1
|
Sai Manogna K, Deva Prasad Raju B, Rajasekhara Reddy G, Kallem P, Shaik MI, John Sushma N. Investigations on anticancer activity of Eu 3+ doped hydroxyapatite nanocomposites against MCF7 and 4T1 breast cancer cell lines: A structural and luminescence Perspective. Heliyon 2024; 10:e25064. [PMID: 38352738 PMCID: PMC10862524 DOI: 10.1016/j.heliyon.2024.e25064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/31/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024] Open
Abstract
Breast cancer remains a significant global health concern, necessitating the development of novel therapeutic approaches. In this study, we investigate the role of Eu3+ doped hydroxyapatite nanocomposites (Han: Eu3+) in the treatment of MCF7 and 4T1 breast cancer cell lines. Furthermore, we explored the structural and luminescent properties of these nanocomposites. Han: Eu3+ were synthesized using a modified co-precipitation method, and their morphology and crystal structure were characterized using scanning electron microscopy (SEM) and X-ray diffraction (XRD) in which the average crystalline size of Han: Eu3+ was found to be 25 nm, rendering them suitable for cellular uptake and targeted therapy. To gain insights into the luminescent properties of Han: Eu3+, their excitation and emission spectra were recorded using photoluminescence spectrometer. The characteristic red emission of Eu3+ ions was observed upon excitation, validating the successful doping of Eu3+ into the Han lattice, which was confirmed by the CIE chromaticity coordinate study. These luminescent properties of Han: Eu3+ hold promise for potential applications in bioimaging. To evaluate the efficacy of Han: Eu3+ in breast cancer treatment, MCF7 and 4T1 cell lines were exposed to varying concentrations of the nanocomposites. Cell viability assays revealed a concentration-dependent reduction in cell viability, indicating the potential anticancer activity of Han: Eu3+. The findings of this study contribute to the expanding field of nanomedicine, bringing targeted breast cancer treatments and us closer to more effective.
Collapse
Affiliation(s)
- K Sai Manogna
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam, (Women's University) Tirupati-517 502, India
| | - B Deva Prasad Raju
- Department of Physics, Sri Venkateswara University, Tirupati - 517501, India
| | - G Rajasekhara Reddy
- School of Mechanical Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Parashuram Kallem
- Environmental Health and Safety Program, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Mannur Ismail Shaik
- Faculty of Fisheries and Food Science, University Malaysia Terengganu, KulaNerus-21030, Terengganu, Malaysia
| | - N John Sushma
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam, (Women's University) Tirupati-517 502, India
| |
Collapse
|
2
|
Malik JA, Ahmed S, Jan B, Bender O, Al Hagbani T, Alqarni A, Anwar S. Drugs repurposed: An advanced step towards the treatment of breast cancer and associated challenges. Biomed Pharmacother 2021; 145:112375. [PMID: 34863612 DOI: 10.1016/j.biopha.2021.112375] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 02/09/2023] Open
Abstract
Breast cancer (BC) is mostly observed in women and is responsible for huge mortality in women subjects globally. Due to the continued development of drug resistance and other contributing factors, the scientific community needs to look for new alternatives, and drug repurposing is one of the best opportunities. Here we light upon the drug repurposing with a major focus on breast cancer. BC is a division of cancer known as the leading cause of death of 2.3 million women globally, with 685,000 fatalities. This number is steadily rising, necessitating the development of a treatment that can extend survival time. All available treatments for BC are very costly as well as show side effects. This unfulfilled requirement of the anti-cancer drugs ignited an enthusiasm for drug repositioning, which means finding out the anti-cancer use of already marketed drugs for other complications. With the advancement in proteomics, genomics, and computational approaches, the drug repurposing process hastens. So many drugs are repurposed for the BC, including alkylating agents, antimetabolite, anthracyclines, an aromatase inhibitor, mTOR, and many more. The drug resistance in breast cancer is rising, so reviewing how the challenges in breast cancer can be combated with drug repurposing. This paper provides the updated information on all the repurposed drugs candidates for breast cancer with the molecular mechanism responsible for their anti-tumor activity. Additionally, all the challenges that occur during the repurposing of the drugs are discussed.
Collapse
Affiliation(s)
- Jonaid Ahmad Malik
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Guwahati, India; Department of Biomedical engineering, Indian Institute of Technology (IIT), Ropar, Punjab, India
| | - Sakeel Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Bisma Jan
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
| | - Onur Bender
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Turki Al Hagbani
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Aali Alqarni
- Pharmaceutical Chemistry Department, Pharmacology unit, College of Clinical Pharmacy, Al Baha University, Saudi Arabia
| | - Sirajudheen Anwar
- Pharmacology and Toxicology Department, College of Pharmacy, University of Hail, Hail, Saudi Arabia.
| |
Collapse
|
3
|
Syed SB, Lin SY, Arya H, Fu IH, Yeh TK, Charles MRC, Periyasamy L, Hsieh HP, Coumar MS. Overcoming vincristine resistance in cancer: Computational design and discovery of piperine-inspired P-glycoprotein inhibitors. Chem Biol Drug Des 2020; 97:51-66. [PMID: 32633857 DOI: 10.1111/cbdd.13758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/05/2020] [Accepted: 06/28/2020] [Indexed: 01/03/2023]
Abstract
P-glycoprotein (P-gp)/MDR-1 plays a major role in the development of multidrug resistance (MDR) by pumping the chemotherapeutic drugs out of the cancer cells and reducing their efficacy. A number of P-gp inhibitors were reported to reverse the MDR when co-administered with chemotherapeutic drugs. Unfortunately, none has approved for clinical use due to toxicity issues. Some of the P-gp inhibitors tested in the clinics are reported to have cross-reactivity with CYP450 drug-metabolizing enzymes, resulting in unpredictable pharmacokinetics and toxicity of co-administered chemotherapeutic drugs. In this study, two piperine analogs (3 and 4) having lower cross-reactivity with CYP3A4 drug-metabolizing enzyme are identified as P-glycoprotein (P-gp) inhibitors through computational design, followed by synthesis and testing in MDR cancer cell lines over-expressing P-gp (KB ChR 8-5, SW480-VCR, and HCT-15). Both the analogs significantly increased the vincristine efficacy in MDR cancer cell lines at low micromole concentrations. Specifically, 3 caused complete reversal of vincristine resistance in KB ChR 8-5 cells and found to act as competitive inhibitor of P-gp as well as potentiated the vincristine-induced NF-KB-mediated apoptosis. Therefore, 3 ((2E,4E)-1-(6,7-dimethoxy-3,4-dihydroisoquinolin-2(1H)-yl)-5-(4-hydroxy-3-methoxyphenyl)penta-2,4-dien-1-one) can serve as a potential P-gp inhibitor for in vivo investigations, to reverse multidrug resistance in cancer.
Collapse
Affiliation(s)
- Safiulla Basha Syed
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, India.,DBT-Interdisciplinary Program in Life Sciences, Pondicherry University, Kalapet, India
| | - Shu-Yu Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Hemant Arya
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, India
| | - I-Hsuan Fu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | | | - Latha Periyasamy
- Department of Biochemistry & Molecular Biology, School of Life Sciences, Pondicherry University, Kalapet, India
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan.,Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
| | - Mohane Selvaraj Coumar
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, India
| |
Collapse
|
4
|
Zhang S, Guo N, Wan G, Zhang T, Li C, Wang Y, Wang Y, Liu Y. pH and redox dual-responsive nanoparticles based on disulfide-containing poly(β-amino ester) for combining chemotherapy and COX-2 inhibitor to overcome drug resistance in breast cancer. J Nanobiotechnology 2019; 17:109. [PMID: 31623608 PMCID: PMC6798417 DOI: 10.1186/s12951-019-0540-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022] Open
Abstract
Background Multidrug resistance (MDR) generally leads to breast cancer treatment failure. The most common mechanism of MDR is the overexpression of ATP-binding cassette (ABC) efflux transporters such as P-glycoprotein (P-gp) that reduce the intracellular accumulation of various chemotherapeutic agents. Celecoxib (CXB), a selective COX-2 inhibitor, can dramatically enhance the cytotoxicity of doxorubicin (DOX) in breast cancer cells overexpressing P-gp. Thus it can be seen that the combination of DOX and CXB maybe obtain synergistic effects against breast cancer by overcoming drug resistance. Results In this study, we designed a pH and redox dual-responsive nanocarrier system to combine synergistic effects of DOX and CXB against drug resistant breast cancer. This nanocarrier system denoted as HPPDC nanoparticles showed good in vitro stability and significantly accelerated drug releases under the acidic and redox conditions. In drug-resistant human breast cancer MCF-7/ADR cells, HPPDC nanoparticles significantly enhanced the cellular uptake of DOX through the endocytosis mediated by CD44/HA specific binding and the down-regulated P-gp expression induced by COX-2 inhibition, and thus notably increased the cytotoxicity and apoptosis-inducing activity of DOX. In MCF-7/ADR tumor-bearing nude mice, HPPDC nanoparticles showed excellent tumor-targeting ability, remarkably enhanced tumor chemosensitivity and reduced COX-2 and P-gp expressions in tumor tissues. Conclusion All results demonstrated that HPPDC nanoparticles can efficiently overcome drug resistance in breast cancer both in vitro and in vivo by combining chemotherapy and COX-2 inhibitor. In a summary, HPPDC nanoparticles show a great potential for combination treatment of drug resistant breast cancer.
Collapse
Affiliation(s)
- Sipei Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences; Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Qixiangtai Road 22, Tianjin, 300070, China
| | - Nan Guo
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences; Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Qixiangtai Road 22, Tianjin, 300070, China
| | - Guoyun Wan
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences; Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Qixiangtai Road 22, Tianjin, 300070, China
| | - Tao Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences; Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Qixiangtai Road 22, Tianjin, 300070, China
| | - Chunyu Li
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences; Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Qixiangtai Road 22, Tianjin, 300070, China
| | - Yongfei Wang
- Choate Rosemary Hall, Class of 2019, Wallingford, CT, 06492, USA
| | - Yinsong Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences; Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Qixiangtai Road 22, Tianjin, 300070, China.
| | - Yuanyuan Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy; Department of Genetics, School of Basic Medical Sciences; Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Qixiangtai Road 22, Tianjin, 300070, China.
| |
Collapse
|
5
|
Liu J, Chang B, Li Q, Xu L, Liu X, Wang G, Wang Z, Wang L. Redox-Responsive Dual Drug Delivery Nanosystem Suppresses Cancer Repopulation by Abrogating Doxorubicin-Promoted Cancer Stemness, Metastasis, and Drug Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801987. [PMID: 31139556 PMCID: PMC6446919 DOI: 10.1002/advs.201801987] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/09/2019] [Indexed: 05/15/2023]
Abstract
Chemotherapy is a major therapeutic option for cancer patients. However, its effectiveness is challenged by chemodrugs' intrinsic pathological interactions with residual cancer cells. While inducing cancer cell death, chemodrugs enhance cancer stemness, invasiveness, and drug resistance of remaining cancer cells through upregulating cyclooxygenase-2/prostaglandin-E2 (COX-2/PGE2) signaling, therefore facilitating cancer repopulation and relapse. Toward tumor eradication, it is necessary to improve chemotherapy by abrogating these chemotherapy-induced effects. Herein, redox-responsive, celecoxib-modified mesoporous silica nanoparticles with poly(β-cyclodextrin) wrapping (MSCPs) for sealing doxorubicin (DOX) are synthesized. Celecoxib, an FDA-approved COX-2 inhibitor, is employed as a structural and functional element to confer MSCPs with redox-responsiveness and COX-2/PGE2 inhibitory activity. MSCPs efficiently codeliver DOX and celecoxib into the tumor location, minimizing systemic toxicity. Importantly, through blocking chemotherapy-activated COX-2/PGE2 signaling, MSCPs drastically enhance DOX's antitumor activity by suppressing enhancement of cancer stemness and invasiveness as well as drug resistance induced by DOX-based chemotherapy in vitro. This is also remarkably achieved in three preclinical tumor models in vivo. DOX-loaded MSCPs effectively inhibit tumor repopulation by blocking COX-2/PGE2 signaling, which eliminates DOX-induced expansion of cancer stem-like cells, distant metastasis, and acquired drug resistance. Thus, this drug delivery nanosystem is capable of effectively suppressing tumor repopulation and has potential clinical translational value.
Collapse
Affiliation(s)
- Jia Liu
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Bingcheng Chang
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Qilin Li
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Luming Xu
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xingxin Liu
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Guobin Wang
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
6
|
Abstract
Breast cancer has a high incidence worldwide. The results of substantial studis reveal that inflammation plays an important role in the initiation, development, and aggressiveness of many malignancies. The use of celecoxib, a novel NSAID, is repetitively associated with the reduced risk of the occurrence and progression of a number of types of cancer, particularly breast cancer. This observation is also substantiated by various meta-analyses. Clinical trials have been implemented on integration treatment of celecoxib and shown encouraging results. Celecoxib could be treated as a potential candidate for antitumor agent. There are, nonetheless, some unaddressed questions concerning the precise mechanism underlying the anticancer effect of celecoxib as well as its activity against different types of cancer. In this review, we discuss different mechanisms of anticancer effect of celecoxib as well as preclinical/clinical results signifying this beneficial effect.
Collapse
Affiliation(s)
- Jieqing Li
- Department of Breast Surgery, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, China.,Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles,CA, USA, ;
| | - Qiongyu Hao
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles,CA, USA, ;
| | - Wei Cao
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles,CA, USA, ; .,Department of Nuclear Medicine, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jaydutt V Vadgama
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles,CA, USA, ; .,David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA, ;
| | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles,CA, USA, ; .,David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA, ;
| |
Collapse
|
7
|
Celecoxib inhibits mitochondrial O2 consumption, promoting ROS dependent death of murine and human metastatic cancer cells via the apoptotic signalling pathway. Biochem Pharmacol 2018; 154:318-334. [DOI: 10.1016/j.bcp.2018.05.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/18/2018] [Indexed: 12/27/2022]
|
8
|
Kozlu S, Sahin A, Ultav G, Yerlikaya F, Calis S, Capan Y. Development and in vitro evaluation of doxorubicin and celecoxib co-loaded bone targeted nanoparticles. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
9
|
MAJDZADEH M, ALIEBRAHIMI S, VATANKHAH M, OSTAD SN. Effects of celecoxib and L-NAME on apoptosis and cell cycle ofMCF-7 CD44+/CD24–/low subpopulation. Turk J Biol 2017. [DOI: 10.3906/biy-1703-101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
10
|
Bakirel T, Alkan FÜ, Üstüner O, Çinar S, Yildirim F, Erten G, Bakirel U. Synergistic growth inhibitory effect of deracoxib with doxorubicin against a canine mammary tumor cell line, CMT-U27. J Vet Med Sci 2016; 78:657-68. [PMID: 26822118 PMCID: PMC4873858 DOI: 10.1292/jvms.15-0387] [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] [Indexed: 12/16/2022] Open
Abstract
Cyclooxygenase (COX) inhibitors have been shown to exert anti-angiogenic and anti-tumor
activities on many types of malignant tumors. These anticancer properties make it
worthwhile to examine the possible benefit of combining COX inhibitors with other
anti-cancer agents. In the present study, we evaluated the potential of deracoxib (DER) in
potentiating antitumor activity of doxorubicin (DOX) in canine mammary carcinoma cells
(CMT-U27). DER (50–250 µM) enhanced the antiproliferative activity of DOX
by reducing the IC50 (approximately 3- to 3.5 fold). Interaction analysis of
the data showed that combinations of DOX at 0.9 µM with DER (100–250
µM) produced synergism in the CMT-U27 cell line, with a ratio index
ranging from 1.98 to 2.33. In additional studies identifying the mechanism of observed
synergistic effect, we found that DER strongly potentiated DOX-caused
G0/G1 arrest in cell cycle progression. Also, DER (100–250
µM) augmented apoptosis induction with approximately 1.35- and 1.37-
fold increases in apoptotic response caused by DOX in the cells. DER enhanced the
antiproliferative effect of DOX in conjunction with induction of apoptosis by modulation
of Bcl-2 expression and changes in the cell cycle of the CMT-U27 cell line. Although the
exact molecular mechanism of the alterations in the cell cycle and apoptosis observed with
DER and DOX combinations require further investigations, the results suggest that the
synergistic effect of DOX and DER combinations in CMT therapy may be achieved at
relatively lower doses of DOX with lesser side effects. Therefore, combining DER with DOX
may prove beneficial in the clinical treatment of canine mammary cancer.
Collapse
Affiliation(s)
- Tülay Bakirel
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University, Istanbul, 34320, Turkey
| | | | | | | | | | | | | |
Collapse
|
11
|
Tsai YT, Lozanski G, Lehman A, Sass EJ, Hertlein E, Salunke SB, Chen CS, Grever MR, Byrd JC, Lucas DM. BRAF V600E induces ABCB1/P-glycoprotein expression and drug resistance in B-cells via AP-1 activation. Leuk Res 2015; 39:S0145-2126(15)30371-4. [PMID: 26350141 PMCID: PMC4779435 DOI: 10.1016/j.leukres.2015.08.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022]
Abstract
A subset of patients with chronic lymphocytic leukemia (CLL) and nearly all patients with classic hairy cell leukemia (HCL) harbor somatic BRAF activating mutations. However, the pathological role of activated BRAF in B-cell leukemia development and progression remains unclear. In addition, although HCL patients respond well to the BRAFV600E inhibitor vemurafenib, relapses are being observed, suggesting the development of drug resistance in patients with this mutation. To investigate the biological role of BRAFV600E in B-cell leukemia, we generated a CLL-like B-cell line, OSUCLL, with doxycycline-inducible BRAFV600E expression. Microarray and real-time PCR analysis showed that ABCB1 mRNA is upregulated in these cells, and P-glycoprotein (P-gp) expression as well as function were confirmed by immunoblot and rhodamine exclusion assays. Additionally, pharmacological inhibition of BRAFV600E and MEK alleviated the BRAFV600E-induced ABCB1/P-gp expression. ABCB1 reporter assays and gel shift assays demonstrated that AP-1 activity is crucial in this mechanism. This study, uncovers a pathological role for BRAFV600E in B-cell leukemia, and provides further evidence that combination strategies with inhibitors of BRAFV600E and MEK can be used to delay disease progression and occurrence of resistance.
Collapse
Affiliation(s)
- Yo-Ting Tsai
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Gerard Lozanski
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Amy Lehman
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Ellen J Sass
- Department of Internal Medicine, College of Medicine; The Ohio State University, Columbus, OH, USA
| | - Erin Hertlein
- Department of Internal Medicine, College of Medicine; The Ohio State University, Columbus, OH, USA
| | - Santosh B Salunke
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Ching-Shih Chen
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Michael R Grever
- Department of Internal Medicine, College of Medicine; The Ohio State University, Columbus, OH, USA
| | - John C Byrd
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA; Department of Internal Medicine, College of Medicine; The Ohio State University, Columbus, OH, USA
| | - David M Lucas
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA; Department of Internal Medicine, College of Medicine; The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
12
|
Doktorova H, Hrabeta J, Khalil MA, Eckschlager T. Hypoxia-induced chemoresistance in cancer cells: The role of not only HIF-1. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2015; 159:166-77. [PMID: 26001024 DOI: 10.5507/bp.2015.025] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/07/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The aim of this review is to provide the information about molecular basis of hypoxia-induced chemoresistance, focusing on the possibility of diagnostic and therapeutic use. RESULTS Hypoxia is a common feature of tumors and represents an independent prognostic factor in many cancers. It is the result of imbalances in the intake and consumption of oxygen caused by abnormal vessels in the tumor and the rapid proliferation of cancer cells. Hypoxia-induced resistance to cisplatin, doxorubicin, etoposide, melphalan, 5-flouoruracil, gemcitabine, and docetaxel has been reported in a number of experiments. Adaptation of tumor cells to hypoxia has important biological effects. The most studied factor responsible for these effects is hypoxia-inducible factor-1 (HIF-1) that significantly contributes to the aggressiveness and chemoresistance of different tumors. The HIF-1 complex, induced by hypoxia, binds to target genes, thereby increasing the expression of many genes. In addition, the expression of hundreds of genes can be also decreased in response to hypoxia in HIF-1 dependent manner, but without the detection of HIF-1 in these genes' promoters. HIF-1 independent mechanisms for drug resistance in hypoxia have been described, however, they are still rarely reported. The first clinical studies focusing on diagnosis of hypoxia and on inhibition of hypoxia-induced changes in cancer cells are starting to yield results. CONCLUSIONS The adaptation to hypoxia requires many genetic and biochemical responses that regulate one another. Hypoxia-induced resistance is a very complex field and we still know very little about it. Different approaches to circumvent hypoxia in tumors are under development.
Collapse
Affiliation(s)
- Helena Doktorova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Jan Hrabeta
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Mohamed Ashraf Khalil
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Tomas Eckschlager
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
13
|
HIĽOVSKÁ LUCIA, JENDŽELOVSKÝ RASTISLAV, FEDOROČKO PETER. Potency of non-steroidal anti-inflammatory drugs in chemotherapy. Mol Clin Oncol 2015; 3:3-12. [PMID: 25469262 PMCID: PMC4251142 DOI: 10.3892/mco.2014.446] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 10/01/2014] [Indexed: 12/19/2022] Open
Abstract
Cancer cell resistance, particularly multidrug resistance (MDR), is the leading cause of chemotherapy failure. A number of mechanisms involved in the development of MDR have been described, including the overexpression of ATP-dependent membrane-bound transport proteins. The enhanced expression of these proteins, referred to as ATP-binding cassette (ABC) transporters, results in an increased cellular efflux of the cytotoxic drug, thereby reducing its intracellular concentration to an ineffective level. Non-steroidal anti-inflammatory drugs (NSAIDs) are the most frequently consumed drugs worldwide. NSAIDs are mainly used to treat pain, fever and inflammation. Numerous studies suggest that NSAIDs also show promise as anticancer drugs. NSAIDs have been shown to reduce cancer cell proliferation, motility, angiogenesis and invasiveness. In addition to these effects, NSAIDs have been shown to induce apoptosis in a wide variety of cancer types. Moreover, several studies have indicated that NSAIDs may sensitise cancer cells to the antiproliferative effects of cytotoxic drugs by modulating ABC transporter activity. Therefore, combining specific NSAIDs with chemotherapeutic drugs may have clinical applications. Such treatments may allow for the use of a lower dose of cytotoxic drugs and may also enhance the effectiveness of therapy. The objective of this review was to discuss the possible role of NSAIDs in the modulation of antitumour drug cytotoxicity. We particularly emphasised on the use of COX-2 inhibitors in combination with chemotherapy and the molecular and cellular mechanisms underlying the alterations in outcome that occur in response to this combination therapy.
Collapse
Affiliation(s)
- LUCIA HIĽOVSKÁ
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, 040 01 Košice, Slovakia
| | - RASTISLAV JENDŽELOVSKÝ
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, 040 01 Košice, Slovakia
| | - PETER FEDOROČKO
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, 040 01 Košice, Slovakia
| |
Collapse
|
14
|
Gong J, Luk F, Jaiswal R, Bebawy M. Microparticles Mediate the Intercellular Regulation of microRNA-503 and Proline-Rich Tyrosine Kinase 2 to Alter the Migration and Invasion Capacity of Breast Cancer Cells. Front Oncol 2014; 4:220. [PMID: 25177548 PMCID: PMC4133752 DOI: 10.3389/fonc.2014.00220] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/01/2014] [Indexed: 11/13/2022] Open
Abstract
The successful treatment of cancer is hampered by drug resistance and metastasis. While these two obstacles were once considered separately, recent evidence associates resistance with an enhanced metastatic capacity. However, the underlying mechanisms remain undefined. We previously described the intercellular transfer of drug resistance via submicron vesicles called microparticles (MPs). We now propose that MPs derived from drug-resistant cells are also involved in the intercellular transfer of components to enhance the migration and invasion capacity of cells. Thus, MPs may be a conduit between resistance and metastasis. We used microarray analysis to identify regulatory microRNAs (miRNAs), which contribute to the dissemination of metastatic traits. miR-503 was downregulated in recipient cells following co-culture with MPs isolated from drug-resistant cells. miR-503 was inversely associated with metastasis, as demonstrated using wound healing/scratch migration assays and Matrigel®-coated transwell invasion assays. Proline-rich tyrosine kinase 2 (PYK2) was upregulated in recipient cells and associated with increased migration and invasion, with these phenotypes being reversed using a pharmacological inhibitor of PYK2 phosphorylation, tyrphostin A9. However, the MP-mediated promotion of metastatic traits was not due to the presence of these effectors in the MP cargo but rather due to down stream effector molecules in these pathways. This is the first demonstration that the role of MPs in trait acquisition extends beyond the direct transfer of vesicle components and also includes transfer of intermediary regulators that induce down stream mediators following transfer to recipient cells. This implicates an expanding role of MPs in cancer pathogenesis.
Collapse
Affiliation(s)
- Joyce Gong
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney , Sydney, NSW , Australia ; Sydney Medical School and Bosch Institute, The University of Sydney , Sydney, NSW , Australia
| | - Frederick Luk
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney , Sydney, NSW , Australia
| | - Ritu Jaiswal
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney , Sydney, NSW , Australia
| | - Mary Bebawy
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney , Sydney, NSW , Australia
| |
Collapse
|
15
|
Giráldez S, Herrero-Ruiz J, Mora-Santos M, Japón MÁ, Tortolero M, Romero F. SCF(FBXW7α) modulates the intra-S-phase DNA-damage checkpoint by regulating Polo like kinase-1 stability. Oncotarget 2014; 5:4370-83. [PMID: 24970797 PMCID: PMC4147330 DOI: 10.18632/oncotarget.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/26/2014] [Indexed: 12/22/2022] Open
Abstract
The intra-S-checkpoint is essential to control cell progression through S phase under normal conditions and in response to replication stress. When DNA lesions are detected, replication fork progression is blocked allowing time for repair to avoid genomic instability and the risk of cancer. DNA replication initiates at many origins of replication in eukaryotic cells, where a series of proteins form pre-replicative complexes (pre-RCs) that are activated to become pre-initiation complexes and ensure a single round of replication in each cell cycle. PLK1 plays an important role in the regulation of DNA replication, contributing to the regulation of pre-RCs formation by phosphorylating several proteins, under both normal and stress conditions. Here we report that PLK1 is ubiquitinated and degraded by SCFFBXW7α/proteasome. Moreover, we identified a new Cdc4 phosphodegron in PLK1, conserved from yeast to humans, whose mutation prevents PLK1 destruction. We established that endogenous SCFFBXW7α degrades PLK1 in the G1 and S phases of an unperturbed cell cycle and in S phase following UV irradiation. Furthermore, we showed that FBXW7α overexpression or UV irradiation prevented the loading of proteins onto chromatin to form pre-RCs and, accordingly, reduced cell proliferation. We conclude that PLK1 degradation mediated by SCFFBXW7α modulates the intra-S-phase checkpoint.
Collapse
Affiliation(s)
- Servando Giráldez
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| | - Joaquín Herrero-Ruiz
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| | - Mar Mora-Santos
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| | - Miguel Á. Japón
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Anatomía Patológica, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Maria Tortolero
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| | - Francisco Romero
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla. Apartado de correos 1095. 41080-Sevilla, Spain
| |
Collapse
|
16
|
Liu Z, Duan ZJ, Chang JY, Zhang ZF, Chu R, Li YL, Dai KH, Mo GQ, Chang QY. Sinomenine sensitizes multidrug-resistant colon cancer cells (Caco-2) to doxorubicin by downregulation of MDR-1 expression. PLoS One 2014; 9:e98560. [PMID: 24901713 PMCID: PMC4047020 DOI: 10.1371/journal.pone.0098560] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/05/2014] [Indexed: 12/28/2022] Open
Abstract
Chemoresistance in multidrug-resistant (MDR) cells over expressing P-glycoprotein (P-gp) encoded by the MDR1 gene, is a major obstacle to successful chemotherapy for colorectal cancer. Previous studies have indicated that sinomenine can enhance the absorption of various P-gp substrates. In the present study, we investigated the effect of sinomenine on the chemoresistance in colon cancer cells and explored the underlying mechanism. We developed multidrug-resistant Caco-2 (MDR-Caco-2) cells by exposure of Caco-2 cells to increasing concentrations of doxorubicin. We identified overexpression of COX-2 and MDR-1 genes as well as activation of the NF-κB signal pathway in MDR-Caco-2 cells. Importantly, we found that sinomenine enhances the sensitivity of MDR-Caco-2 cells towards doxorubicin by downregulating MDR-1 and COX-2 expression through inhibition of the NF-κB signaling pathway. These findings provide a new potential strategy for the reversal of P-gp-mediated anticancer drug resistance.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhi-Jun Duan
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- * E-mail: (ZJD); (QYC)
| | - Jiu-Yang Chang
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhi-feng Zhang
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Rui Chu
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yu-Ling Li
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ke-Hang Dai
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Guang-quan Mo
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qing-Yong Chang
- Department of Neurosurgery, Zhongshan Affiliated Hospital of Dalian University, Dalian, Liaoning, China
- * E-mail: (ZJD); (QYC)
| |
Collapse
|
17
|
Kim HJ, Yim GW, Nam EJ, Kim YT. Synergistic Effect of COX-2 Inhibitor on Paclitaxel-Induced Apoptosis in the Human Ovarian Cancer Cell Line OVCAR-3. Cancer Res Treat 2014; 46:81-92. [PMID: 24520227 PMCID: PMC3918531 DOI: 10.4143/crt.2014.46.1.81] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 05/24/2013] [Indexed: 11/21/2022] Open
Abstract
PURPOSE Celecoxib, a highly selective cyclooxygenase-2 inhibitor, regulates apoptosis of several types of human cancer cells. The purpose of this study was to investigate whether celecoxib in combination with paclitaxel modulates apoptosis of ovarian cancer cells, and to identify the signal pathway by which celecoxib mediates apoptosis. MATERIALS AND METHODS OVCAR-3 cells were exposed to paclitaxel (20 µM) in the absence or presence of celecoxib (10 µM). Cell viability was evaluated using a Cell Counting Kit-8 assay. Apoptosis was evaluated using Annexin-V/7-aminoactinomycin D staining and a cellular DNA fragmentation enzyme-linked immunosorbent assay. Caspase-3, -9, and cleavage of poly ADP-ribose polymerase (PARP) were determined by western blotting. Expression of nuclear factor-κB (NF-κB) and vascular endothelial growth factor (VEGF) and Akt activation were assessed using reverse transcriptase-polymerase chain reaction and western blotting. RESULTS Celecoxib enhanced paclitaxel-induced growth inhibition of OVCAR-3 cells. Celecoxib significantly increased paclitaxel-induced apoptosis of OVCAR-3 cells. Pretreatment with celecoxib also increased activation of caspase-9, -3 and cleaved PARP following paclitaxel-treatment. Exposure of OVCAR-3 cells to celecoxib in combination with paclitaxel resulted in downregulation of NF-κB activation and VEGF expression. Furthermore, combining celecoxib and paclitaxel inhibited phosphorylation of Akt. CONCLUSION OVCAR-3 cells were sensitized to paclitaxel-induced apoptosis by celecoxib through downregulation of NF-κB and Akt activation, suggesting that celecoxib may work synergistically with paclitaxel to inhibit different targets and ultimately produce anticancer effects. Combining celecoxib with paclitaxel may prove beneficial in the clinical treatment of ovarian cancer.
Collapse
Affiliation(s)
- Hee Jung Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ga Won Yim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Ji Nam
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Young Tae Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Hasegawa K, Ishikawa K, Kawai S, Torii Y, Kawamura K, Kato R, Tsukada K, Udagawa Y. Overcoming paclitaxel resistance in uterine endometrial cancer using a COX-2 inhibitor. Oncol Rep 2013; 30:2937-44. [PMID: 24100466 DOI: 10.3892/or.2013.2790] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 09/09/2013] [Indexed: 11/06/2022] Open
Abstract
Cyclooxygenase (COX)-2 inhibitors have been reported to potentially modulate the resistance of cancer cells to chemotherapeutic drugs by affecting multidrug resistance 1 (MDR1) expression. In the present study, we investigated the association between COX-2 and MDR1 expression in endometrial cancers and evaluated the effects of the COX-2 inhibitor, etodolac, in combination with paclitaxel on paclitaxel-resistant endometrial cancer cells. The relationship between COX-2 and MDR1 mRNA expression was examined by quantitative PCR in 36 endometrial cancer specimens. The paclitaxel-resistant cell line OMC-2P was established from OMC-2 cells. Paclitaxel (1 µg/ml) with or without etodolac (10 µg/ml) was added to OMC-2 and OMC-2P cells, and COX-2 and MDR1 mRNA expression levels were examined. The concentration of prostaglandin E2 (PGE2) in the supernatant of each cell line was examined by enzyme-linked immunosorbent assay. The function of MDR1 was determined by intracellular accumulation of rhodamine 123 using flow cytometry, and the concentration of intracellular paclitaxel was determined by high-performance liquid chromatography. We found a positive relationship between COX-2 and MDR1 mRNA expression in endometrial cancer. Both COX-2 mRNA expression and PGE2 production were elevated in resistant OMC-2P cells when compared to non-resistant OMC-2 cells. Additionally, MDR1 mRNA expression was markedly upregulated in OMC-2P cells. In OMC-2 cells, COX-2 and MDR1 mRNA levels were significantly upregulated by paclitaxel treatment and downregulated by co-administration with etodolac. In OMC-2P cells, COX-2 mRNA expression was also significantly upregulated by paclitaxel treatment and tended to be downregulated by co-administration with etodolac. Moreover, co-administration of paclitaxel and etodolac suppressed the induction of MDR1 mRNA. Rhodamine 123 efflux was increased in OMC-2P cells when compared to the efflux in the OMC-2 cells and was increased in response to paclitaxel treatment. Co-administration of paclitaxel and etodolac in both cell lines resulted in decreased rhodamine 123 efflux. The actual concentration of intracellular paclitaxel in OMC-2P cells was significantly lower than that in OMC-2 cells treated with paclitaxel alone and was significantly increased after co-administration of paclitaxel and etodolac. These findings suggest that paclitaxel resistance may be associated with COX-2 and MDR1 expression in cancer cells. Co-administration of COX-2 inhibitors and paclitaxel may have a key role in modulating or overcoming paclitaxel resistance in endometrial cancers.
Collapse
Affiliation(s)
- Kiyoshi Hasegawa
- Department of Obstetrics and Gynecology, Fujita Health University, Banbuntane Hotokukai Hospital, Nagoya, Aichi 454‑8509, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Chow LWC, Tung SY, Ng TY, Im SA, Lee MH, Yip AYS, Toi M, Glück S. Concurrent celecoxib with 5-fluorouracil/epirubicin/cyclophosphamide followed by docetaxel for stages II - III invasive breast cancer: the OOTR-N001 study. Expert Opin Investig Drugs 2013; 22:299-307. [PMID: 23394482 DOI: 10.1517/13543784.2013.766715] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES This prospective study aimed at investigating the efficacy and safety of the concurrent use of celecoxib (CXB) with 5-fluorouracil, epirubicin and cyclophosphamide (FEC), followed by docetaxel (T) in the neoadjuvant setting. PATIENTS AND METHODS A total of 64 invasive breast cancer patients were recruited in the N001 Phase II, multicenter, open-label, single-arm study to receive four cycles of FEC (500, 100, 500 mg/m(2)) followed by four cycles of T (100 mg/m(2)) with concurrent CXB (200 mg b.i.d.) as neoadjuvant therapy (NAT). The combined chemotherapies were administered on day 1 of each cycle every 3 weeks. Primary endpoints were pathologic complete response (pCR) rate and objective response rate (ORR). Quasi-pCR (QpCR), pCR and near pCR (npCR) were discussed considering their similar survival outcomes. ORR included clinical complete response (cCR) and clinical partial response (cPR). Secondary endpoints included safety, breast conservation rate and disease-free survival. RESULTS Between February 2006 and January 2010, 57 of 64 evaluable patients with luminal A (n = 35, 61.4%), luminal B (n = 12, 21.1%), HER-2 positive (n = 8, 14%) and triple-negative (n = 2, 3.5%) breast cancer completed NAT and surgery. QpCR rate was observed in 18 (31.6%) patients. Exclusive of triple-negative subtype, pCR (p = 0.761) did not differ compared to other subtypes, while npCR (p = 0.043) exhibited a difference. Patients with HER-2 overexpression had a significantly higher QpCR than those of the disease attribute (10/20 vs 8/37, p = 0.029). After NAT, 43 (75.4%) and 13 (22.8%) patients achieved cCR and cPR, respectively. Patients responding to FEC were more likely to achieve a better ORR after subsequent T (p = 0.004). Over 80% of all patients received breast-conserving therapy (BCT) after receiving NAT, and 11 of 14 (78.6%) patients with T3 tumor at diagnosis became eligible for BCT after NAT. A total of 60 patients completed ≥ 6 cycles of NAT, followed by surgery; at a median follow-up of 50 months, 80% of the patients are disease-free. Neither drug-induced life-threatening toxicity nor cardiotoxicity was observed. CONCLUSIONS Neoadjuvant use of FEC-T with concurrent CXB is active and safe for treatment of operable invasive breast cancer. The ORR was higher, but QpCR was comparable to other studies. Most patients are still disease-free, and BCT became an option for the females. Further clinical and translational studies on the use of cyclooxygenase-2 inhibitors with neoadjuvant chemotherapy are warranted.
Collapse
Affiliation(s)
- Louis W C Chow
- Organisation for Oncology and Translational Research, Unit A, 9/F, CNT Commercial Building, 302 Queen's Road Central, Hong Kong, China.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Chen C, Xu W, Wang CM. Combination of celecoxib and doxorubicin increases growth inhibition and apoptosis in acute myeloid leukemia cells. Leuk Lymphoma 2013; 54:2517-22. [PMID: 23452119 DOI: 10.3109/10428194.2013.781170] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cyclooxygenase-2 (COX-2) inhibitors have been shown to enhance antitumor activity of therapeutic agents in a variety of solid tumor cells. However, this has not been well established in hematopoietic tumors, especially in acute myeloid leukemia (AML). This study was designed to investigate the effects of the combination of celecoxib, a specific COX-2 inhibitor, and doxorubicin on cell growth and apoptosis in human leukemia cells. Co-treatment with celecoxib and doxorubicin significantly inhibited cell growth and induced cell apoptosis in the acute leukemia cell line HL60 and primary AML cells. The growth inhibition effect was accompanied by down-regulation of the expression of cyclin E and cyclin-dependent kinase 2 (CDK2), the key regulators of cell cycle progression, which was associated with arrest of cells at G0/G1 phase. The pro-apoptotic effect was accompanied by down-regulation of the expression of survivin, an inhibitor of apoptosis protein, which mediated anti-apoptosis in AML cells. These results provide the first evidence that the growth inhibitory and pro-apoptotic effects of celecoxib and doxorubicin on AML cells are synergistic.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hematology, Zaozhuang Municipal Hospital , Zaozhuang , China
| | | | | |
Collapse
|
21
|
Inhibition of COX-2 in colon cancer modulates tumor growth and MDR-1 expression to enhance tumor regression in therapy-refractory cancers in vivo. Neoplasia 2013; 14:624-33. [PMID: 22904679 DOI: 10.1593/neo.12486] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 06/04/2012] [Accepted: 06/06/2012] [Indexed: 12/11/2022] Open
Abstract
Higher cyclooxygenase 2 (COX-2) expression is often observed in aggressive colorectal cancers (CRCs). Here, we attempt to examine the association between COX-2 expression in therapy-refractory CRC, how it affects chemosensitivity, and whether, in primary tumors, it is predictive of clinical outcomes. Our results revealed higher COX-2 expression in chemoresistant CRC cells and tumor xenografts. In vitro, the combination of either aspirin or celecoxib with 5-fluorouracil (5-FU) was capable of improving chemosensitivity in chemorefractory CRC cells, but a synergistic effect with 5-FU could only be demonstrated with celecoxib. To examine the potential clinical significance of these observations, in vivo studies were undertaken, which also showed that the greatest tumor regression was achieved in chemoresistant xenografts after chemotherapy in combination with celecoxib, but not aspirin. We also noted that these chemoresistant tumors with higher COX-2 expression had a more aggressive growth rate. Given the dramatic response to a combination of celecoxib + 5-FU, the possibility that celecoxib may modulate chemosensitivity as a result of its ability to inhibit MDR-1 was examined. In addition, assessment of a tissue microarray consisting of 130 cases of CRCs revealed that, in humans, higher COX-2 expression was associated with poorer survival with a 68% increased risk of mortality, indicating that COX-2 expression is a marker of poor clinical outcome. The findings of this study point to a potential benefit of combining COX-2 inhibitors with current regimens to achieve better response in the treatment of therapy-refractory CRC and in using COX-2 expression as a prognostic marker to help identify individuals who would benefit the greatest from closer follow-up and more aggressive therapy.
Collapse
|
22
|
Zhang W, Chen H, Liu DL, Li H, Luo J, Zhang JH, Li Y, Chen KJ, Tong HF, Lin SZ. Emodin sensitizes the gemcitabine-resistant cell line Bxpc-3/Gem to gemcitabine via downregulation of NF-κB and its regulated targets. Int J Oncol 2013; 42:1189-96. [PMID: 23440366 DOI: 10.3892/ijo.2013.1839] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 02/04/2013] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to evaluate whether emodin can overcome the chemoresistance of the gemcitabine-resistant cancer cell line (Bxpc-3/Gem) in vitro. The cell line Bxpc-3/Gem was derived from the human pancreatic cancer cell line Bxpc-3. We found that Bxpc-3/Gem cells were characterized by a series of morphological changes with a resistance index of 43.51 comparing with the parental cell line. Emodin reduced Bxpc-3/Gem cell proliferation in a dose-dependent manner. Emodin and gemcitabine combination treatments resulted in decreased cell proliferation and increased apoptosis in Bxpc-3/Gem cells. In addition, combination treatments resulted in downregulation of gene and protein expression of MDR-1 (P-gp), NF-κB, XIAP, survivin, as well as inhibition of NF-κB activity and P-gp function. These observations suggest that emodin may sensitize the pancreatic cancer gemcitabine-resistant cell line Bxpc-3/Gem to gemcitabine therapy via inhibition of survival signaling.
Collapse
Affiliation(s)
- Wei Zhang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Synthesis of a dual functional anti-MDR tumor agent PH II-7 with elucidations of anti-tumor effects and mechanisms. PLoS One 2012; 7:e32782. [PMID: 22403708 PMCID: PMC3293869 DOI: 10.1371/journal.pone.0032782] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 02/02/2012] [Indexed: 11/24/2022] Open
Abstract
Multidrug resistance mediated by P-glycoprotein in cancer cells has been a major issue that cripples the efficacy of chemotherapy agents. Aimed for improved efficacy against resistant cancer cells, we designed and synthesized 25 oxindole derivatives based on indirubin by structure-activity relationship analysis. The most potent one was named PH II-7, which was effective against 18 cancer cell lines and 5 resistant cell lines in MTT assay. It also significantly inhibited the resistant xenograft tumor growth in mouse model. In cell cycle assay and apoptosis assay conducted with flow cytometry, PH II-7 induced S phase cell cycle arrest and apoptosis even in resistant cells. Consistently revealed by real-time PCR, it modulates the expression of genes related to the cell cycle and apoptosis in these cells, which may contributes to its efficacy against them. By side-chain modification and FITC-labeling of PH II-7, we were able to show with confocal microscopy that not only it was not pumped by P-glycoprotein, it also attenuated the efflux of Adriamycin by P-glycoprotein in MDR tumor cells. Real-time PCR and western blot analysis showed that PH II-7 down-regulated MDR1 gene via protein kinase C alpha (PKCA) pathway, with c-FOS and c-JUN as possible mediators. Taken together, PH II-7 is a dual-functional compound that features both the cytotoxicity against cancer cells and the inhibitory effect on P-gp mediated drug efflux.
Collapse
|
24
|
Liu DL, Bu H, Li H, Chen H, Guo HC, Wang ZH, Tong HF, Ni ZL, Liu HB, Lin SZ. Emodin reverses gemcitabine resistance in pancreatic cancer cells via the mitochondrial apoptosis pathway in vitro. Int J Oncol 2011; 40:1049-57. [PMID: 22159556 PMCID: PMC3584653 DOI: 10.3892/ijo.2011.1285] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 11/10/2011] [Indexed: 11/24/2022] Open
Abstract
Gemcitabine resistance is a common problem of pancreatic cancer chemotherapy, and how to reverse it plays an important role in the treatment of pancreatic cancer. This study investigated the effect of emodin on the gemcitabine-resistant pancreatic cancer cell line SW1990/Gem, and explored the potential mechanism of its action. SW1990/Gem was obtained by culture of the pancreatic cancer cell line SW1990 in vitro by intermittently increasing the concentration of gemcitabine in the culture medium for 10 months, observing the morphology using inverted microscopy. SW1990/Gem cells were pretreated with emodin (10 μM) for different periods followed by treatment with gemcitabine (20 μM) for 48 h; cell proliferation was tested by MTT assay. SW1990/Gem cells were treated by emodin with different concentrations for 48 h, cell apoptosis was detected by flow cytometry (FCM). The expression of gene and protein, such as MDR-1 (P-gp), NF-κB, Bcl-2, Bax, cytochrome-C (cytosol), caspase-9 and -3 were measured by RT-PCR and Western blotting. The function of P-gp in SW1990/Gem cells was checked by FCM. The results showed that the SW1990/Gem cells changed greatly in morphology and the resistance index was 48.63. Emodin promoted cell apoptosis of the gemcitabine-resistant cell line SW1990/Gem in a dose-dependent manner. Emodin enhanced the SW1990/Gem cell sensitivity to gemcitabine in a time-dependent manner. Emodin monotherapy or combination with gemcitabine both decreased the gene and protein expression levels of MDR-1 (P-gp), NF-κB and Bcl-2 and inhibited the function of P-gp, but increased the expression levels of Bax, cytochrome-C (cytosol), caspase-9 and -3, and promoted cell apoptosis. This demonstrated that emodin had a reversing effect on the gemcitabine-resistant cell line SW1990/Gem, possibly via decreasing the function of P-gp and activating the mitochondrial apoptosis pathway in vitro.
Collapse
Affiliation(s)
- Dian-Lei Liu
- Department of Surgery, The Second Affiliated Hospital of Wenzhou Medical College, No 109, West Xue-yuan Road, Wenzhou 325027, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Yan YX, Li WZ, Huang YQ, Liao WX. The COX-2 inhibitor Celecoxib enhances the sensitivity of KB/VCR oral cancer cell lines to Vincristine by down-regulating P-glycoprotein expression and function. Prostaglandins Other Lipid Mediat 2011; 97:29-35. [PMID: 21835258 DOI: 10.1016/j.prostaglandins.2011.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 07/26/2011] [Indexed: 01/16/2023]
Abstract
Previous studies have indicated that long-term chemotherapy decreases the sensitivity of oral cancer cells to chemotherapeutics while simultaneously increasing resistance to these drugs. COX-2 inhibitors are known to enhance the toxic action of anti-tumor drugs against cancer cells. Using the MTT method, we investigated the influence of the COX-2 selective inhibitor Celecoxib on the proliferation of KB/VCR oral cancer cell lines and analyzed the effect of Celecoxib on the regulation of P-glycoprotein (P-gp) expression and function. Western blot analysis was employed to detect the expression of P-gp, and flow cytometry was used to evaluate P-gp function by detecting the accumulation of the active P-gp functional fluorescence substrate within KB/VCR cells. The results revealed that a low dose of Celecoxib (10 μmol/L) showed no growth inhibitory effects on KB/VCR cell lines. When the concentration of Celecoxib was greater than or equal to 20 μmol/L, the inhibitory effect on KB/VCR cells was significantly enhanced in a time- and dose-dependent manner. The lower dose of Celecoxib (10 μmol/L) significantly enhanced the toxicity of Vincristine (VCR) against KB/VCR cell lines. After the application of Celecoxib plus VCR (10 μmol/L+1.5μmol/L, respectively) treatment for 24, 48 or 72 h, the growth inhibition rates of KB/KBV cells were 37.82 ± 1.60%, 47.84 ± 1.29% and 54.43 ± 2.35%, respectively, which were significantly higher than the rates in the cells treated only with Celecoxib (10 μmol/L) or VCR (1.5 μmol/L) (all P<0.01). P-gp expression levels in KB/KBV cells treated with Celecoxib plus VCR (10 μmol/L+1.5 μmol/L, respectively) were markedly lower than the levels in control cells and those treated with VCR (1.5 μmol/L) (all P<0.01). In addition, the intensity of Rho123 fluorescence of KB/KBV cells in cells treated with Celecoxib plus VCR (10 μmol/L+1.5 μmol/L, respectively) or Celecoxib alone (10 μmol/L) was significantly higher than the intensity observed in control cells and those treated with VCR alone (1.5 μmol/L) (all P<0.01). The underlying mechanism of these phenomena is likely correlated with the down-regulation of the expression and function of P-gp due to Celecoxib, thereby increasing the amount of VCR accumulated in KB/VCR cells.
Collapse
Affiliation(s)
- Yi Xuan Yan
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Road, Guangzhou 510515, China.
| | | | | | | |
Collapse
|
26
|
El-Awady RA, Saleh EM, Ezz M, Elsayed AM. Interaction of celecoxib with different anti-cancer drugs is antagonistic in breast but not in other cancer cells. Toxicol Appl Pharmacol 2011; 255:271-86. [PMID: 21763710 DOI: 10.1016/j.taap.2011.06.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 06/09/2011] [Accepted: 06/22/2011] [Indexed: 01/08/2023]
Abstract
Celecoxib, an inhibitor of cyclooxygenase-2, is being investigated for enhancement of chemotherapy efficacy in cancer clinical trials. This study investigates the ability of cyclooxygenase-2 inhibitors to sensitize cells from different origins to several chemotherapeutic agents. The effect of the drug's mechanism of action and sequence of administration are also investigated. The sensitivity, cell cycle, apoptosis and DNA damage of five different cancer cell lines (HeLa, HCT116, HepG2, MCF7 and U251) to 5-FU, cisplatin, doxorubicin and etoposide±celecoxib following different incubation schedules were analyzed. We found antagonism between celecoxib and the four drugs in the breast cancer cells MCF7 following all incubation schedules and between celecoxib and doxorubicin in all cell lines except for two combinations in HCT116 cells. Celecoxib with the other three drugs in the remaining four cell lines resulted in variable interactions. Mechanistic investigations revealed that celecoxib exerts different molecular effects in different cells. In some lines, it abrogates the drug-induced G2/M arrest enhancing pre-mature entry into mitosis with damaged DNA thus increasing apoptosis and resulting in synergism. In other cells, it enhances drug-induced G2/M arrest allowing time to repair drug-induced DNA damage before entry into mitosis and decreasing cell death resulting in antagonism. In some synergistic combinations, celecoxib-induced abrogation of G2/M arrest was not associated with apoptosis but permanent arrest in G1 phase. These results, if confirmed in-vivo, indicate that celecoxib is not a suitable chemosensitizer for breast cancer or with doxorubicin for other cancers. Moreover, combination of celecoxib with other drugs should be tailored to the tumor type, drug and administration schedule.
Collapse
Affiliation(s)
- Raafat A El-Awady
- Pharmacology unit, Department of Cancer Biology, National, Cancer Institute, Cairo University, Fom El-Khalig, Cairo, Egypt.
| | | | | | | |
Collapse
|
27
|
Singh B, Irving LR, Tai K, Lucci A. Overexpression of COX-2 in celecoxib-resistant breast cancer cell lines. J Surg Res 2010; 163:235-43. [PMID: 20691996 DOI: 10.1016/j.jss.2010.04.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 03/29/2010] [Accepted: 04/30/2010] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cyclooxygenase-2 (COX-2) plays a key role in breast cancer progression and metastasis. Effective therapeutic targeting of COX-2 would require the knowledge of whether a tumor is addicted to COX-2, and if we can counter the potential resistance to anti-COX-2 therapy. Herein we tested the hypothesis that celecoxib-resistance involves selection of cancer cells that overexpress COX-2. MATERIALS AND METHODS We selected celecoxib-resistant (CER) variants from two metastatic cell lines, SUM149 inflammatory breast cancer (IBC) cell line and MDA-MB-231-BSC60 cell line, by culturing them in the presence of celecoxib. We measured the relative levels of COX-2 protein and its network components Bcl-2, Bcl-xL, and Bax in the parental cell lines and their CER variants by Western blotting. To determine whether celecoxib resistance would increase tumorigenicity, we performed an in vitro clonogenicity assay. We determined the statistical significance of differences between the groups using the two-sample t-test. RESULTS Both the celecoxib-resistant cell lines SUM149-CER and BSC60-CER produced significantly higher levels of COX-2 protein than their parental counterparts (P < 0.05). The CER variants produced a reduced level of pro-apoptosis protein Bax (both cell lines) and increased levels of anti-apoptosis proteins Bcl-2 (BSC60) or Bcl-xL (SUM149). Importantly, the CER variants had significantly higher clonogenicity than their parental cell lines (P < 0.05). The siRNA-mediated COX-2 knockdown in SUM149-CER cell line resulted in a significant decrease in clonogenicity and in Bcl-xL and Bcl-2 protein levels, thus supporting our hypothesis. CONCLUSION Celecoxib-resistant variant cells present in breast cancer cell lines overexpress COX-2, which is robustly linked with survival pathways and clonogenicity. Since COX-2 is important in the variant cancer cells of aggressive nature, it represents a good therapeutic target.
Collapse
Affiliation(s)
- Balraj Singh
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|