1
|
Liu S, Zhang X, Wang W, Li X, Sun X, Zhao Y, Wang Q, Li Y, Hu F, Ren H. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol Cancer 2024; 23:261. [PMID: 39574178 PMCID: PMC11580516 DOI: 10.1186/s12943-024-02165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024] Open
Abstract
Metabolic alterations, a hallmark of cancer, enable tumor cells to adapt to their environment by modulating glucose, lipid, and amino acid metabolism, which fuels rapid growth and contributes to treatment resistance. In primary breast cancer, metabolic shifts such as the Warburg effect and enhanced lipid synthesis are closely linked to chemotherapy failure. Similarly, metastatic lesions often display distinct metabolic profiles that not only sustain tumor growth but also confer resistance to targeted therapies and immunotherapies. The review emphasizes two major aspects: the mechanisms driving metabolic resistance in both primary and metastatic breast cancer, and how the unique metabolic environments in metastatic sites further complicate treatment. By targeting distinct metabolic vulnerabilities at both the primary and metastatic stages, new strategies could improve the efficacy of existing therapies and provide better outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Shan Liu
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xingda Zhang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenzheng Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Sun
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuqian Zhao
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qi Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingpu Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Fangjie Hu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - He Ren
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
2
|
Zhou K, Zhang M, Zhai D, Wang Z, Liu T, Xie Y, Shi Y, Shi H, Chen Q, Li X, Xu J, Cai Z, Zhang Y, Shao N, Lin Y. Genomic and transcriptomic profiling of inflammatory breast cancer reveals distinct molecular characteristics to non-inflammatory breast cancers. Breast Cancer Res Treat 2024; 208:441-459. [PMID: 39030466 DOI: 10.1007/s10549-024-07437-0] [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: 02/23/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
PURPOSE Inflammatory breast cancer (IBC), a rare and highly aggressive form of breast cancer, accounts for 10% of breast cancer-related deaths. Previous omics studies of IBC have focused solely on one of genomics or transcriptomics and did not discover common differences that could distinguish IBC from non-IBC. METHODS Seventeen IBC patients and five non-IBC patients as well as additional thirty-three Asian breast cancer samples from TCGA-BRCA were included for the study. We performed whole-exon sequencing (WES) to investigate different somatic genomic alterations, copy number variants, and large structural variants between IBC and non-IBC. Bulk RNA sequencing (RNA-seq) was performed to examine the differentially expressed genes, pathway enrichment, and gene fusions. WES and RNA-seq data were further investigated in combination to discover genes that were dysregulated in both genomics and transcriptomics. RESULTS Copy number variation analysis identified 10 cytobands that showed higher frequency in IBC. Structural variation analysis showed more frequent deletions in IBC. Pathway enrichment and immune infiltration analysis indicated increased immune activation in IBC samples. Gene fusions including CTSC-RAB38 were found to be more common in IBC. We demonstrated more commonly dysregulated RAS pathway in IBC according to both WES and RNA-seq. Inhibitors targeting RAS signaling and its downstream pathways were predicted to possess promising effects in IBC treatment. CONCLUSION We discovered differences unique in Asian women that could potentially explain IBC etiology and presented RAS signaling pathway as a potential therapeutic target in IBC treatment.
Collapse
Affiliation(s)
- Kaiwen Zhou
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mengmeng Zhang
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Duanyang Zhai
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zilin Wang
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ting Liu
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yubin Xie
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yawei Shi
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huijuan Shi
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qianjun Chen
- Department of Breast Oncology, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Xiaoping Li
- Department of Breast Oncology, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Juan Xu
- Department of Breast Oncology, Maternal and Child Health Care Hospital of Guangdong Province, Guangzhou, China
| | - Zhenhai Cai
- Department of Breast Oncology, Jieyang People's Hospital, Jieyang, Guangdong, China
| | - Yunjian Zhang
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Nan Shao
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Ying Lin
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
3
|
Nabawi HMS, Abdelazem AZ, El Rouby WMA, El-Shahawy AAG. A potent formula against triple-negative breast cancer-sorafenib-carbon nanotubes-folic acid: Targeting, apoptosis triggering, and bioavailability enhancing. Biotechnol Appl Biochem 2024. [PMID: 39099309 DOI: 10.1002/bab.2649] [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: 02/19/2024] [Accepted: 07/20/2024] [Indexed: 08/06/2024]
Abstract
Triple-negative breast cancer (TNBC) has short survival rates. This study aimed to prepare a novel formula of sorafenib, carbon nanotubes (CNTs), and folic acid to be tested as a drug delivery system targeting versus TNBC compared with free sorafenib and to evaluate the formula stability, in vitro pharmacodynamic, and in vivo pharmacokinetic properties. The formula preparation was done by the synthesis of polyethylene glycol bis amine linker, CNT PEGylation, folic acid attachment, and sorafenib loading. The prepared formula has been characterized using X-ray diffraction, Flourier-transform infrared, 1HNMR, UV, high resolution-transmission electron microscope, field emission scanning electron microscopy, and Zeta potential. In vitro studies included drug release determination, MTT assay, flow cytometry to determine the apoptotic stage with percent, cell cycle analysis, and apoptotic marker assays for caspase-3, 8, 9, cytochrome c, and BCL-2. The in vivo study was performed to determine bioavailability and half-life in rats. The in vitro MTT antiproliferative assay revealed that the formula was threefold more cytotoxic toward TNBC cells than free sorafenib, and the flow cytometry showed a significant increase in apoptosis and necrosis. The formula has a greater inhibitory effect on BCL-2 and a lessening effect on cytochrome c and caspases 3, 8, and 9 than free sorafenib. In vivo experiments proved that our novel formula was superior to free sorafenib by increasing bioavailability by eight times and prolonging the half-life by three times. These results confirmed the successful preparation of the desired formula with better pharmacodynamic and pharmacokinetic properties. These promising results may show a novel therapeutic strategy for TNBC patients.
Collapse
Affiliation(s)
- Hossam M S Nabawi
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| | - Ahmed Z Abdelazem
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| | - Waleed M A El Rouby
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| | - Ahmed A G El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
4
|
Wang Q, Cheng N, Wang W, Bao Y. Synergistic Action of Benzyl Isothiocyanate and Sorafenib in a Nanoparticle Delivery System for Enhanced Triple-Negative Breast Cancer Treatment. Cancers (Basel) 2024; 16:1695. [PMID: 38730647 PMCID: PMC11083210 DOI: 10.3390/cancers16091695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) presents a therapeutic challenge due to its complex pathology and limited treatment options. Addressing this challenge, our study focuses on the effectiveness of combination therapy, which has recently become a critical strategy in cancer treatment, improving therapeutic outcomes and combating drug resistance and metastasis. We explored a novel combination therapy employing Benzyl isothiocyanate (BITC) and Sorafenib (SOR) and their nanoformulation, aiming to enhance therapeutic outcomes against TNBC. Through a series of in vitro assays, we assessed the cytotoxic effects of BITC and SOR, both free and encapsulated. The BITC-SOR-loaded nanoparticles (NPs) were synthesized using an amphiphilic copolymer, which demonstrated a uniform spherical morphology and favorable size distribution. The encapsulation efficiencies, as well as the sustained release profiles at varied pH levels, were quantified, revealing distinct kinetics that were well-modeled by the Korsmeyer-Peppas equation. The NP delivery system showed a marked dose-dependent cytotoxicity towards TNBC cells, with an IC50 of 7.8 μM for MDA-MB-231 cells, indicating improved efficacy over free drugs, while exhibiting minimal toxicity toward normal breast cells. Furthermore, the NPs significantly inhibited cell migration and invasion in TNBC models, surpassing the effects of free drugs. These findings underscore the potential of BITC-SOR-NPs as a promising therapeutic approach for TNBC, offering targeted delivery while minimizing systemic toxicity.
Collapse
Affiliation(s)
- Qi Wang
- Correspondence: (Q.W.); (Y.B.)
| | | | | | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK
| |
Collapse
|
5
|
Liu S, Liu Y, Chang Q, Celia C, Deng X, Xie Y. pH-Responsive Sorafenib/Iron-Co-Loaded Mesoporous Polydopamine Nanoparticles for Synergistic Ferroptosis and Photothermal Therapy. Biomacromolecules 2024; 25:522-531. [PMID: 38087829 DOI: 10.1021/acs.biomac.3c01173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Ferroptosis has attracted significant attention as a new mechanism of cell death. Sorafenib (SRF) is widely considered a prototypical ferroptosis-inducing drug, particularly for liver cancer treatment. However, the low solubility and hydrophobic nature of SRF, along with the absence of synergistic therapeutic strategies, still limit its application in cancer treatment. Herein, we report a dual therapeutic method incorporating photothermal therapy and ferroptosis by using Fe-doped mesoporous polydopamine nanoparticles (Fe-mPDA@SRF-TPP) as a carrier for loading SRF and targeting triphenylphosphine (TPP). SRF molecules are efficiently encapsulated within the polydopamine nanospheres with a high loading ratio (80%) attributed to the porosity of Fe-mPDA, and the inherent biocompatibility and hydrophilicity of Fe-mPDA@SRF-TPP facilitate the transport of SRF to the target cancer cells. Under the external stimuli of acidic environment (pH 5.0), glutathione (GSH), and laser irradiation, Fe-mPDA@SRF-TPP shows sustained release of SRF and Fe ions with the ratio of 72 and 50% within 48 h. Fe-mPDA@SRF-TPP nanoparticles induce intracellular GSH depletion, inhibit glutathione peroxidase 4 (GPX4) activity, and generate hydroxyl radicals, all of which are essential components of the therapeutic ferroptosis process for killing MDA-MB-231 cancer cells. Additionally, the excellent near-infrared (NIR) light absorption of Fe-mPDA@SRF-TPP nanoparticles demonstrates their capability for photothermal therapy and further enhances the therapeutic efficiency. Therefore, this nanosystem provides a multifunctional therapeutic platform that overcomes the therapeutic limitations associated with standalone ferroptosis and enhances the therapeutic efficacy of SRF for breast cancer.
Collapse
Affiliation(s)
- Shang Liu
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Ying Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Qing Chang
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Christian Celia
- Department of Pharmacy, University of Chieti-Pescara "G. d'Annunzio", Chieti 66100, Italy
| | - Xiaoyong Deng
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yijun Xie
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
6
|
Choi S, Dreyfuss I, Taswell CS, Cyriac J, Butkus M, Takita C. Proton Beam Therapy for Breast Cancer. Crit Rev Oncog 2024; 29:67-82. [PMID: 38683154 DOI: 10.1615/critrevoncog.2023050319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Given the radiobiological and physical properties of the proton, proton beam therapy has the potential to be advantageous for many patients compared with conventional radiotherapy by limiting toxicity and improving patient outcomes in specific breast cancer scenarios.
Collapse
Affiliation(s)
- Seraphina Choi
- Department of Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Isabella Dreyfuss
- Department of Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | | | - Jonathan Cyriac
- Department of Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Michael Butkus
- Department of Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | | |
Collapse
|
7
|
Khafaga DSR, El-Khawaga AM, Elfattah Mohammed RA, Abdelhakim HK. Green synthesis of nano-based drug delivery systems developed for hepatocellular carcinoma treatment: a review. Mol Biol Rep 2023; 50:10351-10364. [PMID: 37817020 PMCID: PMC10676320 DOI: 10.1007/s11033-023-08823-5] [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: 05/14/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023]
Abstract
This review presents an overview of one of the effective strategies for improving the anticancer impact of many drugs including sorafenib using a drug delivery system by employing nanoparticles that is produced through a biological system. The biological process has a lot of benefits, including being inexpensive and safe for the environment. Sorafenib is one of a multi-kinase inhibitor that inhibits molecularly targeted kinases. Because of its poor pharmacokinetic characteristics, such as fast elimination and limited water solubility, the bioavailability of Sorafenib is extremely low. More intelligent nano formulations of sorafenib have been developed to boost both the drug's target ability and bioavailability. Researchers in a wide variety of sectors, including nanomedicine, have recently been interested in the topic of nanotechnology. It is possible for the body to develop resistance to widely used drugs available for treatment of liver cancer, including sorafenib. As a result, our goal of this research is to highlight the efficacy of nanomedicine-based drug delivery system to enhance drug's cancer-fighting properties. Because of their magnetic properties, certain nanoparticle materials can be employed as a carrier for the medicine to the exact place where the cancer is located. This can lower the amount of the drug that is administered with no impact on the normal cells.
Collapse
Affiliation(s)
- Doaa S R Khafaga
- Department of Basic Medical Sciences, Faculty of Medicine, Galala University, Suez, 43511, Egypt.
| | - Ahmed M El-Khawaga
- Department of Basic Medical Sciences, Faculty of Medicine, Galala University, Suez, 43511, Egypt.
| | | | - Heba K Abdelhakim
- Biochemistry Division, Faculty of Science, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
8
|
Gupta N, Kumar H, Gupta S, S M B, Saini K. A Concise Review on Natural Products and Their Derivatives for Breast Cancer Treatment. Chem Biodivers 2023; 20:e202300688. [PMID: 37431959 DOI: 10.1002/cbdv.202300688] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/12/2023]
Abstract
Cancer is a leading cause of death worldwide. Among other cancers, breast cancer has been found to produce maximum number of cases in 2020. Different factors including geographical, genetic, hormonal, oral contraceptives and modern lifestyle could be responsible for the development of breast cancer and different pathways can be targeted for breast cancer treatment. The various conventional approaches used for the treatment of breast cancer including radiotherapy, chemotherapy, hormone and immunotherapy. But due to the side effects associated with these conventional treatments such as non-selectivity, multidrug resistance and bioavailability, there is a need for the development of better therapeutic agents for breast cancer treatment. Several natural products have been explored for breast cancer treatment. However, many of these natural products suffered from the limitations of poor water solubility and possess toxic side effects. To overcome these limitations, several structural analogs of natural products have been synthesized and possess potent anti-breast cancer effects with less side effects over their precursor molecules. In the present manuscript, we describe the pathogenesis of breast cancer, some potent natural products used in the treatment of breast cancer and their selected structural analogs possessing potent anti-breast cancer effects. Database such as Science direct, Pubmed and Google scholar were searched using keywords 'risk factors', 'screening methods','receptors', and 'natural products and derivatives', Registered clinical trials on selected natural products were also analyzed. Present study concludes that eight selected natural products and their derivatives possess wide potential to exhibit anti-breast cancer effects and could be explored further to develop better chemotherapeutic agents against breast cancer.
Collapse
Affiliation(s)
- Nidhi Gupta
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India, 133207
| | - Hitesh Kumar
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India, 133207
| | - Sumeet Gupta
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India, 133207
| | - Basavarajaiah S M
- PG Department of Chemistry, Vijaya College, RV Road, Bengaluru, 560004
| | - Kamal Saini
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India, 133207
| |
Collapse
|
9
|
Altalal A, Almomen A, Alkholief M, Binkhathlan Z, Alzoman NZ, Alshamsan A. Development and validation of a UPLC-MS/MS method for simultaneous detection of doxorubicin and sorafenib in plasma: Application to pharmacokinetic studies in rats. Saudi Pharm J 2023; 31:1317-1326. [PMID: 37323919 PMCID: PMC10267530 DOI: 10.1016/j.jsps.2023.05.025] [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: 03/18/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023] Open
Abstract
An ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was developed for the simultaneous quantitation of doxorubicin (DOX) and sorafenib (SOR) in rat plasma. Chromatographic separation was performed using a reversed-phase column C18 (1.7 μm, 1.0 × 100 mm Acquity UPLC BEH™). The gradient mobile phase system consisted of water containing 0.1% acetic acid (mobile phase A) and methanol (mobile phase B) with a flow rate of 0.40 mL/min over 8 min. Erlotinib (ERL) was used as an internal standard (IS). The quantitation of conversion of [M + H]+, which was the protonated precursor ion, to the corresponding product ions was performed using multiple reaction monitoring (MRM) with a mass-to-charge ratio (m/z) of 544 > 397.005 for DOX, 465.05 > 252.03 for SOR, and 394 > 278 for the IS. Different parameters were used to validate the method including accuracy, precision, linearity, and stability. The developed UPLC-MS/MS method was linear over the concentration ranges of 9-2000 ng/mL and 7-2000 ng/mL with LLOQ of 9 and 7 ng/mL for DOX and SOR, respectively. The intra-day and inter-day accuracy, expressed as % relative standard deviation (RSD%), was below 10% for both DOX and SOR in all QC samples that have drug concentrations above the LLOQ. The intra-day and inter-day precision, expressed as percent relative error (Er %), was within the limit of 15.0% for all concentrations above LLOQ. Four groups of Wistar rats (250-280 g) were used to conduct the pharmacokinetic study. Group I received a single intraperitoneal (IP) injection of DOX (5 mg/kg); Group II received a single oral dose of SOR (40 mg/kg), Group III received a combination of both drugs; and Group IV received sterile water for injection IP and 0.9% w/v sodium chloride solution orally to serve as a control. Non-compartmental analysis was used to calculate the different pharmacokinetic parameters. Data revealed that coadministration of DOX and SOR altered some of the pharmacokinetic parameters of both agents and resulted in an increase in the Cmax and AUC and reduction in the apparent clearance (CL/F). In conclusion, our newly developed method is sensitive, specific, and can reliably be used to simultaneously determine DOX and SOR concentrations in rat plasma. Moreover, the results of the pharmacokinetic study suggest that coadministration of DOX and SOR might cause an increase in exposure of both drugs.
Collapse
Affiliation(s)
- Alanoud Altalal
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Aliyah Almomen
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Musaed Alkholief
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Ziyad Binkhathlan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Nourah Z. Alzoman
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Aws Alshamsan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| |
Collapse
|
10
|
Wawszczyk J, Wolan R, Smolik S, Kapral M. In vitro and in silico study on the effect of carvedilol and sorafenib alone and in combination on the growth and inflammatory response of melanoma cells. Saudi Pharm J 2023; 31:1306-1316. [PMID: 37323921 PMCID: PMC10265481 DOI: 10.1016/j.jsps.2023.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Melanoma is an aggressive skin cancer. Increasing evidence has shown the role of β-adrenergic receptors in the pathogenesis of melanoma. Carvedilol is a widely used non-selective β-AR antagonist with potential anticancer activity. The purpose of the study was to estimate the influence of carvedilol and sorafenib alone and in combination on the growth and inflammatory response of C32 and A2058 melanoma cells. Furthermore, this study also aimed to predict the probable interaction of carvedilol and sorafenib when administered together. Predictive study of the interaction of carvedilol and sorafenib was performed using the ChemDIS-Mixture system. Carvedilol and sorafenib alone and in combination showed a growth inhibitory effect on cells. The greatest synergistic antiproliferative effect on both cell lines was observed at Car 5 μM combined with Sor 5 μM. Analysis in silico identified diseases, proteins, and metabolic pathways that can be affected by the interaction of carvedilol and sorafenib. The results obtained demonstrated that carvedilol and sorafenib modulated the secretion of IL-8 by IL-1β-stimulated by melanoma cell lines but the use of a combination of both drugs did not intensify the effect. In summary, the results presented indicate that the combination of carvedilol and sorafenib may have a promising anticancer effect on melanoma cells.
Collapse
|
11
|
Bhat SM, Prasad PR, Joshi MB. Novel insights into DNA methylation-based epigenetic regulation of breast tumor angiogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 380:63-96. [PMID: 37657860 DOI: 10.1016/bs.ircmb.2023.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Breast tumors are highly vascularized and dependent on angiogenesis for growth, progression and metastasis. Like other solid tumors, vasculature in breast tumors also display leaky and tortuous phenotype and hence inhibit immune cell infiltration, show reduced efficacy to anticancer drugs and radiotherapy. Epigenetic reprogramming including significant alterations in DNA methylation in tumor and stromal cells generate an imbalance in expression of pro- and anti-angiogenic factors and subsequently lead to disordered angiogenesis. Hence, understanding DNA methylation-based regulation of angiogenesis in breast tumors may open new avenues for designing therapeutic targets. Our present review manuscript summarized contemporary knowledge of influence of DNA methylation in regulating angiogenesis. Further, we identified novel set of pro-angiogenic genes enriched in endothelial cells which are coregulated with DNMT isoforms in breast tumors and harboring CpG islands. Our analysis revealed promoters of pro-angiogenic genes were hypomethylated and anti-angiogenic genes were hypermethylated in tumors and further reflected on their expression patterns. Interestingly, promoter DNA methylation intensities of novel set of pro-angiogenic genes significantly correlated to patient survival outcome.
Collapse
Affiliation(s)
- Sharath Mohan Bhat
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Palla Ranga Prasad
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
12
|
Chaurawal N, Misra C, Abul Barkat H, Jatyan R, Chitkara D, Barkat MA, Sharma T, Singh B, Raza K. Oral sorafenib-loaded microemulsion for breast cancer: evidences from the in-vitro evaluations and pharmacokinetic studies. Sci Rep 2022; 12:13746. [PMID: 35962160 PMCID: PMC9374710 DOI: 10.1038/s41598-022-17333-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 07/25/2022] [Indexed: 11/09/2022] Open
Abstract
Sorafenib tosylate (SFB) is a multikinase inhibitor that inhibits tumour growth and proliferation for the management of breast cancer but is also associated with issues like toxicity and drug resistance. Also, being a biopharmaceutical class II (BCS II) drug, its oral bioavailability is the other challenge. Henceforth, this report intended to encapsulate SFB into a biocompatible carrier with biodegradable components, i.e., phospholipid. The microemulsion of the SFB was prepared and characterized for the surface charge, morphology, micromeritics and drug release studies. The cell viability assay was performed on 4T1 cell lines and inferred that the IC50 value of sorafenib-loaded microemulsion (SFB-loaded ME) was enhanced compared to the naïve SFB at the concentrations of about 0.75 µM. More drug was available for the pharmacological response, as the protein binding was notably decreased, and the drug from the developed carriers was released in a controlled manner. Furthermore, the pharmacokinetic studies established that the developed nanocarrier was suitable for the oral administration of a drug by substantially enhancing the bioavailability of the drug to that of the free SFB. The results bring forth the preliminary evidence for the future scope of SFB as a successful therapeutic entity in its nano-form for effective and safer cancer chemotherapy via the oral route.
Collapse
Affiliation(s)
- Nishtha Chaurawal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Charu Misra
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Harshita Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia
| | - Reena Jatyan
- Department of Pharmacy, Birla Institute of Technology and Science (BITS)-Pilani, Pilani Campus, Pilani, Vidya Vihar, Rajasthan, 333031, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS)-Pilani, Pilani Campus, Pilani, Vidya Vihar, Rajasthan, 333031, India
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia
| | - Teenu Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India.,Department of Pharmacy, Chandigarh College of Pharmacy, Landran, Punjab, 140307, India
| | - Bhupinder Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Kaisar Raza
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
13
|
Enhancing the Anticancer Activity of Sorafenib through Its Combination with a Nitric Oxide Photodelivering β-Cyclodextrin Polymer. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061918. [PMID: 35335280 PMCID: PMC8953797 DOI: 10.3390/molecules27061918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/07/2022] [Accepted: 03/12/2022] [Indexed: 11/17/2022]
Abstract
In this contribution, we report a strategy to enhance the therapeutic action of the chemotherapeutic Sorafenib (SRB) through its combination with a multifunctional β-cyclodextrin-based polymer able to deliver nitric oxide (NO) and emit green fluorescence upon visible light excitation (PolyCDNO). The basically water-insoluble SRB is effectively encapsulated in the polymeric host (1 mg mL−1) up to a concentration of 18 μg mL−1. The resulting host-guest supramolecular complex is able to release SRB in sink conditions and to preserve very well the photophysical and photochemical properties of the free PolyCDNO, as demonstrated by the similar values of the NO release and fluorescence emission quantum efficiencies found. The complex PolyCDNO/SRB internalizes in HEP-G2 hepatocarcinoma, MCF-7 breast cancer and ACHN kidney adenocarcinoma cells, localizing in all cases mainly at the cytoplasmic level. Biological experiments have been performed at SRB concentrations below the IC50 and with light doses producing NO at nontoxic concentrations. The results demonstrate exceptional mortality levels for PolyCDNO/SRB upon visible light irradiation in all the different cell lines tested, indicating a clear synergistic action between the chemotherapeutic drug and the NO. These findings can open up exciting avenues to potentiate the anticancer action of SRB and, in principle, to reduce its side effects through its use at low dosages when in combination with the photo-regulated release of NO.
Collapse
|
14
|
Wang HL, Ma X, Guan XY, Song C, Li GB, Yu YM, Yang LL. Potential Synthetic Lethality for Breast Cancer: A Selective Sirtuin 2 Inhibitor Combined with a Multiple Kinase Inhibitor Sorafenib. Pharmacol Res 2021; 177:106050. [PMID: 34973468 DOI: 10.1016/j.phrs.2021.106050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/12/2021] [Accepted: 12/27/2021] [Indexed: 02/08/2023]
Abstract
Sorafenib is a clinically useful multiple kinase inhibitor for the treatment of kidney cancer, liver cancer and acute myelocytic leukemia, while it has shown weak efficacy in suppressing breast cancer. Since sirtuin2 (SIRT2) is an important epigenetic regulator and associated with several cancer types including breast cancer, development and evaluation of new SIRT2 inhibitors to probe their therapeutic potentials is currently desirable. A highly selective SIRT2 inhibitor named I was previously developed by us, which showed activity to inhibit non-small cell lung cancer cell lines in vitro. We herein report expanded screening of I and its structurally similar inactive compound II against other cancer cell lines, and found that I had a wide spectrum of anticancer activity while II had no such effects. The I-sorafenib combination treatment exerted obvious synergistic reduction on cell viability of MCF-7 cells. We observed that the combination treatment could suppress cell proliferation, survival and migration, arrest cell cycle at G0/G1 phase, and induce apoptosis in MCF-7 cells, when compared with the single treatment. In vivo studies revealed that the combination treatment showed stronger tumor growth inhibition (87%), comparing with I-(42.8%) or sorafenib-solely-treated groups (61.1%) in MCF-7 xenograft model. In conclusion, this work clearly revealed a potential synthetic lethality effect for I combined with sorafenib, and will probably offer a new strategy at least for breast cancer treatment.
Collapse
Affiliation(s)
- Hua-Li Wang
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, P. R. China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, P. R. China
| | - Xue Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, P. R. China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, P. R. China
| | - Chen Song
- College of Food and Bioengineering, Xihua University, Sichuan 610039, P.R. China
| | - Guo-Bo Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Sichuan 610041, P. R. China
| | - Ya-Mei Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, P. R. China.
| | - Ling-Ling Yang
- College of Food and Bioengineering, Xihua University, Sichuan 610039, P.R. China.
| |
Collapse
|
15
|
El-Fadl HMA, Hagag NM, El-Shafei RA, Khayri MH, El-Gedawy G, Maksoud AIAE, Mohamed DD, Mohamed DD, El Halfawy I, Khoder AI, Elawdan KA, Elshal MF, Salah A, Khalil H. Effective Targeting of Raf-1 and Its Associated Autophagy by Novel Extracted Peptide for Treating Breast Cancer Cells. Front Oncol 2021; 11:682596. [PMID: 34513674 PMCID: PMC8430328 DOI: 10.3389/fonc.2021.682596] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 08/05/2021] [Indexed: 02/03/2023] Open
Abstract
Breast cancer is one of the most common causes of death in women worldwide and has harmful influence on their psychological state during therapy. Multikinase inhibitors have become effective drugs for treating a variety of cancer diseases such as breast cancer. A purified short peptide (H-P) was isolated from the natural honey and tested for its potential regulatory role in breast cancer cells compared with the effectiveness of the anticancer drug, Sorafenib (SOR), using MCF-7, EFM-19, and MCF-10A cell lines. Furthermore, we investigated the direct connection between Raf-1 activation and cellular autophagy as potential targets of SOR and H-P extract using RNA interference. Interestingly, the treatment with H-P showed competitive regulation of phosphorylated Raf-1, MEK1/2, and matched autophagy-related LC3B without any detectable toxic effects in the non-tumorigenic epithelial cells. Unlike SOR, the regulation of Raf-1 protein and autophagic machinery by the novel H-P extract showed neglected levels of the released proinflammatory cytokine. This regulation of cytokine secretion by H-P resulted in decreasing the expression level of the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) in treated cells. Moreover, the transfection of MCF-7 cells with small interference RNA (siRNA) antagonist Raf-1 expression markedly reduced the expression of LC3B, while it increased the expression of NF-kB1 and NF-kB2, indicating the potential cross-link between Raf-1, autophagy, and NF-kB effector. Collectively, these findings suggest that H-P-mediated Raf-1, MEK1/2, LC3B, and NF-kB provide a novel and efficacious multikinase inhibitor for treating breast cancer without detectable cytotoxic effects.
Collapse
Affiliation(s)
- Hebatullah M. Abou El-Fadl
- Genome Department, Animal Health Research Institute, Cairo, Egypt
- Pharmacology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Naglaa M. Hagag
- Genome Department, Animal Health Research Institute, Cairo, Egypt
| | - Reham A. El-Shafei
- Pharmacology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed H. Khayri
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Gamalat El-Gedawy
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menofyia University, Shebin El-Kom, Egypt
| | - Ahmed I. Abd El Maksoud
- Industrial Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Doaa D. Mohamed
- Industrial Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Dalia D. Mohamed
- Industrial Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Ibrahim El Halfawy
- Department of Molecular Diagnostics, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Ahmed I. Khoder
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Khaled A. Elawdan
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Mohamed F. Elshal
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Ahmed Salah
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Hany Khalil
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| |
Collapse
|
16
|
Huang S, Chen Y, Pan L, Fei C, Wang N, Chu F, Peng D, Duan X, Wang Y. Exploration of the Potential Mechanism of Tao Hong Si Wu Decoction for the Treatment of Breast Cancer Based on Network Pharmacology and In Vitro Experimental Verification. Front Oncol 2021; 11:731522. [PMID: 34513708 PMCID: PMC8427760 DOI: 10.3389/fonc.2021.731522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Tao Hong Si Wu Decoction (THSWD) is a well-known traditional Chinese medicine used clinically alone or combined with drugs to treat breast cancer. However, there has been no study to date on the underlying mechanisms of its therapeutic effects. OBJECTIVES To explore the potential mechanism of THSWD for the treatment of breast cancer using network pharmacology and experimental research. METHODS The active ingredients of THSWD were screened according to Lipinski's rule of five based on the 107 ingredients of THSWD identified by UPLC-Q-TOF-MSE. The targets of THSWD and breast cancer from multiple databases were collected, and a Compound-Target-Pathway network based on protein-protein interaction (PPI) was constructed. Gene ontology (GO) analysis and KEGG pathway analysis were performed via the DAVID server. Molecular docking studies verified the selected key ingredients and key targets. The results of network pharmacology were verified by in vitro experiments. Including the effects of THSWD drug-containing rat serum (THSWD serum) on cell proliferation, and on the targets HRAS, MAPK1, AKT1, GRB2, and MAPK14 were assayed by RT-qPCR and Western blot assays. RESULTS In total, 27 active ingredients including 8 core components, were obtained from 107 ingredients and 218 THSWD target genes for the treatment of breast cancer were identified. THSWD is active in the treatment of breast cancer by targeting Ras, FoxO, PI3K-Akt and other signaling pathways. MCF-7 and MDA-MB-231 cell proliferation was inhibited by THSWD serum in a time and concentration dependent manner. THSWD could regulated the RNA and protein expression of core targets HRAS, MAPK1, AKT1, GRB2, and MAPK14 for treatment of breast cancer. CONCLUSION The results of network pharmacology study showed that THSWD is active against breast cancer by intervening with multiple targets and pathways. Luteolin, kaempferol, senkyunolide E, and other 8 compounds may be the core active ingredients of THSWD in the treatment of breast cancer. THSWD treatment of breast cancer may be related to targeting Ras, FoxO, PI3K-Akt, and other signal pathways associated with the core targets HRAS, MAPK1, AKT1, GRB2, and MAPK14.
Collapse
Affiliation(s)
- Shi Huang
- The First Affiliated Hospital of Anhui University of Chinese Medicinee, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yan Chen
- The First Affiliated Hospital of Anhui University of Chinese Medicinee, Hefei, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lingyu Pan
- The First Affiliated Hospital of Anhui University of Chinese Medicinee, Hefei, China
| | - Changyi Fei
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ni Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Furui Chu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Daiyin Peng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xianchun Duan
- The First Affiliated Hospital of Anhui University of Chinese Medicinee, Hefei, China
| | - Yongzhong Wang
- The First Affiliated Hospital of Anhui University of Chinese Medicinee, Hefei, China
| |
Collapse
|
17
|
The genomic architecture of metastasis in breast cancer: focus on mechanistic aspects, signalling pathways and therapeutic strategies. Med Oncol 2021; 38:95. [PMID: 34268641 DOI: 10.1007/s12032-021-01547-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/03/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is a multifactorial, heterogeneous disease and the second most frequent cancer amongst women worldwide. Metastasis is one of the most leading causes of death in these patients. Early-stage or locally advanced breast cancer is limited to the breast or nearby lymph nodes. When breast cancer spreads to farther tissues/organs from its original site, it is referred to as metastatic or stage IV breast cancer. Normal breast development is regulated by specific genes and signalling pathways controlling cell proliferation, cell death, cell differentiation and cell motility. Dysregulation of genes involved in various signalling pathways not only leads to the formation of primary tumour but also to the metastasis as well. The metastatic cascade is represented by a multi-step process including invasion of the local tumour cell followed by its entry into the vasculature, exit of malignant cells from the circulation and ultimately their colonization at the distant sites. These stages are referred to as formation of primary tumour, angiogenesis, invasion, intravasation and extravasation, respectively. The major sites of metastasis of breast cancer are the lymph nodes, bone, brain and lung. Only about 28% five-year survival rate has been reported for stage IV breast cancer. Metastasis is a serious concern for breast cancer and therefore, various therapeutic strategies such as tyrosine kinase inhibitors have been developed to target specific dysregulated genes and various signalling pathways involved in different steps of metastasis. In addition, other therapies like hyperbaric oxygen therapy, RNA interference and CRISPR/Cas9 are also being explored as novel strategies to cure the stage IV/metastatic breast cancer. Therefore, the current review has been compiled with an aim to evaluate the genetic basis of stage IV breast cancer with a focus on the molecular mechanisms. In addition, the therapeutic strategies targeting these dysregulated genes involved in various signalling pathways have also been discussed. Genome editing technologies that can target specific genes in the affected areas by making knock-in and knock-out alternations and thereby bring significant treatment outcomes in breast cancer have also been summarized.
Collapse
|
18
|
Khojasteh Poor F, Keivan M, Ramazii M, Ghaedrahmati F, Anbiyaiee A, Panahandeh S, Khoshnam SE, Farzaneh M. Mini review: The FDA-approved prescription drugs that target the MAPK signaling pathway in women with breast cancer. Breast Dis 2021; 40:51-62. [PMID: 33896802 DOI: 10.3233/bd-201063] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer (BC) is the most common cancer and the prevalent type of malignancy among women. Multiple risk factors, including genetic changes, biological age, dense breast tissue, and obesity are associated with BC. The mitogen-activated protein kinases (MAPK) signaling pathway has a pivotal role in regulating biological functions such as cell proliferation, differentiation, apoptosis, and survival. It has become evident that the MAPK pathway is associated with tumorigenesis and may promote breast cancer development. The MAPK/RAS/RAF cascade is closely associated with breast cancer. RAS signaling can enhance BC cell growth and progression. B-Raf is an important kinase and a potent RAF isoform involved in breast tumor initiation and differentiation. Depending on the reasons for cancer, there are different strategies for treatment of women with BC. Till now, several FDA-approved treatments have been investigated that inhibit the MAPK pathway and reduce metastatic progression in breast cancer. The most common breast cancer drugs that regulate or inhibit the MAPK pathway may include Farnesyltransferase inhibitors (FTIs), Sorafenib, Vemurafenib, PLX8394, Dabrafenib, Ulixertinib, Simvastatin, Alisertib, and Teriflunomide. In this review, we will discuss the roles of the MAPK/RAS/RAF/MEK/ERK pathway in BC and summarize the FDA-approved prescription drugs that target the MAPK signaling pathway in women with BC.
Collapse
Affiliation(s)
- Fatemeh Khojasteh Poor
- Department of Obstetrics and Gynecology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mona Keivan
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Ramazii
- Kerman University of Medical Sciences, University of Kerman, Kerman, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Samira Panahandeh
- School of Health, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
19
|
Silva BR, Rebelo R, Rodrigues JM, Xavier CPR, Vasconcelos MH, Queiroz MJRP. Synthesis of Novel Methyl 3-(hetero)arylthieno[3,2- b]pyridine-2-carboxylates and Antitumor Activity Evaluation: Studies In Vitro and In Ovo Grafts of Chick Chorioallantoic Membrane (CAM) with a Triple Negative Breast Cancer Cell Line. Molecules 2021; 26:molecules26061594. [PMID: 33805741 PMCID: PMC7999514 DOI: 10.3390/molecules26061594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 11/26/2022] Open
Abstract
A series of novel functionalized methyl 3-(hetero)arylthieno[3,2-b]pyridine-2-carboxylates 2a–2h were synthesized by C-C Pd-catalyzed Suzuki-Miyaura cross-coupling of methyl 3-bromothieno[3,2-b]pyridine-2-carboxylate with (hetero)aryl pinacol boranes, trifluoro potassium boronate salts or boronic acids. Their antitumoral potential was evaluated in two triple negative breast cancer (TNBC) cell lines—MDA-MB-231 and MDA-MB-468, by sulforhodamine B assay. Their effects on the non-tumorigenic MCF-12A cells were also evaluated. The results demonstrated that three compounds caused growth inhibition in both TNBC cell lines, with little or no effect against the non-tumorigenic cells. The most promising compound was further studied concerning possible effects on cell viability (by trypan blue exclusion assay), cell proliferation (by bromodeoxyuridine assay) and cell cycle profile (by flow cytometry). The results demonstrated that the GI50 concentration of compound 2e (13 μM) caused a decreased in MDA-MB-231 cell number, which was correlated with a decreased in the % of proliferating cells. Moreover, this compound increased G0/G1 phase and decreased S phases, when compared to control cells (although was not statistic significant). Interestingly, compound 2e also reduced tumor size using an in ovo CAM (chick chorioallantoic membrane) model. This work highlights the potential antitumor effect of a novel methyl 3-arylthieno[3,2-b]pyridine-2-carboxylate derivative.
Collapse
Affiliation(s)
- Bruna R. Silva
- Centre of Chemistry, University of Minho, 4710-057 Braga, Portugal; (B.R.S.); (J.M.R.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (M.H.V.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rita Rebelo
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (M.H.V.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Juliana M. Rodrigues
- Centre of Chemistry, University of Minho, 4710-057 Braga, Portugal; (B.R.S.); (J.M.R.)
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (M.H.V.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence: (C.P.R.X.); (M.-J.R.P.Q.)
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (M.H.V.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria-João R. P. Queiroz
- Centre of Chemistry, University of Minho, 4710-057 Braga, Portugal; (B.R.S.); (J.M.R.)
- Correspondence: (C.P.R.X.); (M.-J.R.P.Q.)
| |
Collapse
|
20
|
Mechanism and effect of stress granule formation in cancer and its potential roles in breast cancer therapy. Genes Dis 2021; 9:659-667. [PMID: 35782985 PMCID: PMC9243343 DOI: 10.1016/j.gendis.2021.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Stress granules are non-membranous cytoplasmic foci induced by various stress conditions. It is a protective strategy used by cells to suppress overall translation during stress. In cancer cells, it was thought that the formation of stress granules could protect them from apoptosis and induces resistance towards anti-cancer drugs or radiation treatment which makes the stress granules a potential target for cancer treatment. However, most of our understanding of stress granules are still in the stage of molecular and cell biology, and a transitional gap for its actual effect on clinical settings remains. In this review, we summarize the mechanism and effect of stress granules formation in cancer and try to illuminate its potential applications in cancer therapy, using breast cancer as an example.
Collapse
|
21
|
Wen Z, Feng Y, Hu Y, Lian L, Huang H, Guo L, Chen S, Yang Q, Zhang M, Wan L, Xu K, Degejirifu, Yan X. Multiwalled carbon nanotubes co-delivering sorafenib and epidermal growth factor receptor siRNA enhanced tumor-suppressing effect on liver cancer. Aging (Albany NY) 2021; 13:1872-1882. [PMID: 33440348 PMCID: PMC7880368 DOI: 10.18632/aging.103905] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/21/2020] [Indexed: 01/04/2023]
Abstract
OBJECTIVE This study aimed to investigate the effects of multiwalled carbon nanotubes (MWNTs) co-delivering sorafenib (Sor) and epidermal growth factor receptor (EGFR) siRNA (MWNT/Sor/siRNA) on tumor growth in liver cancer (LC). RESULTS MWNT/Sor/siRNA was proved to possess increased Sor release, high siRNA stability, and enhanced cellular uptake. In addition, MWNT treatment has few effects on cell proliferation and apoptosis in HepG2 cells; however, MWNT/Sor/siRNA treatment significantly inhibited clone number and induced cell apoptosis, which shows a more favorable antitumor effect than MWNT/Sor and free Sor and free siRNA in HepG2 cells. Moreover MWNT/Sor/siRNA treatment has the most significant antitumor effect in vivo. CONCLUSIONS MWNT/Sor/siRNA exhibited a superior antitumor effect in vitro and in vivo. METHODS The MWNT/Sor and MWNT/Sor/siRNA were prepared, and then the morphologies of MWNT/Sor/siRNA were analyzed. In vitro Sor release assay, siRNA stability and cellular uptake of MWNT/Sor/siRNA were performed as well. Next, the effects of MWNT, free Sor, free siRNA, MWNT/Sor and MWNT/Sor/siRNA were evaluated by colony-forming assay, and cell apoptosis assay in HepG2 cells. Meanwhile, the level of EGFR and proteins associated with apoptosis was tested. Furthermore, the anti-tumor effects of MWNT/Sor/siRNA on LC xenograft mice were also unraveled.
Collapse
Affiliation(s)
- Zhili Wen
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuliang Feng
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Youwen Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lingyan Lian
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongyan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shanwen Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qian Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Moran Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lijun Wan
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kedong Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Degejirifu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| |
Collapse
|
22
|
Shi Y, Zhao Z, Peng K, Gao Y, Wu D, Kim J, Mitragotri S. Enhancement of Anticancer Efficacy and Tumor Penetration of Sorafenib by Ionic Liquids. Adv Healthc Mater 2021; 10:e2001455. [PMID: 33205621 DOI: 10.1002/adhm.202001455] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Ionic liquids (ILs) possess unique solvation and biological properties for drug delivery. Choline and geranic acid (CAGE) in particular, has been successfully formulated to orally deliver insulin and hydrophobic therapeutics such as sorafenib (SRF). However, relatively little is known about the effect of CAGE on intracellular delivery of drugs. Here the effect of low-concentration CAGE (<2 mg mL-1 ) on the delivery of SRF into cancer cells (4T1, PANC-1, and HT29) as well as intestine epithelium cells (Caco-2) is studied. The anti-cancer effect of SRF is enhanced by up to fivefold in the presence of CAGE (0.5 mg mL-1 ). The effect is mediated not by enhancing the cellular uptake of SRF but by improving intracellular SRF retention by inhibiting exocytosis. Moreover, CAGE improves the anti-tumor effect of SRF by increasing apoptosis and blocking cell-cycle progression. Moreover, CAGE significantly enhances the penetration of SRF into and across multicellular constructs with multiple mechanisms involved. Collectively, the administration of ILs such as CAGE combined with SRF may offer a novel therapy to better inhibit tumor progression.
Collapse
Affiliation(s)
- Yujie Shi
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences Peking University Beijing 100191 P. R. China
| | - Zongmin Zhao
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Kevin Peng
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Yongsheng Gao
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Debra Wu
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Jayoung Kim
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| |
Collapse
|
23
|
Gou L, Yue GGL, Puno PT, Lau CBS. A review on the relationship of mast cells and macrophages in breast cancer - Can herbs or natural products facilitate their anti-tumor effects? Pharmacol Res 2020; 164:105321. [PMID: 33285235 DOI: 10.1016/j.phrs.2020.105321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/05/2020] [Accepted: 11/21/2020] [Indexed: 12/27/2022]
Abstract
Breast cancer is an inflammation-related cancer whose tumor microenvironment is largely infiltrated by inflammatory cells. These inflammatory cells including mast cells and macrophages have been elucidated to be vital participants in breast tumor proliferation, survival, invasion and migration. However, the functions of mast cells and macrophages in breast cancer are quite distinct based on recent data. Mast cells exhibit both anti-tumoral and pro-tumoral functions on breast cancer, while high number of tumor-associated macrophages (TAMs) are strongly correlated with poor prognosis and higher risk of distant metastasis in breast cancer patients. Besides, many natural products/extracts have been reported to regulate mast cells and macrophages. In this review, the roles of mast cells and macrophages play in breast cancer are discussed and a summary of those natural products/herbs regulating the functions of mast cells or macrophages is also presented.
Collapse
Affiliation(s)
- Leilei Gou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Grace Gar-Lee Yue
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, HKSAR, China; State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, HKSAR, China
| | - Pema Tenzin Puno
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
| | - Clara Bik-San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, HKSAR, China; State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, HKSAR, China.
| |
Collapse
|
24
|
Dattachoudhury S, Sharma R, Kumar A, Jaganathan BG. Sorafenib Inhibits Proliferation, Migration and Invasion of Breast Cancer Cells. Oncology 2020; 98:478-486. [PMID: 32434184 DOI: 10.1159/000505521] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 12/17/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Metastatic breast cancer has poor prognosis due to limited therapeutic options. Protein kinase dysregulations have a major role in breast cancer progression and metastasis. In this study, we investigated the anti-cancer activity of sorafenib, a multikinase inhibitor, which targets receptor tyrosine kinases in breast cancer. Although treatment with sorafenib has increased the patient survival and inhibited metastatic migration in hepatocellular carcinoma, its role in breast cancer migration, metastasis, and intracellular signaling modulation is unknown. METHODS Breast cancer cell lines MCF7 and MDA-MB-231 were treated with sorafenib and its effect on proliferation, migration, invasion and gene expression was analyzed. RESULTS We found that sorafenib has an anti-proliferative and cytotoxic effect on breast cancer cells. Importantly, sorafenib inhibited the migration and invasion of breast cancer cells in vitro. Mechanistically, sorafenib increased mitochondrial superoxide production, suppressed breast cancer stem cell self-renewal, inhibited epithelial mesenchymal transition and ERK signaling. CONCLUSION Thus, sorafenib has anti-cancer activity against breast cancer cells and could improve the survival of breast cancer patients by inhibiting their invasive and metastatic properties.
Collapse
Affiliation(s)
- Sreeja Dattachoudhury
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Renu Sharma
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Atul Kumar
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Bithiah Grace Jaganathan
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India,
| |
Collapse
|
25
|
Therapeutic aspects of AMPK in breast cancer: Progress, challenges, and future directions. Biochim Biophys Acta Rev Cancer 2020; 1874:188379. [PMID: 32439311 DOI: 10.1016/j.bbcan.2020.188379] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/06/2020] [Accepted: 05/10/2020] [Indexed: 12/17/2022]
Abstract
Breast cancer is the most ubiquitous type of neoplasms among women worldwide. Molecular aberrations associated with breast development and progressions have been extensively investigated in recent years. An AMP-activated kinase (AMPK) initially identified as a cellular energy sensor that plays a crucial role in cellular energy homeostasis. Intensive research over the last decade about the molecular mechanisms of AMPK has demonstrated that AMPK mediated diverse biological functions are achieved through phosphorylation and regulation of multiple downstream signaling molecules in normal tissue. Downregulation of AMPK activity or decreased level involved in the promotion of breast tumorigenesis, and thus activation of AMPK found to oppose tumor progression. In this review, we epitomize the recent advances in exploring the tumor suppressor function of AMPK pathways. Besides, we discuss the developments in the area of AMPK activator and its molecular mechanisms for breast cancer treatment.
Collapse
|
26
|
Karbownik A, Sobańska K, Grabowski T, Stanisławiak-Rudowicz J, Wolc A, Grześkowiak E, Szałek E. In vivo assessment of the drug interaction between sorafenib and paracetamol in rats. Cancer Chemother Pharmacol 2020; 85:1039-1048. [PMID: 32394097 PMCID: PMC7305075 DOI: 10.1007/s00280-020-04075-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
Purpose Sorafenib is a multi-targeted tyrosine kinase inhibitor (TKI) used for the treatment of advanced renal cell carcinoma, hepatocellular carcinoma and radioactive iodine resistant thyroid carcinoma. Neoplastic diseases are the cause of pain, which may occur regardless of the stage of the disease. Paracetamol is a non-opioid analgesic used alone or in combination with opioids for the treatment of cancer pain. Numerous studies have pointed out changes in the pharmacokinetic parameters of TKIs when co-administered with paracetamol. The aim of the study was to assess drug–drug interactions (DDIs) between sorafenib and paracetamol. Methods Rats were divided into three groups, each consisting of eight animals. The first group received sorafenib (IIS), the second group received sorafenib + paracetamol (IS+PA), whereas the third group received only paracetamol (IIIPA). A single dose of sorafenib (100 mg/kg b.w.) and paracetamol (100 mg/kg b.w.) was administered orally. The plasma concentrations of sorafenib and its metabolite–N-oxide as well as paracetamol and its glucuronide and sulphate metabolites were measured using validated high-performance liquid chromatography (HPLC) method with ultraviolet detection. Results The co-administration of sorafenib and paracetamol increased the maximum concentration (Cmax) of paracetamol by 33% (p = 0.0372). In the IS+ PA group the Cmax of paracetamol glucuronide was reduced by 48% (p = < 0.0001), whereas the Cmax of paracetamol sulphate was higher by 153% (p = 0.0012) than in the IIIPA group. Paracetamol increased sorafenib and sorafenib N-oxide Cmax by 60% (p = 0.0068) and 83% (p = 0.0023), respectively. Conclusions A greater knowledge of DDI between sorafenib and paracetamol may help adjust dose properly and avoid toxicity effects in individual patients.
Collapse
Affiliation(s)
- Agnieszka Karbownik
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, 14 Św. Marii Magdaleny Str., 61-861, Poznań, Poland.
| | - Katarzyna Sobańska
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, 14 Św. Marii Magdaleny Str., 61-861, Poznań, Poland
| | - Tomasz Grabowski
- Polpharma Biologics SA, Trzy Lipy 3 Str., 80-172, Gdańsk, Poland
| | | | - Anna Wolc
- Department of Animal Science, Iowa State University, 239E Kildee Hall, Ames, IA, 50011, USA.,Hy-Line International, 2583 240th Street, Dallas Center, IA, 50063, USA
| | - Edmund Grześkowiak
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, 14 Św. Marii Magdaleny Str., 61-861, Poznań, Poland
| | - Edyta Szałek
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, 14 Św. Marii Magdaleny Str., 61-861, Poznań, Poland
| |
Collapse
|
27
|
Li ZY, Yin YF, Guo Y, Li H, Xu MQ, Liu M, Wang JR, Feng ZH, Duan XC, Zhang S, Zhang SQ, Wang GX, Liao A, Wang SM, Zhang X. Enhancing Anti-Tumor Activity of Sorafenib Mesoporous Silica Nanomatrix in Metastatic Breast Tumor and Hepatocellular Carcinoma via the Co-Administration with Flufenamic Acid. Int J Nanomedicine 2020; 15:1809-1821. [PMID: 32214813 PMCID: PMC7083629 DOI: 10.2147/ijn.s240436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/21/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Because tumor-associated inflammation is a hallmark of cancer treatment, in the present study, sorafenib mesoporous silica nanomatrix (MSNM@SFN) co-administrated with flufenamic acid (FFA, a non-steroidal anti-inflammatory drug (NSAID)) was investigated to enhance the anti-tumor activity of MSNM@SFN. METHODS Metastatic breast tumor 4T1/luc cells and hepatocellular carcinoma HepG2 cells were selected as cell models. The effects of FFA in vitro on cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in 4T1/luc and HepG2 cells were investigated. The in vivo anti-tumor activity of MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) was evaluated in a 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model, respectively. RESULTS The results indicated that FFA could markedly decrease cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in both 4T1/luc and HepG2 cells. The enhanced anti-tumor activity of MSNM@SFN+FFA compared with that of MSNM@SFN was confirmed in the 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model in vivo, respectively. DISCUSSION MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) developed in this study is an alternative strategy for improving the therapeutic efficacy of MSNM@SFN via co-administration with NSAIDs.
Collapse
Affiliation(s)
- Zhuo-Yue Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Yi-Fan Yin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Yang Guo
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Hui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Mei-Qi Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Man Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Jing-Ru Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Zhen-Han Feng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Xiao-Chuan Duan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shuang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shuai-Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Guang-Xue Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Ai Liao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shu-Min Wang
- Department of Ultrasound, Peking University Third Hospital, Peking University, Beijing100191, People’s Republic of China
| | - Xuan Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| |
Collapse
|
28
|
Transferrin-Modified Nanoliposome Codelivery Strategies for Enhancing the Cancer Therapy. J Pharm Sci 2019; 109:2426-2436. [PMID: 31760084 DOI: 10.1016/j.xphs.2019.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/22/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023]
Abstract
Chemotherapy remains one of the most effective treatments for many cancers in a clinic. At present, various targets have been used to modify the PEGylated liposomes for doxorubicin (Dox) delivery, but the antitumor effect of Dox is not satisfactory. Therefore, combination chemotherapeutics has been considered as a promising method to improve tumor treatment. These years, RAF/MEK/ERK-mediated cell signaling pathway has been discovered to inhibit the growth of tumors. Thus, Sorafenib tosylate (Sor) was used in this study, which directly inhibited tumor cell proliferation through blocking RAF/MEK/ERK-mediated cell signaling pathway and indirectly inhibited tumor cell growth through blocking angiogenesis by VEGFR and PDGF. In this article, we develop a "combination delivery system" to deliver the hydrophobic drug (Sor) in phospholipid bilayer and hydrophilic drug (Dox) in inner cores for enhancing the antitumor effect. Moreover, in vitro experiments verified whether the physicochemical properties of carriers were stable and transferrin-modified liposomes displayed the highest uptake. The results of in vivo experiments showed that the codelivery system inhibited the tumor growth more effectively than monotherapy. Overall, this combination delivery system for delivering the hydrophobic and hydrophilic drugs simultaneously may offer a novel strategy for breast cancer treatment and provide a reference for the possibility of clinical usage.
Collapse
|
29
|
Faranda T, Grossi I, Manganelli M, Marchina E, Baiocchi G, Portolani N, Crosatti M, De Petro G, Salvi A. Differential expression profiling of long non-coding RNA GAS5 and miR-126-3p in human cancer cells in response to sorafenib. Sci Rep 2019; 9:9118. [PMID: 31235746 PMCID: PMC6591391 DOI: 10.1038/s41598-019-45604-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) and microRNAs are involved in numerous physio-pathological conditions included cancer. To better understand the molecular mechanism of the oral antitumor multikinase inhibitor sorafenib, we profiled the expression of a panel of lncRNAs and miRNAs by qPCR array in a sorafenib-treated hepatocellular carcinoma (HCC) cell line. Among the most affected ncRNAs, we found that sorafenib mediated the dysregulation of the lncRNAs GAS5, HOTTIP and HOXA-AS2 and the miR-126-3p, in a panel of human cancer cell lines (HCC, renal and breast carcinomas). By luciferase gene reporter assay, we discovered that GAS5 may act as a sponge for miR-126-3p in HCC cells. The expression level of GAS5 and miR-126-3p was verified in human liquid and/or solid biopsies from HCC patients. miR-126-3p expression in HCC tissues was decreased respect to their correspondent peritumoral tissues. The levels of plasmatic circulating miR-126-3p and GAS5 were significantly higher and lower in HCC patients compared to healthy subjects, respectively. This study highlighted the capability of sorafenib to modulate the expression of a wide range of ncRNAs and specifically, GAS5 and miR-126-3p were involved in the response to sorafenib of different cancer cell types.
Collapse
Affiliation(s)
- Teresa Faranda
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Ilaria Grossi
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Michele Manganelli
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Eleonora Marchina
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Gianluca Baiocchi
- Department of Clinical and Experimental Sciences, Surgical Clinic, University of Brescia, Brescia, Italy
| | - Nazario Portolani
- Department of Clinical and Experimental Sciences, Surgical Clinic, University of Brescia, Brescia, Italy
| | | | - Giuseppina De Petro
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Alessandro Salvi
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy.
| |
Collapse
|
30
|
El Sayed R, El Jamal L, El Iskandarani S, Kort J, Abdel Salam M, Assi H. Endocrine and Targeted Therapy for Hormone-Receptor-Positive, HER2-Negative Advanced Breast Cancer: Insights to Sequencing Treatment and Overcoming Resistance Based on Clinical Trials. Front Oncol 2019; 9:510. [PMID: 31281796 PMCID: PMC6597942 DOI: 10.3389/fonc.2019.00510] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/28/2019] [Indexed: 01/30/2023] Open
Abstract
Background: Advanced hormone-receptor positive HER2 negative breast cancer is a common and a very heterogeneous disease. Hormone therapy is the main first line treatment of choice, given alone or in combination with other agents that have shown to improve patient outcomes, Nevertheless, treatment remains generally palliative rather than curative. Sequencing of such treatment remains challenging, especially with resurgence of variable resistance patterns. Multiple attempts have been made to overcome resistance and improve patient survival, yet resistance remains not very well understood and metastatic cancer remains a disease with dismal prognosis. Methods: In this paper, we searched pubmed database as well as local and international meetings for all studies discussing advanced and metastatic hormone-receptor-positive, her2-negative breast cancer, hormonal treatment, resistance to hormonal treatment, mechanism of resistance, and means to overcome such resistance. Conclusion: There does not exist an optimal treatment sequence for hormone-receptor-positive, her2-negative advanced breast cancer. However, after review of literature, a reasonable approach may be starting with tamoxifen, aromatase inhibitors, or fulvestrant in absence of visceral crisis, in addition to ensuring adequate ovarian function suppression in pre/peri-menopausal women. Aromatase inhibitors and fulvestrant seem to be superior. Resistance to such agents is increasing, mostly attributed to genetic and molecular changes. Multiple modalities are addressed to overcome such resistance including use of CKD4/6 inhibitors, mTOR inhibitors and PI3K inhibitors in addition to other agents under study, all with promising results. CDK4/6 inhibitors work best when used in frontline setting. Finally, treatment of breast cancer remains a growing field, and more studies are to be awaited.
Collapse
Affiliation(s)
- Rola El Sayed
- Oncology Division, Department of Internal Medicine, American University of Beirut Medical Center, American University of Beirut, Beirut, Lebanon
| | - Lara El Jamal
- School of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Jeries Kort
- Oncology Division, Department of Internal Medicine, American University of Beirut Medical Center, American University of Beirut, Beirut, Lebanon
| | | | - Hazem Assi
- Oncology Division, Department of Internal Medicine, American University of Beirut Medical Center, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
31
|
Li J, Halfter K, Zhang M, Saad C, Xu K, Bauer B, Huang Y, Shi L, Mansmann UR. Computational analysis of receptor tyrosine kinase inhibitors and cancer metabolism: implications for treatment and discovery of potential therapeutic signatures. BMC Cancer 2019; 19:600. [PMID: 31208363 PMCID: PMC6580552 DOI: 10.1186/s12885-019-5804-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 06/06/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Receptor tyrosine kinase (RTK) inhibitors are frequently used to treat cancers and the results have been mixed, some of these small molecule drugs are highly successful while others show a more modest response. A high number of studies have been conducted to investigate the signaling mechanisms and corresponding therapeutic influence of RTK inhibitors in order to explore the therapeutic potential of RTK inhibitors. However, most of these studies neglected the potential metabolic impact of RTK inhibitors, which could be highly associated with drug efficacy and adverse effects during treatment. METHODS In order to fill these knowledge gaps and improve the therapeutic utilization of RTK inhibitors a large-scale computational simulation/analysis over multiple types of cancers with the treatment responses of RTK inhibitors was performed. The pharmacological data of all eight RTK inhibitor and gene expression profiles of 479 cell lines from The Cancer Cell Line Encyclopedia were used. RESULTS The potential metabolic impact of RTK inhibitors on different types of cancers were analyzed resulting in cancer-specific (breast, liver, pancreas, central nervous system) metabolic signatures. Many of these are in line with results from different independent studies, thereby providing indirect verification of the obtained results. CONCLUSIONS Our study demonstrates the potential of using a computational approach on signature-based-analysis over multiple cancer types. The results reveal the strength of multiple-cancer analysis over conventional signature-based analysis on a single cancer type.
Collapse
Affiliation(s)
- Jian Li
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University München, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kathrin Halfter
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University München, Munich, Germany
| | - Mengying Zhang
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University München, Munich, Germany
| | - Christian Saad
- Department of Computational Science, University of Augsburg, Augsburg, Germany
| | - Kai Xu
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Bernhard Bauer
- Department of Computational Science, University of Augsburg, Augsburg, Germany
| | - Yijiang Huang
- Department of Orthopaedics, Physical Medicine and Rehabilitation, University Hospital, LMU, Munich, Germany
| | - Lei Shi
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Ulrich R. Mansmann
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University München, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
32
|
Elwakeel A, Soudan H, Eldoksh A, Shalaby M, Eldemellawy M, Ghareeb D, Abouseif M, Fayad A, Hassan M, Saeed H. Implementation of the Chou-Talalay method for studying the in vitro pharmacodynamic interactions of binary and ternary drug combinations on MDA-MB-231 triple negative breast cancer cells. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.synres.2019.100047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
33
|
Abdelgalil AA, Alkahtani HM, Al-Jenoobi FI. Sorafenib. PROFILES OF DRUG SUBSTANCES, EXCIPIENTS, AND RELATED METHODOLOGY 2019; 44:239-266. [PMID: 31029219 DOI: 10.1016/bs.podrm.2018.11.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sorafenib (BAY-43-9006), marketed by Bayer as Nexavar® (USA), is anticancer drug approved by US-FDA for the treatment of unresectable hepatocellular carcinoma and advanced renal cell carcinoma. Sorafenib inhibited tumor growth and angiogenesis through targeting both the RAF/MEK/ERK pathway and receptor tyrosine kinases. This study presents a comprehensive profile of sorafenib, including detailed nomenclature, formula, elemental analysis, methods of preparation, physico-chemical characteristics, and methods of analysis (including spectroscopic, electrochemical, and chromatographic methods of analysis). Spectroscopic and spectrometric analyses include UV/vis spectroscopy, vibrational spectroscopy, nuclear magnetic resonance spectrometry ((1)H and (13)C NMR), and mass spectrometry. Chromatographic methods of analyses include thin layer chromatography and high-performance liquid chromatography. Only few stability indicating methods were found for quantification of sorafenib after exposing tablet dosage form to various stress conditions such as hydrolysis, oxidation, thermal stress, photo and UV light. However, none of these described methods were made to separate and quantify the degradation products. Pharmacology studies including pharmacodynamics, mechanism of action, pharmacokinetics and drug-drug interactions were also presented. An appropriate table and figures were attached to each of the above mentioned sections along with total of 55 references.
Collapse
Affiliation(s)
- Ahmed A Abdelgalil
- Central Laboratory, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | - Hamad M Alkahtani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fahad I Al-Jenoobi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Oruç Z, Kaplan MA, Arslan Ç. An update on the currently available and future chemotherapy for treating bone metastases in breast cancer patients. Expert Opin Pharmacother 2018; 19:1305-1316. [PMID: 30129373 DOI: 10.1080/14656566.2018.1504922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Bone metastases in breast cancer patients are a common clinical problem. Many factors influence the treatment decision, including tumor characteristics, previous treatment and tumor burden in the treatment of metastatic breast cancer. AREAS COVERED This present review summarizes the new treatment strategies and the chemotherapeutic agents currently available in the management of metastatic breast cancer with bone metastases. EXPERT OPINION Patients with bone metastases more often have hormone receptor-positive tumours. Although new treatment agents for metastatic breast cancer have been investigated, endocrine therapy is still considered as the treatment of choice for patients with bone metastases although chemotherapy still has an important place. In recent years, new chemotherapeutic agents such as etirinotecan and nab-paclitaxel have been established though there are few studies that have looked at particular types of metastases. In the last decade, therapies for bone metastasis resistant to endocrine therapy have predominantly focused on radiotherapy, surgical resection, chemotherapy, bone-targeting radiopharmaceuticals and targeted therapeutics. New targeted agents include: Src inhibitors, cathepsin K inhibitors, CXCR4 inhibitors, TGF-B blockade and integrin antagonists while drug delivery systems for chemotherapy have also been developed. These new treatment options could be future treatment options for bone metastatic disease if early promising results are confirmed by clinical trials.
Collapse
Affiliation(s)
- Zeynep Oruç
- a Department of Medical Oncology , Mersin City Hospital , Mersin , Turkey
| | - Muhammet Ali Kaplan
- b Department of Medical Oncology , Dicle University Faculty of Medicine , Diyarbakır , Turkey
| | - Çağatay Arslan
- c Department of Medical Oncology , Medical Park Hospital , Izmir , Turkey.,d Faculty of Medicine , Bahcesehir University , Istanbul , Turkey
| |
Collapse
|
35
|
Zanotto-Filho A, Rajamanickam S, Loranc E, Masamsetti VP, Gorthi A, Romero JC, Tonapi S, Gonçalves RM, Reddick RL, Benavides R, Kuhn J, Chen Y, Bishop AJR. Sorafenib improves alkylating therapy by blocking induced inflammation, invasion and angiogenesis in breast cancer cells. Cancer Lett 2018; 425:101-115. [PMID: 29608984 DOI: 10.1016/j.canlet.2018.03.037] [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: 07/12/2017] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022]
Abstract
Molecular targeted compounds are emerging as a strategy to improve classical chemotherapy. Herein, we describe that using low dose of the multikinase inhibitor sorafenib improves cyclophosphamide antitumor activity by inhibiting angiogenesis, metastasis and promoting tumor healing in MDA-MB231 xenografts and the 4T1-12B syngeneic breast cancer metastasis model. Mechanistic studies in MDA-MB231 cells revealed that alkylation upregulates inflammatory genes/proteins such as COX-2, IL8, CXCL2 and MMP1 in a MEK1/2-ERK1/2-dependent manner. These proteins enrich the secretome of cancer cells, stimulating cell invasion and angiogenesis via autocrine and paracrine mechanisms. Sorafenib inhibits MEK1/2-ERK1/2 pathway thereby decreasing inflammatory genes and mitigating cell invasion and angiogenesis at basal and alkylation-induced conditions whereas NRF2 and ER stress pathways involved in alkylation survival are not affected. In non-invasive/non-angiogenic breast cancer cells (SKBR3 and MCF7), alkylation did not elicit inflammatory responses with the only sorafenib effect being ERK1/2-independent ROS-dependent cytotoxicity when using higher drug concentrations. In summary, our data show that alkylating agents may elicit inflammatory responses that seems to contribute to malignant progression in specific breast cancer cells. Identifying and targeting drivers of this phenotype may offer opportunities to optimize combined drug regimens between classical chemotherapeutics and targeted agents.
Collapse
Affiliation(s)
- Alfeu Zanotto-Filho
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Departamento de Farmacologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Subapriya Rajamanickam
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Eva Loranc
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - V Pragathi Masamsetti
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Aparna Gorthi
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - July Carolina Romero
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Sonal Tonapi
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Rosangela Mayer Gonçalves
- Departamento de Farmacologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Robert L Reddick
- Department of Pathology, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Raymond Benavides
- Department of Pathology, University of Texas College of Pharmacy, Austin, TX, USA
| | - John Kuhn
- Department of Pathology, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Pathology, University of Texas College of Pharmacy, Austin, TX, USA
| | - Yidong Chen
- Department of Epidemiology and Biostatistics, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Alexander J R Bishop
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
36
|
Cousins FL, O DF, Gargett CE. Endometrial stem/progenitor cells and their role in the pathogenesis of endometriosis. Best Pract Res Clin Obstet Gynaecol 2018; 50:27-38. [PMID: 29503126 DOI: 10.1016/j.bpobgyn.2018.01.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/23/2018] [Indexed: 01/10/2023]
Abstract
Human endometrium regenerates on a cyclical basis each month, likely mediated by endometrial stem/progenitor cells. Several types of stem/progenitor cells have been identified: CD140b+CD146+ or SUSD2+ endometrial mesenchymal stem cells (eMSCs), N-cadherin+ endometrial epithelial progenitor cells (eEPs), and side population (SP) cells, a heterogeneous population predominantly comprising endothelial cells. eMSCs reside in a perivascular niche and likely mediate angiogenesis and stromal regeneration. Human eEPs are located in the bases of glands in the basalis and are likely more primitive than SSEA-1+ basalis epithelial cells. Endometrial stem/progenitor cells may contribute to the pathogenesis of endometriosis by their retrograde shedding into the pelvic cavity, either after menarche or as a result of neonatal uterine bleeding. eMSCs may have a role in the generation of progesterone-resistant phenotype of endometrial stromal fibroblasts (eSFs) in endometriosis. In future clinical practice, endometrial stem/progenitor cells may be used to establish diagnosis of endometriosis or as therapeutic targets.
Collapse
Affiliation(s)
- Fiona L Cousins
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3800, Australia
| | - Dorien F O
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; KU Leuven-University of Leuven, University Hospitals Leuven, Department of Development and Regeneration, Organ Systems, B-3000, Leuven, Belgium
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
37
|
Zhang J, Yu K, Han X, Zhen L, Liu M, Zhang X, Ren Y, Shi J. Paeoniflorin influences breast cancer cell proliferation and invasion via inhibition of the Notch‑1 signaling pathway. Mol Med Rep 2017; 17:1321-1325. [PMID: 29115554 DOI: 10.3892/mmr.2017.8002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 05/23/2017] [Indexed: 11/06/2022] Open
Abstract
Breast cancer is one of the most frequently occurring malignant tumors affecting women's health. At least one million new cases are diagnosed each year. Therefore, research that aims to identify strategies that inhibit the growth of breast cancer cells has become a primary worldwide focus. Traditional Chinese medicine (TCM) is regarded as a valuable resource in China, and numerous monomer compositions extracted from TCMs have been demonstrated to exhibit antitumor effects. The present study aimed to determine the impact of paeoniflorin (PF) on breast cancer cell proliferation and invasion, and to explore the mechanisms underlying its effects. Different concentrations of PF were applied to MCF‑7 cells at various time points and the Cell Counting kit‑8 assay was used to determine cell proliferation, a transwell invasion assay was employed to determine cell invasion, reverse transcription‑polymerase chain reaction was used to determine notch homolog‑1 (NOTCH‑1) and Hes family basic helix‑loop helix transcription factor (HES)‑1 mRNA expression levels, and western blotting was used to determine NOTCH‑1 and HES‑1 protein expression levels. The results demonstrated that PF inhibited the proliferation of MCF‑7 cells in a dose‑ and time‑dependent manner. Following treatment with different concentrations of PF, the total number of cells present in the PF‑treated groups was significantly lower when compared with the untreated control group (P<0.05). With increasing doses of PF, the rate of cell invasion significantly decreased, indicating a dose‑dependent association. NOTCH‑1 and HES‑1 mRNA expression levels were reduced when compared with the untreated control group, which reached a statistical significance following treatment with 15 and 30 µM PF (P<0.05). NOTCH‑1 and HES‑1 protein levels demonstrated a similar trend to the mRNA levels, whereby an increase in the concentration of PF was associated with a decrease in NOTCH‑1 and HES‑1 protein expression levels. The results of the present study therefore suggest that PF may inhibit the proliferation and invasiveness of breast cancer cells via inhibition of the NOTCH‑1 signaling pathway.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Breast Surgery, Xuzhou Tumor Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Kun Yu
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Xuedong Han
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Linlin Zhen
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Minmin Liu
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Xiwen Zhang
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Yi Ren
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Jianhua Shi
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
38
|
Shang J, Xu S, Zhang J, Ran X, Bai L, Tang H. Efficacy of sorafenib in patients with hepatocellular carcinoma after resection: a meta-analysis. Oncotarget 2017; 8:109723-109731. [PMID: 29312642 PMCID: PMC5752555 DOI: 10.18632/oncotarget.21299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 09/13/2017] [Indexed: 02/05/2023] Open
Abstract
Background The prognosis of hepatocellular carcinoma remains poor even after curative resection and it has no effective adjuvant therapy. Aim This meta-analysis aimed to assess efficacy of sorafenib as adjuvant therapy for patients with hepatocellular carcinoma after resection. Materials and methods A systematic search was conducted of Medline, Embase, Web of Science, Cochrane Library, Chinese Wanfang database, Chinese biological and medical database, China National Knowledgeand the Internet, data from 5 studies that included 296 participants were analyzed. The primary outcome was overall survival. Secondary outcomes included recurrence rate and mortality rate. Results In the comparison of sorafenib versus control, no significant difference in overall survival (hazard ratio 1.39, 95% confidence interval [CI] 0.71-2.74, P = 0.34) or recurrence rate [risk ratio (RR) 0.81, 95% CI; 0.65-1.01, P = 0.06) was found. For mortality rate, subgroup analysis was conducted according to study type, only in subgroup 2, the RR was significantly reduced (0.66, 95% CI; 0.51-0.87, P = 0.003) in studies. Conclusions In this meta-analysis, sorafenib achieves no significant benefit in any of the endpoints except a lower mortality rate in subgroup analysis, indicating that there is no convincing evidence of sorafenib as an effective adjuvant therapy in patients with hepatocellular carcinoma after resection.
Collapse
Affiliation(s)
- Jin Shang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shanling Xu
- Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiaxing Zhang
- Chinese Evidence-based Medicine Center, Sichuan University, Chengdu, China
| | - Xuting Ran
- West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Bronte G, Andreis D, Bravaccini S, Maltoni R, Cecconetto L, Schirone A, Farolfi A, Fedeli A, Serra P, Donati C, Amadori D, Rocca A. Sorafenib for the treatment of breast cancer. Expert Opin Pharmacother 2017; 18:621-630. [PMID: 28335647 DOI: 10.1080/14656566.2017.1309024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Breast cancer treatment includes many options depending on the tumor clinicopathological profile, which groups breast cancer into various subtypes. Bevacizumab is currently the only drug capable of targeting angiogenesis in breast cancer. Sorafenib has also been studied in combination with other agents. Areas covered: Pharmacological aspects of sorafenib, including results from preclinical studies on breast cancer cells; findings about clinical efficacy and safety in both single-arm and randomized clinical trials; ongoing trials. Expert opinion: Since sorafenib as a single agent has shown limited efficacy in breast cancer, its combination with other drugs is under investigation. Dose reduction is the main challenge when sorafenib is combined with chemotherapy or endocrine therapy. Although randomized phase-II trials on sorafenib plus chemotherapy versus chemotherapy alone have shown potential benefits in progression-free survival, preliminary results from a phase-III study in combination with capecitabine are negative. The definitive results of this trial and results from other ongoing phase-II trials will determine further developments of sorafenib in breast cancer. Although these additional data could help determine the most appropriate dose, drug combination and patient settings, a confirmation of the preliminary negative results reported in the phase-III trial are likely to discourage further use of sorafenib in breast cancer, given its non-negligible toxicity, lack of predicting markers, and the number of more promising drugs for breast cancer.
Collapse
Affiliation(s)
- Giuseppe Bronte
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Daniele Andreis
- b Unit of Biostatistics and Clinical Trials , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Sara Bravaccini
- c Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Roberta Maltoni
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Lorenzo Cecconetto
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Alessio Schirone
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Alberto Farolfi
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Anna Fedeli
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Patrizia Serra
- b Unit of Biostatistics and Clinical Trials , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Caterina Donati
- d Oncology Pharmacy , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Dino Amadori
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Andrea Rocca
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| |
Collapse
|