1
|
Wang Y, Zhang T, Song H, Yang C. Lapatinib ameliorates skin fibrosis by inhibiting TGF-β1/Smad and non-Smad signaling pathway. Sci Rep 2025; 15:8444. [PMID: 40069312 PMCID: PMC11897129 DOI: 10.1038/s41598-025-92687-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Skin fibrosis, characterized by excessive accumulation of extracellular matrix (ECM) in the dermis, can lead to hypertrophic scars and impaired mobility. The ErbB family of receptor tyrosine kinases, including ErbB1 and ErbB2, plays a crucial role in organ fibrosis, but their specific impact on skin fibrosis is less understood. This study investigated the role of ErbB1 and ErbB2 in skin fibrosis and the therapeutic potential of lapatinib, a dual ErbB1 and ErbB2 tyrosine kinase inhibitor. Using qPCR, cell culture assays, Western blotting, and in vivo models, we found significant upregulation of ErbB1 and ErbB2 in keloid tissues and fibroblasts. Lapatinib treatment resulted in a dose-dependent decrease in ErbB1 and ErbB2 expression, which suppressed the expression of fibroblast activation markers. Our findings suggest that lapatinib may be a promising therapeutic agent for skin fibrosis by targeting ErbB1/ErbB2 and modulating the TGF-β1/Smad2/3/Erk/Akt signalling pathways. These results warrant further clinical investigation into lapatinib for treating skin fibrosis and related conditions.
Collapse
Affiliation(s)
- Yongping Wang
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Tiantian Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Hao Song
- Frontier Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China.
| |
Collapse
|
2
|
Lozon L, Ramadan WS, Kawaf RR, Al-Shihabi AM, El-Awady R. Decoding cell death signalling: Impact on the response of breast cancer cells to approved therapies. Life Sci 2024; 342:122525. [PMID: 38423171 DOI: 10.1016/j.lfs.2024.122525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/04/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Breast cancer is a principal cause of cancer-related mortality in female worldwide. While many approved therapies have shown promising outcomes in treating breast cancer, understanding the intricate signalling pathways controlling cell death is crucial for optimizing the treatment outcome. A growing body of evidence has unveiled the aberrations in multiple cell death pathways across diverse cancer types, highlighting these pathways as appealing targets for therapeutic interventions. In this review, we provide a comprehensive overview of the current state of knowledge on the cell death signalling mechanisms with a particular focus on their impact on the response of breast cancer cells to approved therapies. Additionally, we discuss the potentials of combination therapies that exploit the synergy between approved drugs and therapeutic agents targeting modulators of cell death pathways.
Collapse
Affiliation(s)
- Lama Lozon
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Wafaa S Ramadan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Rawan R Kawaf
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Aya M Al-Shihabi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Raafat El-Awady
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
3
|
Shaban N, Raevskiy M, Zakharova G, Shipunova V, Deyev S, Suntsova M, Sorokin M, Buzdin A, Kamashev D. Human Blood Serum Counteracts EGFR/HER2-Targeted Drug Lapatinib Impact on Squamous Carcinoma SK-BR-3 Cell Growth and Gene Expression. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:487-506. [PMID: 38648768 DOI: 10.1134/s000629792403009x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/17/2024] [Accepted: 02/20/2024] [Indexed: 04/25/2024]
Abstract
Lapatinib is a targeted therapeutic inhibiting HER2 and EGFR proteins. It is used for the therapy of HER2-positive breast cancer, although not all the patients respond to it. Using human blood serum samples from 14 female donors (separately taken or combined), we found that human blood serum dramatically abolishes the lapatinib-mediated inhibition of growth of the human breast squamous carcinoma SK-BR-3 cell line. This antagonism between lapatinib and human serum was associated with cancelation of the drug induced G1/S cell cycle transition arrest. RNA sequencing revealed 308 differentially expressed genes in the presence of lapatinib. Remarkably, when combined with lapatinib, human blood serum showed the capacity of restoring both the rate of cell growth, and the expression of 96.1% of the genes expression of which were altered by the lapatinib treatment alone. Co-administration of EGF with lapatinib also restores the cell growth and cancels alteration of expression of 95.8% of the genes specific to lapatinib treatment of SK-BR-3 cells. Differential gene expression analysis also showed that in the presence of human serum or EGF, lapatinib was unable to inhibit the Toll-Like Receptor signaling pathway and alter expression of genes linked to the Gene Ontology term of Focal adhesion.
Collapse
Affiliation(s)
- Nina Shaban
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
- The National Medical Research Center for Endocrinology, Moscow, 117036, Russia
| | - Mikhail Raevskiy
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, 119991, Russia.
| | - Galina Zakharova
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, 119991, Russia.
| | - Victoria Shipunova
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Sergey Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia.
- "Biomarker" Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, 420008, Russia
| | - Maria Suntsova
- The National Medical Research Center for Endocrinology, Moscow, 117036, Russia.
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Maksim Sorokin
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia.
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, 1200, Belgium
| | - Anton Buzdin
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
- The National Medical Research Center for Endocrinology, Moscow, 117036, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Dmitri Kamashev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia.
- The National Medical Research Center for Endocrinology, Moscow, 117036, Russia
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| |
Collapse
|
4
|
Fernandes NB, Velagacherla V, Spandana KJ, N B, Mehta CH, Gadag S, Sabhahit JN, Nayak UY. Co-delivery of lapatinib and 5-fluorouracil transfersomes using transpapillary iontophoresis for breast cancer therapy. Int J Pharm 2024; 650:123686. [PMID: 38070658 DOI: 10.1016/j.ijpharm.2023.123686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/18/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023]
Abstract
Combination chemotherapy, involving the intervention of two or more anti-neoplastic agents has been the cornerstone in breast cancer treatment, owing to the applications it holds in contrast to the mono-therapy approach. This research predominantly focussed on proving the synergy between Lapatinib (LPT) and 5-Fluorouracil (5-FU) and further enhancing its localized permeation via transfersome-loaded delivery and iontophoresis to treat breast tumors. The IC50 values for LPT and 5-FU were found to be 19.38 µg/ml and 5.7 µg/ml respectively and their synergistic effect was proven by the Chou-Talalay assay using CompuSyn software. Furthermore, LPT and 5-FU were encapsulated within transfersomes and administered via the transpapillary route. The drug-loaded carriers were characterized for their particle size, polydispersity index, zeta potential, and entrapment efficiency. The ex vivo rat skin permeation studies indicated that when compared to LPT dispersion and 5-FU solution, drug-loaded transfersomes exhibited better permeability and their transpapillary permeation was enhanced on using iontophoresis. Moreover, both LPT and 5-FU transfersomes were found to be stable for 3 months when stored at a temperature of 5 ± 3 °C. The results indicated that this treatment strategy could be an effective approach in contrast to some of the conventional treatments employed to date.
Collapse
Affiliation(s)
- Neha B Fernandes
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Varalakshmi Velagacherla
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - K J Spandana
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru 575018, Karnataka, India
| | - Bhagya N
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru 575018, Karnataka, India
| | - Chetan H Mehta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shivaprasad Gadag
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Jayalakshmi N Sabhahit
- Department of Electrical and Electronics Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
5
|
Yang CY, Lee MY, Chen YL, Shiau JP, Tsai YH, Yang CN, Chang HW, Tseng CH. Synthesis and Anticancer Evaluation of 4-Anilinoquinolinylchalcone Derivatives. Int J Mol Sci 2023; 24:ijms24076034. [PMID: 37047007 PMCID: PMC10094048 DOI: 10.3390/ijms24076034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
A series of 4-anilinoquinolinylchalcone derivatives were synthesized and evaluated for antiproliferative activities against the growth of human cancer cell lines (Huh-7 and MDA-MB-231) and normal lung cells (MRC-5). The results exhibited low cytotoxicity against human lung cells (MRC-5). Among them, (E)-3-{4-{[4-(benzyloxy)phenyl]amino}quinolin-2-yl}-1-(4-methoxyphenyl) prop-2-en-1-one (4a) was found to have the highest cytotoxicity in breast cancer cells and low cytotoxicity in normal cells. Compound 4a causes ATP depletion and apoptosis of breast cancer MDA-MB-231 cells and triggers reactive oxygen species (ROS)-dependent caspase 3/7 activation. In conclusion, it is worth studying 4-anilinoquinolinylchalcone derivatives further as new potential anticancer agents for the treatment of human cancers.
Collapse
Affiliation(s)
- Cheng-Yao Yang
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
| | - Min-Yu Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yung-Hsiang Tsai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 811213, Taiwan
| | - Chia-Ning Yang
- Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chih-Hua Tseng
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung City 80708, Taiwan
- Department of Pharmacy, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City 80145, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung County 912301, Taiwan
| |
Collapse
|
6
|
Jin N, Xia Y, Gao Q. Combined PARP inhibitors and small molecular inhibitors in solid tumor treatment (Review). Int J Oncol 2023; 62:28. [PMID: 36601757 PMCID: PMC9851129 DOI: 10.3892/ijo.2023.5476] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/23/2022] [Indexed: 01/05/2023] Open
Abstract
With the development of precision medicine, targeted therapy has attracted extensive attention. Poly(ADP‑ribose) polymerase inhibitors (PARPi) are critical clinical drugs designed to induce cell death and are major antitumor targeted agents. However, preclinical and clinical data have revealed the limitations of PARPi monotherapy. Therefore, their combination with other targeted drugs has become a research hotspot in tumor treatment. Recent studies have demonstrated the critical role of small molecular inhibitors in multiple haematological cancers and solid tumors via cellular signalling modulation, exhibiting potential as a combined pharmacotherapy. In the present review, studies focused on small molecular inhibitors targeting the homologous recombination pathway were summarized and clinical trials evaluating the safety and efficacy of combined treatment were discussed.
Collapse
Affiliation(s)
- Ning Jin
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Yu Xia
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Qinglei Gao
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
7
|
Lewińska A, Wróbel K, Błoniarz D, Adamczyk-Grochala J, Wołowiec S, Wnuk M. Lapatinib- and fulvestrant-PAMAM dendrimer conjugates promote apoptosis in chemotherapy-induced senescent breast cancer cells with different receptor status. BIOMATERIALS ADVANCES 2022; 140:213047. [PMID: 35917687 DOI: 10.1016/j.bioadv.2022.213047] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/08/2022] [Accepted: 07/22/2022] [Indexed: 06/15/2023]
Abstract
Lapatinib (L) and fulvestrant (F) are used in targeted anticancer therapies, in particular, against phenotypically different breast cancer cells. L, a dual inhibitor of EGFR and HER2 tyrosine kinases, is active against HER2-positive breast cancer cells, while F, a selective estrogen receptor degrader (SERD), is active against ER-positive breast cancer cells. However, the action of L and F can be limited due to their relatively low water solubility and bioavailability. In the present study, poly(amidoamine) (PAMAM) dendrimer G3 was functionalized with L or F or L and F to compare their effects with free L or F against breast cancer cells with different receptor status (ER-positive MCF-7, triple negative MDA-MB-231 and HER2-positive SK-BR-3 cells). L-PAMAM and F-PAMAM conjugates potentiated cytostatic and cytotoxic action of L and F that was accompanied by elevated levels of autophagy. TRDMT1, RNA methyltransferase, was also involved in this response as judged by TRDMT1 nuclear translocation and nano-drug resistance of TRDMT1 gene knockout cells. Nano-drugs also promoted elimination of doxorubicin-induced senescent breast cancer cells by apoptosis-mediated senolysis regardless of receptor status. In conclusion, we propose a novel anticancer approach based on L-PAMAM and F-PAMAM nanoplatforms being effective, at least, against breast cancer cells with different phenotypic features.
Collapse
Affiliation(s)
- Anna Lewińska
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Nature Sciences, University of Rzeszow, Rzeszow, Poland
| | - Konrad Wróbel
- Department of Biochemistry and General Chemistry, Medical College, University of Rzeszow, Rzeszow, Poland
| | - Dominika Błoniarz
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Nature Sciences, University of Rzeszow, Rzeszow, Poland
| | - Jagoda Adamczyk-Grochala
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Nature Sciences, University of Rzeszow, Rzeszow, Poland
| | - Stanisław Wołowiec
- Department of Biochemistry and General Chemistry, Medical College, University of Rzeszow, Rzeszow, Poland
| | - Maciej Wnuk
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Nature Sciences, University of Rzeszow, Rzeszow, Poland.
| |
Collapse
|
8
|
Design and synthesis of some new 6-bromo-2-(pyridin-3-yl)-4-substituted quinazolines as multi tyrosine kinase inhibitors. Bioorg Chem 2022; 128:106099. [PMID: 35994884 DOI: 10.1016/j.bioorg.2022.106099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Abstract
The present study involves design and synthesis of five series of 6-bromo-2-(pyridin-3-yl)-4-substituted quinazolines 9a-l, 11a-e, 13a-c, 14a-f and 15a-e. Candidates 9a-l and 11a-e were evaluated for their EGFR and HER2 inhibitory activity compared to Lapatinib. Compounds 9b, 9d, 9f, 11b and 11c were further screened for their in vitro cytotoxicity against two human breast cancer cell lines: AU-565 and MDA-MB-231 in addition to normal breast cell line MCF10A. Compound 9d revealed a remarkable cytotoxic efficacy against AU-565 cell line (IC50 = 1.54 µM) relative to Lapatinib (IC50 = 0.48 µM), whereas compounds 9d and 11c showed a superior cytotoxicity towards MDA-MB-231 (IC50 = 2.67 and 1.75 µM, respectively) in comparison to Lapatinib (IC50 = 9.29 µM). Moreover, compounds 13a-c, 13a-c, 14a-f and 15a-e were tested for their VEGFR-2 inhibitory activity compared to Sorafenib. Compounds 13a, 14c and 14e exhibited remarkable inhibition (IC50 = 79.80, 50.22 and 78.02 nM, respectively) relative to Sorafenib (IC50 = 51.87 nM). In vitro cytotoxicity of these compounds against HepG2, HCT-116 and normal cell (WISH) revealed a superior cytotoxicity against HepG2, HCT-116 especially 13a (IC50 = 17.51 and 5.56 µM, respectively) and 14c (IC50 = 10.40 and 3.37 µM, respectively) compared to Sorafenib (IC50 = 19.33 and 6.82 µM, respectively). Compounds 9d, 11c and 14c were subjected to cell cycle analysis and apoptotic assay. Molecular docking and ADME prediction studies were fulfilled to illustrate the interaction of the potent derivatives with the hot spots of the active site of EGFR, HER2 and VEGFR-2 along with prediction of their pharmacokinetic and physicochemical properties.
Collapse
|
9
|
Sirinian C, Papanastasiou AD, Degn SE, Frantzi T, Aronis C, Chaniotis D, Makatsoris T, Koutras A, Kalofonos HP. RANK-C Expression Sensitizes ER-Negative, EGFR-Positive Breast Cancer Cells to EGFR-Tyrosine Kinase Inhibitors (TKIs). Genes (Basel) 2021; 12:genes12111686. [PMID: 34828291 PMCID: PMC8619104 DOI: 10.3390/genes12111686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 11/16/2022] Open
Abstract
Background: We have previously shown that overexpression of RANK-c in ER-negative breast cancer cell lines attenuates aggressive properties of cancer cells, partially through a RANK-c/EGFR interaction. EGFR inhibition through TKIs in breast cancer has been tested in triple-negative disease settings with limited clinical benefit for patients. Here we test if expression of RANK-c in ER-negative breast cancer cells in conjunction with treatment with TK inhibitors (erlotinib or gefitinib) can affect survival and colony-forming capacity of cancer cells. Methods: Stably expressing MDA-MB-231-RANK-c and SKBR3-RANK-c cells were employed to test proliferation and colony formation in the presence of TKIs. In addition, Western blot analysis was performed to dissect EGFR related signaling cascades upon TK inhibition in the presence of RANK-c. Results: Interestingly the two RANK-c expressing, ER-negative cells lines presented with a distinct phenotype concerning TKI sensitivity upon treatment. MDA-MB-231-RANK-c cells had a higher sensitivity upon gefitinib treatment, while erlotinib decreased the proliferation rate of SKBR3-RANK-c cells. Further, colony formation assays for MDA-MB-231-RANK-c cells showed a decrease in the number and size of colonies developed in the presence of erlotinib. In addition, RANK-c seems to alter signaling through EGFR after TKI treatment in a cell type-specific manner. Conclusions: Our results indicate that ER-negative breast cancer cells that express RANK-c alter their sensitivity profile against tyrosine kinase inhibitors (erlotinib and gefitinib) in a cell type-specific and culture substrate-dependent manner.
Collapse
Affiliation(s)
- Chaido Sirinian
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece; (T.F.); (C.A.); (T.M.); (A.K.); (H.P.K.)
- Correspondence: or ; Tel.: +30-26-1096-9133
| | | | - Soren E. Degn
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark;
| | - Theodora Frantzi
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece; (T.F.); (C.A.); (T.M.); (A.K.); (H.P.K.)
| | - Christos Aronis
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece; (T.F.); (C.A.); (T.M.); (A.K.); (H.P.K.)
| | - Dimitrios Chaniotis
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (A.D.P.); (D.C.)
| | - Thomas Makatsoris
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece; (T.F.); (C.A.); (T.M.); (A.K.); (H.P.K.)
| | - Angelos Koutras
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece; (T.F.); (C.A.); (T.M.); (A.K.); (H.P.K.)
| | - Haralabos P. Kalofonos
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece; (T.F.); (C.A.); (T.M.); (A.K.); (H.P.K.)
| |
Collapse
|
10
|
Leis-Filho AF, de Faria Lainetti P, Emiko Kobayashi P, Fonseca-Alves CE, Laufer-Amorim R. Effects of Lapatinib on HER2-Positive and HER2-Negative Canine Mammary Carcinoma Cells Cultured In Vitro. Pharmaceutics 2021; 13:897. [PMID: 34204236 PMCID: PMC8235449 DOI: 10.3390/pharmaceutics13060897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 12/19/2022] Open
Abstract
HER2 is a prognostic and predictive marker widely used in breast cancer. Lapatinib is a tyrosine kinase inhibitor that works by blocking the phosphorylation of the receptor HER2. Its use is related to relatively good results in the treatment of women with HER2+ breast cancer. Thus, this study aimed to verify the effects of lapatinib on four canine primary mammary gland carcinoma cell cultures and two paired metastatic cell cultures. Cultures were treated with lapatinib at concentrations of 100, 500, 1000 and 3000 nM for 24 h and the 50% inhibitory concentration (IC50) for each cell culture was determined. In addition, a transwell assay was performed to assess the ability of lapatinib to inhibit cell migration. Furthermore, we verified HER2 expression by RT-qPCR analysis of cell cultures and formalin-fixed paraffin-embedded tissues from samples corresponding to those used in cell culture. Lapatinib was able to inhibit cell proliferation in all cell cultures, but it was not able to inhibit migration in all cell cultures. The higher the expression of HER2 in a culture, the more sensitive the culture was to treatment. This relationship may be an indication that the expression of HER2 may be a predictive factor and opens a new perspective for the treatment of primary and metastatic mammary gland cancer.
Collapse
Affiliation(s)
- Antonio Fernando Leis-Filho
- Department of Veterinary Clinic, Sao Paulo State University-UNESP, Botucatu 18618-681, Brazil; (A.F.L.-F.); (P.E.K.)
| | - Patrícia de Faria Lainetti
- Department of Veterinary Surgery and Animal Reproduction, São Paulo State University-UNESP, Botucatu 18618-681, Brazil; (P.d.F.L.); (C.E.F.-A.)
| | - Priscila Emiko Kobayashi
- Department of Veterinary Clinic, Sao Paulo State University-UNESP, Botucatu 18618-681, Brazil; (A.F.L.-F.); (P.E.K.)
| | - Carlos Eduardo Fonseca-Alves
- Department of Veterinary Surgery and Animal Reproduction, São Paulo State University-UNESP, Botucatu 18618-681, Brazil; (P.d.F.L.); (C.E.F.-A.)
- Institute of Health Sciences, Paulista University-UNIP, Bauru 17048-290, Brazil
| | - Renée Laufer-Amorim
- Department of Veterinary Clinic, Sao Paulo State University-UNESP, Botucatu 18618-681, Brazil; (A.F.L.-F.); (P.E.K.)
| |
Collapse
|
11
|
Expression and purification of a recombinant ELRL-MAP30 with dual-targeting anti-tumor bioactivity. Protein Expr Purif 2021; 185:105893. [PMID: 33933613 DOI: 10.1016/j.pep.2021.105893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/26/2022]
Abstract
MAP30 (Momordica antiviral protein 30kD) is a single-chain Ⅰ-type ribosome inactivating protein with a variety of biological activities, including anti-tumor ability. It was reported that MAP30 would serve as a novel and relatively safe agent for prophylaxis and treatment of liver cancer. To determine whether adding two tumor targeting peptides could improve the antitumor activities of MAP30, we genetically modified MAP30 with an RGD motif and a EGFRi motif, which is a ligand with high affinity for αvβ3 integrins and with high affinity for EGFR. The recombinant protein ELRL-MAP30 (rELRL-MAP30) containing a GST-tag was expressed in E. coli. The rELRL-MAP30 was highly expressed in the soluble fraction after induction with 0.15 mM IPTG for 20 h at 16 °C. The purified rELRL-MAP30 appeared as a band on SDS-PAGE. It was identified by western blotting. Cytotoxicity of recombinant protein to HepG2, MDA-MB-231, HUVEC and MCF-7 cells was detected by MTT analysis. Half maximal inhibitory concentration (IC50) values were 54.64 μg/mL, 70.13 μg/mL, 146 μg/mL, 466.4 μg/mL, respectively. Proliferation inhibition assays indicated that rELRL-MAP30 could inhibit the growth of Human liver cancer cell HepG2 effectively. We found that rELRL-MAP30 significantly induced apoptosis in liver cancer cells, as evidenced by nuclear staining of DAPI. In addition, rELRL-MAP30 induced apoptosis in human liver cancer HepG2 cells by up-regulation of Bax as well as down-regulation of Bcl-2. Migration of cell line were markedly inhibited by rELRL-MAP30 in a dose-dependent manner compared to the recombinant MAP30 (rMAP30). In summary, the fusion protein displaying extremely potent cytotoxicity might be highly effective for tumor therapy.
Collapse
|
12
|
Li B, Kuang Y, Yi Y, Qiao X, Liang L, Ye M. Chemical modifications of ergostane-type triterpenoids from Antrodia camphorata and their cytotoxic activities. Bioorg Med Chem Lett 2021; 43:128066. [PMID: 33915258 DOI: 10.1016/j.bmcl.2021.128066] [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: 02/20/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 11/24/2022]
Abstract
In order to discover potential antitumor agents from natural products, chemical modifications of ergostane-type triterpenoids from Antrodia camphorata yielded ten new compounds. They include nine C-26 amide derivatives of antcin G (1) and a methyl antcin B (4) derivative with hydroxyamino groups at C-3 and C-7. Chemical structures of the new compounds were elucidated by NMR and MS analyses. Furthermore, cytotoxicities of the triterpenoid derivatives were evaluated using four human cancer cell lines (HL60, U251, SW480, and MCF-7). As a result, 1a, 1g, and 4a exhibited potent cytotoxic activities against HL60, U251, and SW480 with IC50 values of 0.7 ± 0.9, 2.9 ± 1.3, and 2.2 ± 0.6 μM, respectively. Molecular docking indicates that 1a, 1g, and 4a have strong binding affinity with DNA topoisomerase IIα (-9.3, -7.9, and -7.4 kcal/mol, respectively), and that they could be potent topoisomerase IIα inhibitors.
Collapse
Affiliation(s)
- Bin Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Yi Kuang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Yang Yi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Xue Qiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Lei Liang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China.
| | - Min Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China.
| |
Collapse
|
13
|
In vitro assessment of the photo(geno)toxicity associated with Lapatinib, a Tyrosine Kinase inhibitor. Arch Toxicol 2020; 95:169-178. [PMID: 32815004 DOI: 10.1007/s00204-020-02880-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/12/2020] [Indexed: 12/20/2022]
Abstract
The epidermal growth factor receptors EGFR and HER2 are the main targets for tyrosine kinase inhibitors (TKIs). The quinazoline derivative lapatinib (LAP) is used since 2007 as dual TKI in the treatment of metastatic breast cancer and currently, it is used as an oral anticancer drug for the treatment of solid tumors such as breast and lung cancer. Although hepatotoxicity is its main side effect, it makes sense to investigate the ability of LAP to induce photosensitivity reactions bearing in mind that BRAF (serine/threonine-protein kinase B-Raf) inhibitors display a considerable phototoxic potential and that afloqualone, a quinazoline-marketed drug, causes photodermatosis. Metabolic bioactivation of LAP by CYP3A4 and CYP3A5 leads to chemically reactive N-dealkylated (N-LAP) and O-dealkylated (O-LAP) derivatives. In this context, the aim of the present work is to explore whether LAP and its N- and O-dealkylated metabolites can induce photosensitivity disorders by evaluating their photo(geno)toxicity through in vitro studies, including cell viability as well as photosensitized protein and DNA damage. As a matter of fact, our work has demonstrated that not only LAP, but also its metabolite N-LAP have a clear photosensitizing potential. They are both phototoxic and photogenotoxic to cells, as revealed by the 3T3 NRU assay and the comet assay, respectively. By contrast, the O-LAP does not display relevant photobiological properties. Remarkably, the parent drug LAP shows the highest activity in membrane phototoxicity and protein oxidation, whereas N-LAP is associated with the highest photogenotoxicity, through oxidation of purine bases, as revealed by detection of 8-Oxo-dG.
Collapse
|
14
|
Ornnork N, Kiriwan D, Lirdprapamongkol K, Choowongkomon K, Svasti J, Eurtivong C. Molecular dynamics, MM/PBSA and in vitro validation of a novel quinazoline-based EGFR tyrosine kinase inhibitor identified using structure-based in silico screening. J Mol Graph Model 2020; 99:107639. [PMID: 32534372 DOI: 10.1016/j.jmgm.2020.107639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/27/2022]
Abstract
EGFR-TK has been a target strongly associated with the development of NSCLCs. A structure-based virtual screening campaign was launched against EGFR-TK by virtual screening a 3D library of 167 commercially available small molecules downloaded from ChemBridge Corporation. The virtual screen identified 12 virtual hit molecules, which were biologically evaluated against an EGFR-TK inhibitor-sensitive NSCLC cell line, A549. A quinazoline-based molecule 1, was most active and displayed ∼58% cytotoxicity at 20 μM single dose. The mode of cell death suggests molecule 1 induced apoptosis, which is characteristic of EGFR-TK pathway inhibition. A 50 ns MD simulation was conducted on three different systems: free EGFR-TK, molecule 1 complexed to EGFR-TK, and the positive control, lapatinib, complexed to EGFR-TK. The MD simulations showed increase in stabilisation of the EGFR-TK structure for the complexed systems, i.e., lower RMSDs and RMSFs for complexed EGFR-TK structures compared to the free EGFR-TK system. The binding affinities were estimated using MM/PBSA in the last 10 ns of the MD simulation that revealed comparable binding free energies between molecule 1 and lapatinib, ΔGbind = -25.0 and -23.9 kcal/mol, respectively. Per residue binding free energy decomposition studies revealed non-polar interactions contributed mostly to the binding free energies. Residues Leu718, Arg841 and Phe856 were predicted to contribute most to the binding free energies for molecule 1.
Collapse
Affiliation(s)
- Narittira Ornnork
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Duangnapa Kiriwan
- Genetic Engineering Interdisciplinary Program, Graduate School, Kasetsart University, Bangkok, 10900, Thailand
| | | | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Jisnuson Svasti
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Chatchakorn Eurtivong
- Program in Chemical Sciences, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, 10210, Thailand.
| |
Collapse
|