1
|
Wang L, Yang H, Huang W, Ran G, He X, Bradley M, Qian S. Tumor-specific cathepsin B-triggered fluorescence imaging and prodrug activation. Eur J Med Chem 2025; 292:117661. [PMID: 40286448 DOI: 10.1016/j.ejmech.2025.117661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Bioorthogonal activation chemistries have great potential in the development of novel drug treatments due to their versatility, tunability, and the ability to generate therapies with improved spatial targeting. The upregulation of Cathepsin B is highly correlated with the development of cancers, however, few fluorescent probes or prodrugs-based on Cathepsin B activity have demonstrated high tumor selectivity, since Cathepsin B is expressed in a variety of normal tissues. In this study, we report a strain-promoted azide-alkyne cycloaddition-activation strategy whereby a para-azido safety-catch linker is triggered by the tumor locating Biotin-TCO (trans-cyclooctene) conjugate, with subsequent tumor-specific Cathepsin B-triggered activation, generating a fluorescent reporter/cytotoxic drug, with high tumor selectivity. Our results suggest that this dual AND-Gate strategy of orthogonal Biotin AND Cathepsin B action would be advantageous for tumor-specific fluorescence labelling, fluorescence-guided surgery and targeted treatment.
Collapse
Affiliation(s)
- Luyang Wang
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China
| | - Houchi Yang
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China
| | - Wanyun Huang
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China
| | - Guojun Ran
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China
| | - Xiaolong He
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China.
| | - Mark Bradley
- Precision Healthcare University Research Institute, Queen Mary University of London, Empire House London E1 1HH, UK
| | - Shan Qian
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China.
| |
Collapse
|
2
|
Descamps A, Arnoux P, Frochot C, Barbault F, Deschamp J, Monteil M, Migianu-Griffoni E, Legigan T, Lecouvey M. Synthesis and preliminary anticancer evaluation of photo-responsive prodrugs of hydroxymethylene bisphosphonate alendronate. Eur J Med Chem 2024; 269:116307. [PMID: 38460269 DOI: 10.1016/j.ejmech.2024.116307] [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: 01/31/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
The antitumoral activity of hydroxymethylene bisphosphonates (HMBP) such as alendronate or zoledronate is hampered by their exceptional bone-binding properties and their short plasmatic half-life which preclude their accumulation in non-skeletal tumors. In this context, the use of lipophilic prodrugs represents a simple and straightforward strategy to enhance the biodistribution of bisphosphonates in these tissues. We describe in this article the synthesis of light-responsive prodrugs of HMBP alendronate. These prodrugs include lipophilic photo-removable nitroveratryl groups which partially mask the highly polar alendronate HMBP scaffold. Photo-responsive prodrugs of alendronate are stable in physiological conditions and display reduced toxicity compared to alendronate against MDA-MB-231 cancer cells. However, the antiproliferative effect of these prodrugs is efficiently restored after cleavage of their nitroveratryl groups upon exposure to UV light. In addition, substitution of alendronate with such photo-responsive substituents drastically reduces its bone-binding properties, thereby potentially improving its biodistribution in soft tissues after i.v. administration. The development of such lipophilic photo-responsive prodrugs is a promising approach to fully exploit the anticancer effect of HMBPs on non-skeletal tumors.
Collapse
Affiliation(s)
- Aurélie Descamps
- Université Sorbonne Paris Nord, Department of Chemistry, UMR-CNRS, 7244, 1 Rue de Chablis, F-93000, Bobigny, France
| | | | - Céline Frochot
- Université de Lorraine, CNRS, LRGP, F-54000, Nancy, France
| | | | - Julia Deschamp
- Université Sorbonne Paris Nord, Department of Chemistry, UMR-CNRS, 7244, 1 Rue de Chablis, F-93000, Bobigny, France
| | - Maelle Monteil
- Université Sorbonne Paris Nord, Department of Chemistry, UMR-CNRS, 7244, 1 Rue de Chablis, F-93000, Bobigny, France
| | - Evelyne Migianu-Griffoni
- Université Sorbonne Paris Nord, Department of Chemistry, UMR-CNRS, 7244, 1 Rue de Chablis, F-93000, Bobigny, France
| | - Thibaut Legigan
- Université Sorbonne Paris Nord, Department of Chemistry, UMR-CNRS, 7244, 1 Rue de Chablis, F-93000, Bobigny, France.
| | - Marc Lecouvey
- Université Sorbonne Paris Nord, Department of Chemistry, UMR-CNRS, 7244, 1 Rue de Chablis, F-93000, Bobigny, France.
| |
Collapse
|
3
|
Zhao Z, Wang X, Wang J, Li Y, Lin W, Lu K, Chen J, Xia W, Mao ZW. A Nanobody-Bioorthogonal Catalyst Conjugate Triggers Spatially Confined Prodrug Activation for Combinational Chemo-immunotherapy. J Med Chem 2023; 66:11951-11964. [PMID: 37590921 DOI: 10.1021/acs.jmedchem.3c00557] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Checkpoint inhibitors have been used with chemotherapy to improve antitumor efficacy. However, overcoming the immunosuppressive effect of chemotherapeutics remains a challenge. We report a nanobody-catalyst conjugate Ru-PD-L1 by fusing a ruthenium catalyst to an anti-PD-L1 nanobody. After administration of Ru-PD-L1 and a doxorubicin (DOX) prodrug, Ru-PD-L1 disrupts the PD-L1/PD-1 interaction and catalyzes the uncaging of the DOX prodrug. The spatially confined release of DOX reduces its systemic toxicity and leads to immunogenic cell death (ICD). The induced ICD triggers antitumor immune responses, which are further amplified by PD-L1 blockade to elicit synergistic chemo-immunotherapy, substantially increasing the number of tumor-infiltrating T-cells by 49.7% compared with the controls, thereby exhibiting high antitumor activity and low cytotoxicity in murine models. The combinational treatment could inhibit the growth of mice tumors by 67.7% compared to the control group. This combinational approach circumvents the negative immunogenic effects of chemotherapeutics and provides a potential chemo-immunotherapy strategy for human cancer treatment.
Collapse
Affiliation(s)
- Zhennan Zhao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, IGCME, Sun Yat-sen University, Guangzhou 510275, China
| | - Xinyu Wang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jinhui Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, IGCME, Sun Yat-sen University, Guangzhou 510275, China
| | - Yiyi Li
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenkai Lin
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, IGCME, Sun Yat-sen University, Guangzhou 510275, China
| | - Kai Lu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, IGCME, Sun Yat-sen University, Guangzhou 510275, China
| | - Jun Chen
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei Xia
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, IGCME, Sun Yat-sen University, Guangzhou 510275, China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, IGCME, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
4
|
Brasino M, Wagnell E, Hamilton S, Ranganathan S, Gomes MM, Branchaud B, Messmer B, Ibsen SD. Turning antibodies off and on again using a covalently tethered blocking peptide. Commun Biol 2022; 5:1357. [PMID: 36496512 PMCID: PMC9741643 DOI: 10.1038/s42003-022-04094-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/11/2022] [Indexed: 12/13/2022] Open
Abstract
In their natural form, antibodies are always in an "on-state" and are capable of binding to their targets. This leads to undesirable interactions in a wide range of therapeutic, analytical, and synthetic applications. Modulating binding kinetics of antibodies to turn them from an "off-state" to an "on-state" with temporal and spatial control can address this. Here we demonstrate a method to modulate binding activity of antibodies in a predictable and reproducible way. We designed a blocking construct that uses both covalent and non-covalent interactions with the antibody. The construct consisted of a Protein L protein attached to a flexible linker ending in a blocking-peptide designed to interact with the antibody binding site. A mutant Protein L was developed to enable photo-triggered covalent crosslinking to the antibody at a specific location. The covalent bond anchored the linker and blocking peptide to the antibody light chain keeping the blocking peptide close to the antibody binding site. This effectively put the antibody into an "off-state". We demonstrate that protease-cleavable and photocleavable moieties in the tether enable controlled antibody activation to the "on-state" for anti-FLAG and cetuximab antibodies. Protein L can bind a range of antibodies used therapeutically and in research for wide applicability.
Collapse
Affiliation(s)
- Michael Brasino
- grid.5288.70000 0000 9758 5690Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201 USA
| | - Eli Wagnell
- grid.5288.70000 0000 9758 5690Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201 USA
| | - Sean Hamilton
- grid.5288.70000 0000 9758 5690Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201 USA ,grid.5288.70000 0000 9758 5690Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201 USA
| | - Srivathsan Ranganathan
- grid.5288.70000 0000 9758 5690Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201 USA
| | - Michelle M. Gomes
- grid.5288.70000 0000 9758 5690Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201 USA
| | - Bruce Branchaud
- grid.5288.70000 0000 9758 5690Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201 USA
| | | | - Stuart D. Ibsen
- grid.5288.70000 0000 9758 5690Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201 USA ,grid.5288.70000 0000 9758 5690Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201 USA
| |
Collapse
|
5
|
Bargathulla I, Babu AA, Shanavas A, Vellaichamy E, Nasar AS. PEGylated bis-indolyl polyurethane dendrimers with anti-cancer activity as carriers for doxorubicin to treat lung cancer cells. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02394-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
6
|
Chen Q, Jia C, Xu Y, Jiang Z, Hu T, Li C, Cheng X. Dual-pH responsive chitosan nanoparticles for improving in vivo drugs delivery and chemoresistance in breast cancer. Carbohydr Polym 2022; 290:119518. [DOI: 10.1016/j.carbpol.2022.119518] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/10/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022]
|
7
|
Johan AN, Li Y. Development of Photoremovable Linkers as a Novel Strategy to Improve the Pharmacokinetics of Drug Conjugates and Their Potential Application in Antibody-Drug Conjugates for Cancer Therapy. Pharmaceuticals (Basel) 2022; 15:655. [PMID: 35745573 PMCID: PMC9230074 DOI: 10.3390/ph15060655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/19/2022] [Accepted: 05/22/2022] [Indexed: 02/04/2023] Open
Abstract
Although there have been extensive research and progress on the discovery of anticancer drug over the years, the application of these drugs as stand-alone therapy has been limited by their off-target toxicities, poor pharmacokinetic properties, and low therapeutic index. Targeted drug delivery, especially drug conjugate, has been recognized as a technology that can bring forth a new generation of therapeutics with improved efficacy and reduced side effects for cancer treatment. The linker in a drug conjugate is of essential importance because it impacts the circulation time of the conjugate and the release of the drug for full activity at the target site. Recently, the light-triggered linker has attracted a lot of attention due to its spatiotemporal controllability and attractive prospects of improving the overall pharmacokinetics of the conjugate. In this paper, the latest developments of UV- and IR-triggered linkers and their application and potential in drug conjugate development are reviewed. Some of the most-well-researched photoresponsive structural moieties, such as UV-triggered coumarin, ortho-nitrobenzyl group (ONB), thioacetal ortho-nitrobenzaldehyde (TNB), photocaged C40-oxidized abasic site (PC4AP), and IR-triggered cyanine and BODIPY, are included for discussion. These photoremovable linkers show better physical and chemical stabilities and can undergo rapid cleavage upon irradiation. Very importantly, the drug conjugates containing these linkers exhibit reduced off-target toxicity and overall better pharmacokinetic properties. The progress on photoactive antibody-drug conjugates, such as antibody-drug conjugates (ADC) and antibody-photoabsorber conjugate (APC), as precision medicine in clinical cancer treatment is highlighted.
Collapse
Affiliation(s)
| | - Yi Li
- Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| |
Collapse
|
8
|
Das A, Banik BK. Advances in heterocycles as DNA intercalating cancer drugs. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2021-0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The insertion of a molecule between the bases of DNA is known as intercalation. A molecule is able to interact with DNA in different ways. DNA intercalators are generally aromatic, planar, and polycyclic. In chemotherapeutic treatment, to suppress DNA replication in cancer cells, intercalators are used. In this article, we discuss the anticancer activity of 10 intensively studied DNA intercalators as drugs. The list includes proflavine, ethidium bromide, doxorubicin, dactinomycin, bleomycin, epirubicin, mitoxantrone, ellipticine, elinafide, and echinomycin. Considerable structural diversities are seen in these molecules. Besides, some examples of the metallo-intercalators are presented at the end of the chapter. These molecules have other crucial properties that are also useful in the treatment of cancers. The successes and limitations of these molecules are also presented.
Collapse
Affiliation(s)
- Aparna Das
- Department of Mathematics and Natural Sciences , College of Sciences and Human Studies, Prince Mohammad Bin Fahd University , Al Khobar 31952 , Kingdom of Saudi Arabia
| | - Bimal Krishna Banik
- Department of Mathematics and Natural Sciences , College of Sciences and Human Studies, Prince Mohammad Bin Fahd University , Al Khobar 31952 , Kingdom of Saudi Arabia
| |
Collapse
|
9
|
Mozaffari S, Salehi D, Mahdipoor P, Beuttler R, Tiwari R, Aliabadi HM, Parang K. Design and application of hybrid cyclic-linear peptide-doxorubicin conjugates as a strategy to overcome doxorubicin resistance and toxicity. Eur J Med Chem 2021; 226:113836. [PMID: 34537446 DOI: 10.1016/j.ejmech.2021.113836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/19/2021] [Accepted: 09/05/2021] [Indexed: 12/15/2022]
Abstract
Doxorubicin (Dox) is used for breast cancer, leukemia, and lymphoma treatment as an effective chemotherapeutic agent. However, Dox use is restricted due to inherent and acquired resistance and an 8-fold increase in the risk of potentially fatal cardiotoxicity. Hybrid cyclic-linear peptide [R5K]W7A and linear peptide R5KW7A were conjugated with Dox through a glutarate linker to afford [R5K]W7A-Dox and R5KW7A-Dox conjugates to generate Dox derivatives. Alternatively, [R5K]W7C was conjugated with Dox via a disulfide linker to generate [R5K]W7C-S-S-Dox conjugate, where S-S is a disulfide bond. Comparative antiproliferative assays between conjugates [R5K]W7A-Dox, [R5K]W7C-S-S-Dox, linear R5KW7A-Dox, the corresponding physical mixtures of the peptides, and Dox were performed in normal and cancer cells. [R5K]W7A-Dox conjugate was 2-fold more efficient than R5KW7A-Dox, and [R5K]W7C-S-S-Dox conjugates in inhibiting the cell proliferation of human leukemia cells (CCRF-CEM). Therefore, hybrid cyclic-linear [R5K]W7A-Dox conjugate was selected for further studies and inhibited the cell viability of CCRF-CEM (84%), ovarian adenocarcinoma (SK-OV-3, 39%), and gastric carcinoma (AGS, 73%) at a concentration of 5 μM after 72 h of incubation, which was comparable to Dox (5 μM) efficacy (CCRF-CEM (85%), SK-OV-3 (33%), and AGS (87%)). While [R5K]W7A-Dox had a significant effect on the viability of cancer cells, it exhibited minimal cytotoxicity to normal kidney (LLC-PK1, 5-7%) and heart cells (H9C2, <9%) at concentrations of 5-10 μM (compared to free Dox at 5 μM that reduced the viability of kidney and heart cells by 85% and 44%, respectively). The fluorescence microscopy images were consistent with the cytotoxicity studies, indicating minimal uptake of the cyclic-linear [R5K]W7A-Dox (5 μM) in H9C2 cells. In comparison, Dox (5 μM) showed significant uptake, reduced cell viability, and changed the morphology of the cells after 24 h. [R5K]W7A-Dox showed 16-fold and 9.5-fold higher activity against Dox-resistant cells MDA231R and MES-SA/MX2 (lethal dose for 50% cell death or LC50 of 2.3 and 4.3 μM, respectively) compared to free Dox (LC50 of 36-41 μM, respectively). These data, along with the results obtained from the cell viability tests, indicate comparable efficiency of [R5K]W7A-Dox to free Dox in leukemia, ovarian, and gastric cancer cells, significantly reduced toxicity in normal kidney LLC-PK1 and heart H9C2 cells, and significantly higher efficiency in Dox-resistant cells. A number of endocytosis inhibitors did not affect the cellular uptake of [R5K]W7A-Dox.
Collapse
Affiliation(s)
- Saghar Mozaffari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA, 92618, USA
| | - David Salehi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA, 92618, USA
| | - Parvin Mahdipoor
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA, 92618, USA
| | - Richard Beuttler
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA, 92618, USA
| | - Rakesh Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA, 92618, USA.
| | - Hamidreza Montazeri Aliabadi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA, 92618, USA.
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA, 92618, USA.
| |
Collapse
|
10
|
Cui X, Du K, Yuan X, Xiao W, Tao Y, Xu D, Hu H. A comparative study of the in vitro antitumor effect of mannose-doxorubicin conjugates with different linkers. Drug Dev Res 2021; 83:646-658. [PMID: 34730851 DOI: 10.1002/ddr.21896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/08/2021] [Accepted: 10/24/2021] [Indexed: 12/27/2022]
Abstract
In this work, five Man-DOX conjugates with different linkers were developed for targeted DOX delivery. The five Man-DOX conjugates with different linkers were characterized by 1 H NMR, HRMS, HPLC, UV-vis, and fluorescence spectroscopy. Man-Suc-DOX, Man-TDG-DOX, and Man-DG-DOX can self-assemble into near-spherical nanoparticles with hydrodynamic diameters of 150-200 nm and negative zeta potentials in deionized water, whereas Man-SS-DOX and Man-SeSe-DOX are hardly dispersed in deionized water. The self-assembly behaviors of Man-Suc-DOX, Man-TDG-DOX, and Man-DG-DOX were studied by dissipative particle dynamics simulation and the results show that Man-Suc-DOX, Man-TDG-DOX, and Man-DG-DOX all self-assemble into spherical particles with Man and linkers on the surfaces and DOX in the interiors. The in vitro drug release study shows that Man-Suc-DOX, Man-TDG-DOX, and Man-DG-DOX exhibit limited drug release, while Man-SS-DOX and Man-SeSe-DOX exhibit glutathione-responsive drug release. The cellular uptake study shows that Man-DG-DOX exhibits the highest cellular uptake amount on HepG2 cells. Finally, Man-DG-DOX exhibits the best in vitro antitumor effect against HepG2 cells among the five Man-DOX conjugates with different linkers. Although the in vitro antitumor activity of Man-DG-DOX is still lower than free DOX, Man-DG-DOX shows significant selectivity toward HepG2 cells. Man-DG-DOX might achieve selective DOX delivery for mannose receptor overexpressed tumors.
Collapse
Affiliation(s)
- Xinxin Cui
- School of Pharmacy, Changzhou University, Changzhou, P. R. China
| | - Kunda Du
- School of Pharmacy, Changzhou University, Changzhou, P. R. China
| | - Xiaoyin Yuan
- School of Pharmacy, Changzhou University, Changzhou, P. R. China
| | - Wen Xiao
- School of Pharmacy, Changzhou University, Changzhou, P. R. China
| | - Yayu Tao
- School of Pharmacy, Changzhou University, Changzhou, P. R. China
| | - Defeng Xu
- School of Pharmacy, Changzhou University, Changzhou, P. R. China
| | - Hang Hu
- School of Pharmacy, Changzhou University, Changzhou, P. R. China
| |
Collapse
|
11
|
Biotinylated chitosan macromolecule based nanosystems: A review from chemical design to biological targets. Int J Biol Macromol 2021; 188:82-93. [PMID: 34363823 DOI: 10.1016/j.ijbiomac.2021.07.197] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/16/2021] [Accepted: 07/31/2021] [Indexed: 12/28/2022]
Abstract
World Health Organization estimates that 30-50% of cancers are preventable by healthy lifestyle choices, early detection and adequate therapy. When the conventional therapeutic strategies are still regulated by the lack of selectivity, multidrug resistance and severe toxic side effects, nanotechnology grants a new frontier for cancer management since it targets cancer cells and spares healthy tissues. This review highlights recent studies using biotin molecule combined with functional nanomaterials used in biomedical applications, with a particular attention on biotinylated chitosan-based nanosystems. Succinctly, this review focuses on five areas of recent advances in biotin engineering: (a) biotin features, (b) biotinylation approaches, (c) biotin functionalized chitosan based nanosystems for drug and gene delivery functions, (d) diagnostic and theranostic perspectives, and (e) author's inputs to the biotin-chitosan based tumour-targeting drug delivery structures. Precisely engineered biotinylated-chitosan macromolecules shaped into nanosystems are anticipated to emerge as next-generation platforms for treatment and molecular imaging modalities applications.
Collapse
|
12
|
Targeted Cancer Therapy Using Compounds Activated by Light. Cancers (Basel) 2021; 13:cancers13133237. [PMID: 34209493 PMCID: PMC8269035 DOI: 10.3390/cancers13133237] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 12/21/2022] Open
Abstract
Cancer chemotherapy is affected by a modest selectivity and toxic side effects of pharmacological interventions. Among novel approaches to overcome this limitation and to bring to therapy more potent and selective agents is the use of light for selective activation of anticancer compounds. In this review, we focus on the anticancer applications of two light-activated approaches still in the experimental phase: photoremovable protecting groups ("photocages") and photoswitches. We describe the structural considerations behind the development of novel compounds and the plethora of assays used to confirm whether the photochemical and pharmacological properties are meeting the stringent criteria for an efficient in vivo light-dependent activation. Despite its immense potential, light activation brings many challenges, and the complexity of the task is very demanding. Currently, we are still deeply in the phase of pharmacological tools, but the vivid research and rapid development bring the light of hope for potential clinical use.
Collapse
|
13
|
Breast Cancer Inhibition by Biosynthesized Titanium Dioxide Nanoparticles Is Comparable to Free Doxorubicin but Appeared Safer in BALB/c Mice. MATERIALS 2021; 14:ma14123155. [PMID: 34201266 PMCID: PMC8229371 DOI: 10.3390/ma14123155] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 12/24/2022]
Abstract
Cancer remains a global health burden prompting affordable, target-oriented, and safe chemotherapeutic agents to reduce its incidence rate worldwide. In this study, a rapid, cost-effective, and green synthesis of titanium dioxide (TiO2) nanoparticles (NPs) has been carried out; Ex vivo and in vivoevaluation of their safety and anti-tumor efficacy compared to doxorubicin (DOX), a highly efficient breast anti-cancer agent but limited by severe cardiotoxicity in many patients.Thereby,TiO2 NPs were eco-friendly synthetized using aqueous leaf extract of the tropical medicinal shrub Zanthoxylum armatum as a reducing agent. Butanol was used as a unique template. TiO2 NPs were physically characterized by ultraviolet-visible (UV-Vis) spectroscopy, dynamic light scattering (DLS), transmission electron microscopy (TEM), scanning electron microscope (SEM), X-ray powder diffraction (XRD), and Fourier-transform infrared spectroscopy (FTIR) as routine state-of-the art techniques. The synthesized TiO2 NPs were then evaluated for their cytotoxicity (by MTT, FACS, and oxidative stress assays) in 4T1 breast tumor cells, and their hemocompatibility (by hemolysis assay). In vivo anti-tumor efficacy and safety of the TiO2 NPs were further assessed using subcutaneous 4T1 breast BALB/c mouse tumor model.The greenly prepared TiO2 NPs were small, spherical, and crystalline in nature. Interestingly, they were hemocompatible and elicited a strong DOX-like concentration-dependent cytotoxicity-induced apoptosis both ex vivo and in vivo (with a noticeable tumor volume reduction). The underlying molecular mechanism was, at least partially, mediated through reactive oxygen species (ROS) generation (lipid peroxidation). Unlike DOX (P < 0.05), it is important to mention that no cardiotoxicity or altered body weight were observed in both the TiO2 NPs-treated tumor-bearing mouse group and the PBS-treated mouse group (P > 0.05). Taken together, Z. armatum-derived TiO2 NPs are cost-effective, more efficient, and safer than DOX. The present findings shall prompt clinical trials using green TiO2 NPs, at least as a possible alternative modality to DOX for effective breast cancer therapy.
Collapse
|
14
|
Gelmi A, Schutt CE. Stimuli-Responsive Biomaterials: Scaffolds for Stem Cell Control. Adv Healthc Mater 2021; 10:e2001125. [PMID: 32996270 PMCID: PMC11468740 DOI: 10.1002/adhm.202001125] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/18/2020] [Indexed: 12/28/2022]
Abstract
Stem cell fate is closely intertwined with microenvironmental and endogenous cues within the body. Recapitulating this dynamic environment ex vivo can be achieved through engineered biomaterials which can respond to exogenous stimulation (including light, electrical stimulation, ultrasound, and magnetic fields) to deliver temporal and spatial cues to stem cells. These stimuli-responsive biomaterials can be integrated into scaffolds to investigate stem cell response in vitro and in vivo, and offer many pathways of cellular manipulation: biochemical cues, scaffold property changes, drug release, mechanical stress, and electrical signaling. The aim of this review is to assess and discuss the current state of exogenous stimuli-responsive biomaterials, and their application in multipotent stem cell control. Future perspectives in utilizing these biomaterials for personalized tissue engineering and directing organoid models are also discussed.
Collapse
Affiliation(s)
- Amy Gelmi
- School of ScienceCollege of Science, Engineering and HealthRMIT UniversityMelbourneVIC3001Australia
| | - Carolyn E. Schutt
- Department of Biomedical EngineeringKnight Cancer Institute Cancer Early Detection Advanced Research Center (CEDAR)Oregon Health and Science UniversityPortlandOR97201USA
| |
Collapse
|
15
|
Zhou N, Zhang N, Zhi Z, Jing X, Liu D, Shao Y, Wang D, Meng L. One-pot synthesis of acid-degradable polyphosphazene prodrugs for efficient tumor chemotherapy. J Mater Chem B 2020; 8:10540-10548. [PMID: 33118582 DOI: 10.1039/d0tb01992e] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
In order to improve the therapeutic efficacy and reduce the side effects of anticancer drugs, stimuli-responsive and biodegradable drug-delivery systems have attracted significant attention in the past three decades. Herein, we report acid-responsive and degradable polyphosphazene nano-prodrugs synthesized via a one-pot cross-linking reaction of 4-hydroxybenzhydrazide-modified doxorubicin (BMD) with hexachlorocyclotriphosphazene (HCCP). The phenol groups in the as-synthesized BMD exhibited a high reactivity towards HCCP and in the presence of a basic catalyst the determined drug loading ratio of the nanoparticles, denoted as HCCP-BMD, was up to 85.64%. Interestingly, the hydrazone bonds in BMD and the skeleton of polyphosphazene tended to break down in acidic environments, and the antitumor active drug DOX was found to be released in an acidic tumor microenvironment (pH ∼ 6.8 for extracellular, and pH ∼ 5.0 for endosomes and lysosomes). The resulting HCCP-BMD prodrug exhibited high cytotoxicity to HeLa cells and could effectively suppress tumor growth, with negligible damage to normal tissues. We therefore believe that this acid- degradable polyphosphazene prodrug may offer great potential in various biomedical fields.
Collapse
Affiliation(s)
- Na Zhou
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Key Laboratory of Sustainable Energy Material Chemistry, Xi'an Jiaotong University, Xi'an 710049, China.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Novel antibody-drug conjugate with UV-controlled cleavage mechanism for cytotoxin release. Bioorg Chem 2020; 111:104475. [PMID: 33798843 DOI: 10.1016/j.bioorg.2020.104475] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/21/2022]
Abstract
Antibody-drug conjugates (ADCs) are being developed worldwide with the potential to revolutionize current cancer treatment strategies. However, off-target toxicity caused by the instability of linkers remains one of the main issues to be resolved. Developing a novel photocontrol-ADC with good stability and photocontrolled release seemed to be an attractive and practical solution. In this study, we designed, for the first time, a novel ultraviolet (UV) light-controlled ADC by carefully integrating the UV-cleavable o-nitro-benzyl structure into the linker. Our preliminary work indicated that the ADC exhibited good stability and photocontrollability while maintaining a targeting effect similar to that of the naked antibody. Upon irradiation with UV light, the ADC rapidly released free cytotoxins and exerted significant cytotoxicity toward drug-resistant tumor cells. Compared to those of the unirradiated cells, the EC50 values of ADCs increased by up to 50-fold. Furthermore, our research confirmed that the degradation products of unirradiated ADC, Cys-1a, were relatively less toxic, thus potentially reducing the off-target toxicity caused by nonspecific uptake of ADCs. The novel design strategy of UV light-controlled ADCs may provide new perspectives for future research on ADCs and promote the development of photocontrol systems.
Collapse
|
17
|
Yang N, Qiu F, Zhu F, Qi L. Therapeutic Potential of Zinc Oxide-Loaded Syringic Acid Against in vitro and in vivo Model of Lung Cancer. Int J Nanomedicine 2020; 15:8249-8260. [PMID: 33149573 PMCID: PMC7602902 DOI: 10.2147/ijn.s272997] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/29/2020] [Indexed: 01/02/2023] Open
Abstract
Introduction Lung cancer is one of the most aggressive forms of cancer that leads to a high mortality rate amongst several cancer types and it is a widely recurrent cancer globally. The use of zinc oxide nanoparticles (ZnONPs) in the formulation of sun cream, food flavors, and colorings due to its varied biological properties. The extensive significance of nanoparticles encourages their production but the approaches are a common challenge in concluding the direct beneficial effect for the disease treatment. Hence, in the present study, zinc oxide-loaded syringic acid (ZnO-SYR) phytochemical was used to elucidate the therapeutic effect against lung cancer. Methods The ZnO-SYR nanoparticles were synthesized and characterized by UV-visible spectroscopy, X-ray diffraction, dynamic light scattering, and FT-IR analysis. The characterized ZnO-SYR was tested on in vivo mouse model of lung cancer (benzo(a)pyrene (BAP)) and in vitro A549 cells. Results The results demonstrated the significant restoration of body weight with attenuated serum marker enzymes compared to BAP-treated animals. In addition, cytokine estimation revealed ameliorated levels of TNF-α, interleukins, IL-6, IL-1β with evidenced histological observations in ZnO-SYR-treated mice compared to BAP-induced lung cancer mice. Discussion Furthermore, cytotoxicity analysis demonstrated the altered mitochondrial membrane potential (MMP), with a profound increase in reactive oxygen species (ROS) levels, and apoptosis mechanism by ZnO-SYR compared to control cells. The conclusions of the present study put forward an evident confirmation of the protective and beneficial effects of zincoxide-loaded syringic acid against the BAP-induced lung cancer model.
Collapse
Affiliation(s)
- Ning Yang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Feng Qiu
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Feng Zhu
- Department of Thoracic Surgery, Shandong Provincial Chest Hospital, Jinan, Shandong 250013, People's Republic of China
| | - Lei Qi
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| |
Collapse
|
18
|
Li J, Xiao D, Liu L, Xie F, Li W, Sun W, Yang X, Zhou X. Design, Synthesis, and In Vitro Evaluation of the Photoactivatable Prodrug of the PARP Inhibitor Talazoparib. Molecules 2020; 25:molecules25020407. [PMID: 31963730 PMCID: PMC7024556 DOI: 10.3390/molecules25020407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 12/13/2022] Open
Abstract
In this article, we report the design, synthesis, photodynamic properties, and in vitro evaluation of photoactivatable prodrug for the poly (ADP-ribose) polymerase 1 (PARP-1) inhibitor Talazoparib. In order to yield a photoactivatable, inactive prodrug, photoactivatable protecting groups (PPGs) were employed to mask the key pharmacophore of Talazoparib. Our study confirmed the good stability and photolytic effect of prodrugs. A PARP-1 enzyme inhibition assay and PARylation experiment showed that the inhibitory activity of the prodrug was reduced 380 times and more than 658 times, respectively, which proved that the prodrug's expected activity was lost after PPG protection. In BRCA1- and BRCA2-deficient cell lines, the inhibitory activity of the compound was significantly restored after ultraviolet (UV) irradiation. The results indicate that the photoactivatable prodrug strategy is an interesting approach for studying PARP inhibitors. Meanwhile, the described photoactivatable prodrug also provided a new biological tool for the mechanism research of PARP.
Collapse
Affiliation(s)
- Jiaguo Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China;
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.X.); (L.L.); (F.X.); (W.L.)
| | - Dian Xiao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.X.); (L.L.); (F.X.); (W.L.)
| | - Lianqi Liu
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.X.); (L.L.); (F.X.); (W.L.)
| | - Fei Xie
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.X.); (L.L.); (F.X.); (W.L.)
| | - Wei Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.X.); (L.L.); (F.X.); (W.L.)
| | - Wei Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China;
- Correspondence: (W.S.); (X.Y.); (X.Z.); Tel.: +86-010-6693-0674 (X.Z.)
| | - Xiaohong Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China;
- Correspondence: (W.S.); (X.Y.); (X.Z.); Tel.: +86-010-6693-0674 (X.Z.)
| | - Xinbo Zhou
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.X.); (L.L.); (F.X.); (W.L.)
- Correspondence: (W.S.); (X.Y.); (X.Z.); Tel.: +86-010-6693-0674 (X.Z.)
| |
Collapse
|
19
|
Tumor targeting vitamin B12 derivatives for X-ray induced treatment of pancreatic adenocarcinoma. Photodiagnosis Photodyn Ther 2019; 30:101637. [PMID: 31899377 DOI: 10.1016/j.pdpdt.2019.101637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND X-Ray induced phototherapy is highly sought after as it provides a deep tissue, synergistic method of treating cancers via standard-of-care radiotherapy. When this is combined with releasable chemotherapy agents, it can provide high target selectivity, with reduced off-target organ effects that limit current systemic therapies. We have recently developed a unique light-activated drug delivery system whereby the drug is conjugated to an alkylcobalamin scaffold. Alkylcobalamins are actively transported into cells by transcobalamin receptors (TCblR), which are overexpressed in a variety of cancer types. We hope to utilize this cobalamin scaffold technology for drug delivery in pancreatic adenocarcinoma (PDAC) cancer. METHODS The ability of the cobalamin scaffold to selectively target PDAC was investigated by treating mice that had MIA PaCa-2 xenografts with an alkylcobalamin labeled with the fluorophore Bodipy650 (Bodipy650-cobalamin). The mice were imaged alive and organs as well as tumors were subsequently imaged ex vivo. In addition, we examined the potential of the cobalamin scaffold to deliver drugs to orthotopic pancreas MIA PaCa-2 tumors with Bodipy650-cobalamin. We determined the light dose required for release of cargo from the cobalamin scaffold by examining the fluorescence increase of Bodipy650-cobalamin in response to red light (650 nm). Finally, we probed the ability of the cobalamin scaffold to release cargo with increasing X-ray doses from a clinical linear accelerator. RESULTS We have found that Bodipy650-cobalamin was shown to localize in MIA PaCa-2 tumors, both in flank and orthotopic models. We quantified a light dose for red light release from the cobalamin scaffold that is within normal clinical doses required for photodynamic therapy. This derivative was also activated with clinical X-ray doses from a linear accelerator. CONCLUSIONS Tumor selectivity combined with fluorescence detection demonstrates the effectiveness of the vitamin B12 scaffold as a theranostic targeting agent. The activation of this scaffold with radiation from a linear accelerator shows potential for action as radiation-induced chemotherapy.
Collapse
|
20
|
Boussada M, Ali RB, Chahbi A, Abdelkarim M, Fradj MKB, Dziri C, Bokri K, Akacha AB, El May MV. A new Thiocyanoacetamide protects rat sperm cells from Doxorubicin-triggered cytotoxicity whereas Selenium shows low efficacy: In vitro approach. Toxicol In Vitro 2019; 61:104587. [PMID: 31271807 DOI: 10.1016/j.tiv.2019.06.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/29/2019] [Accepted: 06/29/2019] [Indexed: 11/26/2022]
Abstract
Doxorubicin (DOX) exhibits a wide-ranging spectrum of antitumor activities which maintain its clinical use despite its devastating impact on highly proliferating cells. The present work was designed to develop a new approach which aims to protect male germ cells from DOX cytotoxicity. Thus, an assessment of the protective potential of a new thioamide analog (thiocyanoacetamide; TA) compared to selenium (Se) was performed in rat sperms exposed to DOX in vitro. Oxygen consumption rate (OCR) was measured after exposure to three different doses (0.5, 1, 1.5 and 2 μM) of DOX, Se or TA, and the suitable concentrations were selected for further studies afterwards. Motility, OCR in a time-dependent manner, glucose extracellular concentration and lipid peroxidation (LPO) were measured. Fatty acid (FA) content was assessed by gas chromatography (GC-FID). Cell death, superoxide anion (O2-), mitochondrial membrane potential (MMP), and DNA damage were evaluated by flow cytometry. TA association with DOX increased OCR and glucose uptake, improved cell survival and decreased DNA damage. The co-administration of DOX with Se increased OCR, significantly prevented O2- overproduction, and decreased LPO. Collected data brought new insights regarding this transformed TA, which showed better efficiency than Se in reducing DOX cytotoxic stress in sperms.
Collapse
Affiliation(s)
- Marwa Boussada
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia.
| | - Ridha Ben Ali
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia; Unity of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Ahlem Chahbi
- Laboratory of Hematology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia.
| | - Mohamed Abdelkarim
- Laboratory of Hematology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Mohamed Kacem Ben Fradj
- UR05/08-08, LR99/ES/11, Department of Biochemistry, Rabta Hospital, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Chadli Dziri
- Unity of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Khouloud Bokri
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092 Tunis, Tunisia
| | - Azaiez Ben Akacha
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092 Tunis, Tunisia
| | - Michèle Véronique El May
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| |
Collapse
|
21
|
Cheng J, Wang X, Qiu L, Li Y, Marraiki N, Elgorban AM, Xue L. Green synthesized zinc oxide nanoparticles regulates the apoptotic expression in bone cancer cells MG-63 cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 202:111644. [PMID: 31770706 DOI: 10.1016/j.jphotobiol.2019.111644] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 09/26/2019] [Accepted: 10/04/2019] [Indexed: 12/26/2022]
Abstract
Management of degenerative spine pathologies frequently leads to the need for bone growth. Rehmanniae Radix (RR), a Chinese herbal formulation was found to exhibit numerous therapeutic properties including its potent effect against cancer cell lines. However, the underlying mechanism through which the Zinc oxide nanoparticles (ZnONPs) synthesized from Rehmanniae Radix exerts its anti-cancer activity against osteosarcoma cell line MG-63 needs to be explored. Therefore, the study was performed to evaluate the anticancer, cytotoxicity and apoptotic effectiveness of ZnONPs from RR against MG-63 cells. Characterization studies such UV-vis spectroscopy, FTIR, TEM and XRD analysis were performed. Cytotoxicity assay, mitochondrial membrane potential (MMP), morphological examination of cells and formation of reactive oxygen species (ROS), and apoptosis inducing ability of RR were evaluated by various procedures. Western blot analysis of apoptotic markers such as Bax, caspase-3 and caspase-9 were also performed. RR was found to inhibit growth of MG-63 cells at increasing dose. AO/EB staining confirmed the apoptotic efficacy of ZnONPs induced by RR in MG-63 cells. ZnONPs was also found to initiate increased generation of ROS and decreased MMP. Decreased MMP has resulted in increased levels of apoptotic proteins Bax, caspase-3 and caspase-9 and induction of apoptosis was substantiated by western blot analysis. The outcomes of the work propose that ZnONPs from RR exhibits strong anticancer action and inducing apoptosis on MG-63 cells via stimulating increased generation of ROS. Thus, ZnONPs from RR might be used as a hopeful drug target against several types of cancer cell lines.
Collapse
Affiliation(s)
- Jun Cheng
- Department of Orthopedics, Chongqing Three Gorges Central Hospital, Chongqing Province 404000, China
| | - Xiaofeng Wang
- Department of Neurosurgery, Weinan Central Hospital, Weinan, Shaanxi Province 714000, China
| | - Lei Qiu
- Department of Oncology, Zhucheng Hospital of Traditional Chinese Medicine, Zhucheng, Shandong Province 262200, China
| | - Yunkai Li
- Emergency Surgery the No.4 Hospital Jinan, Shangdong Province 250031, China
| | - Najat Marraiki
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdallah M Elgorban
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Li Xue
- Department of Orthopaedics, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiao Tong University Medical School, Chengdu, Sichuan Province 610031, China.
| |
Collapse
|
22
|
Nieto C, Vega MA, Enrique J, Marcelo G, Martín Del Valle EM. Size Matters in the Cytotoxicity of Polydopamine Nanoparticles in Different Types of Tumors. Cancers (Basel) 2019; 11:E1679. [PMID: 31671761 PMCID: PMC6896006 DOI: 10.3390/cancers11111679] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
Polydopamine has acquired great relevance in the field of nanomedicine due to its physicochemical properties. Previously, it has been reported that nanoparticles synthetized from this polymer are able to decrease the viability of breast and colon tumor cells. In addition, it is well known that the size of therapeutic particles plays an essential role in their effect. As a consequence, the influence of this parameter on the cytotoxicity of polydopamine nanoparticles was studied in this work. For this purpose, polydopamine nanoparticles with three different diameters (115, 200 and 420 nm) were synthetized and characterized. Their effect on the viability of distinct sorts of human carcinomas (breast, colon, liver and lung) and stromal cells was investigated, as well as the possible mechanisms that could be responsible for such cytotoxicity. Moreover, polydopamine nanoparticles were also loaded with doxorubicin and the therapeutic action of the resulting nanosystem was analyzed. As a result, it was demonstrated that a smaller nanoparticle size is related to a more enhanced antiproliferative activity, which may be a consequence of polydopamine's affinity for iron ions. Smaller nanoparticles would be able to adsorb more lysosomal Fe3+ and, when they are loaded with doxorubicin, a synergistic effect can be achieved.
Collapse
Affiliation(s)
- Celia Nieto
- Departamento de Ingeniería Química y Textil, Facultad de Ciencias Químicas, Universidad de Salamanca, 37008 Salamanca, Spain.
| | - Milena A Vega
- Departamento de Ingeniería Química y Textil, Facultad de Ciencias Químicas, Universidad de Salamanca, 37008 Salamanca, Spain.
| | - Jesús Enrique
- Departamento de Ingeniería Química y Textil, Facultad de Ciencias Químicas, Universidad de Salamanca, 37008 Salamanca, Spain.
| | - Gema Marcelo
- Departamento de Química Analítica, Química Física e Ingeniería Química, Facultad de Farmacia, Universidad de Alcalá, 28801 Alcalá de Henares (Madrid), Spain.
| | - Eva M Martín Del Valle
- Departamento de Ingeniería Química y Textil, Facultad de Ciencias Químicas, Universidad de Salamanca, 37008 Salamanca, Spain.
| |
Collapse
|
23
|
Šťastný M, Štengl V, Štenglová-Netíková I, Šrámová-Slušná M, Janoš P. Removal of anthracycline cytostatics from aquatic environment: Comparison of nanocrystalline titanium dioxide and decontamination agents. PLoS One 2019; 14:e0223117. [PMID: 31603899 PMCID: PMC6788709 DOI: 10.1371/journal.pone.0223117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/13/2019] [Indexed: 11/29/2022] Open
Abstract
Anthracyclines are a class of pharmaceuticals used in cancer treatment have the potential to negatively impact the environment. To study the possibilities of anthracyclines (represented by pirarubicin and valrubicin) removal, chemical inactivation using NaOH (0.01 M) and NaClO (5%) as decontamination agents and adsorption to powdered nanocrystalline titanium dioxide (TiO2) were compared. The titanium dioxide (TiO2) nanoparticles were prepared via homogeneous precipitation of an aqueous solution of titanium (IV) oxy-sulfate (TiOSO4) at different amount (5-120 g) with urea. The as-prepared TiO2 samples were characterized by XRD, HRSEM and nitrogen physisorption. The adsorption process of anthracycline cytostatics was determined followed by high-performance liquid chromatography coupled with mass spectrometry (LC-MS) and an in-situ Diffuse Reflectance Infrared Fourier Transform Spectroscopy (DRIFTS) technique. It was found that NaClO decomposes anthracyclines to form various transformation products (TPs). No TPs were identified after the reaction of valrubicin with a NaOH solution as well as in the presence of TiO2 nanoparticles. The best degree of removal, 100% of pirarubicin and 85% of valrubicin, has been achieved in a sample with 120 grams of TiOSO4 (TIT120) and TiO2 with 60 grams (TIT60), respectively.
Collapse
Affiliation(s)
- Martin Šťastný
- Institute of Inorganic Chemistry of the Czech Academy of Sciences, Řež, Czech Republic
| | - Václav Štengl
- Institute of Inorganic Chemistry of the Czech Academy of Sciences, Řež, Czech Republic
| | | | | | - Pavel Janoš
- Faculty of the Environment, J.E.Purkyně University in Ústí nad Labem, Ústí nad Labem, Czech Republic
| |
Collapse
|
24
|
Hernández Montoto A, Llopis‐Lorente A, Gorbe M, M. Terrés J, Cao‐Milán R, Díaz de Greñu B, Alfonso M, Ibañez J, Marcos MD, Orzáez M, Villalonga R, Martínez‐Máñez R, Sancenón F. Janus Gold Nanostars–Mesoporous Silica Nanoparticles for NIR‐Light‐Triggered Drug Delivery. Chemistry 2019; 25:8471-8478. [DOI: 10.1002/chem.201900750] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Indexed: 01/28/2023]
Affiliation(s)
- Andy Hernández Montoto
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
| | - Antoni Llopis‐Lorente
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Mónica Gorbe
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de, Enfermedades y Nanomedicina ValenciaUniversitat Politècnica de, València, Centro de Investigación Príncipe Felipe València Spain
| | - José M. Terrés
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
| | - Roberto Cao‐Milán
- Facultad de QuímicaUniversidad de la Habana Calle Zapata, s/n 10400 La Habana Cuba
| | - Borja Díaz de Greñu
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - María Alfonso
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Javier Ibañez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
| | - María D. Marcos
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
- Departamento de QuímicaUniversitat Politècnica de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de, Enfermedades y Nanomedicina ValenciaUniversitat Politècnica de, València, Centro de Investigación Príncipe Felipe València Spain
| | - Mar Orzáez
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de, Enfermedades y Nanomedicina ValenciaUniversitat Politècnica de, València, Centro de Investigación Príncipe Felipe València Spain
| | - Reynaldo Villalonga
- Department of Analytical ChemistryFaculty of ChemistryComplutense University of Madrid Av. Complutense, s/n 28040 Madrid Spain
| | - Ramón Martínez‐Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
- Departamento de QuímicaUniversitat Politècnica de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de, Enfermedades y Nanomedicina ValenciaUniversitat Politècnica de, València, Centro de Investigación Príncipe Felipe València Spain
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y, Desarrollo Tecnológico (IDM)Universitat Politècnica de ValènciaUniversitat de València València Spain
- Departamento de QuímicaUniversitat Politècnica de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de, Enfermedades y Nanomedicina ValenciaUniversitat Politècnica de, València, Centro de Investigación Príncipe Felipe València Spain
| |
Collapse
|
25
|
Dupart PS, Mitra K, Lyons CE, Hartman MCT. Photo-controlled delivery of a potent analogue of doxorubicin. Chem Commun (Camb) 2019; 55:5607-5610. [PMID: 31021353 PMCID: PMC6643956 DOI: 10.1039/c9cc02050k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Highly cytotoxic agents have found an important niche in targeted anticancer therapy. Here we develop a new light release strategy for the targeting of one of these agents, 2-pyrrolinodoxorubicin, showing dramatic enhancements in toxicity with light and single digit nM potency.
Collapse
Affiliation(s)
- Patrick S Dupart
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St, Richmond, 23284, VA, USA. and Massey Cancer Center, Virginia Commonwealth University, 401 College St, Richmond, 23219, Virginia, USA
| | - Koushambi Mitra
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St, Richmond, 23284, VA, USA. and Massey Cancer Center, Virginia Commonwealth University, 401 College St, Richmond, 23219, Virginia, USA
| | - Charles E Lyons
- Massey Cancer Center, Virginia Commonwealth University, 401 College St, Richmond, 23219, Virginia, USA
| | - Matthew C T Hartman
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St, Richmond, 23284, VA, USA. and Massey Cancer Center, Virginia Commonwealth University, 401 College St, Richmond, 23219, Virginia, USA
| |
Collapse
|
26
|
Dion J, Minoshima F, Saito S, Kiyoi K, Hasehira K, Tateno H. Photoactivable Elimination of Tumorigenic Human Induced Pluripotent Stem Cells by Using a Lectin-Doxorubicin Prodrug Conjugate. Chembiochem 2019; 20:1606-1611. [PMID: 30737871 DOI: 10.1002/cbic.201900086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Indexed: 12/15/2022]
Abstract
Human pluripotent stem cells (hPSCs) are attractive resources for regenerative medicine, but medical applications are hindered by their tumorigenic potential. Previously, a hPSC-specific lectin probe, rBC2LCN, was identified through comprehensive glycome analysis by using high-density lectin microarrays. Herein, a lectin-doxorubicin (DOX) prodrug conjugate, with controllable photolysis activation for the elimination of tumorigenic human induced pluripotent stem cells, has been developed. rBC2LCN was fused with a biotin-binding protein, tamavidin (BC2Tama), and the fusion protein was expressed in Escherichia coli and purified by means of affinity chromatography. BC2Tama was then conjugated with doxorubicin-photocleavable biotin (DOXPCB). The BC2Tama-DOXPCB conjugates were observed to bind to hPSCs followed by internalization. Upon exposure to ultraviolet light, DOX was released inside the cells, which allowed specific killing of the hPSCs. Thus, BC2Tama-DOXPCB should be useful for the targeted elimination of hPSCs contained in hPSC-derived cell therapy products. This is the first report of the generation of lectin-prodrug conjugates. BC2Tama should be applicable for the targeted delivery of various types of biotinylated compounds into hPSCs.
Collapse
Affiliation(s)
- Johann Dion
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba Central 2, 1-1-1 Umezono, Tsukuba, Ibaraki, 305-8568, Japan
| | - Fumi Minoshima
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba Central 2, 1-1-1 Umezono, Tsukuba, Ibaraki, 305-8568, Japan
| | - Sayoko Saito
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba Central 2, 1-1-1 Umezono, Tsukuba, Ibaraki, 305-8568, Japan
| | - Kayo Kiyoi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba Central 2, 1-1-1 Umezono, Tsukuba, Ibaraki, 305-8568, Japan
| | - Kayo Hasehira
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba Central 2, 1-1-1 Umezono, Tsukuba, Ibaraki, 305-8568, Japan
| | - Hiroaki Tateno
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba Central 2, 1-1-1 Umezono, Tsukuba, Ibaraki, 305-8568, Japan.,Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| |
Collapse
|
27
|
Pereira SGT, Hudoklin S, Kreft ME, Kostevsek N, Stuart MCA, Al-Jamal WT. Intracellular Activation of a Prostate Specific Antigen-Cleavable Doxorubicin Prodrug: A Key Feature Toward Prodrug-Nanomedicine Design. Mol Pharm 2019; 16:1573-1585. [DOI: 10.1021/acs.molpharmaceut.8b01257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sara G. T. Pereira
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, U.K
| | - Samo Hudoklin
- University of Ljubljana, Faculty of Medicine, Institute of Cell Biology, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- University of Ljubljana, Faculty of Medicine, Institute of Cell Biology, Ljubljana, Slovenia
| | - Nina Kostevsek
- Department for Nanostructured Materials, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Marc C. A. Stuart
- Electron Microscopy, University of Groningen, Nijenborgh 7, 9747AG Groningen, The Netherlands
| | - Wafa T. Al-Jamal
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Rd, Belfast BT9 7BL, U.K
| |
Collapse
|
28
|
Tamura R, Balabanova A, Frakes SA, Bargmann A, Grimm J, Koch TH, Yin H. Photoactivatable Prodrug of Doxazolidine Targeting Exosomes. J Med Chem 2019; 62:1959-1970. [PMID: 30703330 DOI: 10.1021/acs.jmedchem.8b01508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Natural lipid nanocarriers, exosomes, carry cell-signaling materials such as DNA and RNA for intercellular communications. Exosomes derived from cancer cells contribute to the progression and metastasis of cancer cells by transferring oncogenic signaling molecules to neighboring and remote premetastatic sites. Therefore, applying the unique properties of exosomes for cancer therapy has been expected in science, medicine, and drug discovery fields. Herein, we report that an exosome-targeting prodrug system, designated MARCKS-ED-photodoxaz, could spatiotemporally control the activation of an exquisitely cytotoxic agent, doxazolidine (doxaz), with UV light. The MARCKS-ED peptide enters a cell by forming a complex with the exosomes in situ at its plasma membrane and in the media. MARCKS-ED-photodoxaz releases doxaz under near-UV irradiation to inhibit cell growth with low nanomolar IC50 values. The MARCKS-ED-photodoxaz system targeting exosomes and utilizing photochemistry will potentially provide a new approach for the treatment of cancer, especially for highly progressive and invasive metastatic cancers.
Collapse
Affiliation(s)
- Ryo Tamura
- Molecular Pharmacology Program , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | | | | | | | - Jan Grimm
- Molecular Pharmacology Program , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | | | - Hang Yin
- School of Pharmaceutical Sciences, Tsinghua University-Peking University Joint Center for Life Sciences , Tsinghua University , Beijing 100082 , China
| |
Collapse
|
29
|
Hernández-Montoto A, Gorbe M, Llopis-Lorente A, Terrés JM, Montes R, Cao-Milán R, Díaz de Greñu B, Alfonso M, Orzaez M, Marcos MD, Martínez-Máñez R, Sancenón F. A NIR light-triggered drug delivery system using core–shell gold nanostars–mesoporous silica nanoparticles based on multiphoton absorption photo-dissociation of 2-nitrobenzyl PEG. Chem Commun (Camb) 2019; 55:9039-9042. [DOI: 10.1039/c9cc04260a] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Gold nanostars coated with a mesoporous silica shell and functionalised with PEG containing photolabile 2-nitrobenzyl moieties released doxorubicin after NIR light irradiation.
Collapse
|
30
|
Gao Y, Hou M, Yang R, Zhang L, Xu Z, Kang Y, Xue P. Transdermal delivery of therapeutics through dissolvable gelatin/sucrose films coated on PEGDA microneedle arrays with improved skin permeability. J Mater Chem B 2019; 7:7515-7524. [DOI: 10.1039/c9tb01994d] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Microneedles are primarily designed for enhancing transdermal drug delivery in a minimally invasive manner.
Collapse
Affiliation(s)
- Ya Gao
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University)
- Ministry of Education
- School of Materials and Energy
- Southwest University
- Chongqing 400715
| | - Mengmeng Hou
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University)
- Ministry of Education
- School of Materials and Energy
- Southwest University
- Chongqing 400715
| | - Ruihao Yang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University)
- Ministry of Education
- School of Materials and Energy
- Southwest University
- Chongqing 400715
| | - Lei Zhang
- Institute of Sericulture and Systems Biology, Southwest University
- Chongqing
- China
| | - Zhigang Xu
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University)
- Ministry of Education
- School of Materials and Energy
- Southwest University
- Chongqing 400715
| | - Yuejun Kang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University)
- Ministry of Education
- School of Materials and Energy
- Southwest University
- Chongqing 400715
| | - Peng Xue
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University)
- Ministry of Education
- School of Materials and Energy
- Southwest University
- Chongqing 400715
| |
Collapse
|
31
|
Foo SC, Yusoff FM, Imam MU, Foo JB, Ismail N, Azmi NH, Tor YS, Khong NMH, Ismail M. Increased fucoxanthin in Chaetoceros calcitrans extract exacerbates apoptosis in liver cancer cells via multiple targeted cellular pathways. ACTA ACUST UNITED AC 2018; 21:e00296. [PMID: 30581767 PMCID: PMC6296166 DOI: 10.1016/j.btre.2018.e00296] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/14/2018] [Accepted: 12/03/2018] [Indexed: 02/08/2023]
Abstract
Both treatments inhibited cancer proliferation in a time and dose dependent manner. FxRF treatment were effective in inducing apoptosis in HepG2 cells than crude extract. Treatments stimulated regulation in cell signalling, apoptotic and antioxidant genes.
In this study, anti-proliferative effects of C. calcitrans extract and its fucoxanthin rich fraction (FxRF) were assessed on human liver HepG2 cancer cell line. Efficacy from each extract was determined by cytotoxicity assay, morphological observation, and cell cycle analysis. Mechanisms of action observed were evaluated using multiplex gene expression analysis. Results showed that CME and FxRF induced cytotoxicity to HepG2 cells in a dose and time-dependent manner. FxRF (IC50: 18.89 μg.mL−1) was found to be significantly more potent than CME (IC50: 87.5 μg.mL−1) (p < 0.05). Gene expression studies revealed that anti-proliferative effects in treated cells by C. calcitrans extracts were mediated partly through the modulation of numerous genes involved in cell signaling (AKT1, ERK1/2, JNK), apoptosis (BAX, BID, Bcl-2, APAF, CYCS) and oxidative stress (SOD1, SOD2, CAT). Overall, C. calcitrans extracts demonstrated effective intervention against HepG2 cancer cells where enhanced apoptotic activities were observed with increased fucoxanthin content.
Collapse
Affiliation(s)
- Su Chern Foo
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.,School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Fatimah Md Yusoff
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.,The International Institute of Aquaculture and Aquatic Science, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor Darul Ehsan, Malaysia.,Department of Aquaculture, Faculty of Agriculture, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia
| | - Mustapha Umar Imam
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.,Department of Medical Biochemistry, College of Health Sciences, Usmanu Danfodio University, Sokoto, Nigeria
| | - Jhi Biau Foo
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.,School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, No. 1 Jalan Taylor's, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Norsharina Ismail
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia
| | - Nur Hanisah Azmi
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.,Department of Cell and Molecular Biology, Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Yin Sim Tor
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.,School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, No. 1 Jalan Taylor's, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Nicholas M H Khong
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia
| | - Maznah Ismail
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
32
|
Borišev I, Mrđanovic J, Petrovic D, Seke M, Jović D, Srđenović B, Latinovic N, Djordjevic A. Nanoformulations of doxorubicin: how far have we come and where do we go from here? NANOTECHNOLOGY 2018; 29:332002. [PMID: 29798934 DOI: 10.1088/1361-6528/aac7dd] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Nanotechnology, focused on discovery and development of new pharmaceutical products is known as nanopharmacology, and one research area this branch is engaged in are nanopharmaceuticals. The importance of being nano has been particularly emphasized in scientific areas dealing with nanomedicine and nanopharmaceuticals. Nanopharmaceuticals, their routes of administration, obstacles and solutions concerning their improved application and enhanced efficacy have been briefly yet comprehensively described. Cancer is one of the leading causes of death worldwide and evergrowing number of scientific research on the topic only confirms that the needs have not been completed yet and that there is a wide platform for improvement. This is undoubtedly true for nanoformulations of an anticancer drug doxorubicin, where various nanocarrriers were given an important role to reduce the drug toxicity, while the efficacy of the drug was supposed to be retained or preferably enhanced. Therefore, we present an interdisciplinary comprehensive overview of interdisciplinary nature on nanopharmaceuticals based on doxorubicin and its nanoformulations with valuable information concerning trends, obstacles and prospective of nanopharmaceuticals development, mode of activity of sole drug doxorubicin and its nanoformulations based on different nanocarriers, their brief descriptions of biological activity through assessing in vitro and in vivo behavior.
Collapse
Affiliation(s)
- Ivana Borišev
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, Novi Sad, Serbia
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Zhang NN, Yu RS, Xu M, Cheng XY, Chen CM, Xu XL, Lu CY, Lu KJ, Chen MJ, Zhu ML, Weng QY, Hui JG, Zhang Q, Du YZ, Ji JS. Visual targeted therapy of hepatic cancer using homing peptide modified calcium phosphate nanoparticles loading doxorubicin guided by T1 weighted MRI. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2167-2178. [PMID: 30017962 DOI: 10.1016/j.nano.2018.06.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/18/2018] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
Abstract
Effective treatment and real-time monitoring of hepatic cancer are essential. A multifunctional calcium phosphate nanoparticles loading chemotherapeutic agent doxorubicin and magnetic resonance imaging contrast agent diethylenetriaminepentaacetic acid gadolinium (A54-CaP/Gd-DTPA/DOX) was developed for visual targeted therapy of hepatic cancer via T1-weighted MRI in real-time. A54-CaP/Gd-DTPA/DOX exhibited a higher longitudinal relaxivity (6.02 mM-1 s-1) than commercial MR contrast agent Gd-DTPA (3.3765 mM-1 s-1). The DOX release from the nanoparticles exhibited a pH dependent behavior. The cellular uptake results showed that the internalization of A54-CaP/Gd-DTPA/DOX into BEL-7402 cells was1.9-fold faster than that of HepG2 cells via A54 binding. In vivo experiments presented that A54-CaP/Gd-DTPA/DOX had higher distribution and longer retention time in tumor tissue than CaP/Gd-DTPA/DOX and free DOX, and also displayed great antitumor efficacy (95.38% tumor inhibition rate) and lower toxicity. Furthermore, the Gd-DTPA entrapped in the nanoparticles could provide T1-weighted MRI for real-time monitoring the progress of tumor treatment.
Collapse
Affiliation(s)
- Nan-Nan Zhang
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China; Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ri-Sheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Min Xu
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China
| | - Xing-Yao Cheng
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China
| | - Chun-Miao Chen
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China
| | - Xiao-Ling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chen-Ying Lu
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China
| | - Kong-Jun Lu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Min-Jiang Chen
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China
| | - Meng-Lu Zhu
- Department of Pharmacy, Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, PR China
| | - Qiao-You Weng
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China
| | - Jun-Guo Hui
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China
| | - Qian Zhang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Jian-Song Ji
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, China.
| |
Collapse
|
34
|
Li Y, Yu Q, Gao P, Yang H, Huang T, Liu S, Zhao Q. Highly efficient organic photosensitizer with aggregation-induced emission for imaging-guided photodynamic ablation of cancer cells. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2018.05.088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
35
|
Guo XJ, Fan XJ, Qiao B, Ge ZQ. A lipophilic prodrug of Danshensu: preparation, characterization, and in vitro and in vivo evaluation. Chin J Nat Med 2018; 15:355-362. [PMID: 28558871 DOI: 10.1016/s1875-5364(17)30056-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Indexed: 01/17/2023]
Abstract
Danshensu [3-(3, 4-dihydroxyphenyl) lactic acid, DSS], one of the significant cardioprotective components, is extracted from the root of Salvia miltiorrhiza. In the present study, an ester prodrug of Danshensu (DSS), palmitoyl Danshensu (PDSS), was synthesized with the aim to improve its oral bioavailability and prolong its half-life. The in vitro experiments were carried out to evaluate the physicochemical properties and stability of PDSS. Although the solubility of PDSS in water was only 0.055 mg·mL-1, its solubility in FaSSIF and FeSSIF reached 4.68 and 9.08 mg·mL-1, respectively. Octanol-water partition coefficient (log P) was increased from -2.48 of DSS to 1.90 of PDSS. PDSS was relatively stable in the aqueous solution in pH range from 5.6 to 7.4. Furthermore, the pharmacokinetics in rats was evaluated after oral administration of PDSS and DSS. AUC and t1/2 of PDSS were enhanced up to 9.8-fold and 2.2-fold, respectively, compared to that of DSS. Cmax was 1.67 ± 0.11 μg·mL-1 for PDSS and 0.81 ± 0.06 μg·mL-1 for DSS. Thus, these results demonstrated that PDSS had much higher oral bioavailability and longer circulation time than its parent drug.
Collapse
Affiliation(s)
- Xue-Jiao Guo
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering, Tianjin 300072, China
| | - Xue-Jiao Fan
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering, Tianjin 300072, China
| | - Bin Qiao
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering, Tianjin 300072, China
| | - Zhi-Qiang Ge
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering, Tianjin 300072, China.
| |
Collapse
|
36
|
Wang X, Zhang Y, Lin C, Zhong W. Sol-gel derived terbium-containing mesoporous bioactive glasses nanospheres: In vitro hydroxyapatite formation and drug delivery. Colloids Surf B Biointerfaces 2017; 160:406-415. [PMID: 28965080 DOI: 10.1016/j.colsurfb.2017.09.051] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/07/2017] [Accepted: 09/22/2017] [Indexed: 11/20/2022]
Abstract
Terbium (Tb) doped mesoporous bioactive glasses (Tb/MBG) nanospheres were successfully synthesized by a facile sol-gel method using cetyl trimethyl ammonium bromide (CTAB) as the template. Results indicated that Tb/MBG had spherical morphology (100-200nm), higher specific surface area (250-350m2/g) and narrow mesopore size distribution (2-3nm). In order to investigate the effects of Tb on the in vitro bioactivity, prepared Tb/MBG nanospheres were soaking in simulated body fluid (SBF) for 3 days, and results indicated incorporation Tb ions in the MBG nanospheres could improve the hydroxyapatite formation ability. In addition, Tb/MBG nanospheres showed controlled release property of anti-cancer drugs (DOX) and distinct degradation in PBS with different pH values. Their release mechanism can be explained by Fickian diffusion according the Higuchi model, and the delivery of DOX from Tb/MBG nanospheres can be dominated by changing the doping concentration of Tb and the values of pH. In addition, the cytotoxicity of Tb/MBG nanospheres was assessed using a cell counting kit-8 (CCK-8), and results showed that the synthesized Tb/MBG nanospheres at low concentration had no significant cytotoxicity in MC3T3 cells. These all note that this material is a promising candidate for the therapy of bone tissue regeneration.
Collapse
Affiliation(s)
- Xiang Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Ying Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China.
| | - Chuan Lin
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Wenxing Zhong
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| |
Collapse
|
37
|
Kong L, Poulcharidis D, Schneider GF, Campbell F, Kros A. Spatiotemporal Control of Doxorubicin Delivery from "Stealth-Like" Prodrug Micelles. Int J Mol Sci 2017; 18:E2033. [PMID: 28937592 PMCID: PMC5666715 DOI: 10.3390/ijms18102033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/14/2017] [Accepted: 09/18/2017] [Indexed: 12/14/2022] Open
Abstract
In the treatment of cancer, targeting of anticancer drugs to the tumor microenvironment is highly desirable. Not only does this imply accurate tumor targeting but also minimal drug release en route to the tumor and maximal drug release once there. Here we describe high-loading, "stealth-like" doxorubicin micelles as a pro-drug delivery system, which upon light activation, leads to burst-like doxorbicin release. Through this approach, we show precise spatiotemporal control of doxorubicin delivery to cells in vitro.
Collapse
Affiliation(s)
- Li Kong
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Dimitrios Poulcharidis
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Gregory F Schneider
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Frederick Campbell
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Alexander Kros
- Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| |
Collapse
|
38
|
Zheng N, Wang X, Wang Y, Xu G, Zhang H, Dai W, He B, Zhang Q, Ji J, Wang X. A sensitive liquid chromatography/electrospray tandem mass spectroscopy method for simultaneous quantification of a disulfide bond doxorubicin conjugation prodrug and activated doxorubicin: Application to cellular pharmacokinetic and catabolism studies. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1065-1066:96-103. [PMID: 28957779 DOI: 10.1016/j.jchromb.2017.09.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/11/2017] [Accepted: 09/20/2017] [Indexed: 11/29/2022]
Abstract
In recent years, drug conjugates as a prodrug strategy have been widely studied, especially combined with nanotechnology. Disulfide-linked doxorubicin drug-drug conjugate (DOX-S-S-DOX) nanoparticles, have recently been developed as a doxorubicin prodrug nanoparticles with greater anticancer activity and less toxicity than doxorubicin in vivo, while its intracellular kinetics and metabolism is unclear which may provide us with a deeper understanding of its pharmacological mechanism and antitumor effect. Hence, in this study, a rapid and sensitive ultra high-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was developed to detect doxorubicin (DOX) activated from DOX-S-S-DOX, as well as the prodrug itself in human breast cancer tumor cells (MCF-7). Sample preparation involved acetonitrile precipitation to extract the analytes simultaneously and bath sonication to remove intercalated DOX from DNA. The calibration range was 3-60ng/mL for DOX and 20-400ng/mL for DOX-S-S-DOX with the correlation coefficients (r2)≥0.99, using daunorubicin as internal standard (IS). The inter- and intra-assay precision (relative standard deviation, RSD%) of quality control samples was in the acceptable range (<15%) and relative error (RE%) for accuracy was between -5.35 and 9.18% for all analytes. Recovery (59.28-69.53% for DOX-S-S-DOX and 99.13-100.10% for DOX) and matrix effect (99.69-111.19%) was consistent, precise, and reproducible at different quality control levels in accordance with FDA guidance. Stability studies showed that DOX-S-S-DOX was unstable both during the bench-top and long-term storage, while the stability during sample preparation and LC-MS runtime was suitable for all the analytes. Hence, the samples should be prepared as soon as possible at the time point to prevent the catabolism of DOX-S-S-DOX. The assay was successfully used in the cellular metabolism and pharmacokinetics study of DOX-S-S-DOX and it may give a clue to explore analytical methods of other prodrug forms of DOX.
Collapse
Affiliation(s)
- Nan Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), National Drug Clinical Trial Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yaoqi Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Guobing Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), National Drug Clinical Trial Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), National Drug Clinical Trial Center, Peking University Cancer Hospital & Institute, Beijing 100142, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
39
|
Schutt C, Ibsen S, Zahavy E, Aryal S, Kuo S, Esener S, Berns M, Esener S. Drug Delivery Nanoparticles with Locally Tunable Toxicity Made Entirely from a Light-Activatable Prodrug of Doxorubicin. Pharm Res 2017; 34:2025-2035. [PMID: 28791550 DOI: 10.1007/s11095-017-2205-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/08/2017] [Indexed: 12/20/2022]
Abstract
PURPOSE A major challenge facing nanoparticle-based delivery of chemotherapy agents is the natural and unavoidable accumulation of these particles in healthy tissue resulting in local toxicity and dose-limiting side effects. To address this issue, we have designed and characterized a new prodrug nanoparticle with controllable toxicity allowing a locally-delivered light trigger to convert the payload of the particle from a low to a high toxicity state. METHODS The nanoparticles are created entirely from light-activatable prodrug molecules using a nanoprecipitation process. The prodrug is a conjugate of doxorubicin and photocleavable biotin (DOX-PCB). RESULTS These DOX-PCB nanoparticles are 30 times less toxic to cells than doxorubicin, but can be activated to release pure therapeutic doxorubicin when exposed to 365 nm light. These nanoparticles have an average diameter of around 100 nm and achieve the maximum possible prodrug loading capacity since no support structure or coating is required to prevent loss of prodrug from the nanoparticle. CONCLUSIONS These light activatable nanoparticles demonstrate tunable toxicity and can be used to facilitate future therapy development whereby light delivered specifically to the tumor tissue would locally convert the nanoparticles to doxorubicin while leaving nanoparticles accumulated in healthy tissue in the less toxic prodrug form.
Collapse
Affiliation(s)
- Carolyn Schutt
- Department of Bioengineering, University of California San Diego, 9500 Gilman Dr. MC 0412, La Jolla, California, 92093-0412, USA.
| | - Stuart Ibsen
- Moores Cancer Center, University of California San Diego, La Jolla, California, 92093, USA
| | - Eran Zahavy
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, P.O. Box 19, 74100, Ness-Ziona, Israel
| | - Santosh Aryal
- Department of Chemistry, Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, Kansas, 66506, USA
| | - Stacey Kuo
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, USA
| | - Selin Esener
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, USA
| | - Michael Berns
- Department of Bioengineering, University of California San Diego, 9500 Gilman Dr. MC 0412, La Jolla, California, 92093-0412, USA
| | - Sadik Esener
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, USA
| |
Collapse
|
40
|
Choi JS, Doh KO, Kim BK, Seu YB. Synthesis of cholesteryl doxorubicin and its anti-cancer activity. Bioorg Med Chem Lett 2017; 27:723-728. [DOI: 10.1016/j.bmcl.2017.01.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/30/2016] [Accepted: 01/14/2017] [Indexed: 01/12/2023]
|
41
|
Štenglová Netíková IR, Slušná M, Tolasz J, Št'astný M, Popelka Š, Štengl V. A new possible way of anthracycline cytostatics decontamination. NEW J CHEM 2017. [DOI: 10.1039/c6nj03051c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cytostatics decontamination based on using an active sorbent of titania, which can adsorb and completely degrade anthracycline anticancer drugs.
Collapse
Affiliation(s)
| | - Michaela Slušná
- Department of Material Chemistry
- Institute of Inorganic Chemistry ASCR v.v.i
- 250 68 Husinec-Rez
- Czech Republic
- Faculty of the Environment
| | - Jakub Tolasz
- Department of Material Chemistry
- Institute of Inorganic Chemistry ASCR v.v.i
- 250 68 Husinec-Rez
- Czech Republic
| | - Martin Št'astný
- Department of Material Chemistry
- Institute of Inorganic Chemistry ASCR v.v.i
- 250 68 Husinec-Rez
- Czech Republic
- Faculty of the Environment
| | - Štěpán Popelka
- Department of Biomaterials and Bioanalogous Polymer Systems
- Institute of Macromolecular Chemistry ASCR v.v.i
- 162 06 Praha 6
- Czech Republic
| | - Václav Štengl
- 1st Faculty of Medicine
- Charles University in Prague
- Department of Oncology
- 116 36 Praha 1
- Czech Republic
| |
Collapse
|
42
|
Preclinical Evaluation of an Epidermal Growth Factor Receptor-Targeted Doxorubicin-Peptide Conjugate: Toxicity, Biodistribution, and Efficacy in Mice. J Pharm Sci 2016; 105:639-649. [PMID: 26869425 DOI: 10.1016/j.xphs.2015.10.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/30/2015] [Accepted: 10/16/2015] [Indexed: 01/14/2023]
Abstract
Doxorubicin (DOX) is known to induce apoptosis and necrosis in healthy tissue resulting in unwanted toxicities. To improve the ability of DOX to more specifically target tumors and minimize undesirable side effects, conjugation of DOX with epidermal growth factor receptor (EGFR)--binding peptide (DOX-EBP) has been developed to deliver DOX to EGFR-overexpressing neoplastic cells. Here, we investigated whether DOX-EBP was able to reduce toxicity and enhance anticancer efficacy in vivo through receptor-mediated targeted delivery system. Nude mice were treated with DOX or DOX-EBP to estimate general toxicity, normal tissue damage, biodistribution, and antitumor efficacy. In addition, the expression levels of EGFR in tumor tissues and normal organs were investigated by Western blotting, and their mRNA expression was analyzed by reverse transcription PCR. This study demonstrated that DOX-EBP was able to effectively decrease the distribution of DOX in normal tissues without EGFR overexpressing and reduce DOX-induced toxicity. On the other hand, the research also confirmed that DOX-EBP was able to preferentially accumulate DOX in EGFR-overexpressing tumor tissues and showed the enhanced anticancer efficacy over free DOX. DOX-EBP could be used for receptor-targeted chemotherapy with less toxicity and greater efficacy of tumor cells overexpressing EGFR. DOX-EBP conjugate is a good therapeutic agent for cancer treatment.
Collapse
|
43
|
Yin Y, Fu C, Li M, Li X, Wang M, He L, Zhang LM, Peng Y. A pH-sensitive hyaluronic acid prodrug modified with lactoferrin for glioma dual-targeted treatment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 67:159-169. [DOI: 10.1016/j.msec.2016.05.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/30/2016] [Accepted: 05/05/2016] [Indexed: 11/25/2022]
|
44
|
Błauż A, Rychlik B, Makal A, Szulc K, Strzelczyk P, Bujacz G, Zakrzewski J, Woźniak K, Plażuk D. Ferrocene-Biotin Conjugates: Synthesis, Structure, Cytotoxic Activity and Interaction with Avidin. Chempluschem 2016; 81:1191-1201. [DOI: 10.1002/cplu.201600320] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Andrzej Błauż
- Cytometry Lab; Department of Molecular Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; 141/143 Pomorska St. 90-236 Łódź Poland
| | - Błażej Rychlik
- Cytometry Lab; Department of Molecular Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; 141/143 Pomorska St. 90-236 Łódź Poland
| | - Anna Makal
- Department of Chemistry; University of Warsaw; Pasteura, 1 02-093 Warszawa Poland
| | - Katarzyna Szulc
- Cytometry Lab; Department of Molecular Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; 141/143 Pomorska St. 90-236 Łódź Poland
| | - Paweł Strzelczyk
- Institute of Technical Biochemistry; Łódź University of Technology; Stefanowskiego 4/10 90-924 Łódź Poland
| | - Grzegorz Bujacz
- Institute of Technical Biochemistry; Łódź University of Technology; Stefanowskiego 4/10 90-924 Łódź Poland
| | - Janusz Zakrzewski
- Department of Organic Chemistry; Faculty of Chemistry; University of Łódź; Tamka 12 41-403 Łódź Poland
| | - Krzysztof Woźniak
- Department of Chemistry; University of Warsaw; Pasteura, 1 02-093 Warszawa Poland
| | - Damian Plażuk
- Department of Organic Chemistry; Faculty of Chemistry; University of Łódź; Tamka 12 41-403 Łódź Poland
| |
Collapse
|
45
|
Peira E, Chirio D, Battaglia L, Barge A, Chegaev K, Gigliotti CL, Ferrara B, Dianzani C, Gallarate M. Solid lipid nanoparticles carrying lipophilic derivatives of doxorubicin: preparation, characterization, andin vitrocytotoxicity studies. J Microencapsul 2016; 33:381-90. [DOI: 10.1080/02652048.2016.1202342] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
46
|
Tupal A, Sabzichi M, Ramezani F, Kouhsoltani M, Hamishehkar H. Dermal delivery of doxorubicin-loaded solid lipid nanoparticles for the treatment of skin cancer. J Microencapsul 2016; 33:372-80. [PMID: 27338131 DOI: 10.1080/02652048.2016.1200150] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Dermal delivery of Doxorubicin (Dox) would be an ideal way in maximising drug efficiency against skin cancer accompanying with minimising side effects. We investigated the potential of Dox-loaded Solid lipid nanoparticles (SLNs) for topical delivery against skin cancer. METHODS In vitro and in vivo cytotoxicity of optimised formulation were evaluated on murine melanoma (B16F10) cells by MTT assay and melanoma induced Balb/C mice, respectively. Animal study followed by histological analysis. RESULTS Optimised formulation showed mean particle size and encapsulation efficiency (EE) of 92 nm and 86% w/w (0.86% w/w value of encapsulated Dox in the lipid matrix), respectively. FTIR experiment confirmed drug-lipid interaction interpreting the observed high EE value for Dox. In vitro and in vivo results indicated the superiority of cytotoxic performance of Dox-loaded SLN compared to Dox solution. CONCLUSION Our findings may open the possibilities for the topical delivery of Dox to the skin cancerous tissues.
Collapse
Affiliation(s)
- Ailar Tupal
- a Biotechnology Research Center and Faculty of Pharmacy , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mehdi Sabzichi
- b Research Center for Pharmaceutical Nanotechnology and Students' Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Fatemeh Ramezani
- c Department of Biochemistry , School of Medicine, Shiraz University of Medical Sciences , Iran
| | - Maryam Kouhsoltani
- d Department of Oral & Maxillofacial Pathology, Faculty of Dentistry , Tabriz University of Medical Science , Tabriz , Iran
| | - Hamed Hamishehkar
- e Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
47
|
Kue CS, Kamkaew A, Burgess K, Kiew LV, Chung LY, Lee HB. Small Molecules for Active Targeting in Cancer. Med Res Rev 2016; 36:494-575. [PMID: 26992114 DOI: 10.1002/med.21387] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/29/2022]
Abstract
For the purpose of this review, active targeting in cancer research encompasses strategies wherein a ligand for a cell surface receptor expressed on tumor cells is used to deliver a cytotoxic or imaging cargo. This area of research is more than two decades old, but in those 20 and more years, how many receptors have been studied extensively? What kinds of the ligands are used for active targeting? Are they mostly naturally occurring molecules such as folic acid, or synthetic substances developed in campaigns for medicinal chemistry efforts? This review outlines the most important receptor or ligand combinations that have been used in active targeting to answer these questions, and therefore to address the most important one of all: is research in active targeting affording diminishing returns, or is this an area for which the potential far exceeds progress made so far?
Collapse
Affiliation(s)
- Chin S Kue
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Anyanee Kamkaew
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX, 77842
| | - Kevin Burgess
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX, 77842
| | - Lik V Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lip Y Chung
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Hong B Lee
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
48
|
Leonidova A, Mari C, Aebersold C, Gasser G. Selective Photorelease of an Organometallic-Containing Enzyme Inhibitor. Organometallics 2016. [DOI: 10.1021/acs.organomet.6b00029] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Anna Leonidova
- Department of Chemistry, University of Zurich, Winterthurerstrasse
190, CH-8057 Zurich, Switzerland
| | - Cristina Mari
- Department of Chemistry, University of Zurich, Winterthurerstrasse
190, CH-8057 Zurich, Switzerland
| | - Christine Aebersold
- Department of Chemistry, University of Zurich, Winterthurerstrasse
190, CH-8057 Zurich, Switzerland
| | - Gilles Gasser
- Department of Chemistry, University of Zurich, Winterthurerstrasse
190, CH-8057 Zurich, Switzerland
| |
Collapse
|
49
|
Leonidova A, Anstaett P, Pierroz V, Mari C, Spingler B, Ferrari S, Gasser G. Induction of Cytotoxicity through Photorelease of Aminoferrocene. Inorg Chem 2015; 54:9740-8. [PMID: 26440628 DOI: 10.1021/acs.inorgchem.5b01332] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Anna Leonidova
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Philipp Anstaett
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Vanessa Pierroz
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
- Institute
of Molecular Cancer Research, University of Zurich, Winterthurerstrasse
190, CH-8057 Zurich, Switzerland
| | - Cristina Mari
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Bernhard Spingler
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Stefano Ferrari
- Institute
of Molecular Cancer Research, University of Zurich, Winterthurerstrasse
190, CH-8057 Zurich, Switzerland
| | - Gilles Gasser
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| |
Collapse
|
50
|
Musa MA, Latinwo LM, Virgile C, Badisa VL, Gbadebo AJ. Synthesis and in vitro evaluation of 3-(4-nitrophenyl)coumarin derivatives in tumor cell lines. Bioorg Chem 2015; 58:96-103. [DOI: 10.1016/j.bioorg.2014.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 11/22/2014] [Accepted: 11/24/2014] [Indexed: 11/26/2022]
|