mitochondrial dynamics dysregulation, mitochondrial dysfunction, increased mitophagy and apoptosis, as well as altered inflammation and VEGF; and 2) combined therapies exert greater damage than monotherapy. Each isolated human ovarian cancer mitochondria (n = 16) or CaOV3 cells (n = 6) were treated with either platinum-based chemotherapy (carboplatin 10 μM and paclitaxel 5 μM), BEV (2 mg/mL) or combined platinum-based chemotherapy and BEV for 60 min or 24 h, respectively. Following the treatment, mitochondrial dynamics, mitochondrial function, mitophagy, apoptosis, cytotoxicity, inflammation and VEGF were determined. Platinum-based chemotherapy caused ovarian cancer mitochondria and cell damage through mitochondrial dysfunction, increased cell death with impairment of membrane integrity, and enhanced VEGF reduction, while BEV did not. BEV caused deterioration of ovarian cancer mitochondria and cells through mitochondrial-dependent apoptosis, but it had no effect on cell viability. Interestingly, combined platinum-based chemotherapy and BEV treatments had no addictive effects on all parameters except mitochondrial maximal respiration, when compared to monotherapy. Collectively, these findings suggest that platinum-based chemotherapy and BEV caused human ovarian cancer mitochondrial and cell damage through different mechanisms.
Collapse
Affiliation(s)
- Chalita Kingnate
- Department of Obstetrics and Gynecology, Lamphun Hospital, Lamphun 51000, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kittipat Charoenkwan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirinart Kumfu
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thidarat Jaiwongkam
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Surapan Khunamornpong
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
17
|
Li D, Finley SD. Mechanistic insights into the heterogeneous response to anti‐VEGF treatment in tumors. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2021. [DOI: 10.1002/cso2.1013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Ding Li
- Department of Biomedical Engineering University of Southern California Los Angeles California USA
| | - Stacey D. Finley
- Departments of Biomedical Engineering, Quantitative and Computational Biology, and Chemical Engineering and Materials Science University of Southern California Los Angeles California USA
| |
Collapse
|
18
|
Jalaleddine N, El-Hajjar L, Dakik H, Shaito A, Saliba J, Safi R, Zibara K, El-Sabban M. Pannexin1 Is Associated with Enhanced Epithelial-To-Mesenchymal Transition in Human Patient Breast Cancer Tissues and in Breast Cancer Cell Lines. Cancers (Basel) 2019; 11:cancers11121967. [PMID: 31817827 PMCID: PMC6966616 DOI: 10.3390/cancers11121967] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Loss of connexin-mediated cell-cell communication is a hallmark of breast cancer progression. Pannexin1 (PANX1), a glycoprotein that shares structural and functional features with connexins and engages in cell communication with its environment, is highly expressed in breast cancer metastatic foci; however, PANX1 contribution to metastatic progression is still obscure. Here we report elevated expression of PANX1 in different breast cancer (BRCA) subtypes using RNA-seq data from The Cancer Genome Atlas (TCGA). The elevated PANX1 expression correlated with poorer outcomes in TCGA BRCA patients. In addition, gene set enrichment analysis (GSEA) revealed that epithelial-to-mesenchymal transition (EMT) pathway genes correlated positively with PANX1 expression. Pharmacological inhibition of PANX1, in MDA-MB-231 and MCF-7 breast cancer cells, or genetic ablation of PANX1, in MDA-MB-231 cells, reverted the EMT phenotype, as evidenced by decreased expression of EMT markers. In addition, PANX1 inhibition or genetic ablation decreased the invasiveness of MDA-MB-231 cells. Our results suggest PANX1 overexpression in breast cancer is associated with a shift towards an EMT phenotype, in silico and in vitro, attributing to it a tumor-promoting effect, with poorer clinical outcomes in breast cancer patients. This association offers a novel target for breast cancer therapy.
Collapse
Affiliation(s)
- Nour Jalaleddine
- Department of Biological and Environmental Sciences, Faculty of Science, Beirut Arab University, Beirut 1107-2809, Lebanon;
| | - Layal El-Hajjar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon;
| | - Hassan Dakik
- University of Tours, EA 7501 GICC, CNRS ERL 7001 LNOx, CEDEX 01, 37032 Tours, France;
| | - Abdullah Shaito
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut 1105, Lebanon;
| | - Jessica Saliba
- Department of Biology, Faculty of Sciences, Lebanese University, Hadath, Beirut 1003, Lebanon;
| | - Rémi Safi
- Department of Dermatology, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon;
| | - Kazem Zibara
- ER045-Laboratory of Stem Cells, PRASE, Department of Biology, Faculty of Sciences, Lebanese University, Hadath, Beirut 1003, Lebanon;
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon;
- Correspondence: ; Tel.: +961-1-350000 (ext. 4765-4766)
| |
Collapse
|
19
|
Uddin MA, Barabutis N. P53: The endothelium defender. J Cell Biochem 2019; 120:10952-10955. [PMID: 30816605 PMCID: PMC6713618 DOI: 10.1002/jcb.28511] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/07/2019] [Indexed: 01/25/2023]
Abstract
P53 represents the paradigm of a multitalented transcription factor, responsible for the cellular defense against a plethora of potentially harmful stimuli. It exercises the ability to strongly oppose both cancer and inflammation, partially due to the fact that both conditions are highly interrelated. Endothelium hyperpermeability is considered the hallmark of severe lung inflammation, and the cardinal feature of the lethal acute respiratory distress syndrome. An emerging body of evidence suggests a strategic role of P53 towards vascular barrier integrity. The "endothelium defender" orchestrates meticulously devised responses; to counteract toxin-induced destructions of endothelium monolayers. The present effort seeks to further our understanding on the expanding P53 universe, discussing the most recent information regarding the involvement of that molecule in the pulmonary function.
Collapse
Affiliation(s)
- Mohammad Afaz Uddin
- School of Basic Pharmaceutical and Toxicological Sciences,
College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences,
College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA.,To whom correspondence should be addressed at:
Nektarios Barabutis, M.Sc., Ph.D., School of Basic Pharmaceutical and
Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe,
Monroe, LA 71201, United States of America, ,
Phone: (318) 342 −1460
| |
Collapse
|
20
|
El-Hajjar L, Shaito A, Jalaleddine N, Zibara K, Kazan JM, El-Saghir J, El-Sabban M. Data on migration of the non-invasive breast cancer cell line, MCF-7 treated with Bevacizumab using Real Time Cell Analyzer (RTCA). Data Brief 2019; 22:635-638. [PMID: 30671510 PMCID: PMC6327074 DOI: 10.1016/j.dib.2018.12.059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/16/2018] [Accepted: 12/17/2018] [Indexed: 11/16/2022] Open
Abstract
Bevacizumab or Avastin® (Av), the recombinant antibody targeting VEGF, improves progression-free but not overall survival of metastatic breast cancer patients due to development of Av resistance. We showed that Av-therapy-induced inflammatory microenvironment contributes to the refractoriness to Av treatment. Here we present data regarding the effect of Av treatment on migration of a non-invasive breast cancer cell line, MCF-7. The data presented hereis related to the research article “Bevacizumab induces inflammation in MDA-MB-231 breast cancer cell line and in a mouse model” (Hajjar et al., 2018).
Collapse
Affiliation(s)
- Layal El-Hajjar
- Department of Biological and Environmental Sciences, Faculty of Science, Beirut Arab University, Beirut, Lebanon
| | - Abdullah Shaito
- Department of Biological and Environmental Sciences, Faculty of Science, Beirut Arab University, Beirut, Lebanon
| | - Nour Jalaleddine
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | - Kazem Zibara
- ER045 - Laboratory of Stem Cells, PRASE, Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Jalal M Kazan
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jamal El-Saghir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|