1
|
Al-Azzawi HMA, Hamza SA, Paolini R, Arshad F, Patini R, O'Reilly L, McCullough M, Celentano A. Towards an emerging role for anticoagulants in cancer therapy: a systematic review and meta-analysis. FRONTIERS IN ORAL HEALTH 2024; 5:1495942. [PMID: 39568788 PMCID: PMC11576436 DOI: 10.3389/froh.2024.1495942] [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: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 11/22/2024] Open
Abstract
Background Anticoagulants, renowned for their role in preventing blood clot formation, have captivated researchers' attention for the exploitation of their potential to inhibit cancer in pre-clinical models. Objectives To undertake a systematic review and meta-analysis of the effects of anticoagulants in murine cancer research models. Further, to present a reference tool for anticoagulant therapeutic modalities relating to future animal pre-clinical models of cancer and their translation into the clinic. Methods Four databases were utilized including Medline (Ovid), Embase (Ovid), Web of science, and Scopus databases. We included studies relating to any cancer conducted in murine models that assessed the effect of traditional anticoagulants (heparin and its derivatives and warfarin) and newer oral anticoagulants on cancer. Results A total of 6,158 articles were identified in an initial multi-database search. A total of 157 records were finally included for data extraction. Studies on heparin species and warfarin demonstrated statistically significant results in favour of tumour growth and metastasis inhibition. Conclusion Our findings constitute a valuable reference guide for the application of anticoagulants in cancer research and explore the promising utilization of non-anticoagulants heparin in preclinical cancer research. Systematic Review Registration PROSPERO [CRD42024555603].
Collapse
Affiliation(s)
| | - Syed Ameer Hamza
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Fizza Arshad
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Romeo Patini
- Head and Neck Department, "Fondazione Policlinico Universitario A. Gemelli-IRCCS" School of Dentistry, Catholic University of Sacred Heart-Rome Largo A. Gemelli, Rome, Italy
| | - Lorraine O'Reilly
- Clinical Translation Centre, Cancer Biology and Stem Cells Division and Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Michael McCullough
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Antonio Celentano
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| |
Collapse
|
2
|
Qadir A, Samad DA, Asif M, Ali MM, Zain S. Investigating the effect of vandetanib and celecoxib combination on angiogenesis. J Taibah Univ Med Sci 2023; 18:1011-1017. [PMID: 36959917 PMCID: PMC10027553 DOI: 10.1016/j.jtumed.2023.02.016] [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: 08/11/2022] [Revised: 01/12/2023] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
Objective Angiogenesis plays an important role in various physiological and pathological conditions and is essential for tumor growth and metastasis. The aim of this study was to evaluate the effect of a combination of vandetanib and celecoxib on angiogenic tube formation and its effect on angiogenic genes (MMP-2 and MMP-9) using an in vitro model of human umbilical vein endothelial cells (HUVECs). Methods HUVECs were cultured and verified by flow cytometry. HUVECs were then treated with vandetanib, celecoxib, and the combination of both drugs. Then, we investigated cell viability and cell apoptosis by MTT assays and flow cytometry. The process of angiogenesis was analyzed by tube formation assays, and the effect on angiogenic genes was determined by RT-qPCR. Results HUVECs were positive for CD144 and negative for CD14. Vandetanib, celecoxib, and their combination inhibited HUVEC viability in a dose-dependent manner (p < 0.001). The rate of apoptosis was 13.1%, 9%, and 23.7% (p < 0.001) when treated with vandetanib, celecoxib, or the combination of both drugs, respectively. Vandetanib inhibited tube formation by 43.7%, celecoxib by 21%, and their combination by 77.3% (p < 0.001), respectively. RT-qPCR revealed that both vandetanib and celecoxib reduced the expression levels of MMP-2 and MMP-9, and their combination resulted in an even greater extent of reduction in expression levels (p < 0.001). Conclusion Celecoxib enhanced the effect of vandetanib in inhibiting in vitro angiogenesis and the combination of these two drugs led to even greater extents of inhibition than vandetanib alone.
Collapse
Affiliation(s)
- Abdul Qadir
- Department of Pharmacology, United Medical and Dental College, Karachi, Pakistan
- Corresponding address: United Medical and Dental College, Department of Pharmacology, Sector 48H Korangi Creek, Karachi, Sindh 75190, Pakistan.
| | | | - Mahayrookh Asif
- Department of Pharmacology, Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Muhammad Mujtaba Ali
- Department of Pharmacology, United Medical and Dental College, Karachi, Pakistan
| | - Syeda Zain
- Department of Pharmacology, United Medical and Dental College, Karachi, Pakistan
| |
Collapse
|
3
|
LMWH and its derivatives represent new rational for cancer therapy: construction strategies and combination therapy. Drug Discov Today 2019; 24:2096-2104. [PMID: 31228613 DOI: 10.1016/j.drudis.2019.06.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/07/2019] [Accepted: 06/17/2019] [Indexed: 02/08/2023]
Abstract
Low-molecular-weight heparin (LMWH) has attracted increasing attention as a tumor treatment because of its board range of physiological functions. Over the past decade, diverse LMWH derivatives have increased the variety of antitumor strategies available, serving not only as anti-tumor agents, but also as drug delivery platforms. In this review, we introduce the basic strategy for structural modification of LMWH to attenuate its antitumor activity while reducing its risk of bleeding and immune responses, as well as highlighting current applications of LMWH and its derivatives in cancer therapy. We select representative drug delivery systems involving LMWH derivatives and discuss the construction principles and therapeutic effects associated with their use. We also analyze progress made in the development of antitumor combination therapies, in which LMWH has shown synergistic or combined effects with other treatment strategies.
Collapse
|
4
|
Ma S, Pradeep S, Hu W, Zhang D, Coleman R, Sood A. The role of tumor microenvironment in resistance to anti-angiogenic therapy. F1000Res 2018; 7:326. [PMID: 29560266 PMCID: PMC5854986 DOI: 10.12688/f1000research.11771.1] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/08/2018] [Indexed: 12/11/2022] Open
Abstract
Anti-angiogenic therapy has been demonstrated to increase progression-free survival in patients with many different solid cancers. Unfortunately, the benefit in overall survival is modest and the rapid emergence of drug resistance is a significant clinical problem. Over the last decade, several mechanisms have been identified to decipher the emergence of resistance. There is a multitude of changes within the tumor microenvironment (TME) in response to anti-angiogenic therapy that offers new therapeutic opportunities. In this review, we compile results from contemporary studies related to adaptive changes in the TME in the development of resistance to anti-angiogenic therapy. These include preclinical models of emerging resistance, dynamic changes in hypoxia signaling and stromal cells during treatment, and novel strategies to overcome resistance by targeting the TME.
Collapse
Affiliation(s)
- Shaolin Ma
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Reproductive Medicine Research Center, Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong province, China
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dikai Zhang
- Reproductive Medicine Research Center, Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong province, China
| | - Robert Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
5
|
Wang X, Jiang C, Qin Y, Peng Y, Wen G, Liang A, Jiang Z. SERS spectral study of HAuCl 4-cysteine nanocatalytic reaction and its application for detection of heparin sodium with label-free VB4r molecular probe. Sci Rep 2017; 7:45979. [PMID: 28378828 PMCID: PMC5380991 DOI: 10.1038/srep45979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/07/2017] [Indexed: 12/23/2022] Open
Abstract
In the presence of nanocatalyst, L-cysteine reduce HAuCl4 rapidly to form gold nanoparticles (AuNP), and a quick nanocatalytic preparation procedure was established for Au/AuNP sol with highly active surface enhanced Raman scattering (SERS) effect and good stability. The nanoreaction was also studied by absorption, resonance Rayleigh scattering (RRS), transmission electron microscopy (TEM) and energy spectra. In the selected conditions, the analyte heparin sodium (HS) could react with victoria blue 4 R (VB4r) to form associated complexes which have very weak SERS effect to make the SERS signals decrease. The SERS signals at 1617 cm−1 reduced linearly with HS concentration increasing. Upon addition of FeCl3, it hydrolyzed to form stable Fe(OH)3 sol platform that carried SERS active Au/AuNPs to enhance the sensitivity. Accordingly, we established a SERS quantitative analysis method in the sol substrate of Fe(OH)3-Au/AuNPs, with a linear range of 0.5–75 ng/mL HS and a detection limit of 0.2 ng/mL. HS in real samples was determined, with a relative standard deviation of 2.65–7.63% and a recovery of 99.3–101%.
Collapse
Affiliation(s)
- Xiaoliang Wang
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection of Ministry Education, Guangxi Key Laboratory of Environmental Pollution Control Theory and Technology, Guangxi Normal University, Guilin 541004, China
| | - Caina Jiang
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection of Ministry Education, Guangxi Key Laboratory of Environmental Pollution Control Theory and Technology, Guangxi Normal University, Guilin 541004, China
| | - Yanna Qin
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection of Ministry Education, Guangxi Key Laboratory of Environmental Pollution Control Theory and Technology, Guangxi Normal University, Guilin 541004, China
| | - Yutao Peng
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection of Ministry Education, Guangxi Key Laboratory of Environmental Pollution Control Theory and Technology, Guangxi Normal University, Guilin 541004, China
| | - Guiqing Wen
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection of Ministry Education, Guangxi Key Laboratory of Environmental Pollution Control Theory and Technology, Guangxi Normal University, Guilin 541004, China
| | - Aihui Liang
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection of Ministry Education, Guangxi Key Laboratory of Environmental Pollution Control Theory and Technology, Guangxi Normal University, Guilin 541004, China
| | - Zhiliang Jiang
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection of Ministry Education, Guangxi Key Laboratory of Environmental Pollution Control Theory and Technology, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
6
|
Kaya TT, Altun A, Turgut NH, Ataseven H, Koyluoglu G. Effects of a Multikinase Inhibitor Motesanib (AMG 706) Alone and Combined with the Selective DuP-697 COX-2 Inhibitor on Colorectal Cancer Cells. Asian Pac J Cancer Prev 2017; 17:1103-10. [PMID: 27039732 DOI: 10.7314/apjcp.2016.17.3.1103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In the present study, we investigated the effects of motesanib (AMG 706), a multikinase inhibitor alone and in combination with DuP-697, an irreversible selective inhibitor of COX-2, on cell proliferation, angiogenesis, and apoptosis induction in a human colorectal cancer cell line (HT29). Real time cell analysis (RTCA, Xcelligence system) was used to determine the effects on colorectal cancer cell proliferation. Apoptosis was assessed with annexin V staining and angiogenesis was determined with chorioallantoic membrane model. We found that motesanib alone exerted antiproliferative, antiangiogenic and apoptotic effects on HT29 colorectal cancer cells. Combination with DUP-697 increased the antiproliferative, antiangiogenic and apoptotic effects. Results of this study indicate that motesanib may be a good choice in treatment of colorectal tumors. In addition, the increased effects of combination of motesanib with DuP-697 raise the possibility of using lower doses of these drugs and therefore avoid/minimize the dose-dependent side effects generally observed.
Collapse
Affiliation(s)
- Tijen Temiz Kaya
- Department of Pharmacology, Faculty of Pharmacy, Cumhuriyet University, Sivas, Turkey E-mail :
| | | | | | | | | |
Collapse
|
7
|
Zhen Y, Zhang W, Liu C, He J, Lu Y, Guo R, Feng J, Zhang Y, Chen J. Exogenous hydrogen sulfide promotes C6 glioma cell growth through activation of the p38 MAPK/ERK1/2-COX-2 pathways. Oncol Rep 2015; 34:2413-22. [PMID: 26351820 DOI: 10.3892/or.2015.4248] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/07/2015] [Indexed: 11/06/2022] Open
Abstract
Hydrogen sulfide (H2S) participates in multifarious physiological and pathophysiologic progresses of cancer both in vitro and in vivo. We have previously demonstrated that exogenous H2S promoted liver cancer cells proliferation/anti‑apoptosis/angiogenesis/migration effects via amplifying the activation of NF-κB pathway. However, the effects of H2S on cancer cell proliferation and apoptosis are controversial and remain unclear in C6 glioma cells. The present study investigated the effects of exogenous H2S on cancer cells growth via activating p38 MAPK/ERK1/2-COX-2 pathways in C6 glioma cells. C6 glioma cells were treated with 400 µmol/l NaHS (a donor of H2S) for 24 h. The expression levels of phosphorylated (p)-p38 MAPK, total (t)-p38 MAPK, p-ERK1/2, t-ERK1/2, cyclooxygenase-2 (COX-2) and caspase-3 were measured by western blotting assay. Cell viability was detected by Cell Counting Kit-8 (CCK-8). Apoptotic cells were observed by Hoechst 33258 staining assay. Cell proliferation was directly detected under fully automatic inverted microscope. Exposure of C6 glioma cells to NaHS resulted in cell proliferation, as evidenced by an increase in cell viability. In addition, NaHS treatment reduced apoptosis, as indicated by the decreased apoptotic percentage and the cleaved caspase-3 expression. Importantly, exposure of the cells to NaHS increased the expression levels of p-p38 MAPK, p-ERK1/2 and COX-2. Notably, co-treatment of C6 glioma cells with 400 µmol/l NaHS and AOAA (an inhibitor of CBS) largely suppressed the above NaHS-induced effects. Combined treatment with NaHS and SB203580 (an inhibitor of p38 MAPK) or PD-98059 (an inhibitor of ERK1/2) resulted in the synergistic reduction of COX-2 expression and increase of caspase-3 expression, a decreased number of apoptotic cells, along with decreased cell viability. Combined treatment with NS-398 (an inhibitor of COX-2) and NaHS also resulted in the synergistic increase of caspase-3, a decreased in the number of apoptotic cells and the decrease in cell viability. The findings of the present study provide novel evidence that p38 MAPK/ERK1/2-COX-2 pathways are involved in NaHS-induced cancer cell proliferation and anti-apoptosis in C6 glioma cells.
Collapse
Affiliation(s)
- Yulan Zhen
- Oncology Center, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Zhang
- Department of Cardiovasology and Cardiac Care Unit (CCU), Huangpu Division of The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chujie Liu
- Department of Neurology, Dalang Hospital, Dongguan, Guangdong 523700, P.R. China
| | - Jing He
- The First People's Hospital of Yueyang, Yueyang, Hunan 414000, P.R. China
| | - Yun Lu
- Department of Infectious Disease I, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Ruixian Guo
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Jianqiang Feng
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Ying Zhang
- Oncology Center, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Jingfu Chen
- Department of Cardiovasology and Cardiac Care Unit (CCU), Huangpu Division of The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|