1
|
Arora M, Singh AK, Kumar A, Singh H, Pathak P, Grishina M, Yadav JP, Verma A, Kumar P. Semisynthetic phytochemicals in cancer treatment: a medicinal chemistry perspective. RSC Med Chem 2024; 15:3345-3370. [PMID: 39430100 PMCID: PMC11484407 DOI: 10.1039/d4md00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer is the uncontrolled proliferation of abnormal cells that invade other areas, spread to other organs, and cause metastases, which is the most common cause of death. A review of all FDA-approved new molecular entities (NMEs) shows that natural products and derivatives account for over one-third of all NMEs. Before 1940, unmodified products and derivatives accounted for 43% and 14% of NME registrations, respectively. Since then, the share of unmodified products has decreased to 9.5% of all approved NMEs, while the share of derivatives has increased to 28%. Since the 1940s, semi-synthetic and synthetic derivatives of natural substances have gained importance, and this trend continues to date. In this study, we have discussed in detail isolated phytoconstituents with chemical modifications that are either FDA-approved or under clinical trials, such as podophyllotoxin, Taxol (paclitaxel, docetaxel), vinca alkaloids (vincristine, vinblastine), camptothecin, genistein, cephalotaxine, rohitukine, and many more, which may act as essential leads to the development of novel anticancer agents. Furthermore, we have also discussed recent developments in the most potent semisynthetic phytoconstituents, their unique properties, and their importance in cancer treatment.
Collapse
Affiliation(s)
- Meghna Arora
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Ankit Kumar Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Adarsh Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Harshwardhan Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Prateek Pathak
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, School of Pharmacy, GITAM (Deemed to be University) Hyderabad Campus India
| | - Maria Grishina
- Laboratory of Computational Modeling of Drugs, Higher Medical and Biological School, South Ural State University Chelyabinsk 454008 Russia
| | - Jagat Pal Yadav
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Pharmacology Research Laboratory, Faculty of Pharmaceutical Sciences, Rama University Kanpur 209217 India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| |
Collapse
|
2
|
Almeida A, Castro F, Resende C, Lúcio M, Schwartz S, Sarmento B. Oral delivery of camptothecin-loaded multifunctional chitosan-based micelles is effective in reduce colorectal cancer. J Control Release 2022; 349:731-743. [PMID: 35905784 DOI: 10.1016/j.jconrel.2022.07.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/01/2022] [Accepted: 07/22/2022] [Indexed: 10/16/2022]
Abstract
Colorectal cancer (CRC) is a heterogeneous disease with high incidence and mortality worldwide. The efficacy of conventional CRC chemotherapy is hampered by poor drug solubility and bioavailability and suboptimal pharmacokinetic profiles. In this work, camptothecin (CPT), a potent anticancer drug, was loaded into an amphiphilic chitosan modified with PEG and oleic acid, to reduce CRC progression after oral administration. While CPT-loaded micelles presented anticancer activity against HCT116, Caco-2 and HT29 CRC cell lines in vitro, empty micelles demonstrated a safe profile when incubated with human blood cells and colorectal cancer cell lines. In a more complex 3D CRC multicellular spheroid model, CPT-loaded micelles also exhibited a significant effect on the spheroid's metabolic activity and size reduction. Remarkably, in vivo studies performed in a HCT116 xenograft model, showed a significant reduction on the tumor growth during and after treatment with CPT-loaded micelles. Moreover, in a more biological relevant in vivo model of chemically-induced CRC, orally administered CPT-loaded micelles demonstrated a significant reduction on tumor incidence and inflammation signs. The findings here reported indicate that CPT-loaded into chitosan-based micelles, by improving drug solubility, alongside its safety profile for normal tissues, may have a promising role CRC chemotherapy.
Collapse
Affiliation(s)
- Andreia Almeida
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Flávia Castro
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Carlos Resende
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Marlene Lúcio
- CF-UM-UP, Centro de Física das Universidades do Minho e Porto, Departamento de Física da Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; CBMA, Centro de Biologia Molecular e Ambiental, Departamento de Biologia, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Simó Schwartz
- Banc de Sang i Teixits, Passeig del Taulat, 116, 08005 Barcelona, Spain
| | - Bruno Sarmento
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central da Gandra, 137, 4585-116 Gandra, Portugal.
| |
Collapse
|
3
|
Topoisomerase I inhibitors: Challenges, progress and the road ahead. Eur J Med Chem 2022; 236:114304. [DOI: 10.1016/j.ejmech.2022.114304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/17/2022]
|
4
|
Martín-Encinas E, Selas A, Palacios F, Alonso C. The design and discovery of topoisomerase I inhibitors as anticancer therapies. Expert Opin Drug Discov 2022; 17:581-601. [PMID: 35321631 DOI: 10.1080/17460441.2022.2055545] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Cancer has been identified as one of the leading causes of death worldwide. The biological target of some anticancer agents is topoisomerase I, an enzyme involved in the relaxation of supercoiled DNA. The synthesis of new compounds with antiproliferative effect and behaving as topoisomerase I inhibitors has become an active field of research. Depending on their mechanism of inhibition, they can be classified as catalytic inhibitors or poisons. AREAS COVERED This review article summarizes the state of the art for the development of selective topoisomerase I inhibitors. Collected compounds showed inhibition of the enzyme, highlighting those approved for clinical use, the combination therapies developed, as well as related drawbacks and future focus. EXPERT OPINION Research related to topoisomerase I inhibitors in cancer therapy started with camptothecin (CPT). This compound was first selected as a good anticancer agent and then topoisomerase I was identified as its therapeutic target. Derivatives of CPT irinotecan, topotecan, and belotecan are the only clinically approved inhibitors. Currently, their limitations are being addressed by different stretegies. Future studies should focus not only on developing other active molecules but also on improving the bioavailability and pharmacokinetics of potent synthetic derivatives.
Collapse
Affiliation(s)
- Endika Martín-Encinas
- Departamento de Química Orgánica I - Centro de Investigación Lascaray, Facultad de Farmacia, Universidad del País Vasco, Paseo de la Universidad 7, 01006 Vitoria, Spain
| | - Asier Selas
- Departamento de Química Orgánica I - Centro de Investigación Lascaray, Facultad de Farmacia, Universidad del País Vasco, Paseo de la Universidad 7, 01006 Vitoria, Spain
| | - Francisco Palacios
- Departamento de Química Orgánica I - Centro de Investigación Lascaray, Facultad de Farmacia, Universidad del País Vasco, Paseo de la Universidad 7, 01006 Vitoria, Spain
| | - Concepción Alonso
- Departamento de Química Orgánica I - Centro de Investigación Lascaray, Facultad de Farmacia, Universidad del País Vasco, Paseo de la Universidad 7, 01006 Vitoria, Spain
| |
Collapse
|
5
|
Investigation to Explain Bioequivalence Failure in Pravastatin Immediate-Release Products. Pharmaceutics 2019; 11:pharmaceutics11120663. [PMID: 31835294 PMCID: PMC6956045 DOI: 10.3390/pharmaceutics11120663] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 11/27/2019] [Accepted: 12/04/2019] [Indexed: 01/19/2023] Open
Abstract
The purpose of this work is to explore the predictive ability of the biopharmaceutics classification system (BCS) biowaiver based on the dissolution methods for two pravastatin test products, where one of them showed bioequivalence (BE) while the other test failed (non-bioequivalence, or NBE), and to explore the reasons for the BE failure. Experimental solubility and permeability data confirmed that pravastatin is a BCS class III compound. The permeability experiments confirmed that the NBE formulation significantly increased pravastatin permeability, and could explain its higher absorption rate and higher Cmax. This finding highlights the relevance of requiring similar excipients for BCS class III drugs. The BCS-based biowaiver dissolution tests at pH 1.2, 4.5, and 6.8, with the paddle apparatus at 50 rpm in 900 mL media, were not able to detect differences in pravastatin products, although the NBE formulation exhibited a more rapid dissolution at earlier sampling times. Dissolution tests conducted in 500 mL did not achieve complete dissolution, and both formulations were dissimilar because the amount dissolved at 15 min was less than 85%. The difference was less than 10% at pH 1.2 and 4.5, while at pH 6.8 f2, results reflected the Cmax rank order.
Collapse
|
6
|
Weng Q, Zhou L, Xia L, Zheng Y, Zhang X, Li F, Li Q. In vitro evaluation of FL118 and 9-Q20 cytotoxicity and cellular uptake in 2D and 3D different cell models. Cancer Chemother Pharmacol 2019; 84:527-537. [DOI: 10.1007/s00280-019-03846-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/22/2019] [Indexed: 12/29/2022]
|
7
|
Liang X, Wu Q, Luan S, Yin Z, He C, Yin L, Zou Y, Yuan Z, Li L, Song X, He M, Lv C, Zhang W. A comprehensive review of topoisomerase inhibitors as anticancer agents in the past decade. Eur J Med Chem 2019; 171:129-168. [PMID: 30917303 DOI: 10.1016/j.ejmech.2019.03.034] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 01/28/2023]
Abstract
The topoisomerase enzymes play an important role in DNA metabolism, and searching for enzyme inhibitors is an important target in the search for new anticancer drugs. Discovery of new anticancer chemotherapeutical capable of inhibiting topoisomerase enzymes is highlighted in anticancer research. Therefore, biologists, organic chemists and medicinal chemists all around the world have been identifying, designing, synthesizing and evaluating a variety of novel bioactive molecules targeting topoisomerase. This review summarizes types of topoisomerase inhibitors in the past decade, and divides them into nine classes by structural characteristics, including N-heterocycles compounds, quinone derivatives, flavonoids derivatives, coumarin derivatives, lignan derivatives, polyphenol derivatives, diterpenes derivatives, fatty acids derivatives, and metal complexes. Then we discussed the application prospect and development of these anticancer compounds, as well as concluded parts of their structural-activity relationships. We believe this review would be invaluable in helping to further search potential topoisomerase inhibition as antitumor agent in clinical usage.
Collapse
Affiliation(s)
- Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China.
| | - Qiang Wu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Shangxian Luan
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Changliang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Zhixiang Yuan
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Min He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Cheng Lv
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Wei Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| |
Collapse
|
8
|
Rocha-Del Castillo E, Gómez-García O, Andrade-Pavón D, Villa-Tanaca L, Ramírez-Apan T, Nieto-Camacho A, Gómez E. Dibutyltin(IV) Complexes Derived from L-DOPA: Synthesis, Molecular Docking, Cytotoxic and Antifungal Activity. Chem Pharm Bull (Tokyo) 2018; 66:1104-1113. [PMID: 30504627 DOI: 10.1248/cpb.c18-00441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A series of organotin(IV) complexes was herein prepared and characterized. A one-pot synthetic strategy afforded reasonable to high yields, depending on the nature of the ligand. All new complexes were fully characterized by spectroscopic techniques, consisting of IR, MS and NMR (1H, 13C and 119Sn). The in vitro cytotoxicity tests demonstrated that the organotin complexes produced a greater inhibition, versus cisplatin (the positive control), of the growth of six human cancer cell lines: U-251 (glioblastoma), K-562 (chronic myelogenous leukemia), HCT-15 (colorectal), MCF-7 (breast), MDA-MB-231 (breast) and SKLU-1 (non-small cell lung). The potency of this cytotoxic activity depended on the nature of the substituent bonded to the aromatic ring. All complexes exhibited excellent IC50 values. The test compounds were also screened in vitro for their antifungal effect against Candida glabrata and Candida albicans, showing minimum inhibitory concentration (MIC) values lower than those obtained for fluconazole. A brine shrimp bioassay was performed to examine the toxic properties. Molecular docking studies demonstrated that the organotin(IV) complexes bind at the active site of topoisomerase I in a similar manner to topotecan, sharing affinity for certain amino acid side chains (Ile535, Arg364 and Asp533), as well as for similar DNA regions (DA113, DC112 and DT10).
Collapse
Affiliation(s)
- Erika Rocha-Del Castillo
- Instituto de Química, Universidad Nacional Autónoma de México (UNAM), Circuito Exterior s/n, Ciudad Universitaria
| | - Omar Gómez-García
- Departamento de Química Orgánica-Laboratorio de Síntesis de Fármacos Heterocíclicos, Escuela Nacional de Ciencias Biológicas-Instituto Politécnico Nacional
| | - Dulce Andrade-Pavón
- Departamento de Microbiología-Laboratorio de Biología Molecular de Bacterias y Levaduras, Escuela Nacional de Ciencias Biológicas-Iinstituto Politécnico Nacional
| | - Lourdes Villa-Tanaca
- Departamento de Microbiología-Laboratorio de Biología Molecular de Bacterias y Levaduras, Escuela Nacional de Ciencias Biológicas-Iinstituto Politécnico Nacional
| | - Teresa Ramírez-Apan
- Instituto de Química, Universidad Nacional Autónoma de México (UNAM), Circuito Exterior s/n, Ciudad Universitaria
| | - Antonio Nieto-Camacho
- Instituto de Química, Universidad Nacional Autónoma de México (UNAM), Circuito Exterior s/n, Ciudad Universitaria
| | - Elizabeth Gómez
- Instituto de Química, Universidad Nacional Autónoma de México (UNAM), Circuito Exterior s/n, Ciudad Universitaria
| |
Collapse
|
9
|
Comparative evaluation of new dihydropyrimidine and dihydropyridine derivatives perturbing mitotic spindle formation. Future Med Chem 2018; 10:2395-2410. [PMID: 30325216 DOI: 10.4155/fmc-2018-0094] [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] [Indexed: 12/12/2022] Open
Abstract
AIM The mitotic spindle plays a key role in cell division which makes it an important target in cancer therapy. In the present study the antiproliferative activity of 4-benzyl-5-phenyl-3,4-dihydropyrimidine-2(1H)-thione (1) and its pyridine bioisoster (2) were evaluated and compared with monastrol (MON), the first known cell-permeable small molecule which disrupts bipolar spindle formation by inhibiting Eg5-kinesin activity. RESULTS Our data revealed that compound 2 showed higher antiproliferative activity than MON against MCF7 and A375 cell lines and comparable reversible cell cycle inhibition in G2/M phase. However, compound 2 produced distinct phenotype from monoastral spindles, and did not affect Eg5 ATPase activity. CONCLUSION The activity of compound 2 may suggest its new promising anticancer mechanism (different than MON), targeting other component required for spindle bipolarity.
Collapse
|
10
|
Zheng J, Ji Y, Shao C, Jing L, Wang Y. Pharmacokinetics and tissue distribution study of 10-methoxycamptothecin in rats following intragastric administration. J Pharm Biomed Anal 2018; 149:564-571. [PMID: 29197298 DOI: 10.1016/j.jpba.2017.11.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/21/2017] [Accepted: 11/26/2017] [Indexed: 11/20/2022]
Abstract
Natural bioactive derivatives of camptothecin (CPT), 10-methoxycamptothecin (MCPT) and 10-hydroxycamptothecin (HCPT) have been confirmed to possess high antitumor activities. MCPT could be metabolized to HCPT in vivo. The HPLC method for the quantification of MCPT and HCPT was established and validated, and the pharmacokinetics and the tissue distribution of MCPT in rats after i.v. administration have been well carried out in our previous studies. To improve the further understanding of the in vivo behavior of MCPT, a rapid and sensitive UPLC-MS/MS method was developed and validated for the quantification of MCPT and HCPT in plasma and tissue samples, and the pharmacokinetics and tissue distribution as well as the bioavailability of MCPT after i.g. were also illustrated. The results showed that MCPT could be highly converted to its active metabolite HCPT in plasma with the AUC0-∞ value of (185.28±61.73) ngh/mL and (717.25±165.67) ngh/mL for MCPT and HCPT, respectively. Meanwhile, MCPT and HCPT were rapidly absorbed and diffused into all the tested tissues (heart, liver, spleen, lung, kidney and brain) after i.g. administration. Similar with the results after i.v. administration of MCPT, MCPT concentration in lung tissue was also extremely higher than in other tested tissues, which implied that MCPT might have a great potential for the treatment of lung cancer.
Collapse
Affiliation(s)
- Jian Zheng
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education/Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin 150040, China
| | - Yong Ji
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education/Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin 150040, China
| | - Changmin Shao
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education/Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin 150040, China
| | - Lijia Jing
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education/Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin 150040, China
| | - Yang Wang
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education/Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
11
|
Self-aggregates of 3,6-O,O’-dimyristoylchitosan derivative are effective in enhancing the solubility and intestinal permeability of camptothecin. Carbohydr Polym 2017; 177:178-186. [DOI: 10.1016/j.carbpol.2017.08.114] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/07/2017] [Accepted: 08/27/2017] [Indexed: 01/27/2023]
|
12
|
González-Alvarez M, Coll C, Gonzalez-Alvarez I, Giménez C, Aznar E, Martínez-Bisbal MC, Lozoya-Agulló I, Bermejo M, Martínez-Máñez R, Sancenón F. Gated Mesoporous Silica Nanocarriers for a "Two-Step" Targeted System to Colonic Tissue. Mol Pharm 2017; 14:4442-4453. [PMID: 29064714 DOI: 10.1021/acs.molpharmaceut.7b00565] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Colon targeted drug delivery is highly relevant not only to treat colonic local diseases but also for systemic therapies. Mesoporous silica nanoparticles (MSNs) have been demonstrated as useful systems for controlled drug release given their biocompatibility and the possibility of designing gated systems able to release cargo only upon the presence of certain stimuli. We report herein the preparation of three gated MSNs able to deliver their cargo triggered by different stimuli (redox ambient (S1), enzymatic hydrolysis (S2), and a surfactant or being in contact with cell membrane (S3)) and their performance in solution and in vitro with Caco-2 cells. Safranin O dye was used as a model drug to track cargo fate. Studies of cargo permeability in Caco-2 monolayers demonstrated that intracellular safranin O levels were significantly higher in Caco-2 monolayers when using MSNs compared to those of free dye. Internalization assays indicated that S2 nanoparticles were taken up by cells via endocytosis. S2 nanoparticles were selected for in vivo tests in rats. For in vivo assays, capsules were filled with S2 nanoparticles and coated with Eudragit FS 30 D to target colon. The enteric coated capsule containing the MSNs was able to deliver S2 nanoparticles in colon tissue (first step), and then nanoparticles were able to deliver safranin O inside the colonic cells after the enzymatic stimuli (second step). This resulted in high levels of safranin O in colonic tissue combined with low dye levels in plasma and body tissues. The results suggested that this combination of enzyme-responsive gated MSNs and enteric coated capsules may improve the absorption of drugs in colon to treat local diseases with a reduction of systemic effects.
Collapse
Affiliation(s)
- Marta González-Alvarez
- Departamento de Ingeniería, Área Farmacia y Tecnología Farmacéutica, Universidad Miguel Hernández , 03550 Elche, Spain
| | - Carmen Coll
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València , Camino de Vera s/n, 46022 Valencia, Spain.,CIBER en Biotecnología, Biomateriales y Nanomedicina (CIBER-BBN) , Spain
| | - Isabel Gonzalez-Alvarez
- Departamento de Ingeniería, Área Farmacia y Tecnología Farmacéutica, Universidad Miguel Hernández , 03550 Elche, Spain
| | - Cristina Giménez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València , Camino de Vera s/n, 46022 Valencia, Spain.,CIBER en Biotecnología, Biomateriales y Nanomedicina (CIBER-BBN) , Spain
| | - Elena Aznar
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València , Camino de Vera s/n, 46022 Valencia, Spain.,CIBER en Biotecnología, Biomateriales y Nanomedicina (CIBER-BBN) , Spain
| | - M Carmen Martínez-Bisbal
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València , Camino de Vera s/n, 46022 Valencia, Spain.,CIBER en Biotecnología, Biomateriales y Nanomedicina (CIBER-BBN) , Spain.,Unidad Mixta de Investigación en Nanomedicina y Sensores Universitat Politècnica de València, IIS La Fe de Valencia , 46026 Valencia, Spain
| | - Isabel Lozoya-Agulló
- Departamento de Ingeniería, Área Farmacia y Tecnología Farmacéutica, Universidad Miguel Hernández , 03550 Elche, Spain
| | - Marival Bermejo
- Departamento de Ingeniería, Área Farmacia y Tecnología Farmacéutica, Universidad Miguel Hernández , 03550 Elche, Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València , Camino de Vera s/n, 46022 Valencia, Spain.,CIBER en Biotecnología, Biomateriales y Nanomedicina (CIBER-BBN) , Spain.,Unidad Mixta de Investigación en Nanomedicina y Sensores Universitat Politècnica de València, IIS La Fe de Valencia , 46026 Valencia, Spain.,Departamento de Química, Universitat Politècnica de València , 46022 Valencia, Spain
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València , Camino de Vera s/n, 46022 Valencia, Spain.,CIBER en Biotecnología, Biomateriales y Nanomedicina (CIBER-BBN) , Spain.,Unidad Mixta de Investigación en Nanomedicina y Sensores Universitat Politècnica de València, IIS La Fe de Valencia , 46026 Valencia, Spain.,Departamento de Química, Universitat Politècnica de València , 46022 Valencia, Spain
| |
Collapse
|
13
|
Bracher F, Tremmel T. From Lead to Drug Utilizing a Mannich Reaction: The Topotecan Story. Arch Pharm (Weinheim) 2016; 350. [DOI: 10.1002/ardp.201600236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/26/2016] [Accepted: 10/06/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Franz Bracher
- Department of Pharmacy, Center for Drug Research; Ludwig-Maximilians University; Munich Germany
| | - Tim Tremmel
- Department of Pharmacy, Center for Drug Research; Ludwig-Maximilians University; Munich Germany
| |
Collapse
|
14
|
Wang L, Xie S, Ma L, Chen Y, Lu W. 10-Boronic acid substituted camptothecin as prodrug of SN-38. Eur J Med Chem 2016; 116:84-89. [PMID: 27060760 DOI: 10.1016/j.ejmech.2016.03.063] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 10/22/2022]
Abstract
Malignant tumor cells have been found to have high levels of reactive oxygen species such as hydrogen peroxide (H2O2), supporting the hypothesis that a prodrug could be activated by intracellular H2O2 and lead to a potential antitumor therapy. In this study, the 7-ethyl-10-boronic acid camptothecin (B1) was synthesized for the first time as prodrug of SN-38, by linking a cleavable aryl carbon-boron bond to the SN-38. Prodrug B1 selectively activated by H2O2, converted rapidly to the active form SN-38 under favorable oxidative conditions in cancer cells with elevated levels of H2O2. The cell survival assay showed that prodrug B1 was equally or more effective in inhibiting the growth of six different cancer cells, as compared to SN-38. Unexpectedly, prodrug B1 displayed even more potent Topo I inhibitory activity than SN-38, suggesting that it was not only a prodrug of SN-38 but also a typical Topo I inhibitor. Prodrug B1 also demonstrated a significant antitumor activity at 2.0 mg/kg in a xenograft model using human brain star glioblastoma cell lines U87MG.
Collapse
Affiliation(s)
- Lei Wang
- School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Shao Xie
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Longjun Ma
- School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Yi Chen
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China.
| | - Wei Lu
- School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China.
| |
Collapse
|
15
|
Compound Library Screening Identified Cardiac Glycoside Digitoxin as an Effective Growth Inhibitor of Gefitinib-Resistant Non-Small Cell Lung Cancer via Downregulation of α-Tubulin and Inhibition of Microtubule Formation. Molecules 2016; 21:374. [PMID: 26999101 PMCID: PMC6274445 DOI: 10.3390/molecules21030374] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/25/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) dominates over 85% of all lung cancer cases. Epidermal growth factor receptor (EGFR) activating mutation is a common situation in NSCLC. In the clinic, molecular-targeting with Gefitinib as a tyrosine kinase inhibitor (TKI) for EGFR downstream signaling is initially effective. However, drug resistance frequently happens due to additional mutation on EGFR, such as substitution from threonine to methionine at amino acid position 790 (T790M). In this study, we screened a traditional Chinese medicine (TCM) compound library consisting of 800 single compounds in TKI-resistance NSCLC H1975 cells, which contains substitutions from leucine to arginine at amino acid 858 (L858R) and T790M mutation on EGFR. Attractively, among these compounds there are 24 compounds CC50 of which was less than 2.5 μM were identified. We have further investigated the mechanism of the most effective one, Digitoxin. It showed a significantly cytotoxic effect in H1975 cells by causing G2 phase arrest, also remarkably activated 5' adenosine monophosphate-activated protein kinase (AMPK). Moreover, we first proved that Digitoxin suppressed microtubule formation through decreasing α-tubulin. Therefore, it confirmed that Digitoxin effectively depressed the growth of TKI-resistance NSCLC H1975 cells by inhibiting microtubule polymerization and inducing cell cycle arrest.
Collapse
|
16
|
Li G, Zhao M, Zhao L. Well-defined hydroxyethyl starch-10-hydroxy camptothecin super macromolecule conjugate: cytotoxicity, pharmacodynamics research, tissue distribution test and intravenous injection safety assessment. Drug Deliv 2016; 23:2860-2868. [PMID: 26836216 DOI: 10.3109/10717544.2015.1110844] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
10-Hydroxy camptothecin (10-HCPT) is an antitumor agent effective in the treatment of several solid tumors but its use is hampered by poor water solubility, low lactone stability, short plasma half-life and dose-limiting toxicity. These limits of 10-HCPT had been overcome by our group through preparing super macromolecule prodrug: 10-HCPT-hydroxyethyl starch (HES) conjugate. In this study, we mainly evaluated in vitro and in vivo behavior of the prodrug, containing cytotoxicity assay, pharmacodynamics study, vascular irritation test, hemolysis experiment and tissue distribution test of rats. The irritation test results achieved much lower irritation than the commercial injection. The tissue distribution results showed that HES-10-HCPT conjugate increased significantly the 10-HCPT concentration in the tumor, liver and spleen site, whereas decreased the drug concentration in the heart and kidney. The hemolysis effect of the prepared conjugate was not obvious. The pharmacodynamics results indicated that HES-10-HCPT prodrug had a better antitumor efficiency against mice with H22 tumor than the commercial injection, and the inhibition ratio of tumor was 85.2% and 31.1%, respectively at the same dosage. These findings suggest that HES-10-HCPT prodrug is a promising drug delivery system providing improved good injection safety, greater tolerance and antitumor effect.
Collapse
Affiliation(s)
- Guofei Li
- a Department of Pharmacy , Shengjing Hospital of China Medical University , Shenyang , China
| | - Mingming Zhao
- a Department of Pharmacy , Shengjing Hospital of China Medical University , Shenyang , China
| | - Limei Zhao
- a Department of Pharmacy , Shengjing Hospital of China Medical University , Shenyang , China
| |
Collapse
|
17
|
Systematic sorption studies of camptothecin on oxidized single-walled carbon nanotubes. Colloids Surf A Physicochem Eng Asp 2016. [DOI: 10.1016/j.colsurfa.2015.11.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
18
|
Topoisomerase I Inhibitors Derived from Natural Products: Structure–Activity Relationships and Antitumor Potency. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/b978-0-444-63603-4.00001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
19
|
Yue Z, Zhang W, Lu Y, Yang Q, Ding Q, Xia J, Chen Y. Prediction of cancer cell sensitivity to natural products based on genomic and chemical properties. PeerJ 2015; 3:e1425. [PMID: 26644976 PMCID: PMC4671159 DOI: 10.7717/peerj.1425] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/02/2015] [Indexed: 12/26/2022] Open
Abstract
Natural products play a significant role in cancer chemotherapy. They are likely to provide many lead structures, which can be used as templates for the construction of novel drugs with enhanced antitumor activity. Traditional research approaches studied structure-activity relationship of natural products and obtained key structural properties, such as chemical bond or group, with the purpose of ascertaining their effect on a single cell line or a single tissue type. Here, for the first time, we develop a machine learning method to comprehensively predict natural products responses against a panel of cancer cell lines based on both the gene expression and the chemical properties of natural products. The results on two datasets, training set and independent test set, show that this proposed method yields significantly better prediction accuracy. In addition, we also demonstrate the predictive power of our proposed method by modeling the cancer cell sensitivity to two natural products, Curcumin and Resveratrol, which indicate that our method can effectively predict the response of cancer cell lines to these two natural products. Taken together, the method will facilitate the identification of natural products as cancer therapies and the development of precision medicine by linking the features of patient genomes to natural product sensitivity.
Collapse
Affiliation(s)
- Zhenyu Yue
- School of Life Sciences, Anhui University , Hefei, Anhui , China
| | - Wenna Zhang
- School of Life Sciences, Anhui University , Hefei, Anhui , China
| | - Yongming Lu
- School of Life Sciences, Anhui University , Hefei, Anhui , China
| | - Qiaoyue Yang
- School of Life Sciences, Anhui University , Hefei, Anhui , China
| | - Qiuying Ding
- School of Life Sciences, Anhui University , Hefei, Anhui , China
| | - Junfeng Xia
- Institute of Health Sciences, Anhui University , Hefei, Anhui , China
| | - Yan Chen
- School of Life Sciences, Anhui University , Hefei, Anhui , China
| |
Collapse
|
20
|
Colón-Useche S, González-Álvarez I, Mangas-Sanjuan V, González-Álvarez M, Pastoriza P, Molina-Martínez I, Bermejo M, García-Arieta A. Investigating the Discriminatory Power of BCS-Biowaiver in Vitro Methodology to Detect Bioavailability Differences between Immediate Release Products Containing a Class I Drug. Mol Pharm 2015; 12:3167-74. [PMID: 26287948 DOI: 10.1021/acs.molpharmaceut.5b00076] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The purpose of this work is to investigate the discriminatory power of the Biopharmaceutics Classification System (BCS)-biowaiver in vitro methodology, i.e., to investigate if a BCS-biowaiver approach would have detected the Cmax differences observed between two zolpidem tablets and to identify the cause of the in vivo difference. Several dissolution conditions were tested with three zolpidem formulations: the reference (Stilnox), a bioequivalent formulation (BE), and a nonbioequivalent formulation (N-BE). Zolpidem is highly soluble at pH 1.2, 4.5, and 6.8. Its permeability in Caco-2 cells is higher than that of metoprolol and its transport mechanism is passive diffusion. None of the excipients (alone or in combination) showed any effect on permeability. All formulations dissolved more than 85% in 15 min in the paddle apparatus at 50 rpm in all dissolution media. However, at 30 rpm the nonbioequivalent formulation exhibited a slower dissolution rate. A slower gastric emptying rate was also observed in rats for the nonbioequivalent formulation. A slower disintegration and dissolution or a delay in gastric emptying might explain the Cmax infra-bioavailability for a highly permeable drug with short half-life. The BCS-biowaiver approach would have declared bioequivalence, although the in vivo study was not conclusive but detected a 14% mean difference in Cmax that precluded the bioequivalence demonstration. Nonetheless, these findings suggest that a slower dissolution rate is more discriminatory and that rotation speeds higher than 50 rpm should not be used in BCS-biowaivers, even if a coning effect occurs.
Collapse
Affiliation(s)
- Sarin Colón-Useche
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University , 03550 Alicante, Spain.,Pharmacokinetics and Pharmaceutical Technology, Complutense University of Madrid , 28040 Madrid, Spain.,Analysis and Control Department, University of Los Andes , Mérida, Venezuela
| | - Isabel González-Álvarez
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University , 03550 Alicante, Spain
| | - Victor Mangas-Sanjuan
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University , 03550 Alicante, Spain
| | - Marta González-Álvarez
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University , 03550 Alicante, Spain
| | - Pilar Pastoriza
- Pharmacokinetics and Pharmaceutical Technology, Complutense University of Madrid , 28040 Madrid, Spain
| | - Irene Molina-Martínez
- Pharmacokinetics and Pharmaceutical Technology, Complutense University of Madrid , 28040 Madrid, Spain
| | - Marival Bermejo
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University , 03550 Alicante, Spain
| | - Alfredo García-Arieta
- Service on Pharmacokinetics and Generic Medicines, Division of Pharmacology and Clinical Evaluation, Department of Human Use Medicines, Spanish Agency for Medicines and Health Care Products , 28022 Madrid, Spain
| |
Collapse
|
21
|
Frik M, Fernández-Gallardo J, Gonzalo O, Mangas-Sanjuan V, González-Alvarez M, Serrano del Valle A, Hu C, González-Alvarez I, Bermejo M, Marzo I, Contel M. Cyclometalated Iminophosphorane Gold(III) and Platinum(II) Complexes. A Highly Permeable Cationic Platinum(II) Compound with Promising Anticancer Properties. J Med Chem 2015; 58:5825-41. [PMID: 26147404 PMCID: PMC4538566 DOI: 10.1021/acs.jmedchem.5b00427] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
![]()
New
organometallic gold(III) and platinum(II) complexes containing
iminophosphorane ligands are described. Most of them are more cytotoxic
to a number of human cancer cell lines than cisplatin. Cationic Pt(II)
derivatives 4 and 5, which differ only in
the anion, Hg2Cl62– or PF6– respectively, display almost identical
IC50 values in the sub-micromolar range (25–335-fold
more active than cisplatin on these cell lines). The gold compounds
induced mainly caspase-independent cell death, as previously reported
for related cycloaurated compounds containing IM ligands. Cycloplatinated
compounds 3, 4, and 5 can also
activate alternative caspase-independent mechanisms of death. However,
at short incubation times cell death seems to be mainly caspase dependent,
suggesting that the main mechanism of cell death for these compounds
is apoptosis. Mercury-free compound 5 does not interact
with plasmid (pBR322) DNA or with calf thymus DNA. Permeability studies
of 5 by two different assays, in vitro Caco-2 monolayers and a rat perfusion model, have revealed a high
permeability profile for this compound (comparable to that of metoprolol
or caffeine) and an estimated oral fraction absorbed of 100%, which
potentially makes it a good candidate for oral administration.
Collapse
Affiliation(s)
- Malgorzata Frik
- †Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,‡Chemistry Ph.D. Program, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
| | - Jacob Fernández-Gallardo
- †Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Oscar Gonzalo
- §Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Víctor Mangas-Sanjuan
- ∥Departamento de Ingeniería, Área Farmacia y Tecnología Farmacéutica, Universidad Miguel Hernández, 03550 San Juan, Alicante, Spain
| | - Marta González-Alvarez
- ∥Departamento de Ingeniería, Área Farmacia y Tecnología Farmacéutica, Universidad Miguel Hernández, 03550 San Juan, Alicante, Spain
| | - Alfonso Serrano del Valle
- §Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Chunhua Hu
- ⊥Chemistry Department, New York University, New York, New York 10003, United States
| | - Isabel González-Alvarez
- ∥Departamento de Ingeniería, Área Farmacia y Tecnología Farmacéutica, Universidad Miguel Hernández, 03550 San Juan, Alicante, Spain
| | - Marival Bermejo
- ∥Departamento de Ingeniería, Área Farmacia y Tecnología Farmacéutica, Universidad Miguel Hernández, 03550 San Juan, Alicante, Spain
| | - Isabel Marzo
- §Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - María Contel
- †Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,‡Chemistry Ph.D. Program, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, New York 10016, United States.,#Biology Ph.D. Program, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
| |
Collapse
|
22
|
Tan H, Wang G, Li J, Meng G, Liu Z, Dong M, Li Y, Ju D, Zhang Q. Synthesis of novel 10-hydroxycamptothecin derivatives utilizing topotecan hydrochloride as ortho-quinonemethide precursor. Bioorg Med Chem 2015; 23:118-25. [DOI: 10.1016/j.bmc.2014.11.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/12/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
|