1
|
Sahoo R, Pattnaik S, Mohanty B, Mir SA, Behera B. Aryl hydrocarbon receptor (AHR) signalling: A double-edged sword guiding both cancer progression and cancer therapy. Biochim Biophys Acta Gen Subj 2025; 1869:130805. [PMID: 40222634 DOI: 10.1016/j.bbagen.2025.130805] [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: 12/20/2024] [Revised: 02/21/2025] [Accepted: 04/05/2025] [Indexed: 04/15/2025]
Abstract
Aryl Hydrocarbon Receptor (AHR) reported to be associated with major carcinogenic signalling cascades which cause cell proliferations, metastasis and invasion as well as immune imbalance. AHR Participates in cellular processes not only through genomic pathways to cause genomic alterations but also via nongenomic pathways to alter various cytoplasmic proteins. In addition, AHR senses a wide range of ligands that modulate its downstream mechanisms that are intricated in cancer induction and prevention. Thus, AHR functions as a two-sided sword where some AHR ligands contribute to enhance cancer whereas few are useful for cancer treatment. Therefore, AHR represent as a regulatory point in cancer progression and treatment. There is a need to reinvestigate the regulatory role of AHR in major intracellular pathways and to explore the potential of AHR ligand for the design of cancer therapeutics. This review emphasizes the interaction of AHR with pro-carcinogenic signalling pathways that modulate cancer induction and progression. Furthermore, it also discusses about the current discovery of AHR ligands for cancer initiation or inhibition. This information could be useful for development of therapeutic strategies for the management of cancer by targeting AHR.
Collapse
Affiliation(s)
- Rahul Sahoo
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Sriya Pattnaik
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Biswajit Mohanty
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Showkat Ahmad Mir
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Birendra Behera
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India.
| |
Collapse
|
2
|
Siddiqui SN, Haider MF, Rahman MA. Innovative approaches in breast cancer therapy: repurposing nanocarriers for enhanced outcomes. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04012-2. [PMID: 40167629 DOI: 10.1007/s00210-025-04012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Breast cancer is one of the most prevalent cancers globally, affecting over 685,000 women annually. While traditional treatment modalities such as surgery, chemotherapy, and radiation therapy have contributing to improved survival rates; however, they are often plagued by limitations such as systemic toxicity, lack of targeted therapy, development of resistance, and collateral damage to healthy tissues. While targeted therapies and endocrine treatment have provided more personalized approaches, challenges like side effects and limited effectiveness in specific subtypes remain. Nanotechnology offers new avenues for addressing these challenges, particularly through the development of advanced nanocarrier systems. Nanocarrier systems are designed to enhance drug targeting, improve bioavailability, reduce side effects, and combat drug resistance. These advanced delivery systems facilitate controlled release, higher drug concentration at target sites, and the potential for combination therapies, thus improving treatment outcomes. Breast cancer clinical trials assess treatment effectiveness, providing critical insights through their statuses and outcomes. The aim of this study is to explore the potential of nanocarrier systems in overcoming the limitation of traditional therapy, enhancing the effectiveness of drug delivery, and enhancing overall treatment outcomes for breast cancer treatment.
Collapse
Affiliation(s)
| | - Md Faheem Haider
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Md Azizur Rahman
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
3
|
Mu J, Li Y, Chen Q, Xiao Y, Hu M, He Z, Zeng J, Ding Y, Song P, He X, Yang X, Zhang X. Revealing the molecular mechanism of baohuoside I for the treatment of breast cancer based on network pharmacology and molecular docking. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118918. [PMID: 39396715 DOI: 10.1016/j.jep.2024.118918] [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: 05/07/2024] [Revised: 09/13/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Traditional Chinese Medicine (TCM), there are many prescriptions for treating breast cancer (BC) that utilize the herb Epimedium brevicornum Maxim, which warms and replenishes kidney yang. Baohuoside I (BI) is a flavonoid compound found in Epimedium brevicornum Maxim. As a single glycoside, it is not easily hydrolyzed in the intestine and is typically absorbed as a precursor. As a natural product with potential anti-cancer properties, studies have shown that BI possesses anti-cancer activity and can inhibit the invasion and migration of BC cells. However, its underlying mechanisms remain unclear, thus further research is needed to validate its modern mechanisms for traditional uses. AIM OF THE STUDY This study aimed to explore the regulatory mechanism of BI in the signaling pathways of BC cells through network pharmacology (NP), molecular docking (MD) techniques and cellular experiments. METHODS Potential targets were predicted using public databases, and a protein-protein interaction (PPI) network was constructed. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed. Key signaling pathways were validated through MD techniques, cellular experiments, RNA interference and Western blot (WB) analysis. RESULTS Treatment-associated targets included SRC, MAPK1, HSP90AA1, PIK3CA, TP53, AKT1, and EGFR. GO enrichment, KEGG enrichment analyses, and MD results indicated that BI exerts its anti-breast cancer effects by inhibiting the tyrosine kinase activity of EGFR, as well as through downstream MAPK signaling pathway and PI3K-Akt signaling pathway pathways. In vitro experiments confirmed that BI primarily induce cell apoptosis through the EGFR-mediated MAPK signaling pathway and PI3K-Akt signaling pathway. CONCLUSION BI can inhibit EGFR activation and promote BC cell apoptosis through the MAPK signaling pathway and PI3K-Akt signaling pathway, thereby exerting therapeutic effects on BC. This study not only provides experimental evidence for the accuracy of NP but also offers an effective approach for rational utilization of Baohuoside I-like flavonoid compounds as anti-breast cancer drugs.
Collapse
Affiliation(s)
- Junjie Mu
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Ying Li
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Qiuxiong Chen
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Yujie Xiao
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Min Hu
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Ziyue He
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Jun Zeng
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China
| | - Yiling Ding
- Pengshui County Forestry Bureau, Chongqing, 409600, PR China
| | - Pengyang Song
- Wansheng Economic and Technological Development Zone Planning and Natural Resources Bureau, Chongqing, 400800, PR China
| | - Xiao He
- Chongqing Three Gorges Medical College, Chongqing, 404120, PR China
| | - Xian Yang
- Engineering Research Center for Biotechnology of Active Substances, Ministry of Education/Chongqing Normal University, Chongqing, 401331, PR China.
| | - Xue Zhang
- Chongqing Medical and Pharmaceutical College, Chongqing, 401331, PR China.
| |
Collapse
|
4
|
Lai Y, Zhang R. Green one-pot synthesis of quinoxaline derivatives using sulfo-anthranilic acid functionalized alginate-MCFe 2O 4 nanostructures: a novel superparamagnetic catalyst with antiproliferative potential. RSC Adv 2025; 15:1698-1712. [PMID: 39835215 PMCID: PMC11744772 DOI: 10.1039/d4ra07892f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
This study reports a green, multi-component synthesis of 2-aminoimidazole-linked quinoxaline Schiff bases using a novel superparamagnetic acid catalyst. The catalyst consists of sulfo-anthranilic acid (SAA) immobilized on MnCoFe2O4@alginate magnetic nanorods (MNRs), achieving high SAA loading (1.8 mmol g-1) and product yields (91-97%). Characterization of the MCFe2O4@Alginate@SAA MNR catalyst revealed an inverse spinel structure (XRD), a saturation magnetization of 31 emu g-1 (VSM), 17.5% organic content (TGA), and a rod-like morphology with diameters of 30-60 nm and lengths of 150-250 nm (SEM). Elemental composition confirmed by EDX analysis indicated successful SAA immobilization and high catalyst purity. The synthesized quinoxaline derivatives were evaluated for antiproliferative activity against SKOV3 and HCT-116 cancer cell lines using the MTT assay. Several compounds, notably 4a, 4s, 4t, 4w, and 4x, exhibited potent activity, inhibiting HCT-116 proliferation by >50% at 50 μg mL-1. Compound 4a demonstrated the most significant inhibition, with 82.3% against SKOV3 cells after 48 h and 69.0% against HCT-116 cells after 24 h, both at 50 μg mL-1. These results suggest the potential of 2-aminoimidazole-linked quinoxaline Schiff bases, particularly 4a, as promising multi-target chemotherapy agents.
Collapse
Affiliation(s)
- Ying Lai
- Department of Life Science and Agriculture, Zhoukou Normal University Zhoukou Henan 466001 China
| | - Ruoyu Zhang
- Department of Life Science and Agriculture, Zhoukou Normal University Zhoukou Henan 466001 China
| |
Collapse
|
5
|
Li X, Wu Z, Yuan L, Chen X, Huang H, Cheng F, Shen W. Hesperidin inhibits colon cancer progression by downregulating SLC5A1 to suppress EGFR phosphorylation. J Cancer 2025; 16:876-887. [PMID: 39781340 PMCID: PMC11705064 DOI: 10.7150/jca.104867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Objective: Hesperidin, an active constituent of traditional Chinese medicine, Chenpi, exhibits anticancer properties across different cancers. This study aimed to clarify the efficacy of Hesperidin against tumors and its mechanisms of action in colon cancer. Method: We assessed the efficacy of Hesperidin on human colon cancer cells (HCT-116 and DLD-1) and normal colonic epithelial cells (NCM460). We quantified cell viability at various Hesperidin concentrations using the CCK8 assay in a series of experiments. We employed clone formation, EdU incorporation, Transwell, and wound healing assays to clarify Hesperidin efficacy on cancer cell proliferation, invasion, and migration. Western blot analyses revealed modulations in epithelial-mesenchymal transition-related proteins, SLC5A1, EGFR, and phosphorylated EGFR levels following Hesperidin exposure. Co-IP assays further validated the interaction between SLC5A1 and EGFR. Our findings were significantly restored following SLC5A1 overexpression in colon cancer cells, highlighting its pivotal role in Hesperidin-induced responses. Results: Hesperidin selectively impaired the viability of HCT-116 and DLD-1 colon cancer cells at specific concentrations while preserving normal NCM460 cells. This flavonoid dose-dependently reduced cancer cell proliferation, migration, and invasion. It significantly suppressed SLC5A1 and phosphorylated EGFR expression. We identified a direct SLC5A1-EGFR interaction essential for regulating EGFR activity in colon cancer. Overexpressing SLC5A1 significantly reduced the inhibitory effects of Hesperidin, highlighting its crucial role in this context. Conclusion: Hesperidin exerts its anticancer effects on colon cancer by inhibiting SLC5A1 expression and consequently downregulating EGFR phosphorylation.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Zhao Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Lebin Yuan
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Xing Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - He Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Fei Cheng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Wei Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| |
Collapse
|
6
|
Uremis MM, Ceylan M, Turkoz Y. Investigation of Apoptotic and Anticancer Effects of 2-substituted Benzothiazoles in Breast Cancer Cell Lines: EGFR Modulation and Mechanistic Insights. Anticancer Agents Med Chem 2025; 25:433-445. [PMID: 39473208 DOI: 10.2174/0118715206335840241018053929] [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: 06/06/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 04/11/2025]
Abstract
BACKGROUND AND OBJECTIVE Benzothiazole derivatives, a class of heterocyclic compounds, exhibited diverse biological activities influenced by substituents in the thiazole ring. This study aimed to synthesize these compounds with two functional groups to investigate their potential as anticancer agents, particularly against breast cancer. While previous research demonstrated the efficacy of 2-substituted benzothiazoles against glioma and cervical and pancreatic cancer cells, there is a gap in studies targeting breast cancer. METHODS The synthesized compounds were tested in vitro using MCF-7, MDA-MB-231, and MCF-10A cell lines, with Doxorubicin as the positive control. Various assays were conducted, including Annexin V/PI, cell cycle analysis, wound healing, and measurement of mitochondrial membrane potential. Protein expression of EGFR and transcription levels of apoptosis-related genes (Bax and Bcl-xL) and cancer progression-related genes (JAK, STAT3, ERK, AKT, mTOR) were analyzed. Additionally, the balance between antioxidants and oxidants was evaluated by measuring TAS and TOS levels. RESULTS Our findings revealed that benzothiazole compounds significantly inhibited breast cancer cell growth by reducing cell motility, disrupting mitochondrial membrane potential, and inducing cell cycle arrest in the sub-G1 phase. These compounds increased reactive oxygen species accumulation, leading to cell death. Furthermore, they decreased EGFR protein levels, increased Bax gene transcription, and downregulated the expression of genes such as JAK, STAT3, ERK, AKT, and mTOR. CONCLUSION In conclusion, benzothiazole derivatives exhibited potent inhibitory effects on breast cancer in vitro by promoting apoptosis, downregulating EGFR activity, and modulating key signaling pathways, including JAK/STAT, ERK/MAPK, and PI3K/Akt/mTOR. These results highlighted the potential of benzothiazole derivatives as novel therapeutic agents for breast cancer treatment.
Collapse
Affiliation(s)
- Muhammed Mehdi Uremis
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Mustafa Ceylan
- Department of Chemistry, Faculty of Science and Letters, Gaziosmanpaşa University, Tokat, Turkey
| | - Yusuf Turkoz
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| |
Collapse
|
7
|
Gahramanov V, Vizeacoumar FS, Morales AM, Bonham K, Sakharkar MK, Kumar S, Vizeacoumar FJ, Freywald A, Sherman MY. Cancer Cell's Achilles Heels: Considerations for Design of Anti-Cancer Drug Combinations. Int J Mol Sci 2024; 25:13495. [PMID: 39769257 PMCID: PMC11676151 DOI: 10.3390/ijms252413495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Loss of function screens using shRNA (short hairpin RNA) and CRISPR (clustered regularly interspaced short palindromic repeats) are routinely used to identify genes that modulate responses of tumor cells to anti-cancer drugs. Here, by integrating GSEA (Gene Set Enrichment Analysis) and CMAP (Connectivity Map) analyses of multiple published shRNA screens, we identified a core set of pathways that affect responses to multiple drugs with diverse mechanisms of action. This suggests that these pathways represent "weak points" or "Achilles heels", whose mild disturbance should make cancer cells vulnerable to a variety of treatments. These "weak points" include proteasome, protein synthesis, RNA splicing, RNA synthesis, cell cycle, Akt-mTOR, and tight junction-related pathways. Therefore, inhibitors of these pathways are expected to sensitize cancer cells to a variety of drugs. This hypothesis was tested by analyzing the diversity of drugs that synergize with FDA-approved inhibitors of the proteasome, RNA synthesis, and Akt-mTOR pathways. Indeed, the quantitative evaluation indicates that inhibitors of any of these signaling pathways can synergize with a more diverse set of pharmaceuticals, compared to compounds inhibiting targets distinct from the "weak points" pathways. Our findings described here imply that inhibitors of the "weak points" pathways should be considered as primary candidates in a search for synergistic drug combinations.
Collapse
Affiliation(s)
- Valid Gahramanov
- Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA;
- Department of Molecular Biology, Ariel University, Ariel 40700, Israel;
| | - Frederick S. Vizeacoumar
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Royal University Hospital, Saskatoon, SK S7N 0W8, Canada; (F.S.V.); (A.M.M.); (A.F.)
| | - Alain Morejon Morales
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Royal University Hospital, Saskatoon, SK S7N 0W8, Canada; (F.S.V.); (A.M.M.); (A.F.)
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
| | - Keith Bonham
- Cancer Research, Saskatchewan Cancer Agency and Division of Oncology, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (K.B.); (F.J.V.)
| | - Meena K. Sakharkar
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada;
| | - Santosh Kumar
- Department of Molecular Biology, Ariel University, Ariel 40700, Israel;
| | - Franco J. Vizeacoumar
- Cancer Research, Saskatchewan Cancer Agency and Division of Oncology, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (K.B.); (F.J.V.)
| | - Andrew Freywald
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Royal University Hospital, Saskatoon, SK S7N 0W8, Canada; (F.S.V.); (A.M.M.); (A.F.)
| | | |
Collapse
|
8
|
Yun Y, Kim S, Lee SN, Cho HY, Choi JW. Nanomaterial-based detection of circulating tumor cells and circulating cancer stem cells for cancer immunotherapy. NANO CONVERGENCE 2024; 11:56. [PMID: 39671082 PMCID: PMC11645384 DOI: 10.1186/s40580-024-00466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Nanomaterials have emerged as transformative tools for detecting circulating tumor cells (CTCs) and circulating cancer stem cells (CCSCs), significantly enhancing cancer diagnostics and immunotherapy. Nanomaterials, including those composed of gold, magnetic materials, and silica, have enhanced the sensitivity, specificity, and efficiency of isolating these rare cells from blood. These developments are of paramount importance for the early detection of cancer and for providing real-time insights into metastasis and treatment resistance, which are essential for the development of personalized immunotherapies. The combination of nanomaterial-based platforms with phenotyping techniques, such as Raman spectroscopy and microfluidics, enables researchers to enhance immunotherapy protocols targeting specific CTC and CCSC markers. Nanomaterials also facilitate the targeted delivery of immunotherapeutic agents, including immune checkpoint inhibitors and therapeutic antibodies, directly to tumor cells. This synergistic approach has the potential to enhance therapeutic efficacy and mitigate the risk of metastasis and relapse. In conclusion, this review critically examines the use of nanomaterial-driven detection systems for detecting CTCs and CCSCs, their application in immunotherapy, and suggests future directions, highlighting their potential to transform the integration of diagnostics and treatment, thereby paving the way for more precise and personalized cancer therapies.
Collapse
Affiliation(s)
- Yeochan Yun
- Department of Bio and Fermentation Convergence Technology, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea
| | - Seewoo Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Sang-Nam Lee
- Uniance Gene Inc., 273, Digital-ro, Guro-gu, Seoul, 08381, Republic of Korea.
| | - Hyeon-Yeol Cho
- Department of Bio and Fermentation Convergence Technology, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea.
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea.
| |
Collapse
|
9
|
Khalil AM, Sabry OM, El-Askary HI, El Zalabani SM, Eltanany BM, Pont L, Benavente F, Mohamed AF, Fayek NM. Uncovering the therapeutic potential of green pea waste in breast cancer: a multi-target approach utilizing LC-MS/MS metabolomics, molecular networking, and network pharmacology. BMC Complement Med Ther 2024; 24:379. [PMID: 39482666 PMCID: PMC11526710 DOI: 10.1186/s12906-024-04669-x] [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: 07/10/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND PISUM SATIVUM: (PS) is a universal legume plant utilized for both human and animal consumption, particularly its seeds, known as green peas. The processing of PS in food industries and households produces a significant amount of waste that needs to be valorized. METHODS In this study, the metabolite profiles of the 70% ethanolic extracts of PS wastes, namely peels (PSP) and a combination of leaves and stems (PSLS), were investigated by liquid chromatography-electrospray ionization-quadrupole time-of-flight tandem mass spectrometry (LC-ESI-QTOF-MS/MS) followed by molecular networking. RESULTS Different classes of metabolites were identified, being flavonoids and their derivatives, along with phenolic acids, the most abundant categories. Additionally, a comprehensive network pharmacology strategy was applied to elucidate potentially active metabolites, key targets, and the pathways involved in cytotoxic activity against breast cancer. This cytotoxic activity was investigated in MCF-7 and MCF-10a cell lines. Results revealed that PSLS extract exhibited a potent cytotoxic activity with a good selectivity index (IC50 = 17.67 and selectivity index of 3.51), compared to the reference drug doxorubicin (IC50 = 2.69 µg/mL and selectivity index of 5.28). Whereas PSP extract appeared to be less potent and selective (IC50 = 32.92 µg/mL and selectivity index of 1.62). A similar performance was also observed for several polyphenolics isolated from the PSLS extract, including methyl cis p-coumarate, trans p-coumaric acid, and liquiritigenin/ 7-methyl liquiritigenin mixture. Methyl cis p-coumarate showed the most potent cytotoxic activity against MCF-7 cell line and the highest selectivity (IC50 = 1.18 µg/mL (6.91 µM) and selectivity index of 27.42). The network pharmacology study revealed that the isolated compounds could interact with several breast cancer-associated protein targets including carbonic anhydrases 1, 2, 4, 9, and 12, as well as aldo-keto reductase family 1 member B1, adenosine A3 receptor, protein tyrosine phosphatase non-receptor type 1, and estrogen receptor 2. CONCLUSION The uncovered therapeutic potential of PSLS and its metabolite constituents pave the way for an efficient and mindful PS waste valorization, calling for further in-vitro and in-vivo research.
Collapse
Affiliation(s)
- Asmaa M Khalil
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Omar M Sabry
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University, Cairo, 4645241, Egypt
| | - Hesham I El-Askary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Soheir M El Zalabani
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Basma M Eltanany
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Laura Pont
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA·UB), University of Barcelona, Barcelona, 08028, Spain
- Serra Húnter Program, Generalitat de Catalunya, Barcelona, 08007, Spain
| | - Fernando Benavente
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA·UB), University of Barcelona, Barcelona, 08028, Spain
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Faculty of Pharmacy, King Salman International University (KSIU), Ras Sedr, 46612, Egypt
| | - Nesrin M Fayek
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
10
|
Arora M, Singh AK, Kumar A, Singh H, Pathak P, Grishina M, Yadav JP, Verma A, Kumar P. Semisynthetic phytochemicals in cancer treatment: a medicinal chemistry perspective. RSC Med Chem 2024; 15:3345-3370. [PMID: 39430100 PMCID: PMC11484407 DOI: 10.1039/d4md00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer is the uncontrolled proliferation of abnormal cells that invade other areas, spread to other organs, and cause metastases, which is the most common cause of death. A review of all FDA-approved new molecular entities (NMEs) shows that natural products and derivatives account for over one-third of all NMEs. Before 1940, unmodified products and derivatives accounted for 43% and 14% of NME registrations, respectively. Since then, the share of unmodified products has decreased to 9.5% of all approved NMEs, while the share of derivatives has increased to 28%. Since the 1940s, semi-synthetic and synthetic derivatives of natural substances have gained importance, and this trend continues to date. In this study, we have discussed in detail isolated phytoconstituents with chemical modifications that are either FDA-approved or under clinical trials, such as podophyllotoxin, Taxol (paclitaxel, docetaxel), vinca alkaloids (vincristine, vinblastine), camptothecin, genistein, cephalotaxine, rohitukine, and many more, which may act as essential leads to the development of novel anticancer agents. Furthermore, we have also discussed recent developments in the most potent semisynthetic phytoconstituents, their unique properties, and their importance in cancer treatment.
Collapse
Affiliation(s)
- Meghna Arora
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Ankit Kumar Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Adarsh Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Harshwardhan Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Prateek Pathak
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, School of Pharmacy, GITAM (Deemed to be University) Hyderabad Campus India
| | - Maria Grishina
- Laboratory of Computational Modeling of Drugs, Higher Medical and Biological School, South Ural State University Chelyabinsk 454008 Russia
| | - Jagat Pal Yadav
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Pharmacology Research Laboratory, Faculty of Pharmaceutical Sciences, Rama University Kanpur 209217 India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| |
Collapse
|
11
|
Keles NA, Dogan S, Dogan A, Sudagidan M, Balci TN, Cetiner O, Kavruk M, Ozalp VC, Tuna BG. Long-term intermittent caloric restriction remodels the gut microbiota in mice genetically prone to breast cancer. Nutrition 2024; 126:112525. [PMID: 39168040 DOI: 10.1016/j.nut.2024.112525] [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: 12/25/2023] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVES Gut microbiota dysbiosis is among the risk factors for breast cancer development, together with genetic background and dietary habits. However, caloric restriction has been shown to remodel the gut microbiota and slow tumor growth. Here, we investigated whether the gut microbiota mediates the preventive effects of long-term chronic or intermittent caloric restriction on breast cancer predisposition. METHODS 10-week-old transgenic breast cancer-prone mice were randomly assigned to dietary groups (ad libitum, chronic caloric restriction, and intermittent caloric restriction groups) and fed up to week 81. Stool samples were collected at weeks 10 (baseline), 17 (young), 49 (adult), and 81 (old). 16S rRNA gene sequencing was performed to identify the gut microbiota profile of the different groups. In order to investigate the breast cancer gut microbiota profile within genetically predisposed individuals regardless of diet, mammary tumor-bearing mice and mammary tumor-free but genetically prone mice were selected from the ad libitum group (n = 6). RESULTS Intermittent caloric restriction increased the microbial diversity of adult mice and modified age-related compositional changes. A total of 13 genera were differentially abundant over time. Pathogenic Mycoplasma was enriched in the re-feeding period of the old intermittent caloric restriction group compared with baseline. Furthermore, mammary tumor-free mice showed shared gut microbiota characteristics with mammary tumor-bearing mice, suggesting an early link between genetic predisposition, gut microbiota, and breast cancer development. CONCLUSIONS Our study revealed the role of gut microbes in the preventive effects of caloric restriction against breast cancer development, implying the significance of diet and microbiome interplay.
Collapse
Affiliation(s)
- Nazim Arda Keles
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Aysenur Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Mert Sudagidan
- Department of Medical Biology, School of Medicine, Atilim University, Ankara, Turkey
| | - Tugce Nur Balci
- Department of Nutrition and Dietetics, School of Health Sciences, Atilim University, Ankara, Turkey
| | - Ozlem Cetiner
- Department of Nutrition and Dietetics, School of Health Sciences, Atilim University, Ankara, Turkey
| | - Murat Kavruk
- Department of Medical Biology, School of Medicine, Istanbul Aydin University, Istanbul, Turkey
| | - Veli Cengiz Ozalp
- Department of Medical Biology, School of Medicine, Atilim University, Ankara, Turkey
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
12
|
Papa F, Grinda T, Rassy E, Cheickh-Hussin R, Ribeiro J, Antonuzzo L, Pistilli B. Long road towards effective HER3 targeting in breast cancer. Cancer Treat Rev 2024; 129:102786. [PMID: 38885540 DOI: 10.1016/j.ctrv.2024.102786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/25/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer is a heterogeneous disease, encompassing multiple different subtypes. Thanks to the increasing knowledge of the diverse biological features of each subtype, most patients receive personalized treatment based on known biomarkers. However, the role of some biomarkers in breast cancer evolution is still unknown, and their potential use as a therapeutic target is still underexplored. HER3 is a member of the human epidermal growth factors receptor family, overexpressed in 50%-70% of breast cancers. HER3 plays a key role in cancer progression, metastasis development, and drug resistance across all the breast cancer subtypes. Owing to its critical role in cancer progression, many HER3-targeting therapies have been developed over the past decade with conflicting findings. Next-generation antibody-drug conjugates have recently shown promising results in solid tumors expressing HER3, including breast cancer. In this review, we discuss the HER3 role in the pathogenesis of breast cancer and its relevance across all subtypes. We also explore the new anti-HER3 treatment strategies, calling into question the significance of HER3 detection as crucial information in breast cancer treatment.
Collapse
Affiliation(s)
- Francesca Papa
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Florence University, Italy
| | - Thomas Grinda
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | | | - Joana Ribeiro
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | | | - Barbara Pistilli
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; INSERM U1279, Gustave Roussy, Villejuif, France.
| |
Collapse
|
13
|
Moharram EA, El-Sayed SM, Ghabbour HA, El-Subbagh HI. Synthesis, molecular modeling simulations and anticancer activity of some new Imidazo[2,1-b]thiazole analogues as EGFR/HER2 and DHFR inhibitors. Bioorg Chem 2024; 150:107538. [PMID: 38861913 DOI: 10.1016/j.bioorg.2024.107538] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
New imidazo[2,1-b]thiazole analogs were designed, synthesized, and biologically evaluated as anticancer agents. In vitro biological evaluation of the anticancer properties of the compounds was performed against different cancer cell lines. Compounds 23 and 39 showed remarkable broad -spectrum cytotoxic potency on most of the tested cell lines. Compounds 23 and 39 exhibited potent activity against the MCF-7 breast cancer cell line, with IC50 values of 1.81 and 4.95 μM, respectively, compared to DOX and SOR (IC50 values of 4.17 and 7.26 μM, respectively). An enzyme inhibition assay was carried out to clarify the possible mode of action of the tested compounds. Compounds 23 and 39 were identified as possible EGFR, HER-2, and DHFR inhibitors. Cell cycle arrest results indicated that compound 23 caused cell cycle arrest at the G0/G1 phase in the MCF-7 cells and at the G2/M phase in the Hep G2 cells. Compound 39 induced cell cycle arrest at the G2/M phase in Hela cells. In vivo testing of the anticancer activity of the two most promising molecules in this study was conducted, and the results indicated that they possess considerable in vivo anticancer activity in mice. Data obtained from the molecular modeling simulation study were consistent with the biological evaluation results.
Collapse
Affiliation(s)
- Esraa A Moharram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Selwan M El-Sayed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt; Pharmacy Center of Scientific Excellence, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Hazem A Ghabbour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Hussein I El-Subbagh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt; Pharmacy Center of Scientific Excellence, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
14
|
Abd El-Lateef HM, Gaafar A, Alqahtani AS, Al-Mutairi AA, Alshaya DS, Elsaid FG, Fayad E, Farouk NA. Design, synthesis, and antiproliferative screening of new quinoline derivatives bearing a cis-vinyl triamide motif as apoptosis activators and EGFR-TK inhibitors. RSC Adv 2024; 14:24781-24790. [PMID: 39114435 PMCID: PMC11305403 DOI: 10.1039/d4ra04915b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
In this work, a congeneric set of quinoline-tethered cis-vinyl triamide hybrids was prepared and evaluated as EGFR tyrosine kinase inhibitors for the management of breast cancer. All of the prepared hybrids were evaluated for their antiproliferative effect against the breast MCF-7 cell line. Among the tested hybrids, compound 6f displayed the most potent antiproliferative activity with an IC50 value of 1.87 μM compared to STU (IC50 = 13.71 μM) as the standard reference. The most promising hybrid, 6f, was found to induce cellular cycle arrest at the G1 phase. Furthermore, the molecular mechanism of this hybrid revealed its ability to induce cellular apoptosis via the mitochondrial-dependent apoptotic pathway. Compound 6f decreased MCF-7 cells' MMP compared to the controls (percentage change value of 57.93%). Further investigation of the selective compound 6f showed that it can inhibit EGFR tyrosine kinase.
Collapse
Affiliation(s)
- Hany M Abd El-Lateef
- Department of Chemistry, College of Science, King Faisal University Al-Ahsa 31982 Saudi Arabia
- Department of Chemistry, Faculty of Science, Sohag University Sohag 82524 Egypt
| | - Ahmed Gaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port Said University Port Said Egypt
| | - Arwa Sultan Alqahtani
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University(IMSIU) P.O. Box 90950 Riyadh 11623 Saudi Arabia
| | - Aamal A Al-Mutairi
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University(IMSIU) P.O. Box 90950 Riyadh 11623 Saudi Arabia
| | - Dalal Sulaiman Alshaya
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University P.O. Box 84428 Riyadh 11671 Saudi Arabia
| | - Fahmy Gad Elsaid
- Department of Biology, College of Science, King Khalid University PO Box 960 Abha 61421 Asir Saudi Arabia
| | - Eman Fayad
- Department of Biotechnology, College of Sciences, Taif University P.O. Box 11099 Taif 21944 Saudi Arabia
| | - N A Farouk
- Department of Chemistry, Faculty of Science, Port Said University Port Said Egypt
| |
Collapse
|
15
|
Mir SA, Mohanta PP, Meher RK, Baitharu I, Behera AK, Raut S, Nayak B. Bioinspired thiazolo-[2,3-b] quinazolin-6-one derivatives as potent anti-cancer agents targeting EGFR: their biological evaluations and in silico assessment. Mol Divers 2024; 28:2479-2494. [PMID: 37395840 DOI: 10.1007/s11030-023-10688-6] [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: 02/27/2023] [Accepted: 06/25/2023] [Indexed: 07/04/2023]
Abstract
Cancer is a challenging and second most deadly disease. The epidermal growth factor receptors (EGFRs) dimerize upon ligand bindings to the extracellular domain that intiates the downstream signaling cascades and activates intracellular kinase domain. Thus, activation of autophosphrylation through kinase domain results in metastasis, cell proliferation, and angiogenesis. In this study, we unravel the binding mechanism of newly synthesized thiazolo-[2,3-b] quinazolin-6-one and evaluate their anti-cancer activity against ovary and prostate carcinoma cell lines (OVCAR-3 and PC-3). Synthesized molecules exhibited promising anti-cancer activity against OVCAR-3 and PC-3 carcinoma cell lines with inhibitory concentrations ranging from 13.4 ± 0.43 to 23.6 ± 1.22 μM and 7.5 ± 0.62 to 67.5 ± 1.24 μM, respectively. These compounds induced apoptosis and resulted in cell cycle arrest at G1 and G2/M transition phases. Next, the nude mice models were taken to investigate the toxicity of the 4bi compound, and in vivo investigations revealed no effects upon examined organs (liver and kidney) treated at different concentrations. Moreover, the combined in silico approaches, molecular docking, molecular dynamics simulations, and MM/PBSA methods were performed to assess the binding affinity and stability of bioinspired synthesized congeners with the epidermal growth factor receptor tyrosine kinase (EGFR-TK). The free binding energy (ΔGbind) of the 4bi molecule was found comparable to Erlotinib drug. The test molecule could be competent for further usage to determine its efficicacy in cancer therapeutics.
Collapse
Affiliation(s)
- Showkat Ahmad Mir
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768019, India.
| | | | - Rajesh Kumar Meher
- Departement of Biotechnology & Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768019, India
| | - Iswar Baitharu
- Departement of Environmental Sciences, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768019, India
| | - Ajaya Kumar Behera
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768019, India
| | - Sangeeta Raut
- Departement of Biotechnology, Siksha 'O' Anusandhan, Deemed University, Bhubaneshwar, Odisha, 751003, India
| | - Binata Nayak
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768019, India.
| |
Collapse
|
16
|
Mhaidly N, Barake N, Trelcat A, Journe F, Saussez S, Descamps G. Bufalin Suppresses Head and Neck Cancer Development by Modulating Immune Responses and Targeting the β-Catenin Signaling Pathway. Cancers (Basel) 2024; 16:2739. [PMID: 39123466 PMCID: PMC11311268 DOI: 10.3390/cancers16152739] [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: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Bufalin, a cardiotonic steroid derived from the Chinese toad (Bufo gargarizans), has demonstrated potent anticancer properties across various cancer types, positioning it as a promising therapeutic candidate. However, comprehensive mechanistic studies specific to head and neck cancers have been lacking. Our study aimed to bridge this gap by investigating bufalin's mechanisms of action in head and neck cancer cells. Using several methods, such as Western blotting, immunofluorescence, and flow cytometry, we observed bufalin's dose-dependent reduction in cell viability, disruption of cell membrane integrity, and inhibition of colony formation in both HPV-positive and HPV-negative cell lines. Bufalin induces apoptosis through the modulation of apoptosis-related proteins, mitochondrial function, and reactive oxygen species production. It also arrests the cell cycle at the G2/M phase and attenuates cell migration while affecting epithelial-mesenchymal transition markers and targeting pivotal signaling pathways, including Wnt/β-catenin, EGFR, and NF-κB. Additionally, bufalin exerted immunomodulatory effects by polarizing macrophages toward the M1 phenotype, bolstering antitumor immune responses. These findings underscore bufalin's potential as a multifaceted therapeutic agent against head and neck cancers, targeting essential pathways involved in proliferation, apoptosis, cell cycle regulation, metastasis, and immune modulation. Further research is warranted to validate these mechanisms and optimize bufalin's clinical application.
Collapse
Affiliation(s)
- Nour Mhaidly
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Noura Barake
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Anne Trelcat
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Fabrice Journe
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium;
| | - Sven Saussez
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Géraldine Descamps
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| |
Collapse
|
17
|
Kumar M, Patil KT, Maity P, Chatterjee J, Singh T, Joshi G, Singh S, Kumar R. Design, synthesis, and anticancer assessment of structural analogues of ( E)-1-((3,4,5-trimethoxybenzylidene)amino)-4-(3,4,5-trimethoxyphenyl)imidazo[1,2- a]quinoxaline-2-carbonitrile (6b), an imidazo[1,2- a]quinoxaline-based non-covalent EGFR inhibitor. RSC Med Chem 2024; 15:2322-2339. [PMID: 39026650 PMCID: PMC11253857 DOI: 10.1039/d4md00237g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/26/2024] [Indexed: 07/20/2024] Open
Abstract
In our quest to find improved anticancer therapeutics, we expedite the lead optimization of (E)-1-((3,4,5-trimethoxybenzylidene)amino)-4-(3,4,5-trimethoxyphenyl)imidazo[1,2-a]quinoxaline-2-carbonitrile (6b), an EGFR inhibitor previously discovered in our laboratory through an in-house screening program. The lead optimization was rationally initiated considering the catalytic site of EGFR. We synthesized twenty-nine new analogues of 6b and assessed their anticancer activities. SAR studies highlighted the role of important groups in controlling anticancer activities. Among all, 5a and 5l were found to exhibit improved EGFR inhibition with anticancer asset potential. In silico studies corroborated with in vitro EGFR inhibitory results. The deeper analysis of 5a and 5l revealed that these synthetics could alter the MMP (ΔΨ m) and significantly reduce the ROS levels in lung cancer cells. This is a vital prerequisite for better plausible EGFR inhibitors devoid of cardiotoxicity. qPCR analysis further revealed that the investigational compounds 5a and 5l were able to downregulate the expression of key oncogenes, viz., KRAS, MAP2K, and EGFR. The downregulation of these genes suggests that the investigational compounds could interact and inhibit key players in the signalling cascade along with the EGFR, which may lead to the inhibition of the growth and prognosis of cancer cells via a holistic approach.
Collapse
Affiliation(s)
- Manvendra Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| | - Kiran T Patil
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| | - Pritam Maity
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| | - Joydeep Chatterjee
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| | - Tashvinder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda - 151401 Punjab India
| | - Gaurav Joshi
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
- Department of Pharmaceutical Science, Hemvati Nandan Bahuguna Garhwal (A Central) University Dist. Garhwal Srinagar 246174 Uttarakhand India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda - 151401 Punjab India
| | - Raj Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab Bathinda 151401 India
| |
Collapse
|
18
|
Damare R, Engle K, Kumar G. Targeting epidermal growth factor receptor and its downstream signaling pathways by natural products: A mechanistic insight. Phytother Res 2024; 38:2406-2447. [PMID: 38433568 DOI: 10.1002/ptr.8166] [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: 08/02/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 03/05/2024]
Abstract
The epidermal growth factor receptor (EGFR) is a transmembrane receptor tyrosine kinase (RTK) that maintains normal tissues and cell signaling pathways. EGFR is overactivated and overexpressed in many malignancies, including breast, lung, pancreatic, and kidney. Further, the EGFR gene mutations and protein overexpression activate downstream signaling pathways in cancerous cells, stimulating the growth, survival, resistance to apoptosis, and progression of tumors. Anti-EGFR therapy is the potential approach for treating malignancies and has demonstrated clinical success in treating specific cancers. The recent report suggests most of the clinically used EGFR tyrosine kinase inhibitors developed resistance to the cancer cells. This perspective provides a brief overview of EGFR and its implications in cancer. We have summarized natural products-derived anticancer compounds with the mechanistic basis of tumor inhibition via the EGFR pathway. We propose that developing natural lead molecules into new anticancer agents has a bright future after clinical investigation.
Collapse
Affiliation(s)
- Rutuja Damare
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, India
| | - Kritika Engle
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, India
| | - Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, India
| |
Collapse
|
19
|
Johnson KK, Koshy P, Kopecky C, Devadason M, Biazik J, Zheng X, Jiang Y, Wang X, Liu Y, Holst J, Yang JL, Kilian KA, Sorrell CC. ROS-mediated anticancer effects of EGFR-targeted nanoceria. J Biomed Mater Res A 2024; 112:754-769. [PMID: 38084898 DOI: 10.1002/jbm.a.37656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/07/2023] [Accepted: 12/01/2023] [Indexed: 03/20/2024]
Abstract
The therapeutic effectiveness of anticancer drugs, including nanomedicines, can be enhanced with active receptor-targeting strategies. Epidermal growth factor receptor (EGFR) is an important cancer biomarker, constitutively expressed in sarcoma patients of different histological types. The present work reports materials and in vitro biomedical analyses of silanized (passive delivery) and/or EGF-functionalized (active delivery) ceria nanorods exhibiting highly defective catalytically active surfaces. The EGFR-targeting efficiency of nanoceria was confirmed by receptor-binding studies. Increased cytotoxicity and reactive oxygen species (ROS) production were observed for EGF-functionalized nanoceria owing to enhanced cellular uptake by HT-1080 fibrosarcoma cells. The uptake was confirmed by TEM and confocal microscopy. Silanized nanoceria demonstrated negligible/minimal cytotoxicity toward healthy MRC-5 cells at 24 and 48 h, whereas this was significant at 72 h owing to a nanoceria accumulation effect. In contrast, considerable cytotoxicity toward the cancer cells was exhibited at all three times points. The ROS generation and associated cytotoxicity were moderated by the equilibrium between catalysis by ceria, generation of cell debris, and blockage of active sites. EGFR-targeting is shown to enhance the uptake levels of nanoceria by cancer cells, subsequently enhancing the overall anticancer activity and therapeutic performance of ceria.
Collapse
Affiliation(s)
- Kochurani K Johnson
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Pramod Koshy
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Chantal Kopecky
- Australian Centre for NanoMedicine, School of Chemistry, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michelle Devadason
- Translational Cancer Metabolism Laboratory, School of Medical Sciences and Prince of Wales Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Joanna Biazik
- Electron Microscope Unit, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Xiaoran Zheng
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Yue Jiang
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Xiaochun Wang
- Prince of Wales Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Yiling Liu
- Australian Centre for NanoMedicine, School of Chemistry, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jeff Holst
- Translational Cancer Metabolism Laboratory, School of Medical Sciences and Prince of Wales Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jia-Lin Yang
- Prince of Wales Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kristopher A Kilian
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
- Australian Centre for NanoMedicine, School of Chemistry, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Charles C Sorrell
- School of Materials Science and Engineering, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
20
|
Crook ZR, Sevilla GP, Young P, Girard EJ, Phi TD, Howard M, Price J, Olson JM, Nairn NW. CYpHER: Catalytic extracellular targeted protein degradation with high potency and durable effect. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.21.581471. [PMID: 38712232 PMCID: PMC11071310 DOI: 10.1101/2024.02.21.581471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Many disease-causing proteins have multiple pathogenic mechanisms, and conventional inhibitors struggle to reliably disrupt more than one. Targeted protein degradation (TPD) can eliminate the protein, and thus all its functions, by directing a cell's protein turnover machinery towards it. Two established strategies either engage catalytic E3 ligases or drive uptake towards the endolysosomal pathway. Here we describe CYpHER (CatalYtic pH-dependent Endolysosomal delivery with Recycling) technology with potency and durability from a novel catalytic mechanism that shares the specificity and straightforward modular design of endolysosomal uptake. By bestowing pH-dependent release on the target engager and using the rapid-cycling transferrin receptor as the uptake receptor, CYpHER induces endolysosomal target delivery while re-using drug, potentially yielding increased potency and reduced off-target tissue exposure risks. The TfR-based approach allows targeting to tumors that overexpress this receptor and offers the potential for transport to the CNS. CYpHER function was demonstrated in vitro with EGFR and PD-L1, and in vivo with EGFR in a model of EGFR-driven non-small cell lung cancer.
Collapse
Affiliation(s)
- Zachary R. Crook
- Cyclera Therapeutics Inc, Seattle, WA 98115, USA. Present address of Z.R.C., G.P.S., and N.W.N
- Blaze Bioscience Inc., Seattle, WA 98109, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Gregory P. Sevilla
- Cyclera Therapeutics Inc, Seattle, WA 98115, USA. Present address of Z.R.C., G.P.S., and N.W.N
- Blaze Bioscience Inc., Seattle, WA 98109, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | | | - Emily J. Girard
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98105, USA. Present address of E.J.G. and J.M.O
| | | | | | - Jason Price
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98105, USA. Present address of E.J.G. and J.M.O
| | - James M. Olson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98105, USA. Present address of E.J.G. and J.M.O
| | - Natalie W. Nairn
- Cyclera Therapeutics Inc, Seattle, WA 98115, USA. Present address of Z.R.C., G.P.S., and N.W.N
- Blaze Bioscience Inc., Seattle, WA 98109, USA
| |
Collapse
|
21
|
YU TW, YAMAMOTO H, MORITA S, FUKUSHIMA R, ELBADAWY M, USUI T, SASAKI K. Comparative pharmacokinetics of tyrosine kinase inhibitor, lapatinib, in dogs and cats following single oral administration. J Vet Med Sci 2024; 86:317-321. [PMID: 38281758 PMCID: PMC10963087 DOI: 10.1292/jvms.23-0448] [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: 10/29/2023] [Accepted: 01/09/2024] [Indexed: 01/30/2024] Open
Abstract
Lapatinib is an orally administered tyrosine kinase inhibitor used to treat human epidermal growth factor receptor 2 (HER2) -overexpressing breast cancers in humans. Recently, the potential of lapatinib treatment against canine urothelial carcinoma or feline mammary tumor was investigated. However, the pharmacokinetic studies of lapatinib in dogs and cats are not well-defined. In the present study, the pharmacokinetic characteristics of lapatinib in both cats and dogs after a single oral administration at a dose of 25 mg/kg were compared with each other. Lapatinib was administered orally to four female laboratory cats and four female beagle dogs. Blood samples were collected over time, and the plasma lapatinib concentrations were analyzed by HPLC. Following a single dose of 25 mg/kg, the averaged maximum plasma concentration (Cmax) of lapatinib in cats was 0.47 μg/mL and achieved at 7.1 hr post-administration, while the Cmax in dogs was 1.63 μg/mL and achieved at 9.5 hr post-administration. The mean elimination half-life was 6.5 hr in cats and 7.8 hr in dogs. The average area under the plasma concentration-time curve of dogs (37.2 hr·μg/mL) was significantly higher than that of cats (7.97 hr·μg/mL). These results exhibited slow absorptions of lapatinib in both animals after oral administration. The Cmax observed in cats was significantly lower and the half-life was shorter than those observed in dogs. Based on these results, a larger dose or shorter dosing intervals might be recommended in cats to achieve similar plasma concentration as dogs.
Collapse
Affiliation(s)
- Ting-Wei YU
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Haru YAMAMOTO
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shohei MORITA
- Animal Emergency Medical Center, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryuji FUKUSHIMA
- Animal Emergency Medical Center, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Mohamed ELBADAWY
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Elqaliobiya, Egypt
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Tatsuya USUI
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kazuaki SASAKI
- Laboratory of Veterinary Pharmacology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
22
|
Li B, Wu J, Cao D, Cao C, Zhang J, Li X, Li H, Shen H, Yu Z. ERBB1 alleviates secondary brain injury induced by experimental intracerebral hemorrhage in rats by modulating neuronal death via PLC-γ/PKC pathway. CNS Neurosci Ther 2024; 30:e14679. [PMID: 38528842 PMCID: PMC10964039 DOI: 10.1111/cns.14679] [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: 11/20/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
AIMS Intracerebral hemorrhage (ICH) is a disease with high rates of disability and mortality. The role of epidermal growth factor receptor 1 (ERBB1) in ICH was elucidated in this study. METHODS ICH model was constructed by injecting autologous arterial blood into the right basal ganglia. The protein level of ERBB1 was detected by western blot analysis. To up- and downregulation of ERBB1 in rats, intraventricular injection of a lentivirus overexpression vector of ERBB1 and AG1478 (a specific inhibitor of ERBB1) was used. The cell apoptosis, neuronal loss, and pro-inflammatory cytokines were assessed by TUNEL, Nissl staining, and ELISA. Meanwhile, behavioral cognitive impairment of ICH rats was evaluated after ERBB1-targeted interventions. RESULTS ERBB1 increased significantly in brain tissue of ICH rats. Overexpression of ERBB1 remarkably reduced cell apoptosis and neuronal loss induced by ICH, as well as pro-inflammatory cytokines and oxidative stress. Meanwhile, the behavioral and cognitive impairment of ICH rats were alleviated after upregulation of ERBB1; however, the secondary brain injury (SBI) was aggravated by AG1478 treatment. Furthermore, the upregulation of PLC-γ and PKC in ICH rats was reversed by AG1478 treatment. CONCLUSIONS ERBB1 can improve SBI and has a neuroprotective effect in experimental ICH rats via PLC-γ/PKC pathway.
Collapse
Affiliation(s)
- Bing Li
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
- Department of Neurosurgery, Yancheng City No. 1 People's Hospital, Yancheng First HospitalAffiliated Hospital of Nanjing University Medical SchoolYanchengJiangsu ProvinceChina
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Demao Cao
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
- Department of NeurosurgeryThe Affiliated Hospital of Yangzhou UniversityYangzhouJiangsu ProvinceChina
| | - Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
- Department of Neurocritical Intensive Care UnitJiangyin Clinical College of Xuzhou Medical CollegeJiangyinJiangsu ProvinceChina
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| |
Collapse
|
23
|
Abramenko N, Vellieux F, Veselá K, Kejík Z, Hajduch J, Masařík M, Babula P, Hoskovec D, Pacák K, Martásek P, Smetana K, Jakubek M. Investigation of the potential effects of estrogen receptor modulators on immune checkpoint molecules. Sci Rep 2024; 14:3043. [PMID: 38321096 PMCID: PMC10847107 DOI: 10.1038/s41598-024-51804-2] [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: 10/03/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Immune checkpoints regulate the immune system response. Recent studies suggest that flavonoids, known as phytoestrogens, may inhibit the PD-1/PD-L1 axis. We explored the potential of estrogens and 17 Selective Estrogen Receptor Modulators (SERMs) as inhibiting ligands for immune checkpoint proteins (CTLA-4, PD-L1, PD-1, and CD80). Our docking studies revealed strong binding energy values for quinestrol, quercetin, and bazedoxifene, indicating their potential to inhibit PD-1 and CTLA-4. Quercetin and bazedoxifene, known to modulate EGFR and IL-6R alongside estrogen receptors, can influence the immune checkpoint functionality. We discuss the impact of SERMs on PD-1 and CTLA-4, suggesting that these SERMs could have therapeutic effects through immune checkpoint inhibition. This study highlights the potential of SERMs as inhibitory ligands for immune checkpoint proteins, emphasizing the importance of considering PD-1 and CTLA-4 inhibition when evaluating SERMs as therapeutic agents. Our findings open new avenues for cancer immunotherapy by exploring the interaction between various SERMs and immune checkpoint pathways.
Collapse
Affiliation(s)
- Nikita Abramenko
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Fréderic Vellieux
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Jan Hajduch
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
| | - Michal Masařík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, 121 08, Prague, Czech Republic
| | - Karel Pacák
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Building 10, Room 1-3140, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Karel Smetana
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00, Prague, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic.
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic.
| |
Collapse
|
24
|
Sombal W, Khan NU, Khan BM, Ismail M, Almutairi MH, Khan S, Khan AU, Mustafa A, Iftikhar B, Ali I. Human epidermal growth receptor polymorphisms ( HER1-rs11543848 and HER2-rs1136201) exhibited significant association with breast cancer risk in Pashtun population of Khyber Pakhtunkhwa, Pakistan. Health Sci Rep 2024; 7:e1846. [PMID: 38317673 PMCID: PMC10839356 DOI: 10.1002/hsr2.1846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/09/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Background and Aims Breast cancer is the most common type of cancer in women. The genetic polymorphism in HER (HER1-rs11543848 and HER2-rs1136201) were found to be associated with breast cancer risk in different ethnicities worldwide with inconsistent results. The aim of this research study was to evaluate the association of HER1-rs11543848 and HER2-rs1136201 polymorphisms as a risk of breast cancer in Pashtun population of Khyber Pakhtunkhwa, Pakistan. Methods A total of 314 women including 164 breast cancer patients and 150 age and gender-matched healthy controls were enrolled from June 2021 to May 2022. All the samples were subjected to DNA extraction followed by Tetra-ARMS-PCR for genotyping and gel electrophoresis. Results Our results indicated that HER1-rs11543848 risk allele A (p = 0.0001) and heterozygous genotype GA (p = 0.0001) displayed highly significant association with breast cancer, while the homozygous mutant genotype AA indicated association but nonsignificant results (odds ratio [OR] = 2.637, 95% confidence interval [CI] = 1.2258-5.6756, p = 0.0833). Similarly, the HER2-rs1136201 risk allele G (p = 0.0023), the heterozygous genotype AG (p = 0.0530) and homozygous mutant genotype GG showed significant association (OR = 2.5946, 95% CI = 0.9876-6.8165, p = 0.0530) with breast cancer risk. Both the SNPs presented a higher but nonsignificant risk of breast cancer in postmenopausal women (OR = 2.242, p = 0.08 and OR = 2.009, p = 0.06). However, both the SNPs showed significant association (p < 0.005) with family history, metastasis, stage, luminal B, and TNBC. Conclusion In conclusion, HER1-rs11543848 and HER2-rs1136201 polymorphisms are significantly associated with the higher risk of breast cancer in Pashtun population of Khyber Pakhtunkhwa, Pakistan. These findings advocate for further exploration with larger datasets, offering promising avenues for personalized approaches in breast cancer research and potentially enhancing clinical practices for better risk assessment and targeted management strategies.
Collapse
Affiliation(s)
- Wafa Sombal
- Institute of Biotechnology & Genetic Engineering (Health Division)The University of Agriculture PeshawarPeshawarPakistan
| | - Najeeb Ullah Khan
- Institute of Biotechnology & Genetic Engineering (Health Division)The University of Agriculture PeshawarPeshawarPakistan
| | - Bibi Maryam Khan
- School of Life ScienceJiangsu UniversityZhejiangJiangsu ProvincePeople's Republic of China
| | | | | | - Samiullah Khan
- Institute of Radiotherapy and Nuclear Medicine (IRNUM)PeshawarPakistan
| | - Aakif Ullah Khan
- Institute of Radiotherapy and Nuclear Medicine (IRNUM)PeshawarPakistan
| | - Adeela Mustafa
- Department of Community MedicineKhyber Medical CollegePeshawarPakistan
| | - Bushra Iftikhar
- Department of Community MedicineKhyber Medical CollegePeshawarPakistan
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics (CAMB)Gulf University for Science and TechnologyHawallyKuwait
| |
Collapse
|
25
|
Nao SC, Kong L, Chan DSH, Liu J, Huang LS, Wu L, Wu J, Wong CY, Wang W, Leung CH. Covalent inhibition of epidermal growth factor receptor using a long-lived iridium(III)-afatinib probe. Int J Biol Macromol 2024; 259:129211. [PMID: 38184034 DOI: 10.1016/j.ijbiomac.2024.129211] [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: 11/16/2023] [Revised: 12/27/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
The overexpression and overactivation of epidermal growth factor receptor (EGFR) are frequently observed in human cancers, including squamous cell carcinoma and adenocarcinoma. In this study, a covalent EGFR probe was developed by conjugating afatinib to an iridium(III) scaffold. Complex 1 showed enhanced luminescence in living epidermoid squamous carcinoma A431 cells compared to other cell lines, via engaging EGFR as confirmed via CETSA and knockdown experiments. Moreover, complex 1 inhibited downstream targets of EGFR in cellulo with repression persisting after removal of the complex, indicating an irreversible mode of inhibition. Finally, complex 1 showed potent antiproliferative activity against A431 cells with comparable potency to afatinib alone. To our knowledge, complex 1 is the first EGFR covalent inhibitor based on an iridium scaffold reported in the literature, with the potential to be further explored as a theranostic agent in the future.
Collapse
Affiliation(s)
- Sang-Cuo Nao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Lingtan Kong
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Daniel Shiu-Hin Chan
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR
| | - Jianhua Liu
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Le-Sheng Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Lei Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Chun-Yuen Wong
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR.
| | - Wanhe Wang
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen 518057, China.
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Taipa, Macau; MoE Frontiers Science Centre for Precision Oncology, University of Macau, Taipa, Macau.
| |
Collapse
|
26
|
Lin Y, Wang S, Zhang L, Yang Q. Elucidating the relationship between breast cancer and brain cortical structure: a Mendelian randomization study. Cereb Cortex 2024; 34:bhad498. [PMID: 38112592 DOI: 10.1093/cercor/bhad498] [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: 11/08/2023] [Revised: 12/02/2023] [Accepted: 12/03/2023] [Indexed: 12/21/2023] Open
Abstract
Cancer-associated cognitive impairment is a significant challenge for individuals who have survived breast cancer, affecting their quality of life. In this study, we conducted an inaugural comprehensive Mendelian randomization analysis discerning the causal relationship between breast cancer, including its two subtypes, and the cerebral cortical structure. Our analysis indicated that estrogen receptor-negative breast cancer significantly decreased surface area (β = -593.01 mm2, 95% CI: -1134.9 to -51.1 mm2, P = 0.032). At the regional level, estrogen receptor-negative breast cancer showed a significant association with surface area and thickness in 17 cortical regions. These regions included the insula, posterior cingulate, superior frontal, precuneus, fusiform, lateral occipital, and rostral middle frontal. Specifically, estrogen receptor-negative breast cancer had a significant impact on decreasing the surface area of the insula without considering global weight (β = -14.09 mm2, 95% CI: -22.91 to -5.27 mm2, P = 0.0017). The results from meta-analysis and LD Score Regression provide support for our findings. This investigation unveils the correlations between breast cancer, its various subcategories, and the cerebral cortical structure. Notably, breast cancer of the estrogen receptor-negative variety may elicit more widespread cerebral atrophy.
Collapse
Affiliation(s)
- Yilong Lin
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361003, China
| | - Songsong Wang
- Department of Urology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Liyi Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361003, China
| | - Qingmo Yang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361003, China
| |
Collapse
|
27
|
Ullah A, Khan BM, Khan NU, Muntaha ST, Khan S, Khan AU, Almutairi MH, Ali I. Assessment of HER1 (rs11543848) and HER2 (rs1136201) polymorphism and their association with colorectal cancer susceptibility in Khyber Pakhtunkhwa, Pakistan. Mol Biol Rep 2023; 51:1. [PMID: 38040921 DOI: 10.1007/s11033-023-08943-y] [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: 08/15/2023] [Accepted: 10/30/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a widespread malignancy characterized by uncontrolled growth in the colon or rectum and remains a leading cause of cancer-related mortality globally. Various genes polymorphisms have been linked with the risk of CRC, but our study aimed to investigate the association between HER1 (rs11543848) and HER2 (rs1136201) polymorphisms with the risk of CRC in the Khyber Pakhtunkhwa (KPK) population of Pakistan. The association of the selected polymorphisms (rs11543848 and rs1136201) with CRC risk has been investigated in various ethnic groups, but their impact remains unexplored in Pakistan, particularly within the KPK population, highlighting the need of the study in this region. METHODS In this study 120 CRC patients and 120 healthy controls were enrolled. The DNA was extracted from the blood by salting-out method and genotyping was done using ARMS-PCR. RESULTS Our investigations provided convincing evidence of a strong association between HER1 (rs11543848) and the risk of CRC. Both the genotypes heterozygous GA (OR = 2.07, CI = 1.18 to 3.64, P = 0.01) and homozygous AA (OR = 6.22, CI = 2.56 to 15.08, P = 0.0001) showed higher risk and significant association with the CRC risk. Similarly, heterozygous genotype AG of HER2 (rs1136201) was significantly associated (OR = 3.16, 95% CI = 1.78 to 5.58, P = 0.0001) while mutant genotype GG showed higher risk but non-significant association (OR = 3.23, 95% CI = 0.84 to 12.43, P = 0.08) with CRC patients. HER1 (rs11543848) demonstrated a significant association (P = 0.003) with the age at diagnosis in CRC patients, while HER2 (rs1136201) showed a non-significant association (P = 0.434). Both the SNPs were non-significantly associated with gender (P = 0.793 and 0.117), metastasis (P = 0.582 and 0.129), location of the tumor (P = 0.555 and 0.993), tumor grade (P = 0.290 and 0.920), tumor size (P = 0.535 and 0.289) and stages of cancer (P = 0.892 and 0.352). CONCLUSION In conclusion, both the polymorphisms rs11543848 and rs1136201 displayed susceptibility with CRC in the KPK population. However, further investigations are recommended while using whole exome sequencing on a larger sample size for more precise results.
Collapse
Affiliation(s)
- Asad Ullah
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, Pakistan
| | | | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, Pakistan.
| | - Sidra Tul Muntaha
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Samiullah Khan
- Institute of Radiotherapy and Nuclear Medicine (IRNUM), Peshawar, Pakistan
| | - Aakif Ullah Khan
- Institute of Radiotherapy and Nuclear Medicine (IRNUM), Peshawar, Pakistan
| | - Mikhlid H Almutairi
- Zoology Department, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Hawally, Kuwait
| |
Collapse
|
28
|
Dakroub R, Huard S, Hajj-Younes Y, Suresh S, Badran B, Fayyad-Kazan H, Dubois T. Therapeutic Advantage of Targeting PRMT5 in Combination with Chemotherapies or EGFR/HER2 Inhibitors in Triple-Negative Breast Cancers. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:785-799. [PMID: 37954171 PMCID: PMC10637385 DOI: 10.2147/bctt.s430513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/04/2023] [Indexed: 11/14/2023]
Abstract
Purpose Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subgroup characterized by a high risk of resistance to chemotherapies and high relapse potential. TNBC shows inter-and intra-tumoral heterogeneity; more than half expresses high EGFR levels and about 30% are classified as HER2-low breast cancers. High PRMT5 mRNA levels are associated with poor prognosis in TNBC and inhibiting PRMT5 impairs the viability of subsets of TNBC cell lines and delays tumor growth in TNBC mice models. TNBC patients may therefore benefit from a treatment targeting PRMT5. The aim of this study was to assess the therapeutic benefit of combining a PRMT5 inhibitor with different chemotherapies used in the clinics to treat TNBC patients, or with FDA-approved inhibitors targeting the HER family members. Methods The drug combinations were performed using proliferation and colony formation assays on TNBC cell lines that were sensitive or resistant to EPZ015938, a PRMT5 inhibitor that has been evaluated in clinical trials. The chemotherapies analyzed were cisplatin, doxorubicin, camptothecin, and paclitaxel. The targeted therapies tested were erlotinib (EGFR inhibitor), neratinib (EGFR/HER2/HER4 inhibitor) and tucatinib (HER2 inhibitor). Results We found that PRMT5 inhibition synergized mostly with cisplatin, and to a lesser extent with doxorubicin or camptothecin, but not with paclitaxel, to impair TNBC cell proliferation. PRMT5 inhibition also synergized with erlotinib and neratinib in TNBC cell lines, especially in those overexpressing EGFR. Additionally, a synergistic interaction was observed with neratinib and tucatinib in a HER2-low TNBC cell line as well as in a HER2-positive breast cancer cell line. We noticed that synergy can be obtained in TNBC cell lines that were resistant to PRMT5 inhibition alone. Conclusion Altogether, our data highlight the therapeutic potential of targeting PRMT5 using combinatorial strategies for the treatment of subsets of TNBC patients.
Collapse
Affiliation(s)
- Rayan Dakroub
- Breast Cancer Biology Group, Translational Research Department, Institut Curie-PSL Research University, Paris, 75005, France
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Hadath, 1003, Lebanon
| | - Solène Huard
- Breast Cancer Biology Group, Translational Research Department, Institut Curie-PSL Research University, Paris, 75005, France
| | - Yara Hajj-Younes
- Breast Cancer Biology Group, Translational Research Department, Institut Curie-PSL Research University, Paris, 75005, France
| | - Samyuktha Suresh
- Breast Cancer Biology Group, Translational Research Department, Institut Curie-PSL Research University, Paris, 75005, France
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Hadath, 1003, Lebanon
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Hadath, 1003, Lebanon
| | - Thierry Dubois
- Breast Cancer Biology Group, Translational Research Department, Institut Curie-PSL Research University, Paris, 75005, France
| |
Collapse
|
29
|
Nifontova G, Kalenichenko D, Kriukova I, Terryn C, Audonnet S, Karaulov A, Nabiev I, Sukhanova A. Impact of Macrophages on the Interaction of Cetuximab-Functionalized Polyelectrolyte Capsules with EGFR-Expressing Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37917654 DOI: 10.1021/acsami.3c10864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Polyelectrolyte capsules (PCs) are a promising tool for anticancer drug delivery and tumor targeting. Surface functionalization of PCs with antibodies is widely used for providing their specific interactions with cancer cells. The efficiency of PC-based targeted delivery systems can be affected by the cellular heterogeneity of the tumor, particularly by the presence of tumor-associated macrophages. We used human epidermoid carcinoma cells and macrophages derived from human leukemia monocytic cells in either monoculture or coculture to analyze the targeting capacity and internalization efficiency of PCs with a mean size of 1.03 ± 0.11 μm. The PCs were functionalized with the monoclonal antibody cetuximab targeting the human epidermal growth factor receptor (EGFR). We have shown that surface functionalization of the PCs with cetuximab ensures a specific interaction with EGFR-expressing cancer cells and promotes capsule internalization. In monoculture, the macrophages derived from human leukemia monocytic cells have been found to internalize both nonfunctionalized PCs and cetuximab-functionalized PCs (Cet-PCs) more intensely compared to epidermoid carcinoma cells. The internalization of Cet-PCs by cancer cells is mediated by lipid rafts of the cell membrane, whereas the PC internalization by macrophages is only slightly influenced by lipid rafts. Experiments with a coculture of human epidermoid carcinoma cells and macrophages derived from human leukemia monocytic cells have shown that Cet-PCs preferentially interact with cancer cells, which are subsequently attacked by macrophages. These data can be used to further improve the strategy of PC functionalization for targeted delivery, with the cellular heterogeneity of the tumor microenvironment taken into consideration.
Collapse
Affiliation(s)
- Galina Nifontova
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51096 Reims, France
| | - Daria Kalenichenko
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51096 Reims, France
| | - Irina Kriukova
- Life Improvement by Future Technologies (LIFT) Center, 143025 Moscow, Russian Federation
- Laboratory of Nano-Bioengineering, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russian Federation
| | - Christine Terryn
- Plateau Technique PICT, Université de Reims Champagne-Ardenne, 51096 Reims, France
| | - Sandra Audonnet
- URCACyt, Flow Cytometry Technical Platform, Université de Reims Champagne-Ardenne, 51096, Reims, France
| | - Alexander Karaulov
- Sechenov First Moscow State Medical University, Sechenov University, 119146 Moscow, Russian Federation
| | - Igor Nabiev
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51096 Reims, France
- Life Improvement by Future Technologies (LIFT) Center, 143025 Moscow, Russian Federation
- Laboratory of Nano-Bioengineering, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russian Federation
- Sechenov First Moscow State Medical University, Sechenov University, 119146 Moscow, Russian Federation
| | - Alyona Sukhanova
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51096 Reims, France
| |
Collapse
|
30
|
Domingues M, Leite Pereira C, Sarmento B, Castro F. Mimicking 3D breast tumor-stromal interactions to screen novel cancer therapeutics. Eur J Pharm Sci 2023; 190:106560. [PMID: 37557927 DOI: 10.1016/j.ejps.2023.106560] [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: 04/11/2023] [Revised: 07/31/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
Most of the 3D breast tumor models used in drug screening studies only comprise tumor cells, keeping out other essential cell players of the tumor microenvironment. Tumor-associated macrophages and fibroblasts are frequently correlated with tumor progression and therapy resistance, and targeting these cells at the tumor site has been appointed as a promising therapeutic strategy. However, the translation of new therapies to the clinic has been hampered by the absence of cellular models that more closely mimic the features of in vivo breast tumor microenvironment. Therefore, the development of innovative 3D models able to provide consistent and predictive responses about the in vivo efficacy of novel therapeutics is still an unmet preclinical need. Herein, we have established an in vitro 3D heterotypic spheroid model including MCF-7 breast tumor cells, human mammary fibroblasts and human macrophages. To establish this model, different cell densities have been combined and characterized through the evaluation of the spheroid size and metabolic activity, as well as histological and immunohistochemistry analysis of the 3D multicellular structures. The final optimized 3D model consisted in a multicellular spheroid seeded at the initial density of 5000 cells and cell ratio of 1:2:1 (MCF-7:monocytes:fibroblasts). Our model recapitulates several features of the breast tumor microenvironment, including the formation of a necrotic core, spatial organization, and extracellular matrix production. Further, it was validated as a platform for drug screening studies, using paclitaxel, a currently approved drug for breast cancer treatment, and Gefitinib, a chemotherapeutic approved for lung cancer and in preclinical evaluation for breast cancer. Generally, the impact on the cell viability of the 3D model was less evident than in 2D model, reinforcing the relevance of such complex 3D models in addressing novel treatment approaches. Overall, the use of a 3D heterotypic spheroid of breast cancer could be a valuable tool to predict the therapeutic effect of new treatments for breast cancer patients, by recapitulating key features of the breast cancer microenvironment.
Collapse
Affiliation(s)
- Mariana Domingues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Doutor Roberto Frias, Porto 4200-465, Portugal
| | - Catarina Leite Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, Gandra 4585-116, Portugal.
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal.
| |
Collapse
|
31
|
Elanany MM, Mostafa D, Hamdy NM. Remodeled tumor immune microenvironment (TIME) parade via natural killer cells reprogramming in breast cancer. Life Sci 2023; 330:121997. [PMID: 37536617 DOI: 10.1016/j.lfs.2023.121997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Breast cancer (BC) is the main cause of cancer-related mortality among women globally. Despite substantial advances in the identification and management of primary tumors, traditional therapies including surgery, chemotherapy, and radiation cannot completely eliminate the danger of relapse and metastatic illness. Metastasis is controlled by microenvironmental and systemic mechanisms, including immunosurveillance. This led to the evolvement of immunotherapies that has gained much attention in the recent years for cancer treatment directed to the innate immune system. The long forgotten innate immune cells known as natural killer (NK) cells have emerged as novel targets for more effective therapeutics for BC. Normally, NK cells has the capacity to identify and eradicate tumor cells either directly or by releasing cytotoxic granules, chemokines and proinflammatory cytokines. Yet, NK cells are exposed to inhibitory signals by cancer cells, which causes them to become dysfunctional in the immunosuppressive tumor microenvironment (TME) in BC, supporting tumor escape and spread. Potential mechanisms of NK cell dysfunction in BC metastasis have been recently identified. Understanding these immunologic pathways driving BC metastasis will lead to improvements in the current immunotherapeutic strategies. In the current review, we highlight how BC evades immunosurveillance by rendering NK cells dysfunctional and we shed the light on novel NK cell- directed therapies.
Collapse
Affiliation(s)
- Mona M Elanany
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Dina Mostafa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| | - Nadia M Hamdy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| |
Collapse
|
32
|
Davodabadi F, Mirinejad S, Fathi-Karkan S, Majidpour M, Ajalli N, Sheervalilou R, Sargazi S, Rozmus D, Rahdar A, Diez-Pascual AM. Aptamer-functionalized quantum dots as theranostic nanotools against cancer and bacterial infections: A comprehensive overview of recent trends. Biotechnol Prog 2023; 39:e3366. [PMID: 37222166 DOI: 10.1002/btpr.3366] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023]
Abstract
Aptamers (Apts) are synthetic nucleic acid ligands that can be engineered to target various molecules, including amino acids, proteins, and pharmaceuticals. Through a series of adsorption, recovery, and amplification steps, Apts are extracted from combinatorial libraries of synthesized nucleic acids. Using aptasensors in bioanalysis and biomedicine can be improved by combining them with nanomaterials. Moreover, Apt-associated nanomaterials, including liposomes, polymeric, dendrimers, carbon nanomaterials, silica, nanorods, magnetic NPs, and quantum dots (QDs), have been widely used as promising nanotools in biomedicine. Following surface modifications and conjugation with appropriate functional groups, these nanomaterials can be successfully used in aptasensing. Advanced biological assays can use Apts immobilized on QD surfaces through physical interaction and chemical bonding. Accordingly, modern QD aptasensing platforms rely on interactions between QDs, Apts, and targets to detect them. QD-Apt conjugates can be used to directly detect prostate, ovarian, colorectal, and lung cancers or simultaneously detect biomarkers associated with these malignancies. Tenascin-C, mucin 1, prostate-specific antigen, prostate-specific membrane antigen, nucleolin, growth factors, and exosomes are among the cancer biomarkers that can be sensitively detected using such bioconjugates. Furthermore, Apt-conjugated QDs have shown great potential for controlling bacterial infections such as Bacillus thuringiensis, Pseudomonas aeruginosa, Escherichia coli, Acinetobacter baumannii, Campylobacter jejuni, Staphylococcus aureus, and Salmonella typhimurium. This comprehensive review discusses recent advancements in the design of QD-Apt bioconjugates and their applications in cancer and bacterial theranostics.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Sonia Fathi-Karkan
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mahdi Majidpour
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Narges Ajalli
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | | | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Dominika Rozmus
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran
| | - Ana M Diez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Quimica Analitica, Quimica Fisica e Ingenieria Quimica, Madrid, Spain
| |
Collapse
|
33
|
Garcia AR, Mendes A, Custódia C, Faria CC, Barata JT, Malhó R, Figueira I, Brito MA. Abrogating Metastatic Properties of Triple-Negative Breast Cancer Cells by EGFR and PI3K Dual Inhibitors. Cancers (Basel) 2023; 15:3973. [PMID: 37568789 PMCID: PMC10416979 DOI: 10.3390/cancers15153973] [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: 05/24/2023] [Revised: 07/25/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a devastating BC subtype. Its aggressiveness, allied to the lack of well-defined molecular targets, usually culminates in the appearance of metastases that account for poor prognosis, particularly when they develop in the brain. Nevertheless, TNBC has been associated with epidermal growth factor receptor (EGFR) overexpression, leading to downstream phosphoinositide 3-kinase (PI3K) signaling activation. We aimed to unravel novel drug candidates for TNBC treatment based on EGFR and/or PI3K inhibition. Using a highly metastatic TNBC cell line with brain tropism (MDA-MB-231 Br4) and a library of 27 drug candidates in silico predicted to inhibit EGFR, PI3K, or EGFR plus PI3K, and to cross the blood-brain barrier, we evaluated the effects on cell viability. The half maximal inhibitory concentration (IC50) of the most cytotoxic ones was established, and cell cycle and death, as well as migration and EGFR pathway intervenient, were further evaluated. Two dual inhibitors emerged as the most promising drugs, with the ability to modulate cell cycle, death, migration and proliferation, morphology, and PI3K/AKT cascade players such as myocyte enhancer factor 2C (MEF2C) and forkhead box P1 (FOXP1). This work revealed EGFR/PI3K dual inhibitors as strong candidates to tackle brain metastatic TNBC cells.
Collapse
Affiliation(s)
- Ana Rita Garcia
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Avilson Mendes
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Carlos Custódia
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Cláudia C. Faria
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Av. Prof. Egas Moniz, 1649-035 Lisbon, Portugal
| | - João T. Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Rui Malhó
- BioISI—Biosystems and Integrative Sciences Institute, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1746-016 Lisbon, Portugal
| | - Inês Figueira
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
- Farm-ID—Faculty of Pharmacy Association for Research and Development, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria Alexandra Brito
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
34
|
Zhao WS, Chen KF, Liu M, Jia XL, Huang YQ, Hao BB, Hu H, Shen XY, Yu Q, Tan MJ. Investigation of targets and anticancer mechanisms of covalently acting natural products by functional proteomics. Acta Pharmacol Sin 2023; 44:1701-1711. [PMID: 36932232 PMCID: PMC10374574 DOI: 10.1038/s41401-023-01072-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/23/2023] [Indexed: 03/19/2023]
Abstract
Eriocalyxin B (EB), 17-hydroxy-jolkinolide B (HJB), parthenolide (PN), xanthatin (XT) and andrographolide (AG) are terpenoid natural products with a variety of promising antitumor activities, which commonly bear electrophilic groups (α,β-unsaturated carbonyl groups and/or epoxides) capable of covalently modifying protein cysteine residues. However, their direct targets and underlying molecular mechanisms are still largely unclear, which limits the development of these compounds. In this study, we integrated activity-based protein profiling (ABPP) and quantitative proteomics approach to systematically characterize the covalent targets of these natural products and their involved cellular pathways. We first demonstrated the anti-proliferation activities of these five compounds in triple-negative breast cancer cell MDA-MB-231. Tandem mass tag (TMT)-based quantitative proteomics showed all five compounds commonly affected the ubiquitin mediated proteolysis pathways. ABPP platform identified the preferentially modified targets of EB and PN, two natural products with high anti-proliferation activity. Biochemical experiments showed that PN inhibited the cell proliferation through targeting ubiquitin carboxyl-terminal hydrolase 10 (USP10). Together, this study uncovered the covalently modified targets of these natural products and potential molecular mechanisms of their antitumor activities.
Collapse
Affiliation(s)
- Wen-Si Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Kai-Feng Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Man Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xing-Long Jia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yu-Qi Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Bing-Bing Hao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hao Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Yan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qiang Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Min-Jia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
35
|
Matou-Nasri S, Aldawood M, Alanazi F, Khan AL. Updates on Triple-Negative Breast Cancer in Type 2 Diabetes Mellitus Patients: From Risk Factors to Diagnosis, Biomarkers and Therapy. Diagnostics (Basel) 2023; 13:2390. [PMID: 37510134 PMCID: PMC10378597 DOI: 10.3390/diagnostics13142390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is usually the most malignant and aggressive mammary epithelial tumor characterized by the lack of expression for estrogen receptors and progesterone receptors, and the absence of epidermal growth factor receptor (HER)2 amplification. Corresponding to 15-20% of all breast cancers and well-known by its poor clinical outcome, this negative receptor expression deprives TNBC from targeted therapy and makes its management therapeutically challenging. Type 2 diabetes mellitus (T2DM) is the most common ageing metabolic disorder due to insulin deficiency or resistance resulting in hyperglycemia, hyperinsulinemia, and hyperlipidemia. Due to metabolic and hormonal imbalances, there are many interplays between both chronic disorders leading to increased risk of breast cancer, especially TNBC, diagnosed in T2DM patients. The purpose of this review is to provide up-to-date information related to epidemiology and clinicopathological features, risk factors, diagnosis, biomarkers, and current therapy/clinical trials for TNBC patients with T2DM compared to non-diabetic counterparts. Thus, in-depth investigation of the diabetic complications on TNBC onset, development, and progression and the discovery of biomarkers would improve TNBC management through early diagnosis, tailoring therapy for a better outcome of T2DM patients diagnosed with TNBC.
Collapse
Affiliation(s)
- Sabine Matou-Nasri
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Maram Aldawood
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Post Graduate and Zoology Department, King Saud University, Riyadh 12372, Saudi Arabia
| | - Fatimah Alanazi
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Abdul Latif Khan
- Tissue Biobank, KAIMRC, MNG-HA, Riyadh 11481, Saudi Arabia
- Pathology and Clinical Laboratory Medicine, King Abdulaziz Medical City (KAMC), Riyadh 11564, Saudi Arabia
| |
Collapse
|
36
|
Yang X, Xu L, Yang L, Xu S. Research progress of STAT3-based dual inhibitors for cancer therapy. Bioorg Med Chem 2023; 91:117382. [PMID: 37369169 DOI: 10.1016/j.bmc.2023.117382] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3), a transcription factor, regulates gene levels that are associated with cell survival, cell cycle, and immune reaction. It is correlated with the grade of malignancy and the development of various cancers and targeting STAT3 protein is a potentially promising therapeutic strategy for tumors. Over the past 20 years, various compounds have been found to directly inhibit STAT3 activity via different strategies. However, numerous difficulties exist in the development of STAT3 inhibitors, such as serious toxic effects, poor therapeutic effects, and intrinsic and acquired drug resistance. STAT3 inhibitors synergistically suppress cancer development with additional anti-tumor drugs, such as indoleamine 2,3-dioxygenase 1 inhibitors (IDO1i), histone deacetylase inhibitors (HDACi), DNA inhibitors, pro-tumorigenic cytokine inhibitors (PTCi), NF-κB inhibitors, and tubulin inhibitors. Therefore, individual molecule- based dual-target inhibitors can be the candidate alternative or complementary treatment to overcome the disadvantages of just STAT3 or other targets as a monotherapy. In this review, we discuss the theoretical basis for formulating STAT3-based dual-target inhibitors and also summarize their structure-activity relationships (SARs).
Collapse
Affiliation(s)
- Xiaojuan Yang
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China.
| | - Lu Xu
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China
| | - Li Yang
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China
| | - Shaohong Xu
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China.
| |
Collapse
|
37
|
Ma J, Chen C, Fan Z, Zhang Y, Ji J, Wei D, Zhang F, Sun B, Huang P, Ren L. CircEGFR reduces the sensitivity of pirarubicin and regulates the malignant progression of triple-negative breast cancer via the miR-1299/EGFR axis. Int J Biol Macromol 2023:125295. [PMID: 37302631 DOI: 10.1016/j.ijbiomac.2023.125295] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
Circular RNAs (circRNAs) have been found to be involved in cancer progression and chemotherapy sensitivity. However, the biological function of circRNAs in triple-negative breast cancer (TNBC) and its effect on the sensitivity to pirarubicin (THP) chemotherapy are still unclear. CircEGFR (hsa_circ_0080220) was screened and verified by bioinformatics analysis, proving it was highly expressed in TNBC cell lines, patient tissues, and plasma exosomes, and was associated with poor prognosis of patients. The expression level of circEGFR in patient tissue has potential diagnostic value to distinguish TNBC tissue from normal breast tissue. In vitro studies confirmed that overexpression of circEGFR promoted the proliferation, migration, invasion, and EMT of TNBC cells and decreased the sensitivity of THP treatment while silencing circEGFR showed the opposite effect. The circEGFR/miR-1299/EGFR pathway was cascaded and verified. CircEGFR regulated malignant progression of TNBC by regulating EGFR via sponging miR-1299. THP can inhibit the malignant phenotype of MDA-MB-231 cells by downregulating the expression of circEGFR. In vivo studies confirmed that overexpression of circEGFR can promote tumor growth and EMT and reduce tumor sensitivity to THP treatment. Silencing circEGFR inhibited the malignant progression of the tumor. These results revealed circEGFR is a promising biomarker for TNBC diagnosis, therapeutic and prognosis.
Collapse
Affiliation(s)
- Jiulong Ma
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Chen Chen
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Zhimin Fan
- General Surgery Center, Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yang Zhang
- Department of Rehabilitation Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiahua Ji
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Dexian Wei
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Fan Zhang
- General Surgery Center, Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Bo Sun
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Peng Huang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China.
| |
Collapse
|
38
|
Wu F, He J, Deng Q, Chen J, Peng M, Xiao J, Zeng Y, Yi L, Li Z, Tian R, Jiang Z. Neuroglobin inhibits pancreatic cancer proliferation and metastasis by targeting the GNAI1/EGFR/AKT/ERK signaling axis. Biochem Biophys Res Commun 2023; 664:108-116. [PMID: 37141638 DOI: 10.1016/j.bbrc.2023.04.080] [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: 03/16/2023] [Revised: 04/11/2023] [Accepted: 04/23/2023] [Indexed: 05/06/2023]
Abstract
Pancreatic cancer is an extremely aggressive malignancy with a very disappointing prognosis. Neuroglobin (NGB), a member of the globin family, has been demonstrated to have a significant role in a variety of tumor forms. The possible role of NGB as a tumor suppressor gene in pancreatic cancer was investigated in this work. Information from the public dataset TCGA combined with GTEx was used to analyze the finding that NGB was commonly downregulated in pancreatic cancer cell lines and tissues, correlating with patient age and prognosis. The expression of NGB in pancreatic cancer was investigated via RT-PCR, qRT-PCR, and Western blot experiments. In-vitro and in-vivo assays, NGB elicited cell cycle arrest in the S phase and apoptosis, hindered migration and invasion, reversed the EMT process, and suppressed cell proliferation and development. The mechanism of action of NGB was predicted via bioinformatics analysis and validated using Western blot and co-IP experiments revealed that NGB inhibited the EGFR/AKT/ERK pathway by binding to and reducing expression of GNAI1 and p-EGFR. In addition, pancreatic cancer cells overexpressing NGB showed increased drug sensitivity to gefitinib (EGFR-TKI). In conclusion, NGB inhibits pancreatic cancer progression by specifically targeting the GNAI1/EGFR/AKT/ERK signaling axis.
Collapse
Affiliation(s)
- Fan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qianxi Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mingyu Peng
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiayi Xiao
- West China School of Medicine and West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, PR China
| | - Yiwei Zeng
- CHINA MEDICAL UNIVERSITY, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Lin Yi
- CHONGQING MEDICAL UNIVERSITY, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, PR China
| | - Zhuoqing Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Rui Tian
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
39
|
Bortnevskaya YS, Shiryaev NA, Zakharov NS, Kitoroage OO, Gradova MA, Karpechenko NY, Novikov AS, Nikolskaya ED, Mollaeva MR, Yabbarov NG, Bragina NA, Zhdanova KA. Synthesis and Biological Properties of EGFR-Targeted Photosensitizer Based on Cationic Porphyrin. Pharmaceutics 2023; 15:pharmaceutics15041284. [PMID: 37111769 PMCID: PMC10145264 DOI: 10.3390/pharmaceutics15041284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Photodynamic therapy (PDT) in oncology is characterized by low invasiveness, minimal side effects, and little tissue scarring. Increasing the selectivity of PDT agents toward a cellular target is a new approach intended to improve this method. This study is devoted to the design and synthesis of a new conjugate based on meso-arylporphyrin with a low-molecular-weight tyrosine kinase inhibitor, Erlotinib. A nano-formulation based on Pluronic F127 micelles was obtained and characterized. The photophysical and photochemical properties and biological activity of the studied compounds and their nano-formulation were studied. A significant, 20-40-fold difference between the dark and photoinduced activity was achieved for the conjugate nanomicelles. After irradiation, the studied conjugate nanomicelles were 1.8 times more toxic toward the EGFR-overexpressing cell line MDA-MB-231 compared to the conditionally normal NKE cells. The IC50 was 0.073 ± 0.014 μM for the MDA-MB-231 cell line and 0.13 ± 0.018 μM for NKE cells after irradiation for the target conjugate nanomicelles.
Collapse
Affiliation(s)
- Yulia S Bortnevskaya
- Institute of Fine Chemical Technology, MIREA-Russian Technological University, Vernadsky pr., 86, 119571 Moscow, Russia
| | - Nikita A Shiryaev
- Institute of Fine Chemical Technology, MIREA-Russian Technological University, Vernadsky pr., 86, 119571 Moscow, Russia
| | - Nikita S Zakharov
- Institute of Fine Chemical Technology, MIREA-Russian Technological University, Vernadsky pr., 86, 119571 Moscow, Russia
| | - Oleg O Kitoroage
- Institute of Fine Chemical Technology, MIREA-Russian Technological University, Vernadsky pr., 86, 119571 Moscow, Russia
| | - Margarita A Gradova
- N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Kosygin St., 4, 119991 Moscow, Russia
| | - Natalia Yu Karpechenko
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe Highway, 24, 115522 Moscow, Russia
- Department of Medical Chemistry and Toxicology, Pirogov National Research Medical University, Ministry of Health of Russia, Ostrovityanova St., 1, 117997 Moscow, Russia
| | - Alexander S Novikov
- Institute of Chemistry, Saint Petersburg State University, Universitetskaya nab. 7-9, 199034 Saint Petersburg, Russia
- Research Institute of Chemistry, Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, 117198 Moscow, Russia
| | - Elena D Nikolskaya
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygina St., 4, 119334 Moscow, Russia
| | - Mariia R Mollaeva
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygina St., 4, 119334 Moscow, Russia
| | - Nikita G Yabbarov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygina St., 4, 119334 Moscow, Russia
| | - Natal'ya A Bragina
- Institute of Fine Chemical Technology, MIREA-Russian Technological University, Vernadsky pr., 86, 119571 Moscow, Russia
| | - Kseniya A Zhdanova
- Institute of Fine Chemical Technology, MIREA-Russian Technological University, Vernadsky pr., 86, 119571 Moscow, Russia
| |
Collapse
|
40
|
Chuang TC, Fang GS, Hsu SC, Lee YJ, Shao WS, Wang V, Lee SL, Kao MC, Ou CC. Baicalein suppresses HER2-mediated malignant transformation of HER2-overexpressing ovarian cancer cells by downregulating HER2 gene expression. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 36988316 DOI: 10.1002/tox.23790] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/02/2022] [Accepted: 03/19/2023] [Indexed: 06/19/2023]
Abstract
The upregulation of the HER2 oncogene is associated with a variety of human cancers and is associated with poor prognosis. Baicalein is reported to have anti-tumor activity, but the molecular mechanism of this effect in HER2-positive cancer cells has not been studied. In this study, our data showed that baicalein can inhibit the proliferation and transformation potential of ovarian cancer cells overexpressing HER2. Baicalein treatment caused a dose-dependent inhibition of HER2 gene expression at the transcriptional level. Baicalein acted on ovarian cancer cells overexpressing HER2 to downregulate the PI3K/Akt signaling pathway downstream of HER2 and inhibit the expression or activity of downstream targets, such as VEGF and cyclin D1 and MMP2. Oral administration of baicalein supplemented with a pharmaceutical excipient significantly inhibited the growth of HER2-overexpressing ovarian SKOV-3 cancer xenografts in mice. These results suggest that downregulation of HER2 gene expression by baicalein at the transcriptional level contributes to inhibit the in vitro and in vivo proliferation and HER2-mediated malignant transformation of HER2-overexpressing ovarian cancer cells.
Collapse
Affiliation(s)
- Tzu-Chao Chuang
- Department of Chemistry, Tamkang University, New Taipei, Taiwan, R.O.C
| | - Guan-Shiun Fang
- Department of Chemistry, Tamkang University, New Taipei, Taiwan, R.O.C
| | - Shih-Chung Hsu
- Department of Early Childhood Care and Education, University of Kang Ning, Taipei, Taiwan, R.O.C
| | - Yi-Jen Lee
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Wei-Syun Shao
- Department of Chemistry, Tamkang University, New Taipei, Taiwan, R.O.C
| | - Vinchi Wang
- Department of Neurology, Cardinal Tien Hospital, New Taipei, Taiwan, R.O.C
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan, R.O.C
| | - Shou-Lun Lee
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Ming-Ching Kao
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Chien-Chih Ou
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, Taipei, Taiwan, R.O.C
| |
Collapse
|
41
|
Wang X, Tang G, Guo H, Ma J, Liu D, Wang Y, Jin R, Li Z, Tang Y. Research Progress on the Anti-Tumor Mechanism and Reversal of Multidrug Resistance of Zuojin Pill and its Main Components, Evodiamine and Berberine. Nat Prod Commun 2023; 18. [DOI: 10.1177/1934578x231161414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Background Cancer is one of the most serious diseases worldwide that threatens human health and leads to death. Chemotherapy is the main clinical method to treat tumors, but, despite the development of new chemotherapeutic drugs, the multidrug resistance (MDR) of cancer cells to conventional chemotherapeutic drugs remains a major cause of failure in cancer prevention and treatment. Therefore, overcoming this resistance has become a major challenge in cancer prevention and treatment. Method With the in-depth study of traditional Chinese medicines (TCMs) for the treatment of tumors, many such medicines have been found that can reverse MDR and enhance the sensitivity of chemotherapy. ZJW is a famous traditional medicine formula from China, recorded first in an ancient medicine book named Danxi Xinfa. It is composed of Huanglian and Wuzhuyu in a ratio of 6:1 by mass. Conclusion ZJW can inhibit proliferation, induce apoptosis, inhibit invasion and metastasis, and reverse MDR of tumor cells through multiple pathways and multiple targets. In this paper, we briefly review recent research on ZJW and its main components, evodiamine and berberine, in the anti-tumor mechanism and reversal of multidrug resistance.
Collapse
Affiliation(s)
- Xinyi Wang
- Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, XianYang, China
| | - Gonghuan Tang
- Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, XianYang, China
| | - Hui Guo
- Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, XianYang, China
| | - Jingjing Ma
- Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, XianYang, China
| | - Dongmei Liu
- No.988 Hospital of Joint Logistic Support Force, Zhengzhou, China
| | - Yuwei Wang
- Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, XianYang, China
| | - Ruyi Jin
- Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, XianYang, China
| | - Zhi Li
- Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, XianYang, China
| | - Yuping Tang
- Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, XianYang, China
| |
Collapse
|
42
|
(Stămat) LRB, Dinescu S, Costache M. Regulation of Inflammasome by microRNAs in Triple-Negative Breast Cancer: New Opportunities for Therapy. Int J Mol Sci 2023; 24:ijms24043245. [PMID: 36834660 PMCID: PMC9963301 DOI: 10.3390/ijms24043245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
During the past decade, researchers have investigated the molecular mechanisms of breast cancer initiation and progression, especially triple-negative breast cancer (TNBC), in order to identify specific biomarkers that could serve as feasible targets for innovative therapeutic strategies development. TNBC is characterized by a dynamic and aggressive nature, due to the absence of estrogen, progesterone and human epidermal growth factor 2 receptors. TNBC progression is associated with the dysregulation of nucleotide-binding oligomerization domain-like receptor and pyrin domain-containing protein 3 (NLRP3) inflammasome, followed by the release of pro-inflammatory cytokines and caspase-1 dependent cell death, termed pyroptosis. The heterogeneity of the breast tumor microenvironment triggers the interest of non-coding RNAs' involvement in NLRP3 inflammasome assembly, TNBC progression and metastasis. Non-coding RNAs are paramount regulators of carcinogenesis and inflammasome pathways, which could help in the development of efficient treatments. This review aims to highlight the contribution of non-coding RNAs that support inflammasome activation and TNBC progression, pointing up their potential for clinical applications as biomarkers for diagnosis and therapy.
Collapse
Affiliation(s)
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania
- Research Institute of the University of Bucharest, 050663 Bucharest, Romania
- Correspondence:
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania
- Research Institute of the University of Bucharest, 050663 Bucharest, Romania
| |
Collapse
|
43
|
Mohiuddin TM, Zhang C, Sheng W, Al-Rawe M, Zeppernick F, Meinhold-Heerlein I, Hussain AF. Near Infrared Photoimmunotherapy: A Review of Recent Progress and Their Target Molecules for Cancer Therapy. Int J Mol Sci 2023; 24:2655. [PMID: 36768976 PMCID: PMC9916513 DOI: 10.3390/ijms24032655] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023] Open
Abstract
Near infrared photoimmunotherapy (NIR-PIT) is a newly developed molecular targeted cancer treatment, which selectively kills cancer cells or immune-regulatory cells and induces therapeutic host immune responses by administrating a cancer targeting moiety conjugated with IRdye700. The local exposure to near-infrared (NIR) light causes a photo-induced ligand release reaction, which causes damage to the target cell, resulting in immunogenic cell death (ICD) with little or no side effect to the surrounding normal cells. Moreover, NIR-PIT can generate an immune response in distant metastases and inhibit further cancer attack by combing cancer cells targeting NIR-PIT and immune regulatory cells targeting NIR-PIT or other cancer treatment modalities. Several recent improvements in NIR-PIT have been explored such as catheter-driven NIR light delivery, real-time monitoring of cancer, and the development of new target molecule, leading to NIR-PIT being considered as a promising cancer therapy. In this review, we discuss the progress of NIR-PIT, their mechanism and design strategies for cancer treatment. Furthermore, the overall possible targeting molecules for NIR-PIT with their application for cancer treatment are briefly summarised.
Collapse
|
44
|
Juzeniene A, Stenberg VY, Bruland ØS, Revheim ME, Larsen RH. Dual targeting with 224Ra/ 212Pb-conjugates for targeted alpha therapy of disseminated cancers: A conceptual approach. Front Med (Lausanne) 2023; 9:1051825. [PMID: 36733936 PMCID: PMC9887039 DOI: 10.3389/fmed.2022.1051825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/05/2022] [Indexed: 01/18/2023] Open
Abstract
Metastases are the primary cause of death among cancer patients and efficacious new treatments are sorely needed. Targeted alpha-emitting radiopharmaceuticals that are highly cytotoxic may fulfill this critical need. The focus of this paper is to describe and explore a novel technology that may improve the therapeutic effect of targeted alpha therapy by combining two radionuclides from the same decay chain in the same solution. We hypothesize that the dual targeting solution containing bone-seeking 224Ra and cell-directed complexes of progeny 212Pb is a promising approach to treat metastatic cancers with bone and soft tissue lesions as well as skeletal metastases of mixed lytic/osteoblastic nature. A novel liquid 224Ra/212Pb-generator for rapid preparation of a dual targeting solution is described. Cancer cell targeting monoclonal antibodies, their fragments, synthetic proteins or peptides can all be radiolabeled with 212Pb in the 224Ra-solution in transient equilibrium with daughter nuclides. Thus, 224Ra targets stromal elements in sclerotic bone metastases and 212Pb-chelated-conjugate targets tumor cells of metastatic prostate cancer or osteosarcoma. The dual targeting solution may also be explored to treat metastatic breast cancer or multiple myeloma after manipulation of bone metastases to a more osteoblastic phenotype by the use of bisphosphonates, denosumab, bortezomib or hormone therapy prior to treatment. This may improve targeting of bone-seeking 224Ra and render an augmented radiation dose deposited within metastases. Our preliminary preclinical studies provide conceptual evidence that the dual 224Ra-solution with bone or tumor-targeted delivery of 212Pb has potential to inhibit cancer metastases without significant toxicity. In some settings, the use of a booster dose of purified 212Pb-conjugate alone could be required to elevate the effect of this tumor cell directed component, if needed, e.g., in a fractionated treatment regimen, where the dual targeting solution will act as maintenance treatment.
Collapse
Affiliation(s)
- Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- ARTBIO AS, Oslo, Norway
| | - Øyvind Sverre Bruland
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Mona-Elisabeth Revheim
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
45
|
The expression and clinical significance of STAMBP in breast cancer. Mol Biol Rep 2023; 50:899-906. [PMID: 36309616 DOI: 10.1007/s11033-022-07964-3] [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: 07/16/2022] [Accepted: 09/21/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Breast cancer is the leading cause of death from cancer in women worldwide. STAMBP functions as a JAMM family deubiquitinating enzyme that modulates the stability of substrate proteins in cells by cleaving ubiquitin moieties. The expression of STAMBP and its clinical significance in breast cancer remain unclear. METHODS AND RESULTS The level of the STAMBP protein in noncancerous and tumor tissues of breast cancer patients was examined by immunohistochemical staining. The expression of STAMBP mRNA in tissues based on healthy individual and breast cancer patient data in the TCGA database was evaluated. The association between the expression of STAMBP mRNA and clinical features and prognosis was evaluated using TCGA database. Cell growth was assessed by Cell Counting Kit-8 (CCK-8) assay, and cell migration and invasion were assessed by wound healing and Transwell assays. Activation of the ERK signaling was detected by Western blotting. The expression of STAMBP was markedly upregulated in the cytoplasm of tumor cells from breast cancer patients. The level of STAMBP was closely associated with the tumor subtype and size and the TNM stage of the breast cancer patients. Importantly, high expression of STAMBP predicted poor overall survival (OS) for breast cancer patients. Furthermore, knockdown of STAMBP expression reduced cell mobility and invasion of breast cancer cells. Notably, the phosphorylation of EGFR and ERK was markedly reduced in STAMBP-knockdown cells. CONCLUSION STAMBP plays a critical role in the progression of breast cancer and may serve as a biomarker to monitor the progression of the disease.
Collapse
|
46
|
Mechanisms and Strategies to Overcome PD-1/PD-L1 Blockade Resistance in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 15:cancers15010104. [PMID: 36612100 PMCID: PMC9817764 DOI: 10.3390/cancers15010104] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a high rate of systemic metastasis, insensitivity to conventional treatment and susceptibility to drug resistance, resulting in a poor patient prognosis. The immune checkpoint inhibitors (ICIs) represented by antibodies of programmed death receptor 1 (PD-1) and programmed death receptor ligand 1 (PD-L1) have provided new therapeutic options for TNBC. However, the efficacy of PD-1/PD-L1 blockade monotherapy is suboptimal immune response, which may be caused by reduced antigen presentation, immunosuppressive tumor microenvironment, interplay with other immune checkpoints and aberrant activation of oncological signaling in tumor cells. Therefore, to improve the sensitivity of TNBC to ICIs, suitable patients are selected based on reliable predictive markers and treated with a combination of ICIs with other therapies such as chemotherapy, radiotherapy, targeted therapy, oncologic virus and neoantigen-based therapies. This review discusses the current mechanisms underlying the resistance of TNBC to PD-1/PD-L1 inhibitors, the potential biomarkers for predicting the efficacy of anti-PD-1/PD-L1 immunotherapy and recent advances in the combination therapies to increase response rates, the depth of remission and the durability of the benefit of TNBC to ICIs.
Collapse
|
47
|
Sonousi A, Hassan RA, Osman EO, Abdou AM, Emam SH. Design and synthesis of novel quinazolinone-based derivatives as EGFR inhibitors with antitumor activity. J Enzyme Inhib Med Chem 2022; 37:2644-2659. [PMID: 36146940 PMCID: PMC9518264 DOI: 10.1080/14756366.2022.2118735] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Nineteen new quinazolin-4(3H)-one derivatives 3a–g and 6a–l were designed and synthesised to inhibit EGFR. The antiproliferative activity of the synthesised compounds was tested in vitro against 60 different human cell lines. The most potent compound 6d displayed superior sub-micromolar antiproliferative activity towards NSC lung cancer cell line NCI-H460 with GI50 = 0.789 µM. It also showed potent cytostatic activity against 40 different cancer cell lines (TGI range: 2.59–9.55 µM). Compound 6d potently inhibited EGFR with IC50 = 0.069 ± 0.004 µM in comparison to erlotinib with IC50 value of 0.045 ± 0.003 µM. Compound 6d showed 16.74-fold increase in total apoptosis and caused cell cycle arrest at G1/S phase in breast cancer HS 578T cell line. Moreover, the most potent derivatives were docked into the EGFR active site to determine their binding mode and confirm their ability to satisfy the pharmacophoric features required for EGFR inhibition.
Collapse
Affiliation(s)
- Amr Sonousi
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rasha A Hassan
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Eman O Osman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Amr M Abdou
- Department of Microbiology and Immunology, National Research Centre, Dokki, Giza, Egypt
| | - Soha H Emam
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
48
|
Sabry MA, Ghaly MA, Maarouf AR, El-Subbagh HI. New thiazole-based derivatives as EGFR/HER2 and DHFR inhibitors: Synthesis, molecular modeling simulations and anticancer activity. Eur J Med Chem 2022; 241:114661. [PMID: 35964425 DOI: 10.1016/j.ejmech.2022.114661] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022]
Abstract
New series of thiazole and imidazo[2,1-b]thiazole derivatives were synthesized and tested for their in vitro anticancer activity. Compounds 27, 34, 39 and 42-44 showed the best anticancer activity against the tested cancer cell lines with high safety profile and selectivity indices, especially MCF-7 breast cancer, compared to sorafenib. As an attempt to reveal their mode of cytotoxicity, EGFR, HER2 kinase and DHFR inhibition assays were performed. Compounds 39 and 43 were the most potent dual EGFR/HER2 kinase inhibitors, with IC50 values of 0.153 (EGFR), 0.108 (HER2) and 0.122 (EGFR), 0.078 (HER2) μM, respectively. 39 and 42 were the best DHFR inhibitors showing IC50 0.291 and 0.123 μM, respectively. 39 and 43 induced their cytotoxicity via cell cycle arrest at G1/S and G1 phases, respectively, and apoptosis rather than necrosis in the MCF-7 breast cancer cell line. In vivo anti-breast cancer assay of 39 and 43 showed significant tumor volume reduction with recovered caspase-3 immunoexpression. Modeling study results proved the importance of the 5-(4-substituted phenyl)-imidazo[2,1-b]thiazole moiety and the hydrazide side chain for the anticancer activity. The most potent compounds showed good drug-likeness features and could be used as prototypes for further optimization. 39 could be an example of a multi-targeting anticancer agent that acts by inhibiting EGFR/HER2 kinase, DHFR enzymes and cellular apoptosis.
Collapse
Affiliation(s)
- Mohamed A Sabry
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt.
| | - Mariam A Ghaly
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt.
| | - Azza R Maarouf
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt
| | - Hussein I El-Subbagh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt.
| |
Collapse
|
49
|
Gandullo-Sánchez L, Ocaña A, Pandiella A. HER3 in cancer: from the bench to the bedside. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:310. [PMID: 36271429 PMCID: PMC9585794 DOI: 10.1186/s13046-022-02515-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
Abstract
The HER3 protein, that belongs to the ErbB/HER receptor tyrosine kinase (RTK) family, is expressed in several types of tumors. That fact, together with the role of HER3 in promoting cell proliferation, implicate that targeting HER3 may have therapeutic relevance. Furthermore, expression and activation of HER3 has been linked to resistance to drugs that target other HER receptors such as agents that act on EGFR or HER2. In addition, HER3 has been associated to resistance to some chemotherapeutic drugs. Because of those circumstances, efforts to develop and test agents targeting HER3 have been carried out. Two types of agents targeting HER3 have been developed. The most abundant are antibodies or engineered antibody derivatives that specifically recognize the extracellular region of HER3. In addition, the use of aptamers specifically interacting with HER3, vaccines or HER3-targeting siRNAs have also been developed. Here we discuss the state of the art of the preclinical and clinical development of drugs aimed at targeting HER3 with therapeutic purposes.
Collapse
Affiliation(s)
- Lucía Gandullo-Sánchez
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Alberto Ocaña
- grid.411068.a0000 0001 0671 5785Hospital Clínico San Carlos and CIBERONC, 28040 Madrid, Spain
| | - Atanasio Pandiella
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
50
|
Abbasian MH, Ardekani AM, Sobhani N, Roudi R. The Role of Genomics and Proteomics in Lung Cancer Early Detection and Treatment. Cancers (Basel) 2022; 14:5144. [PMID: 36291929 PMCID: PMC9600051 DOI: 10.3390/cancers14205144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 08/17/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide, with non-small-cell lung cancer (NSCLC) being the primary type. Unfortunately, it is often diagnosed at advanced stages, when therapy leaves patients with a dismal prognosis. Despite the advances in genomics and proteomics in the past decade, leading to progress in developing tools for early diagnosis, targeted therapies have shown promising results; however, the 5-year survival of NSCLC patients is only about 15%. Low-dose computed tomography or chest X-ray are the main types of screening tools. Lung cancer patients without specific, actionable mutations are currently treated with conventional therapies, such as platinum-based chemotherapy; however, resistances and relapses often occur in these patients. More noninvasive, inexpensive, and safer diagnostic methods based on novel biomarkers for NSCLC are of paramount importance. In the current review, we summarize genomic and proteomic biomarkers utilized for the early detection and treatment of NSCLC. We further discuss future opportunities to improve biomarkers for early detection and the effective treatment of NSCLC.
Collapse
Affiliation(s)
- Mohammad Hadi Abbasian
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 1497716316, Iran
| | - Ali M. Ardekani
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 1497716316, Iran
| | - Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Raheleh Roudi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|