1
|
Mirzanejad S, Bagherzadeh M, Bayrami A, Daneshgar H, Bahrami A, Mahdavi M. Improving the drug delivery performance of ZIF-8 with amine functionalization as a 5-fluorouracil nanocarrier. Sci Rep 2025; 15:18793. [PMID: 40442254 PMCID: PMC12123014 DOI: 10.1038/s41598-025-03542-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 05/21/2025] [Indexed: 06/02/2025] Open
Abstract
This study investigates the effect of amine functional groups in ZIF-8 metal-organic frameworks on the loading and release of 5-fluorouracil (5-FU). The facile and cost-effective solvent-assisted linker exchange (SALE) method was used to exchange 2-methylimidazole (2-MIM) linkers with 3-amino-1,2,4-triazole (Atz) in the ZIF-8 structure, which resulted in a synthesis of ZIF-8A with 22, 53, and 74% Atz exchange, respectively. The prepared nanoparticles were characterized by 1H-NMR, XRD, FT-IR, FE-SEM, UV-Vis spectroscopy, and zeta potential analysis. Drug encapsulation efficiency results showed 12% for 5-FU@ZIF-8 which increased to 48% for 5-FU@ZIF-8A(53%). Also, the results of in-vitro experiments exhibited the pH-responsive behavior of nanocarriers and slower release for 5-FU@ZIF-8A(53%) compared to 5-FU@ZIF-8. The increase in drug encapsulation efficiency and slower release is due to the presence of the amine functional group in the structure, which improves the host-guest interactions between drug molecules and linkers. Moreover, the MTT assay was performed on MCF-7 and HFF-2 cell lines which revealed that 5-FU@ZIF-8A(53%) exhibited more significant cytotoxicity toward cancer cells while less toxicity toward normal cells compared to 5-FU@ZIF-8. These findings highlight the capability of amine-functionalized ZIF-8 as an effective drug delivery system for 5-FU and demonstrate the potential of the facial and low-cost SALE approach as a promising technique in nanocarrier development.
Collapse
Affiliation(s)
- Sahba Mirzanejad
- Department of Chemistry, Sharif University of Technology, PO Box 11155-3615, Tehran, Iran
| | - Mojtaba Bagherzadeh
- Department of Chemistry, Sharif University of Technology, PO Box 11155-3615, Tehran, Iran.
| | - Arshad Bayrami
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin, Iran
| | - Hossein Daneshgar
- Department of Chemistry, Sharif University of Technology, PO Box 11155-3615, Tehran, Iran
| | - Aida Bahrami
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Majid Mahdavi
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| |
Collapse
|
2
|
Wu Y, Li T, Li Z, Lortie F, Bernard J, Binder WH, Chen S, Zhu J. "Mix-and-Match": Self-Sorting Assembly Governed Supramolecular Polymeric Nanomedicine for Boosting Combined Chemo/Phototherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2502416. [PMID: 40190107 DOI: 10.1002/adma.202502416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/24/2025] [Indexed: 05/28/2025]
Abstract
Precise cancer nanomedicine requires rational molecular instructions of therapeutic agents. Harnessing the structure-property-function relationships represents a practical strategy toward smart and effective nanomedicine. A structurally novel hydrogen-bonded (H-bonded) supramolecular nanoformulation generated by orthogonal self-sorting assembly of chemo-prodrug (FPtF) and phototherapeutics (BPeB) is here reported, to reach an autonomous nanomedicine with improved anti-tumor efficacy by combining chemo/phototherapy (CT/PT). The high-fidelity of H-bonding modularity from privileged heterocomplementary diaminopyridine/5-fluorouracil (DAP/FU) and Hamilton wedge/barbiturate (HW/Ba) pairs, respectively enable the precise spatial control of binding interactions toward FPtF and BPeB, in turn allowing the self-sorting process and specific "mix-and-match" capability. To directly stimulate phototherapy from BPeB via near-infrared (NIR) light, spectral matched upconversion nanoparticles (UCNPs, β-NaYF4:Yb,Er) are encapsulated simultaneously. As a result, supramolecular polymeric nanomicelles, i.e., F/B/U@PHDO, are readily fabricated. Moreover, distinct H-bonding association constant (Ka) of DAP/FU (≈102 M-1) and HW/Ba (≈104-5 M-1) pairs reflect different strengths and stabilities of H-bonds, thus endowing the programmable H-bonding dissociation, accompanied with the chemo-prodrug release through pH/thermal-stimuli. Therapeutic regime with appreciated anti-tumor outcomes is ultimately accomplished via combined CT/PT. The privileged opportunities offered by self-sorting design are anticipated to point to new paradigm toward precise nanomedicine for cancer therapy.
Collapse
Affiliation(s)
- Yanggui Wu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ting Li
- Cosme Biotechnology Co., Ltd., Wuhan, 430 074, China
| | - Zeke Li
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Frédéric Lortie
- Universite Claude Bernard Lyon 1, INSA Lyon, Université Jean Monnet, CNRS, UMR 5223, Ingénierie des Matériaux Polymères, Cédex, F-69621, France
| | - Julien Bernard
- Universite Claude Bernard Lyon 1, INSA Lyon, Université Jean Monnet, CNRS, UMR 5223, Ingénierie des Matériaux Polymères, Cédex, F-69621, France
| | - Wolfgang H Binder
- Chair of Macromolecular Chemistry, Faculty of Natural Science II (Chemistry, Physics and Mathematics), Martin Luther University Halle-Wittenberg, von Danckelmann-Platz 4, D-06120, Halle (Saale), Germany
| | - Senbin Chen
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
3
|
Wu L, Chen X, Chen D, Chen Q, Wu F. A novel tRNA-Derived fragment, tRF-20-M0NK5Y93 inhibits the malignant progression of non-small cell lung cancer by mediating PLOD1. Arch Biochem Biophys 2025; 770:110431. [PMID: 40262693 DOI: 10.1016/j.abb.2025.110431] [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: 12/26/2024] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE Non-small cell lung cancer (NSCLC) constitutes a common malignant tumor characterized by substantial mortality rates. Transfer RNA (tRNA)-derived small RNAs (tsRNAs) have been implicated in the progression of various cancers, including NSCLC. However, to date, only a limited number of tsRNAs have been reported to be involved in the development of NSCLC. Hence, the present study aimed to investigate the potential roles of tsRNAs in the progression of NSCLC. MATERIALS AND METHODS A total of forty-six patients with NSCLC who underwent surgical resection were enrolled in this study. The expression patterns of tRNA-derived fragments (tRFs) in tumor and normal tissues of 6 NSCLC cases were investigated using RNA-sequencing assay. Cell viability, proliferation capacity, and migratory ability were measured utilizing the CCK-8, colony formation, and Transwell assays, respectively. Furthermore, cell apoptosis was evaluated by applying flow cytometry. The xenograft tumor model was constructed to observe tumor growth. The relationship between tRF-20-M0NK5Y93 and PLOD1 was clarified using the luciferase and RIP assays. RESULTS The RNA-sequencing assay revealed a significant decrease in the expression of tRF-20-M0NK5Y93 in tumor tissues. In line with this finding, qRT-PCR analysis further confirmed a meaningful downregulation of tRF-20-M0NK5Y93 in 46 patient samples with NSCLC. Importantly, this downregulated expression was strongly correlated with reduced patient survival. Additionally, overexpression of tRF-20-M0NK5Y93 was found to inhibit the proliferation and migration capabilities of NSCLC cells, leading to suppressed tumor growth and accelerated apoptosis. Furthermore, tRF-20-M0NK5Y93 was capable of binding to PLOD1, thereby negatively regulating its expression. Notably, the restoration of PLOD1 expression was able to counteract the inhibitory effects of enforced tRF-20-M0NK5Y93 on NSCLC cell proliferation, migration, and apoptosis. CONCLUSION The expression level of tRF-20-M0NK5Y93 was found to be decreased in NSCLC. Overexpressed tRF-20-M0NK5Y93 exhibited inhibitory effects on NSCLC cell proliferation and migration, accelerated apoptosis, and suppressed tumor growth by targeting PLOD1. These findings highlight tRF-20-M0NK5Y93 as a promising target for NSCLC therapy.
Collapse
Affiliation(s)
- Lixin Wu
- Department of Respiratory Medicine, Zhejiang Rongjun Hospital (The Third Affiliated Hospital of Jiaxing University), Jiaxing, 314000, Zhejiang, China
| | - Xiaojie Chen
- Department of Respiratory Medicine, Zhejiang Rongjun Hospital (The Third Affiliated Hospital of Jiaxing University), Jiaxing, 314000, Zhejiang, China
| | - Dong Chen
- Department of Pathology, Zhejiang Rongjun Hospital (The Third Affiliated Hospital of Jiaxing University), Jiaxing, 314000, Zhejiang, China
| | - Qin Chen
- Department of Respiratory Medicine, Zhejiang Rongjun Hospital (The Third Affiliated Hospital of Jiaxing University), Jiaxing, 314000, Zhejiang, China.
| | - Fengjie Wu
- Department of Respiratory Medicine, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing University, Jiaxing, 314000, Zhejiang, China.
| |
Collapse
|
4
|
Consoli V, Sorrenti V, Gulisano M, Spampinato M, Vanella L. Navigating heme pathways: the breach of heme oxygenase and hemin in breast cancer. Mol Cell Biochem 2025; 480:1495-1518. [PMID: 39287890 PMCID: PMC11842487 DOI: 10.1007/s11010-024-05119-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024]
Abstract
Breast cancer remains a significant global health challenge, with diverse subtypes and complex molecular mechanisms underlying its development and progression. This review comprehensively examines recent advances in breast cancer research, with a focus on classification, molecular pathways, and the role of heme oxygenases (HO), heme metabolism implications, and therapeutic innovations. The classification of breast cancer subtypes based on molecular profiling has significantly improved diagnosis and treatment strategies, allowing for tailored approaches to patient care. Molecular studies have elucidated key signaling pathways and biomarkers implicated in breast cancer pathogenesis, shedding light on potential targets for therapeutic intervention. Notably, emerging evidence suggests a critical role for heme oxygenases, particularly HO-1, in breast cancer progression and therapeutic resistance, highlighting the importance of understanding heme metabolism in cancer biology. Furthermore, this review highlights recent advances in breast cancer therapy, including targeted therapies, immunotherapy, and novel drug delivery systems. Understanding the complex interplay between breast cancer subtypes, molecular pathways, and innovative therapeutic approaches is essential for improving patient outcomes and developing more effective treatment strategies in the fight against breast cancer.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Maria Gulisano
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Mariarita Spampinato
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy.
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy.
| |
Collapse
|
5
|
Somuncu AM, Parlak Somuncu B, Ozbay AD, Cicek I, Suleyman B, Mammadov R, Bulut S, Bal Tastan T, Coban TA, Suleyman H, Aydin A. Potential effects of adenosine triphosphate and melatonin on oxidative and inflammatory optic nerve damage in rats caused by 5-fluorouracil. Int J Ophthalmol 2025; 18:222-228. [PMID: 39967968 PMCID: PMC11754014 DOI: 10.18240/ijo.2025.02.04] [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: 05/19/2024] [Accepted: 11/19/2024] [Indexed: 02/20/2025] Open
Abstract
AIM To investigate the effects of adenosine triphosphate (ATP) and melatonin, which have antioxidant and anti-inflammatory activities, on potential 5-fluorouracil (5-FU)-induced optic nerve damage in rats. METHODS Twenty-four rats were categorized into four groups of six rats: healthy (HG), 5-FU (FUG), ATP+5-FU (AFU), and melatonin+5-FU (MFU). ATP (4 mg/kg) and melatonin (10 mg/kg) were administered intraperitoneally and orally, respectively. One hour after ATP and melatonin administration, rats in the AFU, MFU, and FUG were intraperitoneally injected with 5-FU (100 mg/kg). ATP and melatonin were administered once daily for 10d. 5-FU was administered at a single dose on days 1, 3, and 5 of the experiment. After 10d, the rats were euthanized and optic nerve tissues were extracted. Optic nerve tissues were biochemically and histopathologically examined. RESULTS ATP and melatonin treatments inhibited the increase in malondialdehyde (MDA) and interleukin-6 (IL-6) levels, which were elevated in the FUG. The treatments also prevented the decrease in total glutathione (tGSH) levels and the superoxide dismutase (SOD) and catalase (CAT) activities (P<0.001). This inhibition was higher in the ATP group than in the melatonin group (P<0.001). ATP prevented histopathological damage better than melatonin (P<0.05). CONCLUSION ATP and melatonin have the potential to be used in alleviating 5-FU-induced optic nerve damage. In addition, ATP treatment shows better protective effects than melatonin.
Collapse
Affiliation(s)
- Ahmet Mehmet Somuncu
- Trabzon Kanuni Education and Research Hospital, Health Sciences University, Trabzon 61250, Turkiey
| | - Busra Parlak Somuncu
- Provincial Health Directorate, Ministry of Health of the Republic of Turkey, Trabzon 61040, Turkiey
| | - Ahmet Duhan Ozbay
- Department of Ophthalmology, Erzurum City Hospital, Health Sciences University, Erzurum 25240, Turkiey
| | - Ibrahim Cicek
- Department of Ophthalmology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkiey
| | - Bahadir Suleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkiey
| | - Renad Mammadov
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkiey
| | - Seval Bulut
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkiey
| | - Tugba Bal Tastan
- Department of Histology and Embryology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkiey
| | - Taha Abdulkadir Coban
- Department of Biochemistry, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
| | - Halis Suleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkiey
| | - Aliyev Aydin
- Department of Pharmacology, Azerbaijan Medical University, Baku, AZ1022, Azerbaijan
| |
Collapse
|
6
|
Hanan H, Pervaiz F, Ijaz M, Arshad T, Saeed K, Ahmad R, Bukhari AN, Ain QU, Javaid SM. Thiol functionalized pH-responsive mucoadhesive hydrogel: Potential approach for site-specific delivery of 5 fluorouracil in colon cancer. Int J Biol Macromol 2025; 289:138887. [PMID: 39701245 DOI: 10.1016/j.ijbiomac.2024.138887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
The present study aims to develop a novel thiolated carboxymethyl cellulose (CMC-SH) by the addition of aldehyde groups via oxidation followed by reductive amination and then develop CMC-SH based pH-responsive hydrogel by free radical polymerization approach while assessing its mucoadhesive and permeation-enhancing capabilities. By in-vitro characterization, the intended compound's chemical composition, thermal stability, and amorphous nature were analyzed for CMC-SH polymer. Ellman's assay was utilized to estimate the thiol content and permeation analysis was performed to evaluate its enhanced permeability characteristics. The properties of developed CMC-SH based hydrogel were demonstrated by in-vitro analysis, structural, mechanical stability analysis, morphological evaluation and mucoadhesive properties. Rheological data showed that the viscous modulus and elastic modules of CMC-SH rose by 3.5 and 2.1 times, respectively. Compared to FU, the permeation showed a notable improvement of 4.89-fold with CMC-SH. According to an ex-vivo mucoadhesion analysis, CMC-SH based hydrogel was able to adhere to the intestinal mucosal layer for 210 min. The biocompatibility of CMC-SH and CMC-SH based hydrogel was demonstrated using Caco-2 cell lines. Thiolation of carboxymethyl cellulose significantly enhanced its mucoadhesive and permeation qualities, which makes the hydrogel desirable mucoadhesive and selective cytotoxic biomaterial for colorectal cancer therapy.
Collapse
Affiliation(s)
- Hanasul Hanan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Fahad Pervaiz
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | - Muhammad Ijaz
- School of Veterinary Medicine University College Dublin, Dublin, Ireland; Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Tahreem Arshad
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Komal Saeed
- Department of Botany, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Rizwan Ahmad
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Arshia Noor Bukhari
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Qurat Ul Ain
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Syeda Munazza Javaid
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
7
|
Imai M, Hiramoto K, Tanaka S, Okayama M, Ooi K. Irinotecan-Induced Site-Specific Pigmentation in the Plantar Region of Mice. Biol Pharm Bull 2025; 48:108-114. [PMID: 39909437 DOI: 10.1248/bpb.b24-00662] [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] [Indexed: 02/07/2025]
Abstract
Skin pigmentation is a widely recognized side effect of cancer chemotherapy that can negatively affect patient QOL. However, although numerous case reports have documented pigmentation caused by anticancer drugs, the precise mechanisms remain unclear. Among such pigmentation, that induced by 5-fluorouracil (5-FU) has garnered considerable attention, whereas reports on irinotecan-induced pigmentation are comparatively limited. In this study, we investigated the pigmentation-related effects of irinotecan in colored hairless mice. Mice received intraperitoneal injections of 20 mg/kg irinotecan, and we subsequently examined the pigmentation of the plantar and buttock regions. The results indicated that irinotecan specifically induces pigmentation in the plantar region, with no pigmentation observed on the buttocks. In contrast, pigmentation was noted on the buttocks, although not in the plantar region, in the control mice treated with 5-FU and cytarabine. Furthermore, irinotecan treatment promoted a marked elevation in the expression of tyrosinase, cAMP response element binding protein (CREB), and microphthalmia-associated transcription factor (MITF) in the plantar region, whereas no significant changes were observed in the buttocks. These findings indicate that irinotecan leads to site-specific pigmentation in the sole of the foot, thereby highlighting the potential for anticancer drugs to cause localized pigmentation.
Collapse
Affiliation(s)
- Masashi Imai
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8607, Japan
| | - Keiichi Hiramoto
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8607, Japan
| | - Shota Tanaka
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8607, Japan
| | - Mei Okayama
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8607, Japan
| | - Kazuya Ooi
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8607, Japan
| |
Collapse
|
8
|
Salerno L, Notaro A, Consoli V, Affranchi F, Pittalà V, Sorrenti V, Vanella L, Giuliano M, Intagliata S. Evaluation of the anticancer effects exerted by 5-fluorouracil and heme oxygenase-1 inhibitor hybrids in HTC116 colorectal cancer cells. J Enzyme Inhib Med Chem 2024; 39:2337191. [PMID: 38634597 PMCID: PMC11028004 DOI: 10.1080/14756366.2024.2337191] [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: 10/17/2023] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
Colon cancer remains a clinical challenge in industrialised countries. Its treatment with 5-Flurouracil (5-FU) develops many side effects and resistance. Thus, several strategies have been undertaken so far, including the use of drug cocktails and polypharmacology. Heme oxygenase-1 (HO-1) is an emerging molecular target in the treatment of various cancers. We recently demonstrated that a combination of HO-1 inhibitors with 5-FU and the corresponding hybrids SI1/17, SI1/20, and SI1/22, possessed anticancer activity against prostate and lung cancer cells. In this work, we evaluated these hybrids in a model of colon cancer and found that SI1/22 and the respective combo have greater potency than 5-FU. Particularly, compounds inhibit HO-1 activity in cell lysates, increase ROS and the expression of HO-1, SOD, and Nrf2. Moreover, we observed a decrease of pro-caspase and an increase in cleaved PARP-1 and p62, suggesting apoptotic and autophagic cell death and potential application of these drugs as anticancer agents.
Collapse
Affiliation(s)
- Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Antonietta Notaro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Federica Affranchi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Molecular Medicine, Arabian Gulf University, Manama, Bahrain
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Michela Giuliano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | | |
Collapse
|
9
|
Heise NV, Meyer SJ, Csuk R, Mueller T. Dehydroabietylamine-substituted trifluorobenzene sulfonamide rhodamine B hybrids as anticancer agents overcoming drug resistance. Eur J Med Chem 2024; 276:116667. [PMID: 38996651 DOI: 10.1016/j.ejmech.2024.116667] [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: 06/18/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Attachment of a conjugate assembled from a novel fluorinated carbonic anhydrase inhibitor and rhodamine B onto dehydroabietylamine (DHA) or cyclododecylamine led to first-in-class conjugates of good cytotoxicity; thereby IC50 values (from SRB assays; employed tumor cell lines A2780, A2780Cis, A549, HT29, MCF7, and non-malignant human fibroblasts CCD18Co) between 0.2 and 0.7 μM were found. Both conjugates showed similar cytotoxic activity but the dehydroabietylamine derived conjugate outperformed its cyclododecyl analog in terms of tumor cell/non-tumor cell selectivity. Both conjugates accumulate intracellular, and the DHA conjugate was able to overcome drug resistance which is effective independent of the expression status of carbonic anhydrase IX.
Collapse
Affiliation(s)
- Niels V Heise
- Organic Chemistry, Martin-Luther University Halle-Wittenberg, Kurt-Mothes-Str. 2, D-06120, Halle (Saale), Germany
| | - Sven J Meyer
- Organic Chemistry, Martin-Luther University Halle-Wittenberg, Kurt-Mothes-Str. 2, D-06120, Halle (Saale), Germany
| | - René Csuk
- Organic Chemistry, Martin-Luther University Halle-Wittenberg, Kurt-Mothes-Str. 2, D-06120, Halle (Saale), Germany.
| | - Thomas Mueller
- University Clinic for Internal Medicine IV, Hematology/Oncology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Str. 40, D-06120, Halle (Saale), Germany
| |
Collapse
|
10
|
Bai SR, Zhao BX, Zhao Q, Ge YC, Li M, Zhao CG, Wu XJ, Wang XB. Oleanolic acid improves 5-fluorouracil-induced intestinal damage and inflammation by alleviating intestinal senescence. Sci Rep 2024; 14:21852. [PMID: 39300121 DOI: 10.1038/s41598-024-72536-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
5-Fluorouracil (5-FU) is used as a standard first-line drug for colorectal cancer malignancy (CRC), but it brings a series of side effects such as severe diarrhea and intestinal damage. Our previous study found that a large number of senescent cells increased while 5-Fu induced intestinal damage, and anti-senescence drugs can alleviate its side effects of inflammatory damage. Oleanolic acid (OA) is a common pentacyclic triterpenoid mainly derived from food fungi and medicinal plants, and studies have shown that it mainly possesses hepatoprotective, enzyme-lowering, anti-inflammatory, and anti-tumor effects. But its role in senescence is still unclear. In the present study, we demonstrated for the first time that OA ameliorated 5-Fu-induced human umbilical vein endothelial cells (HUVECs) and human normal intestinal epithelial cells (NCM460) in a 5-Fu-induced cellular senescence model by decreasing the activity of SA-β-gal-positive cells, and the expression of senescence-associated proteins (p16), senescence-associated genes (p53 and p21), and senescence-associated secretory phenotypes (SASPs: IL-1β, IL-6, IL-8, IFN-γ and TNF-α). Meanwhile, in this study, in a BALB/c mouse model, we demonstrated that 5-FU induced intestinal inflammatory response and injury, which was also found to be closely related to the increase of senescent cells, and that OA treatment was effective in ameliorating these adverse phenomena. Furthermore, our in vivo and in vitro studies showed that OA could alleviate senescence by inhibiting mTOR. In colon cancer cell models, OA also enhanced the ability of 5-FU to kill HCT116 cells and SW480 cells. Overall, this study demonstrates for the first time the potential role of OA in counteracting the side effects of 5-FU chemotherapy, providing a new option for the treatment of colorectal cancer to progressively achieve the goal of high efficacy and low toxicity of chemotherapy.
Collapse
Affiliation(s)
- Shi-Rui Bai
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Bing-Xiang Zhao
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Qi Zhao
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Yu-Chen Ge
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Man Li
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
| | - Cheng-Gang Zhao
- Microbiology Research Institute, Guangxi Academy of Agricultural Science, Nanning, 530007, Guangxi Province, China
| | - Xiao-Jian Wu
- Microbiology Research Institute, Guangxi Academy of Agricultural Science, Nanning, 530007, Guangxi Province, China.
| | - Xiao-Bo Wang
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China.
| |
Collapse
|
11
|
Zhang N, Zang L. MiR-22-3p Inhibits 5-Fluorouracil Resistance in Cholangiocarcinoma Cells Through PTEN/PI3K/AKT Axis. Assay Drug Dev Technol 2024; 22:217-228. [PMID: 38967602 DOI: 10.1089/adt.2024.007] [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] [Indexed: 07/06/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a prevalent and highly lethal form of cancer globally. Although microRNAs (miRNAs) have been implicated in the advancement of CCA, their potential influence on 5-fluorouracil (5-Fu) resistance in CCA remains to be fully elucidated. Here, in this study, we investigated the impact of miR-22-3p on CCA resistance. Our investigation involved bioinformatics analysis, which revealed an association between miR-22-3p and the progression, diagnosis, and patient survival of CCA. Furthermore, we validated a notable downregulation of miR-22-3p expression in CCA cell lines. Elevated levels of miR-22-3p inhibit the activity and proliferation of 5-Fu-resistant CCA cell lines. In addition, we confirmed that phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a target gene of miR-22-3p, and its expression correlates with the survival of CCA patients. Reduced PTEN expression enhances apoptosis in 5-Fu-resistant CCA cells. Meanwhile, we verified the existence of the miR-22-3p/PTEN/phosphatidylinositol-3 kinase (PI3K)/Protein kinase B (AKT) regulatory networks in CCA, influencing the sensitivity of CCA cells to 5-Fu. In conclusion, our findings suggest that miR-22-3p acts as a tumor suppressor. Its overexpression inhibits the PTEN/PI3K/AKT axis, promoting cell apoptosis and enhancing CCA sensitivity to 5-Fu.
Collapse
Affiliation(s)
- Ningrong Zhang
- Department of Pharmacy, Ningbo Haishu People's Hospital, Ningbo, Republic of China
| | - Li Zang
- Department of Pharmacy, Ningbo Haishu People's Hospital, Ningbo, Republic of China
| |
Collapse
|
12
|
Qu M, Chen J, Xu B, Shi Q, Zhao S, Wang Z, Li Z, Ma B, Xu H, Ye Q, Xie J. Assessing genotoxic effects of chemotherapy agents by a robust in vitro assay based on mass spectrometric quantification of γ-H2AX in HepG2 cells. Front Pharmacol 2024; 15:1356753. [PMID: 38962306 PMCID: PMC11219945 DOI: 10.3389/fphar.2024.1356753] [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: 12/16/2023] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Chemotherapy has already proven widely effective in treating cancer. Chemotherapeutic agents usually include DNA damaging agents and non-DNA damaging agents. Assessing genotoxic effect is significant during chemotherapy drug development, since the ability to attack DNA is the major concern for DNA damaging agents which relates to the therapeutic effect, meanwhile genotoxicity should also be evaluated for chemotherapy agents' safety especially for non-DNA damaging agents. However, currently applicability of in vitro genotoxicity assays is hampered by the fact that genotoxicity results have comparatively high false positive rates. γ-H2AX has been shown to be a bifunctional biomarker reflecting both DNA damage response and repair. Previously, we developed an in vitro genotoxicity assay based on γ-H2AX quantification using mass spectrometry. Here, we employed the assay to quantitatively assess the genotoxic effects of 34 classic chemotherapy agents in HepG2 cells. Results demonstrated that the evaluation of cellular γ-H2AX could be an effective approach to screen and distinguish types of action of different classes of chemotherapy agents. In addition, two crucial indexes of DNA repair kinetic curve, i.e., k (speed of γ-H2AX descending) and t50 (time required for γ-H2AX to drop to half of the maximum value) estimated by our developed online tools were employed to further evaluate nine representative chemotherapy agents, which showed a close association with therapeutic index or carcinogenic level. The present study demonstrated that mass spectrometric quantification of γ-H2AX may be an appropriate tool to preliminarily evaluate genotoxic effects of chemotherapy agents.
Collapse
Affiliation(s)
- Minmin Qu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jia Chen
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Bin Xu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Qinyun Shi
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Shujing Zhao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhaoxia Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhi Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Bo Ma
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Hua Xu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, China
| | - Jianwei Xie
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
13
|
TAHIYA EC, ISLAM AA, HATTA M, LUSIKOOY RE, PRIHANTONO P, RUDIMAN R, WIDIANA IK, PATELONGI I, BUKHARI AS. 5-Fluorouracil for colorectal cancer: mechanism of action and metabolism. GAZZETTA MEDICA ITALIANA ARCHIVIO PER LE SCIENZE MEDICHE 2024; 183. [DOI: 10.23736/s0393-3660.23.05249-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
14
|
Sarkar S, Kiren S, Gmeiner WH. Review of Prodrug and Nanodelivery Strategies to Improve the Treatment of Colorectal Cancer with Fluoropyrimidine Drugs. Pharmaceutics 2024; 16:734. [PMID: 38931855 PMCID: PMC11206923 DOI: 10.3390/pharmaceutics16060734] [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: 04/24/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Fluoropyrimidine (FP) drugs are central components of combination chemotherapy regimens for the treatment of colorectal cancer (CRC). FP-based chemotherapy has improved survival outcomes over the last several decades with much of the therapeutic benefit derived from the optimization of dose and delivery. To provide further advances in therapeutic efficacy, next-generation prodrugs and nanodelivery systems for FPs are being developed. This review focuses on recent innovative nanodelivery approaches for FP drugs that display therapeutic promise. We summarize established, clinically useful FP prodrug strategies, including capecitabine, which exploit tumor-specific enzyme expression for optimal anticancer activity. We then describe the use of FP DNA-based polymers (e.g., CF10) for the delivery of activated FP nucleotides as a nanodelivery approach with proven activity in pre-clinical models and with clinical potential. Multiple nanodelivery systems for FP delivery show promise in CRC pre-clinical models and we review advances in albumin-mediated FP delivery, the development of mesoporous silica nanoparticles, emulsion-based nanoparticles, metal nanoparticles, hydrogel-based delivery, and liposomes and lipid nanoparticles that display particular promise for therapeutic development. Nanodelivery of FPs is anticipated to impact CRC treatment in the coming years and to improve survival for cancer patients.
Collapse
Affiliation(s)
- Santu Sarkar
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA;
| | - Sezgin Kiren
- Department of Chemistry, Winston-Salem State University, Winston-Salem, NC 27110, USA;
| | - William H. Gmeiner
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA;
| |
Collapse
|
15
|
Azevedo A, Coelho MP, Pinho JO, Soares PIP, Reis CP, Borges JP, Gaspar MM. An alternative hybrid lipid nanosystem combining cytotoxic and magnetic properties as a tool to potentiate antitumor effect of 5-fluorouracil. Life Sci 2024; 344:122558. [PMID: 38471621 DOI: 10.1016/j.lfs.2024.122558] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
AIMS Colorectal cancer is the third most frequent type of cancer and the second leading cause of cancer-related deaths worldwide. The majority of cases are diagnosed at a later stage, leading to the need for more aggressive treatments such as chemotherapy. 5-Fluorouracil (5-FU), known for its high cytotoxic properties has emerged as a chemotherapeutic agent. However, it presents several drawbacks such as lack of specificity and short half-life. To reduce these drawbacks, several strategies have been designed namely chemical modification or association to drug delivery systems. MATERIALS AND METHODS Current research was focused on the design, physicochemical characterization and in vitro evaluation of a lipid-based system loaded with 5-FU. Furthermore, aiming to maximize preferential targeting and release at tumour sites, a hybrid lipid-based system, combining both therapeutic and magnetic properties was developed and validated. For this purpose, liposomes co-loaded with 5-FU and iron oxide (II, III) nanoparticles were accomplished. KEY FINDINGS The characterization of the developed nanoformulation was performed in terms of incorporation parameters, mean size and surface charge. In vitro studies assessed in a murine colon cancer cell line confirmed that 5-FU antiproliferative activity was preserved after incorporation in liposomes. In same model, iron oxide (II, III) nanoparticles did not exhibit cytotoxic properties. Additionally, the presence of these nanoparticles was shown to confer magnetic properties to the liposomes, allowing them to respond to external magnetic fields. SIGNIFICANCE Overall, a lipid nanosystem loading a chemotherapeutic agent displaying magnetic characteristics was successfully designed and physicochemically characterized, for further in vivo applications.
Collapse
Affiliation(s)
- Afonso Azevedo
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Mariana P Coelho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Jacinta O Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Paula I P Soares
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Catarina P Reis
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal; IBEB, Institute of Biophysics and Biomedical Engineering, Faculty of Sciences, Universidade de Lisboa, Lisboa, Portugal
| | - João P Borges
- i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University Lisbon, Campus de Caparica, 2829-516 Caparica, Portugal
| | - M Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal; IBEB, Institute of Biophysics and Biomedical Engineering, Faculty of Sciences, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
16
|
da Cunha de Medeiros P, Nunes EA, Barcelos GRM, Perobelli JE. Genotoxicity and cytotoxicity of antineoplastic drugs at environmentally relevant concentrations after long-term exposure. Toxicol Res (Camb) 2024; 13:tfae049. [PMID: 38533178 PMCID: PMC10962016 DOI: 10.1093/toxres/tfae049] [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: 12/12/2023] [Revised: 02/20/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Introduction 5-fluorouracil (5-FU) and methotrexate (MTX) are the antineoplastic drugs most commonly used worldwide. Considered cytotoxic, these pharmaceuticals exhibit low specificity, causing damage not only to cancer cells but also to healthy cells in organisms. After being consumed and metabolized, these drugs are excreted through urine and feces, followed by wastewater treatment. However, conventional treatments do not have the capacity to completely remove these substances, risking their introduction into freshwater systems. This could pose a risk to human health even at low concentrations. Aims Thus, the present study aimed to investigate the genotoxicity, cytotoxicity, and mutagenicity of 5-FU and MTX at environmentally relevant concentrations after a long-term exposure, using adult male rats as an experimental model. Methods Male Wistar rats (70 days old) were distributed into 4 groups (n = 10/group): control, received only vehicle; MTX, received methotrexate at 10ngL-1; 5-FU received 5-fluorouracil at 10ngL-1; and MTX + 5-FU, received a combination of MTX and 5-FU at 10ngL-1 each. The period of exposure was from postnatal day (PND) 70 to PND 160, through drinking water. After that, the animals were euthanized and the samples (liver, testis, femoral bone marrow, and peripheral blood) were obtained. Results Increased DNA fragmentation was observed in the peripheral blood, liver, and testis, altering the parameters of the tail moment and tail intensity in the Comet assay. Besides, the change in the ratio between PCE and NCE indicates bone marrow suppression. Conclusion These findings warn the adverse effects for the general population worldwide chronically exposed to these drugs at trace concentration unintentionally.
Collapse
Affiliation(s)
- P da Cunha de Medeiros
- Laboratory of Experimental Toxicology – LATOEX, Universidade Federal de São Paulo, Instituto do Mar, Carvalho de Mendonça, 144, Santos 11070-100, SP, Brazil
| | - E A Nunes
- Department of Biosciences, Laboratory of Gene-Environmental Interactions in Toxicology – GENINTOX, Universidade Federal de São Paulo, XV de novembro 195, sala 614, Santos 11.010-151, SP, Brazil
| | - G R M Barcelos
- Department of Biosciences, Laboratory of Gene-Environmental Interactions in Toxicology – GENINTOX, Universidade Federal de São Paulo, XV de novembro 195, sala 614, Santos 11.010-151, SP, Brazil
| | - J E Perobelli
- Laboratory of Experimental Toxicology – LATOEX, Universidade Federal de São Paulo, Instituto do Mar, Carvalho de Mendonça, 144, Santos 11070-100, SP, Brazil
| |
Collapse
|
17
|
Chen H, Wang Z, Gong L, Chen J, Huang Y, Guo W, Zhang Q, Li Y, Bao G, Li D, Chen Y. Attenuation effect of a polysaccharide from large leaf yellow tea by activating autophagy. Int J Biol Macromol 2024; 265:130697. [PMID: 38490395 DOI: 10.1016/j.ijbiomac.2024.130697] [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: 09/06/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024]
Abstract
Chemotherapy, the most common class of anticancer drugs, is considerably limited owing to its adverse side effects. In this study, we aimed to evaluate the protective effect and mechanism of action of large-leaf yellow tea polysaccharides (ULYTP-1, 1.29 × 104 Da) against chemotherapeutic 5-fluorouracil (5-Fu). Structural characterisation revealed that ULYTP-1 was a β-galactopyranouronic acid. Furthermore, ULYTP-1 promoted autolysosome formation, activating autophagy and reducing the oxidative stress and inflammation caused by 5-Fu. Our in vivo study of 4 T1 tumour-bearing mice revealed that ULYTP-1 also attenuated 5-Fu toxicity through modulation of the gut microbiota. Moreover, ULYTP-1 effectively protected immune organs and the liver from 5-Fu toxicity, while promoting its tumour-inhibitory properties. The current findings provide a new strategy for optimising chemotherapy regimens in the clinic.
Collapse
Affiliation(s)
- Hao Chen
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Zhuang Wang
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Lei Gong
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Jielin Chen
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Yuzhe Huang
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Wenqiang Guo
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Qiang Zhang
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Yong Li
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Guanhu Bao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.
| | - Daxiang Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.
| | - Yan Chen
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China.
| |
Collapse
|
18
|
Panjeta A, Kaur K, Sharma R, Verma I, Preet S. Human Intestinal Defensin 5 Ameliorates the Sensitization of Colonic Cancer Cells to 5-Fluorouracil. Arch Med Res 2024; 55:102966. [PMID: 38330831 DOI: 10.1016/j.arcmed.2024.102966] [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: 09/11/2023] [Revised: 12/12/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND AND AIM The increasing dilemma of multidrug-resistant cancer cells in response to currently available chemotherapeutic drugs and their associated side effect(s), calls for the investigation of alternative anticancer advances and molecules. Therefore, the present study aimed to elucidate the combinatorial potential against colon cancer of human defensin 5 in combination with 5-fluorouracil (5-FU), and against 5-FU resistant colon tumor cells. METHODS The in vivo combinatorial potential of HD-5 with 5-FU was elucidated in terms of tumor morphometrics, apoptosis assay, surface morphology histology of the colon(s), and transcriptional alterations. Changes in membrane dynamics with mucin expression were evaluated by fluorescence microscopy and histochemistry. The in vitro activity of the peptide/drug conjunction was explored by phase contrast microscopy, MTT, LDH assay, and AO/EtBr staining. Chemoresistance to 5-FU was determined by phase contrast microscopy, MTT assay, annexin V-FITC/PI flow cytometry, and MDR-1, Bak, and Bax expression. RESULTS In vivo decreases in tumor parameters, with a marked increase in apoptosis and neutrophil infiltrations indicated restoration of normal architecture with improved mucin content in the treated colons. This happened with substantial changes in key molecular markers of the intrinsic apoptotic cascade. Membrane dynamics revealed that peptides and chemotherapeutic drugs could bind to cancerous cells by taking advantage of altered levels of membrane fluidity. CONCLUSION Peptide treatment of drug-resistant Caco-2 cells promotes enhanced 5-FU uptake, in contrast to when cells were treated with 5-FU alone. Hence, HD-5 as an adjunct to 5-FU, exhibited strong cancer cell killing even against 5-FU-resistant tumorigenic cells.
Collapse
Affiliation(s)
- Anshul Panjeta
- Department of Biophysics, BMS Block II, South Campus, Panjab University, Chandigarh, India
| | - Khushpreet Kaur
- Department of Biochemistry, Research Block-A, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rinkle Sharma
- Department of Biochemistry, Research Block-A, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Indu Verma
- Department of Biochemistry, Research Block-A, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Simran Preet
- Department of Biophysics, BMS Block II, South Campus, Panjab University, Chandigarh, India.
| |
Collapse
|
19
|
Peng F, Hong W, Wang Y, Peng Y, Fang Z. Mechanism of herb pair containing Astragali Radix and Spatholobi Caulis in the treatment of myelosuppression based on network pharmacology and experimental investigation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117178. [PMID: 37741472 DOI: 10.1016/j.jep.2023.117178] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Astragali Radix and Spatholobi Caulis herb pair (ARSC) is one of the most commonly used herbal combinations for bone marrow suppression. According to traditional Chinese medicine, Astragali Radix strengthens the spleen and replenishes qi, while Spatholobi Caulis is a hematinic agent that promotes blood circulation and enrichment. The compatibility of the two helps the body to tonify the spleen and kidneys and compensate for visceral deficiencies. However, the multi-target mechanism of ARSC in bone marrow suppression has remained largely unknown. AIM OF THE STUDY The aim of this study is to explore the key targets and signaling pathways of the traditional Chinese herbal pair ARSC for the treatment of bone marrow suppression. MATERIALS AND METHODS The active components of ARSC and targets for myelosuppression were screened using network databases. Cytoscape 3.8.0 was used to construct compound-target, compound-disease-target and protein-protein interaction (PPI) networks. Go-function and pathway enrichment analyses were performed to explore the potential mechanism. In vivo animal experiments were conducted to verify the molecular mechanisms. RESULTS The 36 active compounds were identified from the ARSC, and a total of 108 genes involved in myelosuppression were screened. VEGFA, IL6, TNF, JUN, STAT3, PTGS2, CASP3 and MMP9 genes were identified as potential drug targets in the PPI network analyzed by CytoHubba. Enrichment analysis indicated that ARSC may treat myelosuppression through various biological processes, such as apoptosis, TNF-α signaling pathway via NF-κB, PI3K/AKT/mTOR signaling pathway, IL6/JAK/STAT3 signaling pathway, P53 signaling pathway and G2/M checkpoint signaling pathway. The results of the experiment showed that the aqueous extract of ARSC significantly alleviated myelosuppression, reduced the apoptosis rate of bone marrow cells, upregulated the mRNA expression levels of TNF-α, IL-6 and VEGF, and promoted NF-κB phosphorylation in myelosuppressed mice. CONCLUSIONS This study identified the active components and relevant mechanisms of ARSC in the treatment of myelosuppression. Our findings predicted that ARSC could treat bone marrow suppression through multiple components, multiple targets and multiple pathways. Pharmacological experiments showed that ARSC alleviated fluorouracil-induced myelosuppression by reducing the apoptosis rate of bone marrow cells and regulating the TNF-α/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Fei Peng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| | - Wanying Hong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| | - Yingyu Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| | - Yunru Peng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| | - Zhijun Fang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
20
|
Preitakaitė V, Barasa P, Aučynaitė A, Plakys G, Koplūnaitė M, Zubavičiūtė S, Meškys R. Bacterial amidohydrolases and modified 5-fluorocytidine compounds: Novel enzyme-prodrug pairs. PLoS One 2023; 18:e0294696. [PMID: 38032917 PMCID: PMC10688628 DOI: 10.1371/journal.pone.0294696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
Gene-directed enzyme prodrug therapy is an emerging strategy for cancer treatment based on the delivery of a gene that encodes an enzyme that is able to convert a prodrug into a potent cytotoxin exclusively in target cancer cells. However, it is limited by the lack of suitable enzyme variants and a scarce choice of chemical bonds that could be activated. Therefore, this study is aimed to determine the capability of bacterial amidohydrolases YqfB and D8_RL to activate novel prodrugs and the effect such system has on the viability of eukaryotic cancer cells. We have established cancer cell lines that stably express the bacterial amidohydrolase genes and selected several N4-acylated cytidine derivatives as potential prodrugs. A significant decrease in the viability of HCT116 human colon cancer cell lines expressing either the YqfB or the D8_RL was observed after exposure to the novel prodrugs. The data we acquired suggests that bacterial YqfB and D8_RL amidohydrolases, together with the modified cytidine-based prodrugs, may serve as a promising enzyme-prodrug system for gene-directed enzyme prodrug therapy.
Collapse
Affiliation(s)
- Viktorija Preitakaitė
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Povilas Barasa
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agota Aučynaitė
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Gediminas Plakys
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Martyna Koplūnaitė
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Simona Zubavičiūtė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Rolandas Meškys
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
21
|
Mironiuk-Puchalska E, Karatsai O, Żuchowska A, Wróblewski W, Borys F, Lehka L, Rędowicz MJ, Koszytkowska-Stawińska M. Development of 5-fluorouracil-dichloroacetate mutual prodrugs as anticancer agents. Bioorg Chem 2023; 140:106784. [PMID: 37639758 DOI: 10.1016/j.bioorg.2023.106784] [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: 06/02/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
5-Fluorouracil (5-FU) is one of the most widely applied chemotherapeutic agents with a broad spectrum of activity. However, despite this versatile activity, its use poses many limitations. Herein, novel derivatives of 5-FU and dichloroacetic acid have been designed and synthesized as a new type of codrugs, also known as mutual prodrugs, to overcome the drawbacks of 5-FU and enhance its therapeutic efficiency. The stability of the obtained compounds has been tested at various pH values using different analytical techniques, namely HPLC and potentiometry. The antiproliferative activity of the new 5-FU derivatives was assessed in vitro on SK-MEL-28 and WM793 human melanoma cell lines in 2D culture as well as on A549 human lung carcinoma, MDA-MB-231 breast adenocarcinoma, LL24 normal lung tissue, and HMF normal breast tissue as a multicellular 3D spheroid model cultured in standard (static) conditions and with the use of microfluidic systems, which to a great extent resembles the in vivo environment. In all cases, new mutual prodrugs showed a higher cytotoxic activity toward cancer models and lower to normal cell models than the parent 5-FU itself.
Collapse
Affiliation(s)
- Ewa Mironiuk-Puchalska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland.
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Agnieszka Żuchowska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Wojciech Wróblewski
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Filip Borys
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Lilya Lehka
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | | |
Collapse
|
22
|
Elbanan ME, Amer ME, El-Missiry MA, Othman AI, Shabana SM. Melatonin protected against kidney impairment induced by 5-fluorouracil in mice. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2023; 339:777-787. [PMID: 37395484 DOI: 10.1002/jez.2728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/04/2023]
Abstract
The utility of 5-fluorouracil (5-FU) as a successful chemotherapeutic drug for several cancers is limited by the induction of kidney injury and dysfunction due to redox imbalance, inflammation, and apoptosis. Meanwhile, melatonin (MLT) is a potent antioxidant and anti-inflammatory natural compound with a wide safety range. The current study aimed to investigate MLT's protective effect against 5-FU-induced kidney impairment. Male mice were given multiple doses of 5-FU at 25 and 100 mg/kg, as well as MLT at 20 mg/kg. MLT treatment alleviated the toxic effect of 5-FU by normalizing blood urea and creatinine levels and preserving the histological structure, indicating MLT's nephroprotective ability. This is accompanied by body weight maintenance, an increase in survival percentage, and preserved hematological parameters in comparison to the 5-FU-treated mice. MLT's renoprotective effect was explained by improvements in C-reactive protein, IL-6, and caspase-3 in kidney tissue, indicating MLT's anti-inflammatory and antiapoptotic ability. Furthermore, MLT inhibited 5-FU-induced lipid peroxidation by maintaining the activity of superoxide dismutase and catalase, as well as glutathione levels in kidney tissue from mice treated with both doses of 5-FU. The current findings show that MLT has a novel protective effect against 5-FU-induced renal injury and renal impairment.
Collapse
Affiliation(s)
- Mona E Elbanan
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Maggie E Amer
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt
| | | | - Azza I Othman
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Sameh M Shabana
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
23
|
da Silva KS, Abboud KY, Schiebel CS, de Oliveira NMT, Bueno LR, de Mello Braga LLV, da Silveira BC, Santos IWFD, Gomes EDS, Gois MB, Cordeiro LMC, Maria Ferreira D. Polysaccharides from Passion Fruit Peels: From an Agroindustrial By-Product to a Viable Option for 5-FU-Induced Intestinal Damage. Pharmaceuticals (Basel) 2023; 16:912. [PMID: 37513823 PMCID: PMC10383750 DOI: 10.3390/ph16070912] [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: 05/17/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Gastrointestinal mucositis is a serious and dose-limiting toxic side effect of oncologic treatment. Interruption of cancer treatment due to gastrointestinal mucositis leads to a significant decrease in cure rates and consequently to the deterioration of a patient's quality of life. Natural polysaccharides show a variety of beneficial effects, including a gastroprotective effect. Treatment with soluble dietary fiber (SDF) from yellow passion fruit (Passiflora edulis) biomass residues protected the gastric and intestinal mucosa in models of gastrointestinal injury. In this study, we investigated the protective therapeutic effect of SDF on 5-FU-induced mucositis in male and female mice. Oral treatment of the animals with SDF did not prevent weight loss but reduced the disease activity index and preserved normal intestinal function by alleviating diarrhea and altered gastrointestinal transit. SDF preserved the length of the colon and histological damage caused by 5-FU. SDF significantly restored the oxidative stress and inflammation in the intestine and the enlargement and swelling of the spleen induced by 5-FU. In conclusion, SDF may be a promising adjuvant strategy for the prevention and treatment of intestinal mucositis induced by 5-FU.
Collapse
Affiliation(s)
- Karien Sauruk da Silva
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Kahlile Youssef Abboud
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba 81531-980, Brazil
| | - Carolina Silva Schiebel
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Laryssa Regis Bueno
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Lara Luisa Valerio de Mello Braga
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Bruna Carla da Silveira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Isabella Wzorek França Dos Santos
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Everton Dos Santos Gomes
- Programa de Pós-Graduação em Imunologia, Universidade Federal da Bahia, Salvador 40231-300, Brazil
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78736-900, Brazil
| | - Marcelo Biondaro Gois
- Programa de Pós-Graduação em Imunologia, Universidade Federal da Bahia, Salvador 40231-300, Brazil
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78736-900, Brazil
| | | | - Daniele Maria Ferreira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| |
Collapse
|
24
|
Salerno L, Sorrenti V, Pittalà V, Consoli V, Modica MN, Romeo G, Marrazzo A, Giuliano M, Zajdel P, Vanella L, Intagliata S. Discovery of SI 1/20 and SI 1/22 as Mutual Prodrugs of 5-Fluorouracil and Imidazole-Based Heme Oxygenase 1 Inhibitor with Improved Cytotoxicity in DU145 Prostate Cancer Cells. ChemMedChem 2023; 18:e202300047. [PMID: 36756924 DOI: 10.1002/cmdc.202300047] [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: 01/28/2023] [Accepted: 02/08/2023] [Indexed: 02/10/2023]
Abstract
In this work, we extend the concept of 5-fluorouracil/heme oxygenase 1 (5-FU/HO-1) inhibitor hybrid as an effective strategy for enhancing 5-FU-based anticancer therapies. For this purpose, we designed and synthesized new mutual prodrugs, named SI 1/20 and SI 1/22, in which the two active parent drugs (i. e., 5-FU and an imidazole-based HO-1 inhibitor) were connected through an easily cleavable succinic linker. Experimental hydrolysis rate, and in silico ADMET predictions were indicative of good drug-likeness and pharmacokinetic properties. Novel hybrids significantly reduced the viability of prostate DU145 cancer cells compared to the parent compounds 5-FU and HO-1 inhibitor administered alone or in combination. Interestingly, both compounds showed statistically significant lower toxicity, than 5-FU at the same dose, against non-tumorigenic human benign prostatic hyperplasia (BPH-1) cell line. Moreover, the newly synthesized mutual prodrugs inhibited the HO-1 activity both in a cell-free model and in vitro, as well as downregulated the HO-1 expression and increased the reactive oxygen species (ROS) levels.
Collapse
Affiliation(s)
- Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al Jawhara Centre for Molecular Medicine, Arabian Gulf University, Manama, 329, Bahrain
| | - Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Maria N Modica
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Michela Giuliano
- Department of Biological, Chemical and Pharmaceutical Sciences, and Technologies (STEBICEF), University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Paweł Zajdel
- Department of Organic Chemistry, Jagiellonian University Medical College, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
25
|
Shibata T, Narita T, Suto Y, Yasmin H, Kabashima T. A Facile Fluorometric Assay of Orotate Phosphoribosyltransferase Activity Using a Selective Fluorogenic Reaction for Orotic Acid. SENSORS (BASEL, SWITZERLAND) 2023; 23:2507. [PMID: 36904710 PMCID: PMC10007123 DOI: 10.3390/s23052507] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 06/18/2023]
Abstract
Orotate phosphoribosyltransferase (OPRT) exists as a bifunctional enzyme, uridine 5'-monophosphate synthase, in mammalian cells and plays an important role in pyrimidine biosynthesis. Measuring OPRT activity has been considered important for understanding biological events and development of molecular-targeting drugs. In this study, we demonstrate a novel fluorescence method for measuring OPRT activity in living cells. The technique utilizes 4-trifluoromethylbenzamidoxime (4-TFMBAO) as a fluorogenic reagent, which produces selective fluorescence for orotic acid. To perform the OPRT reaction, orotic acid was added to HeLa cell lysate, and a portion of the enzyme reaction mixture was heated at 80 °C for 4 min in the presence of 4-TFMBAO under basic conditions. The resulting fluorescence was measured using a spectrofluorometer, which reflects the consumption of orotic acid by the OPRT. After optimization of the reaction conditions, the OPRT activity was successfully determined in 15 min of enzyme reaction time without further procedures such as purification of OPRT or deproteination for the analysis. The activity obtained was compatible with the value measured by the radiometric method with [3H]-5-FU as the substrate. The present method provides a reliable and facile measurement of OPRT activity and could be useful for a variety of research fields targeting pyrimidine metabolism.
Collapse
Affiliation(s)
- Takayuki Shibata
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi 371-8514, Japan
| | - Tomohiro Narita
- Department of Pharmaceutical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Yutaka Suto
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 37-1 Nakaoruimachi, Takasaki 370-0033, Japan
| | - Hasina Yasmin
- Department of Pharmacy, BRAC University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | - Tsutomu Kabashima
- Graduate School of Pharmaceutical Sciences, Nagasaki International University, 2825-7 Huis Ten Bosch, Sasebo 859-3298, Japan
| |
Collapse
|
26
|
Singh M, Pal P, Dutta RS, Marbaniang D, Ray S, Mazumder B. Nanodiamond Mediated Molecular Targeting in Pancreatic Ductal Adenocarcinoma: Disrupting the Tumor-stromal Cross-talk, Next Hope on the Horizon? Curr Cancer Drug Targets 2023; 23:620-633. [PMID: 36843367 DOI: 10.2174/1568009623666230227120837] [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: 03/11/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 02/28/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the foremost causes of cancer-related morbidities worldwide. Novel nanotechnology-backed drug delivery stratagems, including molecular targeting of the chemotherapeutic payload, have been considered. However, no quantum leap in the gross survival rate of patients with PDAC has been realized. One of the predominant causes behind this is tumor desmoplasia, a dense and heterogenous stromal extracellular matrix of the tumor, aptly termed tumor microenvironment (TME). It plays a pivotal role in the tumor pathogenesis of PDAC as it occupies most of the tumor mass, making PDAC one of the most stromal-rich cancers. The complex crosstalk between the tumor and dynamic components of the TME impacts tumor progression and poses a potential barrier to drug delivery. Understanding and deciphering the complex cascade of tumorstromal interactions are the need of the hour so that we can develop neoteric nano-carriers to disrupt the stroma and target the tumor. Nanodiamonds (NDs), due to their unique surface characteristics, have emerged as a promising nano delivery system in various pre-clinical cancer models and have the potential to deliver the chemotherapeutic payload by moving beyond the dynamic tumor-stromal barrier. It can be the next revolution in nanoparticle-mediated pancreatic cancer targeting.
Collapse
Affiliation(s)
- Mohini Singh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Paulami Pal
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Rajat Subhra Dutta
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Daphisha Marbaniang
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Subhabrata Ray
- Dr. B.C. Roy College of Pharmacy & AHS, Durgapur, WB, India
| | - Bhaskar Mazumder
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| |
Collapse
|
27
|
Long Intergenic Non-Protein Coding RNA 173 in Human Cancers. Cancers (Basel) 2022; 14:cancers14235923. [PMID: 36497407 PMCID: PMC9737410 DOI: 10.3390/cancers14235923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Long non-coding RNAs belong to non-coding RNAs (ncRNAs) with a length of more than 200 nucleotides and limited protein-coding ability. Growing research has clarified that dysregulated lncRNAs are correlated with the development of various complex diseases, including cancer. LINC00173 has drawn researchers' attention as one of the recently discovered lncRNAs. Aberrant expression of LINC00173 affects the initiation and progression of human cancers. In the present review, we summarize the recent considerable research on LINC00173 in 11 human cancers. Through the summary of the abnormal expression of LINC00173 and its potential molecular regulation mechanism in cancers, this article indicates that LINC00173 may serve as a potential diagnostic biomarker and a target for drug therapy, thus providing novel clues for future related research.
Collapse
|
28
|
Wang S, Li T, Wang Y, Wang M, Liu Y, Zhang X, Zhang L. 5-Fluorouracil and actinomycin D lead to erythema multiforme drug eruption in chemotherapy of invasive mole: Case report and literature review. Medicine (Baltimore) 2022; 101:e31678. [PMID: 36451432 PMCID: PMC9704884 DOI: 10.1097/md.0000000000031678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
RATIONALE 5-Fluorouracil (5-FU) and actinomycin D (ActD) are often used in chemotherapy for various cancers. Side effects are more common in bone marrow suppression, liver function impairment, and gastrointestinal responses. Skin effects are rare and easy to be ignored by doctors and patients, which can lead to life-threatening consequence. PATIENT CONCERNS We reported a 45-year-old woman patient developed skin erythema and fingernail belt in chemotherapy of 5-FU and ActD. DIAGNOSIS Erythema multiforme drug eruption. INTERVENTIONS Laboratory tests including blood and urine routine, liver and kidney function, electrolytes and coagulation function and close observation. OUTCOMES The rash was gone and the nail change returned. LESSONS Delays in diagnosis or treatment may lead to serious consequence. We should pay attention to the dosage of 5-FU and ActD, monitor adverse reactions strictly, to reduce occurrence of skin malignant events.
Collapse
Affiliation(s)
- Shan Wang
- Departments of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Tengfei Li
- Departments of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yuan Wang
- Departments of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mengdi Wang
- Departments of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yibin Liu
- Departments of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xiaoguang Zhang
- Departments of Dermatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Lijuan Zhang
- Departments of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- * Correspondence: Lijuan Zhang, Departments of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China (e-mail: )
| |
Collapse
|
29
|
Guo K, Ma X, Li J, Zhang C, Wu L. Recent advances in combretastatin A-4 codrugs for cancer therapy. Eur J Med Chem 2022; 241:114660. [PMID: 35964428 DOI: 10.1016/j.ejmech.2022.114660] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
CA4 is a potent microtubule polymerization inhibitor and vascular disrupting agent. However, the in vivo efficiency of CA4 is limited owing to its poor pharmacokinetics resulting from its high lipophilicity and low water solubility. To improve the water solubility, CA4 phosphate (CA4P) has been developed and shows potent antivascular and antitumor effects. CA4P had been evaluated as a vascular disrupting agent in previousc linical trials. However, it had been discontinued due to the lack of a meaningful improvement in progression-free survival and unfavorable partial response data. Codrug is a drug design approach to chemically bind two or more drugs to improve therapeutic efficiency or decrease adverse effects. This review describes the progress made over the last twenty years in developing CA4-based codrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues. It also discusses the existing problems and the developmental prospects of CA4 codrugs.
Collapse
Affiliation(s)
- Kerong Guo
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xin Ma
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jian Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chong Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liqiang Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
30
|
In Vitro Antioxidant and Pancreatic Anticancer Activity of Novel 5-Fluorouracil-Coumarin Conjugates. Pharmaceutics 2022; 14:pharmaceutics14102152. [DOI: 10.3390/pharmaceutics14102152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
Molecular hybridization consists of the combination of two or more non-identical pharmacophores in a single molecule. It has emerged as a promising strategy that allows the design of molecular frameworks with enhanced activity and affinity compared to their parent drugs. In this work, two novel hybrids that combine the well-known anticancer chemotherapeutic agent 5-fluorouracil with antioxidant coumarin derivatives have been synthesized and characterized by means of a copper-catalyzed azide-alkyne cycloaddition (CuAAC). The conjugates showed good antioxidant properties and a high tendency to aggregate and form stable nanoparticles in aqueous media, with regular shape and uniform size. These materials have proven to be preferential cytotoxic agents in vitro against human pancreatic cancer cells PANC-1, with an activity superior to free 5-fluorouracil. These results open up the possibility of exploiting the synergistic combination between 5-fluorouracil and coumarin derivatives and warrant further investigation of these hybrids as promising pancreatic anticancer agents.
Collapse
|
31
|
López S, Rodríguez-López J, García MT, Rodríguez JF, Pérez-Ortiz JM, Ramos MJ, Gracia I. Self-assembled coumarin- and 5-fluorouracil-PEG micelles as multifunctional drug delivery systems. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
32
|
The Promise of Nanotechnology in Personalized Medicine. J Pers Med 2022; 12:jpm12050673. [PMID: 35629095 PMCID: PMC9142986 DOI: 10.3390/jpm12050673] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Both personalized medicine and nanomedicine are new to medical practice. Nanomedicine is an application of the advances of nanotechnology in medicine and is being integrated into diagnostic and therapeutic tools to manage an array of medical conditions. On the other hand, personalized medicine, which is also referred to as precision medicine, is a novel concept that aims to individualize/customize therapeutic management based on the personal attributes of the patient to overcome blanket treatment that is only efficient in a subset of patients, leaving others with either ineffective treatment or treatment that results in significant toxicity. Novel nanomedicines have been employed in the treatment of several diseases, which can be adapted to each patient-specific case according to their genetic profiles. In this review, we discuss both areas and the intersection between the two emerging scientific domains. The review focuses on the current situation in personalized medicine, the advantages that can be offered by nanomedicine to personalized medicine, and the application of nanoconstructs in the diagnosis of genetic variability that can identify the right drug for the right patient. Finally, we touch upon the challenges in both fields towards the translation of nano-personalized medicine.
Collapse
|
33
|
Floresta G, Fallica AN, Patamia V, Sorrenti V, Greish K, Rescifina A, Pittalà V. From Far West to East: Joining the Molecular Architecture of Imidazole-like Ligands in HO-1 Complexes. Pharmaceuticals (Basel) 2021; 14:ph14121289. [PMID: 34959690 PMCID: PMC8704944 DOI: 10.3390/ph14121289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
HO-1 overexpression has been reported in several cases/types of human malignancies. Unfortunately, poor clinical outcomes are reported in most of these cases, and the inhibition of HO-1 is considered a valuable and proven anticancer approach. To identify novel hit compounds suitable as HO-1 inhibitors, we report here a fragment-based approach where ligand joining experiments were used. The two most important parts of the classical structure of the HO-1 inhibitors were used as a starting point, and 1000 novel compounds were generated and then virtually evaluated by structure and ligand-based approaches. The joining experiments led us to a novel series of indole-based compounds. A synthetic pathway for eight selected molecules was designed, and the compounds were synthesized. The biological activity revealed that some molecules reach the micromolar activity, whereas molecule 4d inhibits the HO-1 with an IC50 of 1.03 μM. This study suggested that our joining approach was successful, and a novel hit compound was generated. These results are ongoing for further development.
Collapse
Affiliation(s)
- Giuseppe Floresta
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
- Department of Analytics, Environmental & Forensics, King’s College London, London SE1 9NH, UK
| | - Antonino Nicolò Fallica
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
| | - Vincenzo Patamia
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
| | - Khaled Greish
- Department of Molecular Medicine and Nanomedicine Unit, Princess Al-Jawhara Center for Molecular Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain;
| | - Antonio Rescifina
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
- Correspondence: (A.R.); (V.P.)
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
- Correspondence: (A.R.); (V.P.)
| |
Collapse
|