1
|
O'Sullivan Coyne G, Kummar S, Rubinstein LV, Wilsker D, Moore N, Hogu M, Piekarz R, Covey J, Beumer JH, Ferry-Galow KV, Villaruz LC, Hollingshead MG, Holleran JL, Deppas JJ, Pommier Y, Ko B, Johnson BC, Parchhment RE, Ivy P, Doroshow JH, Chen AP. Phase 1 studies of the indenoisoquinolines LMP776 and LMP744 in patients with solid tumors and lymphomas. Cancer Chemother Pharmacol 2025; 95:58. [PMID: 40439882 PMCID: PMC12122562 DOI: 10.1007/s00280-025-04778-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 05/03/2025] [Indexed: 06/02/2025]
Abstract
PURPOSE Indenoisoquinolines are a class of topoisomerase I (TOP1) inhibitors designed to overcome clinical limitations of camptothecins. Three indenoisoquinolines (LMP400, LMP776, and LMP744) demonstrated activity in murine models and a comparative canine lymphoma study. Clinical data for LMP400 were previously reported (NCT01051635). The maximum tolerated dose (MTD), safety, and clinical data from phase 1 studies of LMP776 (NCT01051635) and LMP744 (NCT03030417) are reported herein. METHODS Patients ≥ 18 years of age with advanced, refractory solid tumors or lymphomas received either LMP776 (n = 34) or LMP744 (n = 35) intravenously following a Simon accelerated titration design. Both LMP776 and LMP744 were administered daily for 5 days (QDx5) in 28-day cycles. Adverse events and clinical responses were evaluated according to CTCAE and RECIST v1.1 criteria, respectively. Pharmacokinetic and pharmacodynamic changes were evaluated. RESULTS The MTD of LMP776 was 12 mg/m2/day and that of LMP744 was 190 mg/m2/day. Dose-limiting toxicities (DLTs) for LMP776 included hypercalcemia, anemia, and hyponatremia; DLTs for LMP744 included hypokalemia, anemia, and weight loss. There was 1 confirmed partial response (cPR) among 35 patients receiving LMP744 (overall response rate 3%) and no objective responses in patients receiving LMP776. Tumor biopsies from the patient with cPR demonstrated high baseline expression of SLFN11 and a unique pattern of pharmacodynamic responses, including increased RAD51, phosphorylated KAP1 (pKAP1), γH2AX, and cleaved caspase-3 (cCasp3). CONCLUSION MTDs and safety profiles are reported for LMP776 and LMP744. Target engagement by an indenoisoquinoline was measured for the first time in human samples.
Collapse
Affiliation(s)
- Geraldine O'Sullivan Coyne
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | | | - Deborah Wilsker
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nancy Moore
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA
| | - Murielle Hogu
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA
| | - Richard Piekarz
- Cancer Therapeutics Evaluation Program, NCI, NIH, Bethesda, MD, USA
| | - Joe Covey
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA
| | - Jan H Beumer
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katherine V Ferry-Galow
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | | | | | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Brian Ko
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA
| | - Barry C Johnson
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA
| | - Ralph E Parchhment
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Percy Ivy
- Cancer Therapeutics Evaluation Program, NCI, NIH, Bethesda, MD, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA
- Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Building 31, Room 3A44, Bethesda, MD, 20892, USA.
| |
Collapse
|
2
|
Sarsar O, Macar O, Kalefetoğlu Macar T, Çavuşoğlu K, Yalçın E, Acar A. Multifaceted investigation of esfenvalerate-induced toxicity on Allium cepa L. Sci Rep 2025; 15:16977. [PMID: 40374842 PMCID: PMC12081867 DOI: 10.1038/s41598-025-01638-3] [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: 01/09/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
The objective of this study was to assess the toxicity of the insecticide esfenvalerate in Allium cepa, employing a multifaceted methodology. For this purpose, A. cepa bulbs were organized into four groups, one of which served as the control. The control group was exposed to tap water, while the remaining three groups were exposed to esfenvalerate at concentrations of 0.33 mg/L, 0.64 mg/L and 0.98 mg/L, respectively. The application of the highest dose of 0.98 mg/L esfenvalerate resulted in a significant decrease in physiological parameters, including a 51% reduction in rooting percentage, an 85.3% decrease in root elongation, and a 54.3% decrease in weight gain (p < 0.05). In the esfenvalerate-treated group (0.98 mg/L), a 45.7% decrease in mitotic index was observed, while a significant increase in chromosomal aberrations and micronucleus formation was observed compared to the control group (p < 0.05). The most frequently observed chromosomal abnormalities due to esfenvalerate were sticky chromosome, vagrant chromosome, fragment, unequal distribution of chromatin, bridge, vacuolated nucleus, reverse polarization and multipolar anaphase. Insecticide application could significantly increase the percentage of DNA tails up to 48.3%, as determined by the Comet test (p < 0.05). Exposure to 0.98 mg/L esfenvalerate increased malondialdehyde level (2.75-fold), proline level (1.96-fold), superoxide dismutase activity (1.35-fold), and catalase activity (1.69-fold) while reducing chlorophyll a level (58.18%) and chlorophyll b level (70.35%) (p < 0.05). Molecular docking analysis revealed that esfenvalerate can interact with tubulins, DNA topoisomerases, glutamate-1-semialdehyde aminotransferase, protochlorophyllide reductase and DNA molecules. Epidermis and cortex cell damages, cortex cell wall thickening, material accumulation in cortex cells and flattened cell nucleus were recorded as meristematic cell damages due to esfenvalerate. The toxicological profile of esfenvalerate on A. cepa exhibited dose dependence. While esfenvalerate-induced oxidative stress is the most probable cause of toxicity, direct interaction with DNA and other molecules that play a crucial role in maintaining cell integrity may also be among the mechanisms of toxicity. The study's findings emphasize that esfenvalerate poses a risk to non-target organisms, underscoring the need for a reassessment of its regulations and further research into its toxicity.
Collapse
Affiliation(s)
- Onur Sarsar
- Department of Biology Giresun, Faculty of Science and Art, Giresun University, Giresun, Turkey
| | - Oksal Macar
- Department of Biology Giresun, Faculty of Science and Art, Giresun University, Giresun, Turkey.
| | - Tuğçe Kalefetoğlu Macar
- Department of Food Technology, Şebinkarahisar School of Applied Sciences, Giresun University, Giresun, Turkey
| | - Kültiğin Çavuşoğlu
- Department of Biology Giresun, Faculty of Science and Art, Giresun University, Giresun, Turkey
| | - Emine Yalçın
- Department of Biology Giresun, Faculty of Science and Art, Giresun University, Giresun, Turkey
| | - Ali Acar
- Department of Medical Services and Techniques, Vocational School of Health Services, Giresun University, Giresun, Turkey
| |
Collapse
|
3
|
Li YL, Mao J, Cheng Z, Zhou XY, Zhang DN, Li YZ, Cao ZX, Ren JX. Identification of low-toxicity DNA topoisomerase I inhibitors with potential blood-brain barrier penetrability for glioblastoma therapy: structure-based virtual screening reveals promising novel Scaffolds. Mol Divers 2025:10.1007/s11030-025-11185-8. [PMID: 40237873 DOI: 10.1007/s11030-025-11185-8] [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: 02/05/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025]
Abstract
Due to the blood-brain barrier (BBB), DNA topoisomerase I (Topo I) inhibitors often cause dose-limiting toxicity in glioblastoma (GBM) treatment. Therefore, developing low-toxicity Topo I inhibitors with enhanced BBB permeability holds a significant promise for improving GBM treatment outcomes. In this study, structure-based virtual screening methods combined with biological evaluations successfully identified three potent Topo I inhibitors, which exhibited IC50 values of approximately 25 µM against A172 cells. Structural similarity analysis showed that these compounds have novel scaffolds. Compounds F1260-0895 and F2557-0012 exhibited negligible cytotoxicity on HK-2 cells. The most active compound, F2557-0012, directly targets human Topo I. Clonal formation assays and growth inhibition curves demonstrated the sustained inhibitory effects of F2557-0012 on A172 cells. The flow cytometric analysis showed that F2557-0012 effectively inhibits cell proliferation with minimal effect on apoptosis. Molecular dynamic simulations demonstrated that compound F2557-0012 exhibits stable binding to the Topo I-DNA complex. Two new easily synthesized compounds, demonstrating improved BBB permeability and reduced hematotoxicity, were derived from F1260-0895 and F2557-0012 through structural optimization utilizing the OptADMET platform. Molecular docking analyses indicated that the two novel compounds exhibited a significantly stronger interaction with the Topo I-DNA complex. Further investigations are warranted to synthesize optimized compounds and evaluate their anti-GBM activity both in vitro and in vivo.
Collapse
Affiliation(s)
- Ya-Lin Li
- State Key Laboratory of Macromolecular Drugs and Large-Scale Preparation, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, People's Republic of China
| | - Jun Mao
- College of Chemistry, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Zhong Cheng
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030607, People's Republic of China
| | - Xin-Yu Zhou
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Duan-Na Zhang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Yu-Zhi Li
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Zhi-Xing Cao
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China.
| | - Ji-Xia Ren
- State Key Laboratory of Macromolecular Drugs and Large-Scale Preparation, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, People's Republic of China.
| |
Collapse
|
4
|
Lv WQ, Gao J, Guo X. Molecular mechanism and therapeutic strategies for embryonal tumors with multilayered rosettes in children (Review). Mol Clin Oncol 2025; 22:30. [PMID: 39926370 PMCID: PMC11803348 DOI: 10.3892/mco.2025.2825] [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: 09/14/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025] Open
Abstract
Embryonal tumors with multilayered rosettes (ETMR) are relatively rare but highly aggressive intracranial tumors that mainly occur in children under four years of age. Despite high-intensity and multi-modal treatment, the five-year overall survival rate of patients with ETMR remains <30%. Therefore, it is necessary to improve understanding of the molecular biological changes in ETMR. The present review presents an overview of the recent molecular and biological characteristics of ETMR in children, the current recommended treatments, and research into potential targeted strategies based on these findings. ETMR are molecularly characterized by distinct DNA methylation signatures and dysregulated expression of oncogenic miRNAs. Despite increased knowledge of the novel molecular characteristics of ETMR in children, treatment outcomes have only marginally improved. Thus, there is an urgent need to translate these new insights in ETMR biology into more effective treatment.
Collapse
Affiliation(s)
- Wen-Qiong Lv
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ju Gao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xia Guo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
5
|
Topatan ZŞ, Kalefetoğlu Macar T, Macar O, Yalçin E, Çavuşoğlu K, Acar A, Kutluer F. Alleviatory efficacy of achillea millefolium L. in etoxazole-mediated toxicity in allium cepa L. Sci Rep 2024; 14:31674. [PMID: 39738374 PMCID: PMC11686124 DOI: 10.1038/s41598-024-81586-6] [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: 08/07/2024] [Accepted: 11/27/2024] [Indexed: 01/02/2025] Open
Abstract
The application of pesticides may adversely impact a variety of non-target organisms. The use of side-effect-free herbal remedies to protect against the toxicity of harmful pesticides such as etoxazole has gained attention in recent times. The current study aimed to reveal the potential mitigating efficacy of Achillea millefolium L. extract against etoxazole toxicity in Allium cepa L. A. cepa bulbs in the control group were applied with tap water, while bulbs in the treatment groups were applied with etoxazole at dose of 0.5 m/L and two different doses of A. millefolium extract (200 mg/L and 400 mg/L). The impact of the treatments on certain parameters was evaluated. The molecular docking analysis was employed to investigate the potential interactions of etoxazole with DNA species, DNA topoisomerases, tubulin proteins, glutamate-1-semialdehyde aminotransferase, and protochlorophyllide reductase. The phenolic profile of A. millefolium was assessed. Etoxazole exposure reduced rooting percentage, root length, weight gain, mitotic index, and levels of chlorophyll a and chlorophyll b. Conversely, etoxazole treatment led to an increase in chromosomal aberrations and micronuclei occurrence. The most frequently observed chromosomal aberrations induced by etoxazole, which serve as bioindicators of genotoxicity, were fragment, vagrant chromosome, sticky chromosome, unequal chromatin distribution, bridge, reverse polarization, and vacuolated nucleus. The levels of malondialdehyde and antioxidant enzyme (superoxide dismutase and catalase) activities were also elevated. Epidermis cell damage, flattened cell nucleus, thickened cortex cell wall, and thickened conduction tissue were the meristematic cell disorders triggered by etoxazole. Molecular docking studies showed that etoxazole can interact directly with DNA, tubulins, and the enzymes mentioned above. A. millefolium extract was found to contain a substantial quantity of phenolic compounds. A. millefolium extract, when co-administered with etoxazole, attenuated all toxic effects of etoxazole dose-dependently. In conclusion, A. millefolium may potentially serve as a reliable pharmacological shield against the toxicity of pesticides in non-target organisms.
Collapse
Affiliation(s)
- Zeynep Şule Topatan
- Department of Biology, Faculty of Science and Art, Giresun University, Giresun, Turkey
| | - Tuğçe Kalefetoğlu Macar
- Department of Food Technology, Şebinkarahisar School of Applied Sciences, Giresun University, Giresun, Turkey.
| | - Oksal Macar
- Department of Biology, Faculty of Science and Art, Giresun University, Giresun, Turkey
| | - Emine Yalçin
- Department of Biology, Faculty of Science and Art, Giresun University, Giresun, Turkey
| | - Kültiğin Çavuşoğlu
- Department of Biology, Faculty of Science and Art, Giresun University, Giresun, Turkey
| | - Ali Acar
- Department of Medical Services and Techniques, Vocational School of Health Services, Giresun University, Giresun, Turkey
| | - Fatih Kutluer
- Department of Herbal and Animal Production, Kırıkkale Vocational School, Kırıkkale University, Kırıkkale, Turkey
| |
Collapse
|
6
|
Borg KN, Shetty A, Cheng G, Zhu S, Wang T, Yuan W, Ho HP, Knudsen BR, Tesauro C, Ho YP. Hydrogel bead-based isothermal detection (BEAD-ID) for assessing the activity of DNA-modifying enzymes. iScience 2024; 27:111332. [PMID: 39640584 PMCID: PMC11617385 DOI: 10.1016/j.isci.2024.111332] [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: 06/20/2024] [Revised: 09/25/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
DNA-modifying enzymes are crucial in biological processes and have significant clinical implications. Traditional quantification methods often overlook enzymatic activity, the true determinants of enzymes' functions. We present hydrogel Bead-based Isothermal Detection (BEAD-ID), utilizing uniform hydrogel bead-based microreactors to evaluate DNA-modifying enzyme activity on-bead. We fabricated homogeneous oligo-conjugated polyacrylamide (oligo-PAA) beads via droplet microfluidics, optimized for capturing and amplifying enzyme-modified nanosensors. By incorporating DNA oligos within the hydrogel network, BEAD-ID retains isothermally amplified products, facilitating in situ detection of enzyme activities on-bead. We validate BEAD-ID by quantifying human topoisomerase I (TOP1) and restriction endonuclease EcoRI, showing a direct correlation between enzyme concentration and fluorescence intensity, demonstrating the platform's sensitivity (6.25 nM TOP1, 6.25 U/μL EcoRI) and reliability in food matrix (25 U/μL EcoRI). Additionally, a customized flow cytometry-mimicking setup allows high-throughput detection at 352 Hz with objective assessment. BEAD-ID, offering flexibility and scalability, is a promising tool for studying DNA-modifying enzymes.
Collapse
Affiliation(s)
- Kathrine Nygaard Borg
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 00000, China
| | - Ayush Shetty
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 00000, China
| | - Guangyao Cheng
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 00000, China
| | - Shaodi Zhu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 00000, China
| | - Tianle Wang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 00000, China
| | - Wu Yuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 00000, China
| | - Ho Pui Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 00000, China
| | - Birgitta Ruth Knudsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Cinzia Tesauro
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 00000, China
- Centre for Biomaterials, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, Hong Kong SAR, China
- State Key Laboratory of Marine Pollution, City University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Ali DME, Aziz HA, Bräse S, Al Bahir A, Alkhammash A, Abuo-Rahma GEDA, Elshamsy AM, Hashem H, Abdelmagid WM. Unveiling the Anticancer Potential of a New Ciprofloxacin-Chalcone Hybrid as an Inhibitor of Topoisomerases I & II and Apoptotic Inducer. Molecules 2024; 29:5382. [PMID: 39598770 PMCID: PMC11596536 DOI: 10.3390/molecules29225382] [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: 08/29/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
The current study has yielded promising results in the evaluation of a new ciprofloxacin-chalcone hybrid (CP derivative) for its anticancer activity as potential Topoisomerases (Topo) I and II inhibitors. The in vitro results showed that the CP derivative significantly suppressed the growth of HCT-116 and LOX IMVI cells, with IC50 values of 5.0 μM and 1.3 μM, respectively, outperforming Staurosporine, which had IC50 values of 8.4 μM and 1.6 μM, respectively. Flow cytometry analysis revealed that the new CP derivative triggered apoptosis and cell cycle arrest at the G2/M phase, associated with the up-regulation of pro-apoptotic genes (Bax and Caspase 9) and downregulation of the anti-apoptotic gene (Bcl-2). Further investigations showed that the CP derivative inhibited Topo I and II enzymes, as expected molecular targets; docking studies further supported its dual inhibitory action on Topo I and II. These findings suggest that the ciprofloxacin-chalcone hybrid could be a promising lead compound for developing new anticancer therapy.
Collapse
Affiliation(s)
| | - Hossameldin A. Aziz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, New Valley University, New Valley 72511, Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems—Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Kaiserstrasse 12, 76131 Karlsruhe, Germany
| | - Areej Al Bahir
- Chemistry Department, Faculty of Science, King Khalid University, Abha 64734, Saudi Arabia
| | - Abdullah Alkhammash
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Gamal El-Din A. Abuo-Rahma
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, New Minia City 61768, Egypt
| | - Ali M. Elshamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, New Minia City 61768, Egypt
| | - Hamada Hashem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Walid M. Abdelmagid
- Medicinal Chemistry and Drug Discovery Research Centre, Swenam College, 210-6125 Sussex Avenue, Burnaby, BC V5H 4G1, Canada;
| |
Collapse
|
8
|
Badr M, Elmongy EI, Elkhateeb D, Moemen YS, Khalil A, Ali H, Binsuwaidan R, Awadallah F, El Sayed IET. In Silico and In Vitro Investigation of Cytotoxicity and Apoptosis of Acridine/Sulfonamide Hybrids Targeting Topoisomerases I and II. Pharmaceuticals (Basel) 2024; 17:1487. [PMID: 39598399 PMCID: PMC11597879 DOI: 10.3390/ph17111487] [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: 09/25/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Sulfonamide acridine derivatives have garnered significant attention from medicinal chemists due to their diverse range of biological activities. METHODS In this study, eleven compounds were synthesized according to the literature, and their impact on cell growth inhibition, induction of apoptosis, and cell cycle distribution were assessed in three different cell lines. Their inhibitory effects on the topoisomerase (Topo) I and II were investigated in vitro. Molecular docking studies were conducted to predict the binding affinities of these compounds for crystallized downloaded topoisomerases. RESULTS The compounds were examined in vitro for their anticancer activity against human hepatic (HepG2) colon (HCT-8) and breast (MCF-7) carcinoma cell lines. Compound 8b was the most active against HepG2, HCT-116, and MCF-7 with IC50 14.51, 9.39, and 8.83 µM, respectively, compared to Doxorubicin as reference. In addition, it demonstrated the highest potency among the tested compounds against Topo-I, with an IC50 value of 3.41 µg/mL compared to the control camptothecin (IC50 of 1.46 μM). Compound 7c displayed a significant inhibitory effect on Topo-II, with an IC50 of 7.33 μM, compared to an IC50 value of 6.49 μM via Doxorubicin, the control. Compounds 7c and 8b were assessed against topoisomerases showing induction of apoptosis and a reduction in the S phase of the cell cycle. Molecular docking demonstrated interaction with the active site as with those exhibited by the co-crystallized ligands of the crystallized proteins in both topoisomerases. CONCLUSION Compounds 7c and 8b hold promise as potential anticancer drugs due to their anti-proliferative and proapoptotic effects, which are mediated by their action on the topoisomerase enzyme, particularly Topo II.
Collapse
Affiliation(s)
- Mohamed Badr
- Department of Biochemistry, Faculty of Pharmacy, Menoufia University, Shebin El-Kom 6131567, Egypt;
| | - Elshaymaa I. Elmongy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo P.O. Box 11795, Egypt
| | - Doaa Elkhateeb
- Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Kom 32511, Egypt; (D.E.); (H.A.); (F.A.); (I.E.T.E.S.)
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom 32511, Egypt;
| | - Yasmine S. Moemen
- Clinical Pathology Department, National Liver Institute, Menoufia University, Shebin El-Kom 32511, Egypt;
| | - Ashraf Khalil
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom 32511, Egypt;
| | - Hadeer Ali
- Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Kom 32511, Egypt; (D.E.); (H.A.); (F.A.); (I.E.T.E.S.)
| | - Reem Binsuwaidan
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Feby Awadallah
- Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Kom 32511, Egypt; (D.E.); (H.A.); (F.A.); (I.E.T.E.S.)
- Clinical pathology Department, Menoufia University Hospital, Shebin El-Kom 32511, Egypt
| | - Ibrahim El Tantawy El Sayed
- Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Kom 32511, Egypt; (D.E.); (H.A.); (F.A.); (I.E.T.E.S.)
| |
Collapse
|
9
|
Bagattin A, Tammaccaro SL, Chiral M, Makinistoglu MP, Zimmermann N, Lerner J, Garbay S, Kuperwasser N, Pontoglio M. HNF1β bookmarking involves Topoisomerase 1 activation and DNA topology relaxation in mitotic chromatin. Cell Rep 2024; 43:114805. [PMID: 39388351 DOI: 10.1016/j.celrep.2024.114805] [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: 12/04/2023] [Revised: 07/03/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
HNF1β (HNF1B) is a transcription factor frequently mutated in patients with developmental renal disease. It binds to mitotic chromatin and reactivates gene expression after mitosis, a phenomenon referred to as bookmarking. Using a crosslinking method that circumvents the artifacts of formaldehyde, we demonstrate that HNF1β remains associated with chromatin in a sequence-specific way in both interphase and mitosis. We identify an HNF1β-interacting protein, BTBD2, that enables the interaction and activation of Topoisomerase 1 (TOP1) exclusively during mitosis. Our study identifies a shared microhomology domain between HNF1β and TOP1, where a mutation, found in "maturity onset diabetes of the young" patients, disrupts their interaction. Importantly, HNF1β recruits TOP1 and induces DNA relaxation around HNF1β mitotic chromatin sites, elucidating its crucial role in chromatin remodeling and gene reactivation after mitotic exit. These findings shed light on how HNF1β reactivates target gene expression after mitosis, providing insights into its crucial role in maintenance of cellular identity.
Collapse
Affiliation(s)
- Alessia Bagattin
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France.
| | - Salvina Laura Tammaccaro
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Magali Chiral
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Munevver Parla Makinistoglu
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Nicolas Zimmermann
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Jonathan Lerner
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Serge Garbay
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Nicolas Kuperwasser
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Marco Pontoglio
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France.
| |
Collapse
|
10
|
Mohamed MA, Elsaman T, Elderdery AY, Alsrhani A, Ghanem HB, Alruwaili MM, Hamza SMA, Mekki SEI, Alotaibi HA, Mills J. Unveiling the Anticancer Potential: Computational Exploration of Nitrogenated Derivatives of (+)-Pancratistatin as Topoisomerase I Inhibitors. Int J Mol Sci 2024; 25:10779. [PMID: 39409108 PMCID: PMC11476810 DOI: 10.3390/ijms251910779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer poses a substantial global health challenge, driving the need for innovative therapeutic solutions that offer improved effectiveness and fewer side effects. Topoisomerase I (Topo I) has emerged as a validated molecular target in the pursuit of developing anticancer drugs due to its critical role in DNA replication and transcription. (+)-Pancratistatin (PST), a naturally occurring compound found in various Amaryllidaceae plants, exhibits promising anticancer properties by inhibiting Topo I activity. However, its clinical utility is hindered by issues related to limited chemical availability and aqueous solubility. To address these challenges, molecular modelling techniques, including virtual screening, molecular docking, molecular mechanics with generalised born and surface area solvation (MM-GBSA) calculations, and molecular dynamics simulations were utilised to evaluate the binding interactions and energetics of PST analogues with Topo I, comparing them with the well-known Topo I inhibitor, Camptothecin. Among the compounds screened for this study, nitrogenated analogues emerged as the most encouraging drug candidates, exhibiting improved binding affinities, favourable interactions with the active site of Topo I, and stability of the protein-ligand complex. Structural analysis pinpointed key molecular determinants responsible for the heightened potency of nitrogenated analogues, shedding light on essential structural modifications for increased activity. Moreover, in silico absorption, distribution, metabolism, excretion, and toxicity (ADMET) predictions highlighted favourable drug-like properties and reduced toxicity profiles for the most prominent nitrogenated analogues, further supporting their potential as effective anticancer agents. In summary, this screening study underscores the significance of nitrogenation in augmenting the anticancer efficacy of PST analogues targeting Topo I. The identified lead compounds exhibit significant potential for subsequent experimental validation and optimisation, thus facilitating the development of novel and efficacious anticancer therapeutics with enhanced pharmacological profiles.
Collapse
Affiliation(s)
- Magdi Awadalla Mohamed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Tilal Elsaman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Heba Bassiony Ghanem
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Majed Mowanes Alruwaili
- Nursing Administration & Education Department, College of Nursing, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Siddiqa M. A. Hamza
- Department of Pathology, College of Medicine, Umm Alqura University, Algunfudah 21912, Saudi Arabia;
| | | | | | - Jeremy Mills
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK;
| |
Collapse
|
11
|
Doan NQH, Tran HN, Nguyen NTM, Pham TM, Nguyen QDK, Vu TT. Synthesis, Antimicrobial - Cytotoxic Evaluation, and Molecular Docking Studies of Quinolin-2-one Hydrazones Containing Nitrophenyl or Isonicotinoyl/Nicotinoyl Moiety. Chem Biodivers 2024; 21:e202401142. [PMID: 39032128 DOI: 10.1002/cbdv.202401142] [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: 05/04/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 07/22/2024]
Abstract
By applying the hybrid molecular strategy, in this study, we reported the synthesis of fifteen quinolin-2-one hydrazones containing nitrophenyl or nicotinonyl/isonicotinoyl moiety, followed by in vitro and in silico evaluations of their potential antimicrobial and anticancer activities. In vitro antimicrobial evaluation of the target compounds on seven pathogenic strains, applying the broth microdilution method, revealed that compound 4a demonstrated the most potential antifungal activity against C. albicans (MIC 512 μg mL-1) and C. krusei (MIC 128 μg mL-1). In vitro cytotoxic evaluation of the target compounds on three human cancer cell lines, employing the MTT method, suggested that compound 5c exhibited the most potential cytotoxicities against HepG2 (IC50 10.19 μM), A549 (IC50 20.43 μM), and MDA-MB-231 (IC50 16.82 μM) cells. Additionally, molecular docking studies were performed to investigate the binding characteristics of compounds 4a and 5c with fungal lanosterol 14α-demethylase and human topoisomerase I-II, respectively, thereby contributing to the elucidation of their in vitro antifungal and cytotoxic properties. Furthermore, compounds 4a and 5c, via SwissADME prediction, could exhibit favorable physicochemical and pharmacokinetic properties. In conclusion, this study provides valuable insights into the potential of quinolin-2-one hydrazones as promising candidates for the development of novel antimicrobial and anticancer agents in the future.
Collapse
Affiliation(s)
- Nam Q H Doan
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Hoan N Tran
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Nhu T M Nguyen
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Thu M Pham
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Quyen D K Nguyen
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Thanh-Thao Vu
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 41-43 Dinh Tien Hoang Street, Ben Nghe Ward, District 1, Ho Chi Minh City, 70000, Vietnam
| |
Collapse
|
12
|
Nitiss KC, Bandak A, Berger JM, Nitiss JL. Genome Instability Induced by Topoisomerase Misfunction. Int J Mol Sci 2024; 25:10247. [PMID: 39408578 PMCID: PMC11477040 DOI: 10.3390/ijms251910247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Topoisomerases alter DNA topology by making transient DNA strand breaks (DSBs) in DNA. The DNA cleavage reaction mechanism includes the formation of a reversible protein/DNA complex that allows rapid resealing of the transient break. This mechanism allows changes in DNA topology with minimal risks of persistent DNA damage. Nonetheless, small molecules, alternate DNA structures, or mutations in topoisomerase proteins can impede the resealing of the transient breaks, leading to genome instability and potentially cell death. The consequences of high levels of enzyme/DNA adducts differ for type I and type II topoisomerases. Top1 action on DNA containing ribonucleotides leads to 2-5 nucleotide deletions in repeated sequences, while mutant Top1 enzymes can generate large deletions. By contrast, small molecules that target Top2, or mutant Top2 enzymes with elevated levels of cleavage lead to small de novo duplications. Both Top1 and Top2 have the potential to generate large rearrangements and translocations. Thus, genome instability due to topoisomerase mis-function is a potential pathogenic mechanism especially leading to oncogenic progression. Recent studies support the potential roles of topoisomerases in genetic changes in cancer cells, highlighting the need to understand how cells limit genome instability induced by topoisomerases. This review highlights recent studies that bear on these questions.
Collapse
Affiliation(s)
- Karin C. Nitiss
- Pharmaceutical Sciences Department, University of Illinois Chicago, Rockford, IL 61107, USA;
| | - Afif Bandak
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD 20215, USA; (A.B.); (J.M.B.)
| | - James M. Berger
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD 20215, USA; (A.B.); (J.M.B.)
| | - John L. Nitiss
- Pharmaceutical Sciences Department, University of Illinois Chicago, Rockford, IL 61107, USA;
| |
Collapse
|
13
|
Westhorpe R, Roske JJ, Yeeles JTP. Mechanisms controlling replication fork stalling and collapse at topoisomerase 1 cleavage complexes. Mol Cell 2024; 84:3469-3481.e7. [PMID: 39236719 PMCID: PMC7617106 DOI: 10.1016/j.molcel.2024.08.004] [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: 03/08/2024] [Revised: 06/14/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024]
Abstract
Topoisomerase 1 cleavage complexes (Top1-ccs) comprise a DNA-protein crosslink and a single-stranded DNA break that can significantly impact the DNA replication machinery (replisome). Consequently, inhibitors that trap Top1-ccs are used extensively in research and clinical settings to generate DNA replication stress, yet how the replisome responds upon collision with a Top1-cc remains obscure. By reconstituting collisions between budding yeast replisomes, assembled from purified proteins, and site-specific Top1-ccs, we have uncovered mechanisms underlying replication fork stalling and collapse. We find that stalled replication forks are surprisingly stable and that their stability is influenced by the template strand that Top1 is crosslinked to, the fork protection complex proteins Tof1-Csm3 (human TIMELESS-TIPIN), and the convergence of replication forks. Moreover, nascent-strand mapping and cryoelectron microscopy (cryo-EM) of stalled forks establishes replisome remodeling as a key factor in the initial response to Top1-ccs. These findings have important implications for the use of Top1 inhibitors in research and in the clinic.
Collapse
Affiliation(s)
- Rose Westhorpe
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Johann J Roske
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Joseph T P Yeeles
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
14
|
Tian JR, Qiao YH, Zhuang QB, Fan R, Li Z, Zhang XM, Zhang FM, Tu YQ. Organo-cation catalyzed enantioselective α-hydroxylation of pyridinone-fused lactones: asymmetric synthesis of SN-38 and irinotecan. Chem Commun (Camb) 2024; 60:9954-9957. [PMID: 39177032 DOI: 10.1039/d4cc03580a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
A catalytic asymmetric α-hydroxylation of pyridinone-fused lactones, containing the core structure of camptothecin, is described. Development of a novel spiropyrrolidine amide (SPA) derived triazolium bromide organo-cation catalyst is crucial for a highly enantioselective oxidation, which also accommodates a wide array of lactones with various substituents. The resulting tricyclic tertiary alcohol with an oxa-quaternary carbon center can be further applied in the synthesis of SN-38 and irinotecan, two anti-cancer drugs derived from camptothecin.
Collapse
Affiliation(s)
- Jin-Rui Tian
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China.
| | - Yu-Hao Qiao
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China.
| | - Qing-Bo Zhuang
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China.
- School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rong Fan
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China.
| | - Zhen Li
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China.
| | - Xiao-Ming Zhang
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China.
| | - Fu-Min Zhang
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China.
| | - Yong-Qiang Tu
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China.
| |
Collapse
|
15
|
Kutluer F, Özkan B, Yalçin E, Çavuşoğlu K. Direct and indirect toxicity mechanisms of the natural insecticide azadirachtin based on in-silico interactions with tubulin, topoisomerase and DNA. CHEMOSPHERE 2024; 364:143006. [PMID: 39098344 DOI: 10.1016/j.chemosphere.2024.143006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Natural pesticides, which attract attention with safe properties, pose a threat to many non-target organisms, so their toxic effects should be studied extensively. In this study, the toxic effects of Azadirachtin, a natural insecticide derived from Azadirachta indica, were investigated by in-vivo and in-silico methods. In-vivo toxic effects were determined using the Allium test and bulbs were treated with 5 mg/L (0.5x EC50), 10 mg/L (EC50), and 20 mg/L (2xEC50) Azadirachtin. In the groups treated with Azadirachtin, there was a decline in germination-related parameters and accordingly growth was delayed. This regression may be related to oxidative stress in the plant, and the increase in malondialdehyde and proline levels in Azadirachtin-applied groups confirms oxidative stress. Azadirachtin toxicity increased dose-dependently and the most significant toxic effect was observed in the group administered 20 mg/L Azadirachtin. In this group, the mitotic index decreased by 43.4% and sticky chromosomes, vagrant chromosomes and fragments were detected at rates of 83.1 ± 4.01, 72.7 ± 3.46 and 65.1 ± 3.51, respectively. By comet analysis, it was determined that Azadirachtin caused DNA fragmentation, and tail DNA, which was 0.10 ± 0.32% in the control group, increased to 34.5 ± 1.35% in the Azadirachtin -treated groups. These cytotoxic and genotoxic effects of Azadirachtin may be due to direct interaction with macromolecules as well as induced oxidative stress. Azadirachtin has been found to interact in-silico with alpha-tubulin, beta-tubulin, topoisomerase I and II, and various DNA sequences. Possible deteriorations in macromolecular structure and functions as a result of these interactions may cause cytotoxic and genotoxic effects. These results suggest that natural insecticides may also be unreliable for non-target organisms, and the toxic effects of compounds presented as "natural" should also be investigated.
Collapse
Affiliation(s)
- Fatih Kutluer
- Department of Herbal and Animal Production, Kırıkkale Vocational School, Kırıkkale University, Kırıkkale, Turkiye.
| | - Burak Özkan
- Department of Biology, Institute of Science, Giresun University, Giresun, Turkiye.
| | - Emine Yalçin
- Department of Biology, Faculty of Science and Art, Giresun University, Giresun, Turkiye.
| | - Kültiğin Çavuşoğlu
- Department of Biology, Faculty of Science and Art, Giresun University, Giresun, Turkiye.
| |
Collapse
|
16
|
El-Hady NAAA, ElSayed AI, Wadan KM, El-Saadany SS, El-Sayed ASA. Bioprocessing of camptothecin from Alternaria brassicicola, an endophyte of Catharanthus roseus, with a strong antiproliferative activity and inhibition to Topoisomerases. Microb Cell Fact 2024; 23:214. [PMID: 39060918 PMCID: PMC11282713 DOI: 10.1186/s12934-024-02471-5] [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: 12/21/2023] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Suppression of fungal camptothecin (CPT) biosynthesis with the preservation and successive subculturing is the challenge that impedes fungi from the industrial application, so, screening for a novel fungal isolate with a conceivable stable producing potency of CPT was the main objective of this work. Catharanthus roseus with diverse contents of bioactive metabolites could have a plethora of novel endophytes with unique metabolic properties. Among the endophytes of C. roseus, Alternaria brassicicola EFBL-NV OR131587.1 was the highest CPT producer (96.5 μg/L). The structural identity of the putative CPT was verified by HPLC, FTIR, HNMR and LC-MS/MS, with a molecular mass 349 m/z, and molecular fragmentation patterns that typically identical to the authentic one. The purified A. brassicicola CPT has a strong antiproliferative activity towards UO-31 (0.75 μM) and MCF7 (3.2 μM), with selectivity index 30.8, and 7.1, respectively, in addition to resilient activity to inhibit Topo II (IC50 value 0.26 nM) than Topo 1 (IC50 value 3.2 nM). The purified CPT combat the wound healing of UO-31 cells by ~ 52%, stops their matrix formation, cell migration and metastasis. The purified CPT arrest the cellular division of the UO-31 at the S-phase, and inducing their cellular apoptosis by ~ 20.4 folds, compared to the control cells. Upon bioprocessing with the surface response methodology, the CPT yield by A. brassicicola was improved by ~ 3.3 folds, compared to control. The metabolic potency of synthesis of CPT by A. brassicicola was attenuated with the fungal storage and subculturing, losing ~ 50% of their CPT productivity by the 6th month of storage and 6th generation. Practically, the CPT productivity of the attenuated A. brassicicola was restored by addition of 1% surface sterilized leaves of C. roseus, ensuring the eliciting of cryptic gene cluster of A. brassicicola CPT via the plant microbiome-A. brassicicola interactions. So, for the first time, a novel endophytic isolate A. brassicicola, from C. roseus, was explored to have a relatively stable CPT biosynthetic machinery, with an affordable feasibility to restore their CPT productivity using C. roseus microbiome, in addition to the unique affinity of the extracted CPT to inhibit Topoisomerase I and II.
Collapse
Affiliation(s)
- Nouran A A Abd El-Hady
- Enzymology and Fungal Biotechnology Lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Abdelaleim I ElSayed
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Khalid M Wadan
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Sayed S El-Saadany
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Ashraf S A El-Sayed
- Enzymology and Fungal Biotechnology Lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
17
|
Correa de Moraes M, Frassini R, Roesch-Ely M, Reisdorfer de Paula F, Barcellos T. Novel Coumarin-Nucleobase Hybrids with Potential Anticancer Activity: Synthesis, In Vitro Cell-Based Evaluation, and Molecular Docking. Pharmaceuticals (Basel) 2024; 17:956. [PMID: 39065804 PMCID: PMC11279566 DOI: 10.3390/ph17070956] [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: 05/16/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
A new series of compounds planned by molecular hybridization of the nucleobases uracil and thymine, or the xanthine theobromine, with coumarins, and linked through 1,2,3-triazole heterocycles were evaluated for their in vitro anticancer activity against the human tumor cell lines: colon carcinoma (HCT116), laryngeal tumor cells (Hep-2), and lung carcinoma cells (A549). The hybrid compound 9a exhibited better activity in the series, showing an IC50 of 24.19 ± 1.39 μM against the HCT116 cells, with a selectivity index (SI) of 6, when compared to the cytotoxicity against the non-tumor cell line HaCat. The in silico search for pharmacological targets was achieved through molecular docking studies on all active compounds, which suggested that the synthesized compounds possess a high affinity to the Topoisomerase 1-DNA complex, supporting their antitumor activity. The in silico toxicity prediction studies suggest that the compounds present a low risk of causing theoretical mutagenic and tumorigenic effects. These findings indicate that molecular hybridization from natural derivative molecules is an interesting approach to seek new antitumor candidates.
Collapse
Affiliation(s)
- Maiara Correa de Moraes
- Laboratório de Biotecnologia de Produtos Naturais e Sintéticos, Universidade de Caxias do Sul, Francisco Getúlio Vargas St., 1130, Caxias do Sul 95070-560, RS, Brazil;
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Sul—Campus Caxias do Sul, Avelino Antônio de Souza, 1730, Caxias do Sul 95043-700, RS, Brazil
| | - Rafaele Frassini
- Laboratório de Genômica, Proteômica e Reparo de DNA, Universidade de Caxias do Sul, Francisco Getúlio Vargas St., 1130, Caxias do Sul 95070-560, RS, Brazil; (R.F.); (M.R.-E.)
| | - Mariana Roesch-Ely
- Laboratório de Genômica, Proteômica e Reparo de DNA, Universidade de Caxias do Sul, Francisco Getúlio Vargas St., 1130, Caxias do Sul 95070-560, RS, Brazil; (R.F.); (M.R.-E.)
| | - Favero Reisdorfer de Paula
- Laboratório de Desenvolvimento e Controle de Qualidade em Medicamentos, Universidade Federal do Pampa, Campus Uruguaiana, BR 472, Km 592, Uruguaiana 97508-000, RS, Brazil;
| | - Thiago Barcellos
- Laboratório de Biotecnologia de Produtos Naturais e Sintéticos, Universidade de Caxias do Sul, Francisco Getúlio Vargas St., 1130, Caxias do Sul 95070-560, RS, Brazil;
| |
Collapse
|
18
|
Fedorov II, Bubis JA, Kazakova EM, Lobas AA, Ivanov MV, Emekeeva DD, Tarasova IA, Nazarov AA, Gorshkov MV. On the utility of ultrafast MS1-only proteomics in drug target discovery studies based on thermal proteome profiling method. Anal Bioanal Chem 2024; 416:4083-4089. [PMID: 38744720 DOI: 10.1007/s00216-024-05330-9] [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: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024]
Abstract
Advances in high-throughput high-resolution mass spectrometry and the development of thermal proteome profiling approach (TPP) have made it possible to accelerate a drug target search. Since its introduction in 2014, TPP quickly became a method of choice in chemical proteomics for identifying drug-to-protein interactions on a proteome-wide scale and mapping the pathways of these interactions, thus further elucidating the unknown mechanisms of action of a drug under study. However, the current TPP implementations based on tandem mass spectrometry (MS/MS), associated with employing lengthy peptide separation protocols and expensive labeling techniques for sample multiplexing, limit the scaling of this approach for the ever growing variety of drug-to-proteomes. A variety of ultrafast proteomics methods have been developed in the last couple of years. Among them, DirectMS1 provides MS/MS-free quantitative proteome-wide analysis in 5-min time scale, thus opening the way for sample-hungry applications, such as TPP. In this work, we demonstrate the first implementation of the TPP approach using the ultrafast proteome-wide analysis based on DirectMS1. Using a drug topotecan, which is a known topoisomerase I (TOP1) inhibitor, the feasibility of the method for identifying drug targets at the whole proteome level was demonstrated for an ovarian cancer cell line.
Collapse
Affiliation(s)
- Ivan I Fedorov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Leninsky Pr. 38, Bld.2, 119334, Moscow, Russia
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - Julia A Bubis
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Leninsky Pr. 38, Bld.2, 119334, Moscow, Russia
| | - Elizaveta M Kazakova
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Leninsky Pr. 38, Bld.2, 119334, Moscow, Russia
| | - Anna A Lobas
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Leninsky Pr. 38, Bld.2, 119334, Moscow, Russia
| | - Mark V Ivanov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Leninsky Pr. 38, Bld.2, 119334, Moscow, Russia
| | - Daria D Emekeeva
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Leninsky Pr. 38, Bld.2, 119334, Moscow, Russia
| | - Irina A Tarasova
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Leninsky Pr. 38, Bld.2, 119334, Moscow, Russia
| | - Alexey A Nazarov
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, 119991, Moscow, Russia
- Faculty of Chemistry of the National Research University Higher School of Economics, Vavilova Str. 7, 101000, Moscow, Russia
| | - Mikhail V Gorshkov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Leninsky Pr. 38, Bld.2, 119334, Moscow, Russia.
| |
Collapse
|
19
|
Patil DM, Hunasagi BS, Raghu AV, Kulkarni RV, Akamanchi KG. Optimisation of enzyme-assisted extraction of camptothecin from Nothapodytes nimmoniana and its characterisation. PHYTOCHEMICAL ANALYSIS : PCA 2024; 35:825-839. [PMID: 38351290 DOI: 10.1002/pca.3331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION Efficient extraction of camptothecin (CPT), an anticancer agent from the commercial source Nothapodytes nimmoniana (J. Graham) Mabb in India, is of paramount importance. CPT is present in the highest concentration in the stem portion, and the stem can be readily harvested without uprooting the plant. The fluorescence microscopy mapping of the bark matrix for CPT revealed its presence in a free form within both the outer (epidermal and cortical tissues) and inner (xylem and phloem tissues) sections. The bark matrix primarily consists of cellulose, hemicellulose, and lignin, rendering it woody, rigid, and resistant to efficient solvent penetration for CPT extraction. We proposed a hypothesis that subjecting it to disruption through treatment with hydrolytic enzymes like cellulase and xylanase could enhance solvent diffusion, thereby enabling a swift and effective extraction of CPT. OBJECTIVE The present study was aimed at enzyme-assisted extraction, using cellulase and xylanase for hydrolytic disruption of the cells to readily access CPT from the stem of the plant N. nimmoniana (J. Graham) Mabb. METHODOLOGY The hydrolytic cell disruption of ground powder from the stem bark was studied using cellulase and xylanase enzymes. The enzymatically pretreated stem bark powder was subsequently recovered by filtration, dried, and subjected to extraction with methanol to isolate CPT. This process was optimised through a Box-Behnken design, employing a one-factor-at-a-time approach to assess parameters such as enzyme concentration (2-10% w/w), pH (3-7), incubation time (6-24 h), and solid-to-solvent ratio (1:30-1:70 g/mL). CPT was characterised using proton nuclear magnetic resonance (1H-NMR) and Fourier transform infrared (FTIR) spectra, and a high-performance liquid chromatography (HPLC) method was developed for quantification. RESULTS The cellulase and xylanase treatment resulted in the highest yields of 0.285% w/w and 0.343% w/w, with efficiencies of 67% and 81%, respectively, achieved in a significantly shorter time compared to the untreated material, which yielded 0.18% with an efficiency of only 42%. Extraction by utilising the predicted optimised process parameters, a nearly two-fold increase in the yield, was observed for xylanase, with incubation and solvent extraction times set at 16 and 2 h, respectively. Scanning electron microscopy (SEM) images of the spent material indicated perforations attributed to enzymatic action, suggesting that this could be a primary factor contributing to the enhanced extraction. CONCLUSION Enzyme-mediated hydrolytic cell disruption could be a potential approach for efficient and rapid isolation of CPT from the bark of N. nimmoniana.
Collapse
Affiliation(s)
- Dhiraj M Patil
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Basavaraj S Hunasagi
- Department of Pharmacognosy, BLDEA's SSM College of Pharmacy and Research Centre, Vijayapura, Karnataka, India
| | - Anjanapura V Raghu
- Science and Technology, BLDE (Deemed-to-be University), Vijayapura, Karnataka, India
| | - Raghavendra V Kulkarni
- Department of Pharmaceutics, BLDEA's SSM College of Pharmacy and Research Centre, Vijayapura, Karnataka, India
| | - Krishnacharya G Akamanchi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
- Department of Allied Health Sciences, BLDE (Deemed-to-be University), Vijayapura, Karnataka, India
| |
Collapse
|
20
|
Liu J, Xue Y, Bai K, Yan F, Long X, Guo H, Yan H, Huang G, Zhou J, Tang Y. Experimental and computational study on anti-gastric cancer activity and mechanism of evodiamine derivatives. Front Pharmacol 2024; 15:1380304. [PMID: 38783957 PMCID: PMC11113551 DOI: 10.3389/fphar.2024.1380304] [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: 02/01/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction: Human topoisomerase 1 (TOP1) is an important target of various anticancer compounds. The design and discovery of inhibitors targeting TOP1 are of great significance for the development of anticancer drugs. Evodiamine and thieno [2,3-d] pyridine hybrids show potential antitumor activity. Herein, the anti-gastric cancer activities of these hybrids were investigated. Methods: The inhibitory effects of different concentrations of ten evodiamine derivatives on the gastric cancer cell line SGC-7901 were assessed using a methyl thiazolyl tetrazolium assay. Compounds EVO-1 and EVO-6 strongly inhibited gastric cancer cell proliferation, with inhibition rates of 81.17% ± 5.08% and 80.92% ± 2.75%, respectively. To discover the relationship between the structure and activity of these two derivatives, density functional theory was used to investigate their optimized geometries, natural population charges, frontier molecular orbitals, and molecular electrostatic potentials. To clarify their anti-gastric cancer mechanisms, molecular docking, molecular dynamics simulations, and binding free energy calculations were performed against TOP1. Results: The results demonstrated that these compounds could intercalate into the cleaved DNA-binding site to form a TOP1-DNA-ligand ternary complex, and the ligand remained secure at the cleaved DNA-binding site to form a stable ternary complex. As the binding free energy of compound EVO-1 with TOP1 (-38.33 kcal·mol-1) was lower than that of compound EVO-6 (-33.25 kcal·mol-1), compound EVO-1 could be a more potent anti-gastric cancer agent than compound EVO-6. Discussion: Thus, compound EVO-1 could be a promising anti-gastric cancer drug candidate. This study may facilitate the design and development of novel TOP1 inhibitors.
Collapse
Affiliation(s)
- Jingli Liu
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yingying Xue
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Kaidi Bai
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Fei Yan
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xu Long
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Hui Guo
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Hao Yan
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Guozheng Huang
- College of Chemistry and Chemical Engineering, Anhui University of Technology, Ma’anshan, Anhui, China
| | - Jing Zhou
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yuping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
21
|
Cho JE, Shaltz S, Yakovleva L, Shuman S, Jinks-Robertson S. Deletions initiated by the vaccinia virus TopIB protein in yeast. DNA Repair (Amst) 2024; 137:103664. [PMID: 38484460 PMCID: PMC10994728 DOI: 10.1016/j.dnarep.2024.103664] [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: 10/06/2023] [Revised: 02/14/2024] [Accepted: 03/01/2024] [Indexed: 04/06/2024]
Abstract
The type IB topoisomerase of budding yeast (yTop1) generates small deletions in tandem repeats through a sequential cleavage mechanism and larger deletions with random endpoints through the nonhomologous end-joining (NHEJ) pathway. Vaccinia virus Top1 (vTop1) is a minimized version of the eukaryal TopIB enzymes and uniquely has a strong consensus cleavage sequence: the pentanucleotide (T/C)CCTTp↓. To define the relationship between the position of TopIB cleavage and mutagenic outcomes, we expressed vTop1 in yeast top1Δ strains containing reporter constructs with a single CCCTT site, tandem CCCTT sites, or CCCTT sites separated by 42 bp. vTop1 cleavage at a single CCCTT site was associated with small, NHEJ-dependent deletions. As observed with yTop1, vTop1 generated 5-bp deletions at tandem CCCTT sites. In contrast to yTop1-initiated deletions, however, 5-bp deletions associated with vTop1 expression were not affected by the level of ribonucleotides in genomic DNA. vTop1 expression was associated with a 47-bp deletion when CCCTT sites were separated by 42 bp. Unlike yTop1-initiated large deletions, the vTop1-mediated 47-bp deletion did not require NHEJ, consistent with a model in which re-ligation of enzyme-associated double-strand breaks is catalyzed by vTop1.
Collapse
Affiliation(s)
- Jang Eun Cho
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Samantha Shaltz
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lyudmila Yakovleva
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stewart Shuman
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sue Jinks-Robertson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
22
|
Bocci M, Zana A, Principi L, Lucaroni L, Prati L, Gilardoni E, Neri D, Cazzamalli S, Galbiati A. In vivo activation of FAP-cleavable small molecule-drug conjugates for the targeted delivery of camptothecins and tubulin poisons to the tumor microenvironment. J Control Release 2024; 367:779-790. [PMID: 38346501 DOI: 10.1016/j.jconrel.2024.02.014] [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: 12/08/2023] [Revised: 01/25/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
Small molecule-drug conjugates (SMDCs) are increasingly considered as a therapeutic alternative to antibody-drug conjugates (ADCs) for cancer therapy. OncoFAP is an ultra-high affinity ligand of Fibroblast Activation Protein (FAP), a stromal tumor-associated antigen overexpressed in a wide variety of solid human malignancies. We have recently reported the development of non-internalizing OncoFAP-based SMDCs, which are activated by FAP thanks to selective proteolytic cleavage of the -GlyPro- linker with consequent release of monomethyl auristatin E (MMAE) in the tumor microenvironment. In this article, we describe the generation and the in vivo characterization of FAP-cleavable OncoFAP-drug conjugates based on potent topoisomerase I inhibitors (DXd, SN-38, and exatecan) and an anti-tubulin payload (MMAE), which are already exploited in clinical-stage and approved ADCs. The Glycine-Proline FAP-cleavable technology was directly benchmarked against linkers found in Adcetris™, Enhertu™, and Trodelvy™ structures by means of in vivo therapeutic experiments in mice bearing tumors with cellular or stromal FAP expression. OncoFAP-GlyPro-Exatecan and OncoFAP-GlyPro-MMAE emerged as the most efficacious anti-cancer therapeutics against FAP-positive cellular models. OncoFAP-GlyPro-MMAE exhibited a potent antitumor activity also against stromal models, and was therefore selected for clinical development.
Collapse
Affiliation(s)
- Matilde Bocci
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland.
| | - Aureliano Zana
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | | | - Laura Lucaroni
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | - Luca Prati
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | | | - Dario Neri
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences, Zurich CH-8093, Switzerland; Philogen S.p.A., Siena 53100, Italy
| | | | - Andrea Galbiati
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland.
| |
Collapse
|
23
|
Wang Y, Liu Y, Zhang J, Peng Q, Wang X, Xiao X, Shi K. Nanomaterial-mediated modulation of the cGAS-STING signaling pathway for enhanced cancer immunotherapy. Acta Biomater 2024; 176:51-76. [PMID: 38237711 DOI: 10.1016/j.actbio.2024.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/30/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024]
Abstract
Despite the current promise of immunotherapy, many cancer patients still suffer from challenges such as poor immune response rates, resulting in unsatisfactory clinical efficacy of existing therapies. There is an urgent need to combine emerging biomedical discoveries and innovations in traditional therapies. Modulation of the cGAS-STING signalling pathway represents an important innate immunotherapy pathway that serves as a crucial DNA sensing mechanism in innate immunity and viral defense. It has attracted increasing attention as an emerging target for cancer therapy. The recent advancements in nanotechnology have led to the significant utilization of nanomaterials in cancer immunotherapy, owing to their exceptional physicochemical properties such as large specific surface area and efficient permeability. Given the rapid development of cancer immunotherapy driven by the cGAS-STING activation, this study reviews the latest research progress in employing nanomaterials to modulate this signaling pathway. Based on the introduction of the main activation mechanisms of cGAS-STING pathway, this review focuses on nanomaterials that mediate the agonists involved and effectively activate this signaling pathway. In addition, combination nanotherapeutics based on the activation of the cGAS-STING signaling pathway are also discussed, including emerging strategies combining nanoformulated agonists with chemotherapy, radiotherapy as well as other immunomodulation in tumor targeting therapy. STATEMENT OF SIGNIFICANCE: Given the rapid development of cancer immunotherapy driven by the cGAS / STING activation, this study reviews the latest research advances in the use of nanomaterials to modulate this signaling pathway. Based on the introduction of key cGAS-STING components and their activation mechanisms, this review focuses on nanomaterials that can mediate the corresponding agonists and effectively activate this signaling pathway. In addition, combination nanotherapies based on the activation of the cGAS-STING signaling pathway are also discussed, including emerging strategies combining nanoformulated agonists with chemotherapy, radiotherapy as well as immunomodulation in cancer therapy,.
Collapse
Affiliation(s)
- Yaxin Wang
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Yunmeng Liu
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Jincheng Zhang
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Qikai Peng
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Xingdong Wang
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Xiyue Xiao
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Kai Shi
- College of Pharmacy, Nankai University, Tianjin 300350, PR China.
| |
Collapse
|
24
|
Sharma NK, Bahot A, Sekar G, Bansode M, Khunteta K, Sonar PV, Hebale A, Salokhe V, Sinha BK. Understanding Cancer's Defense against Topoisomerase-Active Drugs: A Comprehensive Review. Cancers (Basel) 2024; 16:680. [PMID: 38398072 PMCID: PMC10886629 DOI: 10.3390/cancers16040680] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, the emergence of cancer drug resistance has been one of the crucial tumor hallmarks that are supported by the level of genetic heterogeneity and complexities at cellular levels. Oxidative stress, immune evasion, metabolic reprogramming, overexpression of ABC transporters, and stemness are among the several key contributing molecular and cellular response mechanisms. Topo-active drugs, e.g., doxorubicin and topotecan, are clinically active and are utilized extensively against a wide variety of human tumors and often result in the development of resistance and failure to therapy. Thus, there is an urgent need for an incremental and comprehensive understanding of mechanisms of cancer drug resistance specifically in the context of topo-active drugs. This review delves into the intricate mechanistic aspects of these intracellular and extracellular topo-active drug resistance mechanisms and explores the use of potential combinatorial approaches by utilizing various topo-active drugs and inhibitors of pathways involved in drug resistance. We believe that this review will help guide basic scientists, pre-clinicians, clinicians, and policymakers toward holistic and interdisciplinary strategies that transcend resistance, renewing optimism in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Nilesh Kumar Sharma
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Anjali Bahot
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Gopinath Sekar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Mahima Bansode
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Kratika Khunteta
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Priyanka Vijay Sonar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Ameya Hebale
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Vaishnavi Salokhe
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Birandra Kumar Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
25
|
Singh T, Kim TW, Murthy ASN, Paul M, Sepay N, Jeong Kong H, Sung Ryu J, Rim Koo N, Yoon S, Song KH, Jun Baek M, Jeon S, Im J. Tumor-homing peptide iRGD-conjugate enhances tumor accumulation of camptothecin for colon cancer therapy. Eur J Med Chem 2024; 265:116050. [PMID: 38128233 DOI: 10.1016/j.ejmech.2023.116050] [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: 06/08/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Poor intracellular uptake of therapeutics in the tumor parenchyma is a key issue in cancer therapy. We describe a novel approach to enhance tumor targeting and achieve targeted delivery of camptothecin (CPT) based on a tumor-homing internalizing RGD peptide (iRGD). We synthesized an iRGD-camptothecin conjugate (iRGD-CPT) covalently coupled by a heterobifunctional linker and evaluated its in vitro and in vivo activity in human colon cancer cells. In vitro studies revealed that iRGD-CPT penetrated cells efficiently and reduced colon cancer cell viability to a significantly greater extent at micromolar concentrations than did the parent drug. Furthermore, iRGD-CPT showed high distribution toward tumor tissue, effectively suppressed tumor progression, and showed enhanced antitumor effects relative to the parent drug in a mouse model, demonstrating that iRGD-CPT is effective in vivo cancer treatment. These results suggest that intracellular delivery of CPT via the iRGD peptide is a promising drug delivery strategy that will facilitate the development of CPT derivatives and prodrugs with improved efficacy.
Collapse
Affiliation(s)
- Tejinder Singh
- Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Tae Wan Kim
- Department of Medical Life Science, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Akula S N Murthy
- Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Mohuya Paul
- Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Nasim Sepay
- Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Hye Jeong Kong
- Department of Medical Life Science, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Jae Sung Ryu
- Department of Medical Life Science, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Na Rim Koo
- Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Sujeong Yoon
- Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Keon-Hyoung Song
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Moo Jun Baek
- Department of Surgery, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, 31151, Republic of Korea
| | - Seob Jeon
- Department of Obstetrics and Gynecology, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, 31151, Republic of Korea.
| | - Jungkyun Im
- Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea; Department of Chemical Engineering, Soonchunhyang University, Asan, 31538, Republic of Korea.
| |
Collapse
|
26
|
El-Sayed ASA, ElSayed AI, Wadan KM, El-Saadany SS, Abd El-Hady NAA. Camptothecin bioprocessing from Aspergillus terreus, an endophyte of Catharanthus roseus: antiproliferative activity, topoisomerase inhibition and cell cycle analysis. Microb Cell Fact 2024; 23:15. [PMID: 38183118 PMCID: PMC10768243 DOI: 10.1186/s12934-023-02270-4] [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: 09/13/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
Attenuation of camptothecin (CPT) productivity by fungi with preservation and subculturing is the challenge that halts fungi to be an industrial platform of CPT production. Thus, screening for novel endophytic fungal isolates with metabolic stability for CPT production was the objective. Catharanthus roseus is one of the medicinal plants with diverse bioactive metabolites that could have a plethora of novel endophytes with unique metabolites. Among the endophytes of C. roseus, Aspergillus terreus EFBL-NV OR131583.1 had the most CPT producing potency (90.2 μg/l), the chemical identity of the putative CPT was verified by HPLC, FT-IR, NMR and LC-MS/MS. The putative A. terreus CPT had the same molecular mass (349 m/z), and molecular fragmentation patterns of the authentic one, as revealed from the MS/MS analyses. The purified CPT had a strong activity against MCF7 (5.27 μM) and UO-31 (2.2 μM), with a potential inhibition to Topo II (IC50 value 0.52 nM) than Topo 1 (IC50 value 6.9 nM). The CPT displayed a high wound healing activity to UO-31 cells, stopping their metastasis, matrix formation and cell immigration. The purified CPT had a potential inducing activity to the cellular apoptosis of UO-31 by ~ 17 folds, as well as, arresting their cellular division at the S-phase, compared to the control cells. Upon Plackett-Burman design, the yield of CPT by A. terreus was increased by ~ 2.6 folds, compared to control. The yield of CPT by A. terreus was sequentially suppressed with the fungal storage and subculturing, losing ~ 50% of their CPT productivity by 3rd month and 5th generation. However, the productivity of the attenuated A. terreus culture was completely restored by adding 1% surface sterilized leaves of C. roseus, and the CPT yield was increased over-the-first culture by ~ 3.2 folds (315.2 μg/l). The restoring of CPT productivity of A. terreus in response to indigenous microbiome of C. roseus, ensures the A. terreus-microbiome interactions, releasing a chemical signal that triggers the CPT productivity of A. terreus. This is the first reports exploring the potency of A. terreus, endophyte of C. roseus" to be a platform for industrial production of CPT, with an affordable sustainability with addition of C. roseus microbiome.
Collapse
Affiliation(s)
- Ashraf S A El-Sayed
- Enzymology and Fungal Biotechnology Lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt.
| | - Abdelaleim I ElSayed
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Khalid M Wadan
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Sayed S El-Saadany
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Nouran A A Abd El-Hady
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
27
|
Cui Q, Huang C, Liu JY, Zhang JT. Small Molecule Inhibitors Targeting the "Undruggable" Survivin: The Past, Present, and Future from a Medicinal Chemist's Perspective. J Med Chem 2023; 66:16515-16545. [PMID: 38092421 PMCID: PMC11588358 DOI: 10.1021/acs.jmedchem.3c01130] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Survivin, a homodimeric protein and a member of the IAP family, plays a vital function in cell survival and cycle progression by interacting with various proteins and complexes. Its expression is upregulated in cancers but not detectable in normal tissues. Thus, it has been regarded and validated as an ideal cancer target. However, survivin is "undruggable" due to its lack of enzymatic activities or active sites for small molecules to bind/inhibit. Academic and industrial laboratories have explored different strategies to overcome this hurdle over the past two decades, with some compounds advanced into clinical testing. These strategies include inhibiting survivin expression, its interaction with binding partners and homodimerization. Here, we provide comprehensive analyses of these strategies and perspective on different small molecule survivin inhibitors to help drug discovery targeting "undruggable" proteins in general and survivin specifically with a true survivin inhibitor that will prevail in the foreseeable future.
Collapse
Affiliation(s)
- Qingbin Cui
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Caoqinglong Huang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jing-Yuan Liu
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| |
Collapse
|
28
|
Lica JJ, Heldt M, Wieczór M, Chodnicki P, Ptaszyńska N, Maciejewska N, Łęgowska A, Brankiewicz W, Gucwa K, Stupak A, Pradhan B, Gitlin-Domagalska A, Dębowski D, Milewski S, Bieniaszewska M, Grabe GJ, Hellmann A, Rolka K. Dual-Activity Fluoroquinolone-Transportan 10 Conjugates Offer Alternative Leukemia Therapy during Hematopoietic Cell Transplantation. Mol Pharmacol 2023; 105:39-53. [PMID: 37977824 DOI: 10.1124/molpharm.123.000735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/01/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023] Open
Abstract
Hematopoietic cell transplantation (HCT) is often considered a last resort leukemia treatment, fraught with limited success due to microbial infections, a leading cause of mortality in leukemia patients. To address this critical issue, we explored a novel approach by synthesizing antileukemic agents containing antibacterial substances. This innovative strategy involves conjugating fluoroquinolone antibiotics, such as ciprofloxacin (CIP) or levofloxacin (LVX), with the cell-penetrating peptide transportan 10 (TP10). Here, we demonstrate that the resultant compounds display promising biologic activities in preclinical studies. These novel conjugates not only exhibit potent antimicrobial effects but are also selective against leukemia cells. The cytotoxic mechanism involves rapid disruption of cell membrane asymmetry leading to membrane damage. Importantly, these conjugates penetrated mammalian cells, accumulating within the nuclear membrane without significant effect on cellular architecture or mitochondrial function. Molecular simulations elucidated the aggregation tendencies of TP10 conjugates within lipid bilayers, resulting in membrane disruption and permeabilization. Moreover, mass spectrometry analysis confirmed efficient reduction of disulfide bonds within TP10 conjugates, facilitating release and activation of the fluoroquinolone derivatives. Intriguingly, these compounds inhibited human topoisomerases, setting them apart from traditional fluoroquinolones. Remarkably, TP10 conjugates generated lower intracellular levels of reactive oxygen species compared with CIP and LVX. The combination of antibacterial and antileukemic properties, coupled with selective cytostatic effects and minimal toxicity toward healthy cells, positions TP10 derivatives as promising candidates for innovative therapeutic approaches in the context of antileukemic HCT. This study highlights their potential in search of more effective leukemia treatments. SIGNIFICANCE STATEMENT: Fluoroquinolones are commonly used antibiotics, while transportan 10 (TP10) is a cell-penetrating peptide (CPP) with anticancer properties. In HCT, microbial infections are the primary cause of illness and death. Combining TP10 with fluoroquinolones enhanced their effects on different cell types. The dual pharmacological action of these conjugates offers a promising proof-of-concept solution for leukemic patients undergoing HCT. Strategically designed therapeutics, incorporating CPPs with antibacterial properties, have the potential to reduce microbial infections in the treatment of malignancies.
Collapse
Affiliation(s)
- Jan Jakub Lica
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Mateusz Heldt
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Milosz Wieczór
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Pawel Chodnicki
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Natalia Ptaszyńska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Natalia Maciejewska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Anna Łęgowska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Wioletta Brankiewicz
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Katarzyna Gucwa
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Anna Stupak
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Bhaskar Pradhan
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Agata Gitlin-Domagalska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Dawid Dębowski
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Sławomir Milewski
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Maria Bieniaszewska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Grzegorz Jan Grabe
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Andrzej Hellmann
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Krzysztof Rolka
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| |
Collapse
|
29
|
Bednarek E, Bocian W, Sitkowski J, Urbanowicz M, Kozerski L. New 5-Substituted SN38 Derivatives: A Stability Study and Interaction with Model Nicked DNA by NMR and Molecular Modeling Methods. Int J Mol Sci 2023; 24:17445. [PMID: 38139274 PMCID: PMC10743537 DOI: 10.3390/ijms242417445] [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: 10/26/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The new 5-substituted SN-38 derivatives, 5(R)-(N-pyrrolidinyl)methyl-7-ethyl-10-hydroxycamptothecin (1) and its diastereomer 5(S) (2), were investigated using a combination of nuclear magnetic resonance (NMR) spectroscopy and molecular modeling methods. The chemical stability, configuration stability, and propensity to aggregate as a function of concentration were determined using 1H NMR. The calculated self-association constants (Ka) were found to be 6.4 mM-1 and 2.9 mM-1 for 1 and 2, respectively. The NMR experiments were performed to elucidate the interaction of each diastereomer with a nicked decamer duplex, referred to as 3. The calculated binding constants were determined to be 76 mM-1 and 150 mM-1 for the 1-3 and 2-3 complexes, respectively. NMR studies revealed that the interaction between 1 or 2 and the nicked decamer duplex occurred at the site of the DNA strand break. To complement these findings, molecular modeling methods and calculation protocols were employed to establish the interaction mode and binding constants and to generate molecular models of the DNA/ligand complexes.
Collapse
Affiliation(s)
- Elżbieta Bednarek
- National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland; (W.B.); (J.S.); (M.U.); (L.K.)
| | | | | | | | | |
Collapse
|
30
|
Rady AM, El-Sayed ASA, El-Baz AF, Abdel-Fattah GG, Magdeldin S, Ahmed E, Osama A, Hassanein SE, Saed H, Yassin M. Proteomics and metabolomics analyses of camptothecin-producing Aspergillus terreus reveal the integration of PH domain-containing proteins and peptidylprolyl cis/trans isomerase in restoring the camptothecin biosynthesis. Microbiol Spectr 2023; 11:e0228123. [PMID: 37855596 PMCID: PMC10714794 DOI: 10.1128/spectrum.02281-23] [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: 05/31/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE Decreasing the camptothecin productivity by fungi with storage and subculturing is the challenge that halts their further implementation to be an industrial platform for camptothecin (CPT) production. The highest differentially abundant proteins were Pleckstrin homology (PH) domain-containing proteins and Peptidyl-prolyl cis/trans isomerase that fluctuated with the subculturing of A. terreus with a remarkable relation to CPT biosynthesis and restored with addition of F. elastica microbiome.
Collapse
Affiliation(s)
- Amgad M. Rady
- Enzymology and Fungal Biotechnology Lab (EFBL), Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
| | - Ashraf S. A. El-Sayed
- Enzymology and Fungal Biotechnology Lab (EFBL), Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Ashraf F. El-Baz
- Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | | | - Sameh Magdeldin
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children’s Cancer Hospital, Cairo, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Eman Ahmed
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children’s Cancer Hospital, Cairo, Egypt
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Aya Osama
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children’s Cancer Hospital, Cairo, Egypt
| | - Sameh E. Hassanein
- Agricultural Genetic Engineering Research Institute (AGERI), Agriculture Research Center, Cairo, Egypt
| | - Hend Saed
- Microbiology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Marwa Yassin
- Enzymology and Fungal Biotechnology Lab (EFBL), Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
31
|
Yu C, Huang F, Wang K, Liu M, Chow WA, Ling X, Li F, Causey JL, Huang X, Cook-Wiens G, Cui X. Single protein encapsulated SN38 for tumor-targeting treatment. J Transl Med 2023; 21:897. [PMID: 38072965 PMCID: PMC10712105 DOI: 10.1186/s12967-023-04778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The alkaloid camptothecin analog SN38 is a potent antineoplastic agent, but cannot be used directly for clinical application due to its poor water solubility. Currently, the prodrug approach on SN38 has resulted in 3 FDA-approved cancer therapeutics, irinotecan, ONIVYDE, and Trodelvy. However, only 2-8% of irinotecan can be transformed enzymatically in vivo into the active metabolite SN38, which severely limits the drug's efficacy. While numerous drug delivery systems have been attempted to achieve effective SN38 delivery, none have produced drug products with antitumor efficacy better than irinotecan in clinical trials. Therefore, novel approaches are urgently needed for effectively delivering SN38 to cancer cells with better efficacy and lower toxicity. METHODS Based on the unique properties of human serum albumin (HSA), we have developed a novel single protein encapsulation (SPE) technology to formulate cancer therapeutics for improving their pharmacokinetics (PK) and antitumor efficacy and reducing their side effects. Previous application of SPE technology to doxorubicin (DOX) formulation has led to a promising drug candidate SPEDOX-6 (FDA IND #, 152154), which will undergo a human phase I clinical trial. Using the same SPE platform on SN38, we have now produced two SPESN38 complexes, SPESN38-5 and SPESN38-8. We conducted their pharmacological evaluations with respect to maximum tolerated dose, PK, and in vivo efficacy against colorectal cancer (CRC) and soft tissue sarcoma (STS) in mouse models. RESULTS The lyophilized SPESN38 complexes can dissolve in aqueous media to form clear and stable solutions. Maximum tolerated dose (MTD) of SPESN38-5 is 250 mg/kg by oral route (PO) and 55 mg/kg by intravenous route (IV) in CD-1 mice. SPESN38-8 has the MTD of 45 mg/kg by IV in the same mouse model. PK of SPESN38-5 by PO at 250 mg/kg gave mouse plasma AUC0-∞ of 0.05 and 4.5 nmol × h/mL for SN38 and SN38 glucuronidate (SN38G), respectively, with a surprisingly high molar ratio of SN38G:SN38 = 90:1. However, PK of SPESN38-5 by IV at 55 mg/kg yielded much higher mouse plasma AUC0-∞ of 19 and 28 nmol × h/mL for SN38 and SN38G, producing a much lower molar ratio of SN38G:SN38 = 1.5:1. Antitumor efficacy of SPESN38-5 and irinotecan (control) was evaluated against HCT-116 CRC xenograft tumors. The data indicates that SPESN38-5 by IV at 55 mg/kg is more effective in suppressing HCT-116 tumor growth with lower systemic toxicity compared to irinotecan at 50 mg/kg. Additionally, SPESN38-8 and DOX (control) by IV were evaluated in the SK-LMS-1 STS mouse model. The results show that SPESN38-8 at 33 mg/kg is highly effective for inhibiting SK-LMS-1 tumor growth with low toxicity, in contrast to DOX's insensitivity to SK-LMS-1 with high toxicity. CONCLUSION SPESN38 complexes provide a water soluble SN38 formulation. SPESN38-5 and SPESN38-8 demonstrate better PK values, lower toxicity, and superior antitumor efficacy in mouse models, compared with irinotecan and DOX.
Collapse
Affiliation(s)
- Changjun Yu
- Department of Chemistry, California Institute of Technology, Pasadena, CA, 91125, USA.
- Sunstate Biosciences, LLC, 870 S. Myrtle Ave, Monrovia, CA, 91016, USA.
| | - Faqing Huang
- Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, MS, 39406, USA.
| | - Kinsley Wang
- Sunstate Biosciences, LLC, 870 S. Myrtle Ave, Monrovia, CA, 91016, USA
| | - Mengmeng Liu
- Sunstate Biosciences, LLC, 870 S. Myrtle Ave, Monrovia, CA, 91016, USA
| | - Warren A Chow
- Division of Hematology/Oncology, Department of Medicine, UCI Health, Orange, CA, 92868, USA
| | - Xiang Ling
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
- Canget BioTekpharma, LLC, 701 Ellicott Street, Buffalo, NY, 14203, USA
| | - Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Jason L Causey
- Department of Computer Sciences, Arkansas State University, Jonesboro, AR, 72467, USA
| | - Xiuzhen Huang
- Department of Computational Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Galen Cook-Wiens
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Xiaojiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
32
|
Gasser SM, Stutz F. SUMO in the regulation of DNA repair and transcription at nuclear pores. FEBS Lett 2023; 597:2833-2850. [PMID: 37805446 DOI: 10.1002/1873-3468.14751] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 10/09/2023]
Abstract
Two related post-translational modifications, the covalent linkage of Ubiquitin and the Small Ubiquitin-related MOdifier (SUMO) to lysine residues, play key roles in the regulation of both DNA repair pathway choice and transcription. Whereas ubiquitination is generally associated with proteasome-mediated protein degradation, the impact of sumoylation has been more mysterious. In the cell nucleus, sumoylation effects are largely mediated by the relocalization of the modified targets, particularly in response to DNA damage. This is governed in part by the concentration of SUMO protease at nuclear pores [Melchior, F et al. (2003) Trends Biochem Sci 28, 612-618; Ptak, C and Wozniak, RW (2017) Adv Exp Med Biol 963, 111-126]. We review here the roles of sumoylation in determining genomic locus positioning relative to the nuclear envelope and to nuclear pores, to facilitate repair and regulate transcription.
Collapse
Affiliation(s)
- Susan M Gasser
- Department of Fundamental Microbiology, University of Lausanne, Switzerland
- ISREC Foundation, Agora Cancer Research Center, Lausanne, Switzerland
| | - Françoise Stutz
- Department of Molecular and Cellular Biology, University of Geneva, Switzerland
| |
Collapse
|
33
|
Zakharenko AL, Dyrkheeva NS, Luzina OA, Filimonov AS, Mozhaitsev ES, Malakhova AA, Medvedev SP, Zakian SM, Salakhutdinov NF, Lavrik OI. Usnic Acid Derivatives Inhibit DNA Repair Enzymes Tyrosyl-DNA Phosphodiesterases 1 and 2 and Act as Potential Anticancer Agents. Genes (Basel) 2023; 14:1931. [PMID: 37895279 PMCID: PMC10606488 DOI: 10.3390/genes14101931] [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: 08/03/2023] [Revised: 09/04/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Tyrosyl-DNA phosphodiesterase 1 and 2 (Tdp1 and Tdp2) are DNA repair enzymes that repair DNA damage caused by various agents, including anticancer drugs. Thus, these enzymes resist anticancer therapy and could be the reason for resistance to such widely used drugs such as topotecan and etoposide. In the present work, we found compounds capable of inhibiting both enzymes among derivatives of (-)-usnic acid. Both (+)- and (-)-enantiomers of compounds act equally effectively against Tdp1 with IC50 values in the range of 0.02-0.2 μM; only (-)-enantiomers inhibited Tdp2 with IC50 values in the range of 6-9 μM. Surprisingly, the compounds protect HEK293FT wild type cells from the cytotoxic effect of etoposide (CC50 3.0-3.9 μM in the presence of compounds and 2.4 μM the presence of DMSO) but potentiate it against Tdp2 knockout cells (CC50 1.2-1.6 μM in the presence of compounds against 2.3 μM in the presence of DMSO). We assume that the sensitizing effect of the compounds in the absence of Tdp2 is associated with the effective inhibition of Tdp1, which could take over the functions of Tdp2.
Collapse
Affiliation(s)
- Alexandra L. Zakharenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (N.S.D.); (O.I.L.)
| | - Nadezhda S. Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (N.S.D.); (O.I.L.)
| | - Olga A. Luzina
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 9 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (O.A.L.); (A.S.F.); (E.S.M.); (N.F.S.)
| | - Aleksandr S. Filimonov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 9 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (O.A.L.); (A.S.F.); (E.S.M.); (N.F.S.)
| | - Evgenii S. Mozhaitsev
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 9 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (O.A.L.); (A.S.F.); (E.S.M.); (N.F.S.)
| | - Anastasia A. Malakhova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (A.A.M.); (S.P.M.); (S.M.Z.)
| | - Sergey P. Medvedev
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (A.A.M.); (S.P.M.); (S.M.Z.)
| | - Suren M. Zakian
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (A.A.M.); (S.P.M.); (S.M.Z.)
| | - Nariman F. Salakhutdinov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 9 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (O.A.L.); (A.S.F.); (E.S.M.); (N.F.S.)
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Akademika Lavrentieva Ave., 630090 Novosibirsk, Russia; (N.S.D.); (O.I.L.)
| |
Collapse
|
34
|
Zhang M, Zhu LZ, Yang CJ, Yan JX, Wang ZP, Bai YP, Peng LZ, Luo HB, Zhang ZJ, Li L, Xu CR, Liu YQ. Improved anti-tumor activity of fluorinated camptothecin derivatives 9-fluorocamptothecin and 7-ethyl-9-fluorocamptothecin on hepatocellular carcinoma by targeting topoisomerase I. Bioorg Chem 2023; 139:106652. [PMID: 37390632 DOI: 10.1016/j.bioorg.2023.106652] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/25/2023] [Accepted: 06/01/2023] [Indexed: 07/02/2023]
Abstract
Primary liver cancer is one of the most common malignant cancers of the digestive system that lacks effective chemotherapeutic drugs in clinical settings. Camptothecin (CPT) and its derivatives have been approved for cancer treatment; however, their application is limited by their systemic toxicity. For lead optimization in new drug discovery stages, fluorination is an effective and robust approach to increase the bioavailability and optimize the pharmacokinetics of candidate compounds, thereby improving their efficacy. To obtain new and highly active CPT derivatives, we designed, synthesized, and evaluated two new fluorinated CPT derivatives, 9-fluorocamptothecin (A1) and 7-ethyl-9-fluorocamptothecin (A2), in this study. In vitro, A1 and A2 exhibited more robust anti-tumor activity than topotecan (TPT) in various cancer cells, particularly hepatocellular carcinoma (HCC) cells. In vivo, A1 and A2 exhibited greater anti-tumor activity than TPT in both AKT/Met induced primary HCC mouse models and implanted HepG2 cell xenografts. Acute toxicity tests revealed that A1 and A2 were not lethal and did not cause significant body weight loss at high doses. Moreover, A1 and A2 exhibited no significant toxicity in the mouse liver, heart, lung, spleen, kidney, and hematopoietic systems at therapeutic doses. Mechanistically, A1 and A2 blocked HCC cell proliferation by inhibiting the enzymatic activity of Topo I, subsequently inducing DNA damage, cell cycle arrest, and apoptosis. In summary, our results indicate that fluorination improves the anti-tumor activity of CPT while decreasing its toxicity and highlight the application potential of fluorination products A1 and A2 in clinical settings.
Collapse
Affiliation(s)
- Mi Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Li-Zu Zhu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Cheng-Jie Yang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Jia-Xuan Yan
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Zhi-Ping Wang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, PR China
| | - Yin-Peng Bai
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Li-Zeng Peng
- Jinan AISI Pharmaceutical Technology Co Ltd, Jinan 250104, PR China
| | - Hong-Bo Luo
- Department of Urology, The Second Hospital of Huangshi, Huangshi, Hubei 435000, PR China
| | - Zhi-Jun Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China
| | - Lei Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| | - Chuan-Rui Xu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| | - Ying-Qian Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
35
|
Wang Z, Li H, Gou L, Li W, Wang Y. Antibody-drug conjugates: Recent advances in payloads. Acta Pharm Sin B 2023; 13:4025-4059. [PMID: 37799390 PMCID: PMC10547921 DOI: 10.1016/j.apsb.2023.06.015] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/30/2023] [Accepted: 06/23/2023] [Indexed: 10/05/2023] Open
Abstract
Antibody‒drug conjugates (ADCs), which combine the advantages of monoclonal antibodies with precise targeting and payloads with efficient killing, show great clinical therapeutic value. The ADCs' payloads play a key role in determining the efficacy of ADC drugs and thus have attracted great attention in the field. An ideal ADC payload should possess sufficient toxicity, low immunogenicity, high stability, and modifiable functional groups. Common ADC payloads include tubulin inhibitors and DNA damaging agents, with tubulin inhibitors accounting for more than half of the ADC drugs in clinical development. However, due to clinical limitations of traditional ADC payloads, such as inadequate efficacy and the development of acquired drug resistance, novel highly efficient payloads with diverse targets and reduced side effects are being developed. This perspective summarizes the recent research advances of traditional and novel ADC payloads with main focuses on the structure-activity relationship studies, co-crystal structures, and designing strategies, and further discusses the future research directions of ADC payloads. This review also aims to provide valuable references and future directions for the development of novel ADC payloads that will have high efficacy, low toxicity, adequate stability, and abilities to overcome drug resistance.
Collapse
Affiliation(s)
- Zhijia Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| | - Hanxuan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lantu Gou
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| |
Collapse
|
36
|
Eldeghidy A, Abdel-Fattah G, El-Sayed ASA, Abdel-Fattah GG. Production, bioprocessing and antiproliferative activity of camptothecin from Aspergillus terreus, endophyte of Cinnamomum camphora: restoring their biosynthesis by indigenous microbiome of C. camphora. Microb Cell Fact 2023; 22:143. [PMID: 37533061 PMCID: PMC10399021 DOI: 10.1186/s12934-023-02158-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023] Open
Abstract
Fungal producing potency of camptothecin (CPT) raise the hope for their usage to be a platform for industrial production of CPT, nevertheless, attenuation of their productivity of CPT with the subculturing and preservation is the challenge. So, screening for novel endophytic fungal isolates with a reliable CPT-biosynthetic stability was the objective. Among the isolated endophytic fungi from the tested medicinal plants, Aspergillus terreus OQ642314.1, endophyte of Cinnamomum camphora, exhibits the highest yield of CPT (89.4 μg/l). From the NMR, FT-IR and LC-MS/MS analyses, the extracted CPT from A. terreus gave the same structure and molecular mass fragmentation pattern of authentic CPT (349 m/z). The putative CPT had a significant activity against MCF7 (0.27 µM) and HEPG-2 (0.8 µM), with a strong affinity to inhibits the human Topoisomerase 1 activity (IC50 0.362 μg/ml) as revealed from the Gel-based DNA relaxation assay. The purified CPT displayed a strong antimicrobial activity for various bacterial (E. coli and B. cereus) and fungal (A. flavus and A. parasiticus) isolates, ensuring the unique tertiary, and stereo-structure of A. terreus for penetrating the microbial cell walls and targeting the topoisomerase I. The higher dual activity of the purified CPT as antimicrobial and antitumor, emphasize their therapeutic efficiency, especially with growth of the opportunistic microorganisms due to the suppression of human immune system with the CPT uses in vivo. The putative CPT had an obvious activity against the tumor cell (MCF7) metastasis, and migration as revealed from the wound healing assay. The overall yield of A. terreus CPT was maximized with the Blackett-Burman design by twofolds increment (164.8 μg/l). The CPT yield by A. terreus was successively diminished with the multiple fungal subculturing, otherwise, the CPT productivity of A. terreus was restored, and increased over the zero culture upon coculturing with C. camphora microbiome (1.5% w/v), ensuring the restoring of CPT biosynthetic potency of A. terreus by the plant microbiome-derived chemical signals "microbial communication". This is the first report exploring the feasibility of A. terreus "endophyte of C. camphora" to be a preliminary platform for commercial production of CPT with a reliable sustainability upon uses of indigenous C. camphora microbiome.
Collapse
Affiliation(s)
- Abeer Eldeghidy
- Botany and Microbiology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Gamal Abdel-Fattah
- Botany and Microbiology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Ashraf S A El-Sayed
- Enzymology and Fungal Biotechnology Lab, Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt.
| | - Ghada G Abdel-Fattah
- Botany and Microbiology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
37
|
Yu CJ, Huang F, Wang K, Liu M, Chow WA, Ling X, Li F, Causey JL, Huang X, Cook-Wiens G, Cui X. Single Protein Encapsulated SN38 for Tumor-Targeting Treatment. RESEARCH SQUARE 2023:rs.3.rs-3154635. [PMID: 37546894 PMCID: PMC10402254 DOI: 10.21203/rs.3.rs-3154635/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Background The alkaloid camptothecin analog SN38 is a potent antineoplastic agent, but cannot be used directly for clinical application due to its poor water solubility. Currently, the prodrug approach on SN38 has resulted in 3 FDA-approved cancer therapeutics, irinotecan, ONIVYDE, and Trodelvy. However, only 2-8% of irinotecan can be transformed enzymatically in vivo into the active metabolite SN38, which severely limits the drug's efficacy. While numerous drug delivery systems have been attempted to achieve effective SN38 delivery, none have produced drug products with antitumor efficacy better than irinotecan in clinical trials. Therefore, novel approaches are urgently needed for effectively delivering SN38 to cancer cells with better efficacy and lower toxicity. Methods Based on the unique properties of human serum albumin (HSA), we have developed a novel single protein encapsulation (SPE) technology to formulate cancer therapeutics for improving their pharmacokinetics (PK) and antitumor efficacy and reducing their side effects. Previous application of SPE technology to doxorubicin (DOX) formulation has led to a promising drug candidate SPEDOX-6 (FDA IND #, 152154), which will undergo a human phase I clinical trial. Using the same SPE platform on SN38, we have now produced two SPESN38 complexes, SPESN38-5 and SPESN38-8. We conducted their pharmacological evaluations with respect to maximum tolerated dose, PK, and in vivo efficacy against colorectal cancer (CRC) and soft tissue sarcoma (STS) in mouse models. Results The lyophilized SPESN38 complexes can dissolve in aqueous media to form clear and stable solutions. Maximum tolerated dose (MTD) of SPESN38-5 is 250 mg/kg by oral route (PO) and 55 mg/kg by intravenous route (IV) in CD-1 mice. SPESN38-8 has the MTD of 45 mg/kg by IV in the same mouse model. PK of SPESN38-5 by PO at 250 mg/kg gave mouse plasma AUC0-∞ of 0.0548 and 4.5007 (nmol × h/mL) for SN38 and SN38 glucuronidate (SN38G), respectively, with a surprisingly high molar ratio of SN38G:SN38 = 82:1. However, PK of SPESN38-5 by IV at 55 mg/kg yielded much higher mouse plasma AUC0-∞ of 18.80 and 27.78 nmol × h/mL for SN38 and SN38G, producing a much lower molar ratio of SN38G:SN38 = 1.48:1. Antitumor efficacy of SPESN38-5 and irinotecan (control) was evaluated against HCT-116 CRC xenograft tumors. The data indicates that SPESN38-5 by IV at 55 mg/kg is more effective in suppressing HCT-116 tumor growth with lower systemic toxicity compared to irinotecan at 50 mg/kg. Additionally, SPESN38-8 and DOX (control) by IV were evaluated in the SK-LMS-1 STS mouse model. The results show that SPESN38-8 at 33 mg/kg is highly effective for inhibiting SK-LMS-1 tumor growth with low toxicity, in contrast to DOX's insensitivity to SK-LMS-1 with high toxicity. Conclusion SPESN38 complexes provide a water soluble SN38 formulation. SPESN38-5 and SPESN38-8 demonstrate better PK values, lower toxicity, and superior antitumor efficacy in mouse models, compared with irinotecan and DOX.
Collapse
Affiliation(s)
| | - Faqing Huang
- University of Southern Mississippi Center For Tobacco Prevention and Health Promotion: University of Southern Mississippi
| | | | | | - Warren A Chow
- University of California Irvine Department of Medicine
| | - Xiang Ling
- Roswell Park Comprehensive Cancer Center
| | - Fengzhi Li
- Roswell Park Comprehensive Cancer Center
| | | | | | | | | |
Collapse
|
38
|
El-Kalyoubi S, Elbaramawi SS, Eissa AG, Al-Ageeli E, Hobani YH, El-Sharkawy AA, Mohamed HT, Al-Karmalawy AA, Abulkhair HS. Design and synthesis of novel uracil-linked Schiff bases as dual histone deacetylase type II/topoisomerase type I inhibitors with apoptotic potential. Future Med Chem 2023; 15:937-958. [PMID: 37381751 DOI: 10.4155/fmc-2023-0112] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023] Open
Abstract
Aim: The previously reported dual histone deacetylase type II (HDAC II) / topoisomerase type I (Topo I) inhibitors suffer pharmacokinetic limitations because of their huge molecular weights. Materials & methods: We report the design and synthesis of a smarter novel set of uracil-linked Schiff bases (19-30) as dual HDAC II/Topo I inhibitors keeping the essential pharmacophoric features. Cytotoxicity of all compounds was assessed against three cancer cell lines. Studies of their effects on the apoptotic BAX and antiapoptotic BCL2 genes, molecular docking studies, and absorption, distribution, metabolism and excretion studies were conducted. Results: Compounds 22, 25 and 30 exhibited significant activities. The bromophenyl derivative 22 displayed the best selectivity index, with IC50 values against HDAC II and Topo I of 1.12 and 13.44 μM, respectively. Conclusion: Compound 22 could be considered a lead HDAC II/Topo I inhibitor.
Collapse
Affiliation(s)
- Samar El-Kalyoubi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Port Said University, Port Said, 42511, Egypt
| | - Samar S Elbaramawi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Ahmed G Eissa
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Essam Al-Ageeli
- Department of Clinical Biochemistry (Medical Genetics), Faculty of Medicine, Jazan University, Jazan, 82621, Saudi Arabia
| | - Yahya Hasan Hobani
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Jazan University, Jazan, 82621, Saudi Arabia
| | - Aya Ali El-Sharkawy
- Zoology Department, Faculty of Science, Cairo University, Cairo, 12613, Egypt
| | - Hossam Taha Mohamed
- Zoology Department, Faculty of Science, Cairo University, Cairo, 12613, Egypt
- Faculty of Biotechnology, October University for Modern Sciences & Arts, Giza, 12451, Egypt
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12566, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, International Coastal Road, New Damietta, 34518, Egypt
| |
Collapse
|
39
|
Marini V, Nikulenkov F, Samadder P, Juul S, Knudsen BR, Krejci L. MUS81 cleaves TOP1-derived lesions and other DNA-protein cross-links. BMC Biol 2023; 21:110. [PMID: 37194054 PMCID: PMC10189953 DOI: 10.1186/s12915-023-01614-1] [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: 09/16/2022] [Accepted: 05/04/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND DNA-protein cross-links (DPCs) are one of the most deleterious DNA lesions, originating from various sources, including enzymatic activity. For instance, topoisomerases, which play a fundamental role in DNA metabolic processes such as replication and transcription, can be trapped and remain covalently bound to DNA in the presence of poisons or nearby DNA damage. Given the complexity of individual DPCs, numerous repair pathways have been described. The protein tyrosyl-DNA phosphodiesterase 1 (Tdp1) has been demonstrated to be responsible for removing topoisomerase 1 (Top1). Nevertheless, studies in budding yeast have indicated that alternative pathways involving Mus81, a structure-specific DNA endonuclease, could also remove Top1 and other DPCs. RESULTS This study shows that MUS81 can efficiently cleave various DNA substrates modified by fluorescein, streptavidin or proteolytically processed topoisomerase. Furthermore, the inability of MUS81 to cleave substrates bearing native TOP1 suggests that TOP1 must be either dislodged or partially degraded prior to MUS81 cleavage. We demonstrated that MUS81 could cleave a model DPC in nuclear extracts and that depletion of TDP1 in MUS81-KO cells induces sensitivity to the TOP1 poison camptothecin (CPT) and affects cell proliferation. This sensitivity is only partially suppressed by TOP1 depletion, indicating that other DPCs might require the MUS81 activity for cell proliferation. CONCLUSIONS Our data indicate that MUS81 and TDP1 play independent roles in the repair of CPT-induced lesions, thus representing new therapeutic targets for cancer cell sensitisation in combination with TOP1 inhibitors.
Collapse
Affiliation(s)
- Victoria Marini
- Department of Biology, Masaryk University, Kamenice 5/B07, Brno, 62500, Czech Republic
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital Brno, Pekařská 53, Brno, 60200, Czech Republic
| | - Fedor Nikulenkov
- Department of Biology, Masaryk University, Kamenice 5/B07, Brno, 62500, Czech Republic
| | - Pounami Samadder
- Department of Biology, Masaryk University, Kamenice 5/B07, Brno, 62500, Czech Republic
| | - Sissel Juul
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, Aarhus, 8000, Denmark
| | - Birgitta R Knudsen
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, Aarhus, 8000, Denmark
| | - Lumir Krejci
- Department of Biology, Masaryk University, Kamenice 5/B07, Brno, 62500, Czech Republic.
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital Brno, Pekařská 53, Brno, 60200, Czech Republic.
- National Centre for Biomolecular Research, Masaryk University, Kamenice 5/C04, Brno, 625 00, Czech Republic.
| |
Collapse
|
40
|
Noireterre A, Serbyn N, Bagdiul I, Stutz F. Ubx5-Cdc48 assists the protease Wss1 at DNA-protein crosslink sites in yeast. EMBO J 2023:e113609. [PMID: 37144685 DOI: 10.15252/embj.2023113609] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023] Open
Abstract
DNA-protein crosslinks (DPCs) pose a serious threat to genome stability. The yeast proteases Wss1, 26S proteasome, and Ddi1 are safeguards of genome integrity by acting on a plethora of DNA-bound proteins in different cellular contexts. The AAA ATPase Cdc48/p97 is known to assist Wss1/SPRTN in clearing DNA-bound complexes; however, its contribution to DPC proteolysis remains unclear. Here, we show that the Cdc48 adaptor Ubx5 is detrimental in yeast mutants defective in DPC processing. Using an inducible site-specific crosslink, we show that Ubx5 accumulates at persistent DPC lesions in the absence of Wss1, which prevents their efficient removal from the DNA. Abolishing Cdc48 binding or complete loss of Ubx5 suppresses sensitivity of wss1∆ cells to DPC-inducing agents by favoring alternate repair pathways. We provide evidence for cooperation of Ubx5-Cdc48 and Wss1 in the genotoxin-induced degradation of RNA polymerase II (RNAPII), a described candidate substrate of Wss1. We propose that Ubx5-Cdc48 assists Wss1 for proteolysis of a subset of DNA-bound proteins. Together, our findings reveal a central role for Ubx5 in DPC clearance and repair.
Collapse
Affiliation(s)
- Audrey Noireterre
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Nataliia Serbyn
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Ivona Bagdiul
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Françoise Stutz
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
41
|
Keller JG, Petersen KV, Mizielinski K, Thiesen C, Bjergbæk L, Reguera RM, Pérez-Pertejo Y, Balaña-Fouce R, Trejo A, Masdeu C, Alonso C, Knudsen BR, Tesauro C. Gel-Free Tools for Quick and Simple Screening of Anti-Topoisomerase 1 Compounds. Pharmaceuticals (Basel) 2023; 16:ph16050657. [PMID: 37242440 DOI: 10.3390/ph16050657] [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: 02/21/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
With the increasing need for effective compounds against cancer or pathogen-borne diseases, the development of new tools to investigate the enzymatic activity of biomarkers is necessary. Among these biomarkers are DNA topoisomerases, which are key enzymes that modify DNA and regulate DNA topology during cellular processes. Over the years, libraries of natural and synthetic small-molecule compounds have been extensively investigated as potential anti-cancer, anti-bacterial, or anti-parasitic drugs targeting topoisomerases. However, the current tools for measuring the potential inhibition of topoisomerase activity are time consuming and not easily adaptable outside specialized laboratories. Here, we present rolling circle amplification-based methods that provide fast and easy readouts for screening of compounds against type 1 topoisomerases. Specific assays for the investigation of the potential inhibition of eukaryotic, viral, or bacterial type 1 topoisomerase activity were developed, using human topoisomerase 1, Leishmania donovani topoisomerase 1, monkeypox virus topoisomerase 1, and Mycobacterium smegmatis topoisomerase 1 as model enzymes. The presented tools proved to be sensitive and directly quantitative, paving the way for new diagnostic and drug screening protocols in research and clinical settings.
Collapse
Affiliation(s)
| | | | | | - Celine Thiesen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Lotte Bjergbæk
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Rosa M Reguera
- Department of Biomedical Sciences, Faculty of Veterinary Medicine, University of León, 24071 León, Spain
| | - Yolanda Pérez-Pertejo
- Department of Biomedical Sciences, Faculty of Veterinary Medicine, University of León, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Department of Biomedical Sciences, Faculty of Veterinary Medicine, University of León, 24071 León, Spain
| | - Angela Trejo
- Department of Organic Chemistry, Faculty of Pharmacy, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
| | - Carme Masdeu
- Department of Organic Chemistry, Faculty of Pharmacy, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
| | - Concepcion Alonso
- Department of Organic Chemistry, Faculty of Pharmacy, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
| | - Birgitta R Knudsen
- VPCIR Biosciences ApS, 8000 Aarhus C, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | | |
Collapse
|
42
|
El-Kalyoubi S, Elbaramawi SS, Zordok WA, Malebari AM, Safo MK, Ibrahim TS, Taher ES. Design and synthesis of uracil/thiouracil based quinoline scaffolds as topoisomerases I/II inhibitors for chemotherapy: A new hybrid navigator with DFT calculation. Bioorg Chem 2023; 136:106560. [PMID: 37121108 DOI: 10.1016/j.bioorg.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: 02/21/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/02/2023]
Abstract
In this work, a novel promising hybrid mode of uracil/thiouracil based quinoline pharmacophore i.e. 5a-f was rationalized and synthesized based on rigidification and lipophilic principles, and following the reported pharmacophoric features of camptothecin & doxorubicin. Concurrently, a non-rigid mode pharmacophore i.e. 7a-f was also designed and synthesized. The anti-proliferative activity of the compounds was assessed against three different cancer cell lines, namely A549 lung cancer, MCF-7 breast adenocarcinoma, and HepG-2 hepatic carcinoma. Further, promising candidates were evaluated against A549, and MCF-7 and for their ability to inhibit topoisomerases I &II. Compound 5f was observed to be the most active congener, displaying the highest cell inhibition of 84.4% for topoisomerase I and 92%, for topoisomerase II at a concentration of 100 µM. When its cytotoxicity was evaluated against A549 cells, 5f arrested the cell cycle at the S phase and increased the apoptosis ratio by 46.31%. DFT calculation of 5f showed higher dipole moment and greater negative energy values (-247531.510 kcal/mol) with positive & negative poles, and better stability reflection. Furthermore, molecular docking of 5f to both enzymes showed good agreement with the biological assessment. This study has given insight for further consideration of the highly promising hybrid 5f.
Collapse
Affiliation(s)
- Samar El-Kalyoubi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Port Said University, 42511 Port Said, Egypt.
| | - Samar S Elbaramawi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| | - Wael A Zordok
- Department of Chemistry (Physical Chemistry Division), Faculty of Science, Zagazig University, Zagazig 44519, Egypt.
| | - Azizah M Malebari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Martin K Safo
- Institute for Structural Biology, Drug Discovery and Development, Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA.
| | - Tarek S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ehab S Taher
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt; Research School of Chemistry, Institute of Advanced Studies, The Australian National University, Canberra, Australian Capital Territory 2601, Australia.
| |
Collapse
|
43
|
Baptista D, Ferreira PG, Rocha M. A systematic evaluation of deep learning methods for the prediction of drug synergy in cancer. PLoS Comput Biol 2023; 19:e1010200. [PMID: 36952569 PMCID: PMC10072473 DOI: 10.1371/journal.pcbi.1010200] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 04/04/2023] [Accepted: 02/08/2023] [Indexed: 03/25/2023] Open
Abstract
One of the main obstacles to the successful treatment of cancer is the phenomenon of drug resistance. A common strategy to overcome resistance is the use of combination therapies. However, the space of possibilities is huge and efficient search strategies are required. Machine Learning (ML) can be a useful tool for the discovery of novel, clinically relevant anti-cancer drug combinations. In particular, deep learning (DL) has become a popular choice for modeling drug combination effects. Here, we set out to examine the impact of different methodological choices on the performance of multimodal DL-based drug synergy prediction methods, including the use of different input data types, preprocessing steps and model architectures. Focusing on the NCI ALMANAC dataset, we found that feature selection based on prior biological knowledge has a positive impact-limiting gene expression data to cancer or drug response-specific genes improved performance. Drug features appeared to be more predictive of drug response, with a 41% increase in coefficient of determination (R2) and 26% increase in Spearman correlation relative to a baseline model that used only cell line and drug identifiers. Molecular fingerprint-based drug representations performed slightly better than learned representations-ECFP4 fingerprints increased R2 by 5.3% and Spearman correlation by 2.8% w.r.t the best learned representations. In general, fully connected feature-encoding subnetworks outperformed other architectures. DL outperformed other ML methods by more than 35% (R2) and 14% (Spearman). Additionally, an ensemble combining the top DL and ML models improved performance by about 6.5% (R2) and 4% (Spearman). Using a state-of-the-art interpretability method, we showed that DL models can learn to associate drug and cell line features with drug response in a biologically meaningful way. The strategies explored in this study will help to improve the development of computational methods for the rational design of effective drug combinations for cancer therapy.
Collapse
Affiliation(s)
- Delora Baptista
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Pedro G Ferreira
- Department of Computer Science, Faculty of Sciences, University of Porto, Porto, Portugal
- INESC TEC, Porto, Portugal
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- i3s - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
| | - Miguel Rocha
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
44
|
Lamba S, Roy A. Demystifying the potential of inhibitors targeting DNA topoisomerases in unicellular protozoan parasites. Drug Discov Today 2023; 28:103574. [PMID: 37003515 DOI: 10.1016/j.drudis.2023.103574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/09/2023] [Accepted: 03/25/2023] [Indexed: 04/01/2023]
Abstract
DNA topoisomerases are a group of enzymes omnipresent in all organisms. They maintain the DNA topology during replication, repair, recombination, and transcription. However, the structure of topoisomerase in protozoan parasites differs significantly from that of human topoisomerases; thus, this enzyme acts as a crucial target in drug development against parasitic diseases. Although the therapeutic potential of drugs targeting the parasitic topoisomerase is well known, to manage the shortcomings of currently available therapeutics and the emergence of drug resistance, the discovery of novel antiparasitic molecules is an urgent need. In this review, we describe various investigational and repurposed topoisomerase inhibitors developed against protozoan parasites over the past few years. Teaser: Fatal parasitic diseases are an increasing cause for concern; here, we provide a compilation of different inhibitors targeting DNA topoisomerases, enzymes that are essential for, and unique to, protozoan parasites; therefore, inhibitors are efficient and have few adverse effects.
Collapse
Affiliation(s)
- Swati Lamba
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Amit Roy
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India.
| |
Collapse
|
45
|
Bailly C, Vergoten G. Interaction of Camptothecin Anticancer Drugs with Ribosomal Proteins L15 and L11: A Molecular Docking Study. Molecules 2023; 28:molecules28041828. [PMID: 36838813 PMCID: PMC9967338 DOI: 10.3390/molecules28041828] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 01/31/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The antitumor drug topotecan (TPT) is a potent inhibitor of topoisomerase I, triggering DNA breaks lethal for proliferating cancer cells. The mechanism is common to camptothecins SN38 (the active metabolite of irinotecan) and belotecan (BLT). Recently, TPT was shown to bind the ribosomal protein L15, inducing an antitumor immune activation independent of topoisomerase I. We have modeled the interaction of four camptothecins with RPL15 derived from the 80S human ribosome. Two potential drug-binding sites were identified at Ile135 and Phe129. SN38 can form robust RPL15 complexes at both sites, whereas BLT essentially gave stable complexes with site Ile135. The empirical energy of interaction (ΔE) for SN38 binding to RPL15 is similar to that determined for TPT binding to the topoisomerase I-DNA complex. Molecular models with the ribosomal protein L11 sensitive to topoisomerase inhibitors show that SN38 can form a robust complex at a single site (Cys25), much more stable than those with TPT and BLT. The main camptothecin structural elements implicated in the ribosomal protein interaction are the lactone moiety, the aromatic system and the 10-hydroxyl group. The study provides guidance to the design of modulators of ribosomal proteins L11 and L15, both considered anticancer targets.
Collapse
Affiliation(s)
- Christian Bailly
- Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, University of Lille, 3 rue du Professeur Laguesse, BP-83, F-59006 Lille, France
- CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, University of Lille, F-59000 Lille, France
- OncoWitan, Consulting Scientific Office, Wasquehal, F-59290 Lille, France
- Correspondence:
| | - Gérard Vergoten
- Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, University of Lille, 3 rue du Professeur Laguesse, BP-83, F-59006 Lille, France
| |
Collapse
|
46
|
Buchtova T, Lukac D, Skrott Z, Chroma K, Bartek J, Mistrik M. Drug-Drug Interactions of Cannabidiol with Standard-of-Care Chemotherapeutics. Int J Mol Sci 2023; 24:ijms24032885. [PMID: 36769206 PMCID: PMC9917508 DOI: 10.3390/ijms24032885] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Cannabidiol (CBD) is an easily accessible and affordable Marijuana (Cannabis sativa L.) plant derivative with an extensive history of medical use spanning thousands of years. Interest in the therapeutic potential of CBD has increased in recent years, including its anti-tumour properties in various cancer models. In addition to the direct anticancer effects of CBD, preclinical research on numerous cannabinoids, including CBD, has highlighted their potential use in: (i) attenuating chemotherapy-induced adverse effects and (ii) enhancing the efficacy of some anticancer drugs. Therefore, CBD is gaining popularity as a supportive therapy during cancer treatment, often in combination with standard-of-care cancer chemotherapeutics. However, CBD is a biologically active substance that modulates various cellular targets, thereby possibly resulting in unpredictable outcomes, especially in combinations with other medications and therapeutic modalities. In this review, we summarize the current knowledge of CBD interactions with selected anticancer chemotherapeutics, discuss the emerging mechanistic basis for the observed biological effects, and highlight both the potential benefits and risks of such combined treatments. Apart from the experimental and preclinical results, we also indicate the planned or ongoing clinical trials aiming to evaluate the impact of CBD combinations in oncology. The results of these and future trials are essential to provide better guidance for oncologists to judge the benefit-versus-risk ratio of these exciting treatment strategies. We hope that our present overview of this rapidly advancing field of biomedicine will inspire more preclinical and clinical studies to further our understanding of the underlying biology and optimize the benefits for cancer patients.
Collapse
Affiliation(s)
- Tereza Buchtova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77 147 Olomouc, Czech Republic
| | - David Lukac
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77 147 Olomouc, Czech Republic
| | - Zdenek Skrott
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77 147 Olomouc, Czech Republic
| | - Katarina Chroma
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77 147 Olomouc, Czech Republic
| | - Jiri Bartek
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Genome Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77 147 Olomouc, Czech Republic
- Correspondence:
| |
Collapse
|
47
|
Wang XY, Zhang LN. RNA binding protein SAMD4: current knowledge and future perspectives. Cell Biosci 2023; 13:21. [PMID: 36732864 PMCID: PMC9893680 DOI: 10.1186/s13578-023-00968-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/22/2023] [Indexed: 02/04/2023] Open
Abstract
SAMD4 protein family is a class of novel RNA-binding proteins that can mediate post-transcriptional regulation and translation repression in eukaryotes, which are highly conserved from yeast to humans during evolution. In mammalian cells, SAMD4 protein family consists of two members including SAMD4A/Smaug1 and SAMD4B/Smaug2, both of which contain common SAM domain that can specifically bind to different target mRNAs through stem-loop structures, also known as Smaug recognition elements (SREs), and regulate the mRNA stability, degradation and translation. In addition, SAMD4 can form the cytoplasmic mRNA silencing foci and regulate the translation of SRE-containing mRNAs in neurons. SAMD4 also can form the cytosolic membrane-less organelles (MLOs), termed as Smaug1 bodies, and regulate mitochondrial function. Importantly, many studies have identified that SAMD4 family members are involved in various pathological processes including myopathy, bone development, neural development, and cancer occurrence and progression. In this review, we mainly summarize the structural characteristics, biological functions and molecular regulatory mechanisms of SAMD4 protein family members, which will provide a basis for further research and clinical application of SAMD4 protein family.
Collapse
Affiliation(s)
- Xin-Ya Wang
- grid.28703.3e0000 0000 9040 3743Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, 100124 Beijing, People’s Republic of China
| | - Li-Na Zhang
- grid.28703.3e0000 0000 9040 3743Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of Environment and Life, Beijing University of Technology, 100124 Beijing, People’s Republic of China
| |
Collapse
|
48
|
Baiju S, Afzal A, Shahin Thayyil M, S.Al-Otaibi J, Kashif Ali S. Computational Studies on Anticancerous Camptothecin and it’s derivative Camp-10 by Density Functional Theory. RESULTS IN CHEMISTRY 2023. [DOI: 10.1016/j.rechem.2023.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
49
|
Chepanova AA, Zakharenko AL, Dyrkheeva NS, Chernyshova IA, Zakharova OD, Ilina ES, Luzina OA, Salakhutdinov NF, Lavrik OI. Influence of Tyrosyl-DNA Phosphodiesterase 1 Inhibitor on the Proapoptotic and Genotoxic Effects of Anticancer Agent Topotecan. DOKL BIOCHEM BIOPHYS 2023; 508:25-30. [PMID: 36653585 PMCID: PMC10042932 DOI: 10.1134/s1607672922700077] [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: 11/28/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 01/20/2023]
Abstract
To date, various strategies have been proposed to increase the efficiency of cancer therapy. It is known that the action of DNA repair system can determine the resistance of cancer cells to DNA-damaging chemotherapy and radiotherapy, and one of these ways to increase therapeutic efficiency is the search for inhibitors of enzymes of the DNA repair system. Inhibition of the DNA repair enzyme tyrosyl-DNA phosphodiesterase1 (Tdp1) leads to an increase in the effectiveness of the topoisomerase 1 (Top1) inhibitor, the anticancer drug topotecan. Covalent complexes Top1-DNA, which are normally short-lived and are not a threat to the cell, are stabilized under the influence of topotecan and lead to cell death. Tdp1 eliminates such stabilized complexes and thus weaken the effect of topotecan therapy. We have previously shown that the use of the usnic acid hydrazonothiazole derivative OL9-119 in combination with topotecan increased the antitumor and antimetastatic efficacy of the latter in a mouse model of Lewis lung carcinoma. In this work, it was shown that the combined use of topotecan and Tdp1 inhibitor, the hydrazonothiazole derivative of usnic acid OL9-119, leads to an increase in the DNA-damaging effect of topotecan which is used in the clinic for the treatment of cancer. The study of the proapoptotic effect of the compound OL9-119 showed that the compound itself does not induce apoptosis, but increases the proapoptotic effect of topotecan. The results of the study could be used to improve the effectiveness of anticancer therapy and/or to reduce the therapeutic dose of topotecan and, therefore, the severity of side effects.
Collapse
Affiliation(s)
- A A Chepanova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A L Zakharenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - N S Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - I A Chernyshova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - O D Zakharova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E S Ilina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - O A Luzina
- Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - N F Salakhutdinov
- Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - O I Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.
| |
Collapse
|
50
|
Zhang L, Zhu L, Tang L, Xie J, Gao Y, Yu C, Shang K, Han H, Liu C, Lu Y. Glutathione-Responsive Nanoparticles of Camptothecin Prodrug for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205246. [PMID: 36442854 PMCID: PMC9875659 DOI: 10.1002/advs.202205246] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/17/2022] [Indexed: 05/28/2023]
Abstract
Camptothecin (CPT) is a potent chemotherapeutic agent for various cancers, but the broader application of CPT is still hindered by its poor bioavailability and systemic toxicity. Here, a prodrug that releases CPT in response to glutathione (GSH), which is commonly overexpressed by cancer cells is reported. Through assembling with PEGylated lipids, the prodrug is incorporated within as-assembled nanoparticles, affording CPT with a prolonged half-life in blood circulation, enhanced tumor targetingability, and improved therapeutic efficacy. Furthermore, such prodrug nanoparticles can also promote dendritic cell maturation and tumor infiltration of CD8+ T cells, providing a novel strategy to improve the therapeutic efficacy of CPT.
Collapse
Affiliation(s)
- Lingpu Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Lin Zhu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Lin Tang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Jiayi Xie
- Department of AutomaticTsinghua UniversityPeking University Third HospitalBeijing Key Laboratory of Magnetic Resonance Imaging Devices and TechnologyBeijing100191P. R. China
| | - Yajuan Gao
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Changyuan Yu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Kun Shang
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Hongbin Han
- Department of RadiologyPeking University Third HospitalInstitute of Medical TechnologyPeking University Health Science CenterBeijing100019P. R. China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Yunfeng Lu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| |
Collapse
|