51
|
Collins SL, Patterson AD. The gut microbiome: an orchestrator of xenobiotic metabolism. Acta Pharm Sin B 2020; 10:19-32. [PMID: 31998605 PMCID: PMC6984741 DOI: 10.1016/j.apsb.2019.12.001] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/21/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Microbes inhabiting the intestinal tract of humans represent a site for xenobiotic metabolism. The gut microbiome, the collection of microorganisms in the gastrointestinal tract, can alter the metabolic outcome of pharmaceuticals, environmental toxicants, and heavy metals, thereby changing their pharmacokinetics. Direct chemical modification of xenobiotics by the gut microbiome, either through the intestinal tract or re-entering the gut via enterohepatic circulation, can lead to increased metabolism or bioactivation, depending on the enzymatic activity within the microbial niche. Unique enzymes encoded within the microbiome include those that reverse the modifications imparted by host detoxification pathways. Additionally, the microbiome can limit xenobiotic absorption in the small intestine by increasing the expression of cell-cell adhesion proteins, supporting the protective mucosal layer, and/or directly sequestering chemicals. Lastly, host gene expression is regulated by the microbiome, including CYP450s, multi-drug resistance proteins, and the transcription factors that regulate them. While the microbiome affects the host and pharmacokinetics of the xenobiotic, xenobiotics can also influence the viability and metabolism of the microbiome. Our understanding of the complex interconnectedness between host, microbiome, and metabolism will advance with new modeling systems, technology development and refinement, and mechanistic studies focused on the contribution of human and microbial metabolism.
Collapse
Key Words
- 5-ASA, 5-aminosalicylic acid
- 5-FU, 5-fluorouracil
- AHR, aryl Hydrocarbon Receptor
- ALDH, aldehyde dehydrogenase
- Absorption
- BDE, bromodiphenyl ether
- BRV, brivudine
- BVU, bromovinyluracil
- Bioactivation
- CAR, constitutive androgen receptor
- CV, conventional
- CYP, cytochrome P450
- ER, estrogen receptor
- Enterohepatic circulation
- FXR, farnesoid X receptor
- GF, germ-free
- GUDCA, glycoursodeoxycholic acid
- Gastrointestinal tract
- Gut microbiome
- NSAID, non-steroidal anti-inflammatory drug
- PABA, p-aminobenzenesulphonamide
- PAH, polycyclic aromatic hydrocarbon
- PCB, polychlorinated biphenyl
- PD, Parkinson's disease
- PFOS, perfluorooctanesulfonic acid
- PXR, pregnane X receptor
- Pharmacokinetics
- SCFA, short chain fatty acid
- SN-38G, SN-38 glucuronide
- SULT, sulfotransferase
- TCDF, 2,3,7,8-tetrachlorodibenzofuran
- TUDCA, tauroursodeoxycholic acid
- UGT, uracil diphosphate-glucuronosyltransferase
- Xenobiotic metabolism
- cgr, cytochrome glycoside reductase
Collapse
Affiliation(s)
- Stephanie L. Collins
- Department of Biochemistry, Microbiology, and Molecular Biology, the Pennsylvania State University, University Park, PA 16802, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Science, the Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
52
|
Stanković T, Dinić J, Podolski-Renić A, Musso L, Burić SS, Dallavalle S, Pešić M. Dual Inhibitors as a New Challenge for Cancer Multidrug Resistance Treatment. Curr Med Chem 2019; 26:6074-6106. [PMID: 29874992 DOI: 10.2174/0929867325666180607094856] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Dual-targeting in cancer treatment by a single drug is an unconventional approach in relation to drug combinations. The rationale for the development of dualtargeting agents is to overcome incomplete efficacy and drug resistance frequently present when applying individual targeting agents. Consequently, -a more favorable outcome of cancer treatment is expected with dual-targeting strategies. METHODS We reviewed the literature, concentrating on the association between clinically relevant and/or novel dual inhibitors with the potential to modulate multidrug resistant phenotype of cancer cells, particularly the activity of P-glycoprotein. A balanced analysis of content was performed to emphasize the most important findings and optimize the structure of this review. RESULTS Two-hundred and forty-five papers were included in the review. The introductory part was interpreted by 9 papers. Tyrosine kinase inhibitors' role in the inhibition of Pglycoprotein and chemosensitization was illustrated by 87 papers. The contribution of naturalbased compounds in overcoming multidrug resistance was reviewed using 92 papers, while specific dual inhibitors acting against microtubule assembling and/or topoisomerases were described with 55 papers. Eleven papers gave an insight into a novel and less explored approach with hybrid drugs. Their influence on P-glycoprotein and multidrug resistance was also evaluated. CONCLUSION These findings bring into focus rational anticancer strategies with dual-targeting agents. Most evaluated synthetic and natural drugs showed a great potential in chemosensitization. Further steps in this direction are needed for the optimization of anticancer treatment.
Collapse
Affiliation(s)
- Tijana Stanković
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Jelena Dinić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Loana Musso
- DeFENS, Department of Food, Environmental and Nutritional Sciences, Universita degli Studi di Milano, Milano, Italy
| | - Sonja Stojković Burić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Sabrina Dallavalle
- DeFENS, Department of Food, Environmental and Nutritional Sciences, Universita degli Studi di Milano, Milano, Italy
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| |
Collapse
|
53
|
Nandi A, Ghosh C, Basu S. Polymer conjugated graphene-oxide nanoparticles impair nuclear DNA and Topoisomerase I in cancer. NANOSCALE ADVANCES 2019; 1:4965-4971. [PMID: 36133106 PMCID: PMC9417292 DOI: 10.1039/c9na00617f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/24/2019] [Indexed: 05/11/2023]
Abstract
Cancer chemotherapy had been dominated by the use of small molecule DNA damaging drugs. Eventually, the emergence of DNA damage repair machinery in cancer cells has led to combination therapy with the DNA topology controlling enzyme, topoisomerase I inhibitor along with DNA impairing agents. However, integrating multiple drugs having diverse water solubility and hence bio-distribution effectively for cancer treatment remains a significant challenge, which can be addressed by using suitable nano-scale materials. Herein, we have chemically conjugated graphene oxide (GO) with biocompatible and hydrophilic polymers [polyethylene glycol (PEG) and ethylene-diamine modified poly-isobutylene-maleic anhydride (PMA-ED)], which can encompass highly hydrophobic topoisomerase I inhibitor, SN38. Interestingly, these sheet structured GO-polymer-SN38 composites self-assembled into spherical nanoparticles in water after complexing with a hydrophilic DNA damaging drug, cisplatin. These nanoparticles showed much improved colloidal stability in water compared to their drug-loaded non-polymeric counterpart. These SN38 and cisplatin laden GO-polymer nanoparticles were taken up by HeLa cancer cells through clathrin-dependent endocytosis to home into lysosomes within 6 h, as confirmed by confocal microscopy. A combination of gel electrophoresis, flow cytometry, and fluorescence microscopy showed that these nanoparticles damaged nuclear DNA and induced topoisomerase I inhibition leading to apoptosis and finally improved HeLa cell death. These self-assembled GO-polymer nanoparticles can be used for strategic impairment of multiple cellular targets involving hydrophobic and hydrophilic drugs for effective combination therapy.
Collapse
Affiliation(s)
- Aditi Nandi
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune Dr Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Chandramouli Ghosh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune Dr Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Sudipta Basu
- Discipline of Chemistry, Indian Institute of Technology (IIT)-Gandhinagar Palaj Gandhinagar Gujarat 382355 India
| |
Collapse
|
54
|
McDaid WJ, Greene MK, Johnston MC, Pollheimer E, Smyth P, McLaughlin K, Van Schaeybroeck S, Straubinger RM, Longley DB, Scott CJ. Repurposing of Cetuximab in antibody-directed chemotherapy-loaded nanoparticles in EGFR therapy-resistant pancreatic tumours. NANOSCALE 2019; 11:20261-20273. [PMID: 31626255 PMCID: PMC6861736 DOI: 10.1039/c9nr07257h] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The anti-Epidermal Growth Factor Receptor (EGFR) antibody Cetuximab (CTX) has demonstrated limited anti-cancer efficacy in cells overexpressing EGFR due to activating mutations in RAS in solid tumours, such as pancreatic cancer. The utilisation of antibodies as targeting components of antibody-drug conjugates, such as trastuzumab emtansine (Kadcyla), demonstrates that antibodies may be repurposed to direct therapeutic agents to antibody-resistant cancers. Here we investigated the use of CTX as a targeting agent for camptothecin (CPT)-loaded polymeric nanoparticles (NPs) directed against KRAS mutant CTX-resistant cancer cells. CPT was encapsulated within poly(lactic-co-glycolic acid) (PLGA) NPs using the solvent evaporation method. CTX conjugation improved NP binding and delivery of CPT to CTX-resistant cancer cell lines. CTX successfully targeted CPT-loaded NPs to mutant KRAS PANC-1 tumours in vivo and reduced tumour growth. This study highlights that CTX can be repurposed as a targeting agent against CTX-resistant cancers and that antibody repositioning may be applicable to other antibodies restricted by resistance.
Collapse
Affiliation(s)
- William J McDaid
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Michelle K Greene
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Michael C Johnston
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Ellen Pollheimer
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Peter Smyth
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Kirsty McLaughlin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | | | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, NY 14260-1200, USA
| | - Daniel B Longley
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
55
|
Kuwahara M, Fujita H, Kataoka Y, Nakajima Y, Yamada M, Sugimoto N. In situ condensation of an anti-cancer drug into fibrin gel enabling effective inhibition of tumor cell growth. Chem Commun (Camb) 2019; 55:11679-11682. [PMID: 31524892 DOI: 10.1039/c9cc06418d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We show herein the highly effective inhibition of tumor cell growth using a gel consisting of a fibrin polymer formed with the in situ condensation of a camptothecin (CPT) derivative as an anti-cancer drug, which is efficiently conveyed with a carrier aptamer from a solution to the gel in a phenomenon, called selective oligonucleotide entrapment in fibrin polymers (SOEF).
Collapse
Affiliation(s)
- Masayasu Kuwahara
- Graduate School of Integrated Basic Sciences, Nihon University, 3-25-40 Sakurajosui, Setagaya-ku, Tokyo 156-8550, Japan.
| | - Hiroto Fujita
- Graduate School of Integrated Basic Sciences, Nihon University, 3-25-40 Sakurajosui, Setagaya-ku, Tokyo 156-8550, Japan.
| | - Yuka Kataoka
- Graduate School of Integrated Basic Sciences, Nihon University, 3-25-40 Sakurajosui, Setagaya-ku, Tokyo 156-8550, Japan.
| | - Yasuyo Nakajima
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Graduate School of Medicine, Gunma University, 3-39-15 Showa-machi, Maebashi, 371-8511, Japan
| | - Masanobu Yamada
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Graduate School of Medicine, Gunma University, 3-39-15 Showa-machi, Maebashi, 371-8511, Japan
| | - Naoki Sugimoto
- Frontier Institute for Biomolecular Engineering Research (FIBER), Konan University, 7-1-20 Minatojima-minamimachi, Kobe, 650-0047, Japan and Graduate School of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, 7-1-20 Minatojima-minamimachi, Kobe, 650-0047, Japan
| |
Collapse
|
56
|
Li Y, Zhang T, Liu Q, He J. PEG-Derivatized Dual-Functional Nanomicelles for Improved Cancer Therapy. Front Pharmacol 2019; 10:808. [PMID: 31379579 PMCID: PMC6659352 DOI: 10.3389/fphar.2019.00808] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/24/2019] [Indexed: 02/05/2023] Open
Abstract
Polymeric micelles have attracted considerable attention for effective delivery of poorly water-soluble cancer drugs. Polyethylene glycol (PEG), which has been approved for human use by the US Food and Drug Administration, is the most commonly used hydrophilic component of polymeric micelles because it is biocompatible and biodegradable. One disadvantage of traditional polymeric micelles is that they include a large amount of inert carrier materials, which do not contribute to therapeutic activity but increase cost and toxicity risk. A better alternative may be "dual-functional" micellar carriers, in which the hydrophobic carrier material (conjugated to PEG) has intrinsic therapeutic activity that complements, or even synergizes with, the antitumor activity of the drug cargo. This review summarizes recent progress in the development of PEG-derivatized dual-functional nanomicelles and surveys the evidence of their feasibility and promise for cancer therapy.
Collapse
Affiliation(s)
- Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Ting Zhang
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Jinhan He
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China.,Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
57
|
Pusuluri A, Krishnan V, Wu D, Shields CW, Wang LW, Mitragotri S. Role of synergy and immunostimulation in design of chemotherapy combinations: An analysis of doxorubicin and camptothecin. Bioeng Transl Med 2019; 4:e10129. [PMID: 31249879 PMCID: PMC6584462 DOI: 10.1002/btm2.10129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/09/2019] [Accepted: 03/26/2019] [Indexed: 01/03/2023] Open
Abstract
Combination chemotherapy is often employed to improve therapeutic efficacies of drugs. However, traditional combination regimens often utilize drugs at or near-their maximum tolerated doses (MTDs), elevating the risk of dose-related toxicity and impeding their clinical success. Further, high doses of adjuvant or neoadjuvant chemotherapies can cause myeloablation, which compromises the immune response and hinders the efficacy of chemotherapy as well as accompanying treatments such as immunotherapy. Clinical outcomes can be improved if chemotherapy combinations are designed to reduce the overall doses without compromising their therapeutic efficacy. To this end, we investigated a combination of camptothecin (CPT) with doxorubicin (DOX) as a low-dose treatment option for breast cancer. DOX-CPT combinations were synergistic in several breast cancer cell lines in vitro and one particular ratio displayed extremely high synergy on human triple negative breast cancer cells (MDA-MB-231). This combination led to excellent long-term survival of mice bearing MDA-MB-231 tumors at doses roughly five-fold lower than the reported MTD values of its constituent drugs. Impact of low dose DOX-CPT treatment on local tumor immune environment was assessed in immunocompetent mice bearing breast cancer (4T1) tumors. The combination was not only superior in inhibiting the disease progression compared to individual drugs, but it also generated a more favorable antitumor immunogenic response. Engineering DOX and CPT ratios to manifest synergy enables treatment at doses much lower than their MTDs, which could ultimately facilitate their translation into the clinic as a promising combination for breast cancer treatment.
Collapse
Affiliation(s)
- Anusha Pusuluri
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityCambridgeMA 02138
- Wyss Institute of Biologically Inspired Engineering, Harvard UniversityBostonMA 02115
- Department of Chemical EngineeringUniversity of CaliforniaSanta BarbaraCA 93106
| | - Vinu Krishnan
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityCambridgeMA 02138
- Wyss Institute of Biologically Inspired Engineering, Harvard UniversityBostonMA 02115
| | - Debra Wu
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityCambridgeMA 02138
- Wyss Institute of Biologically Inspired Engineering, Harvard UniversityBostonMA 02115
| | - C. Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityCambridgeMA 02138
- Wyss Institute of Biologically Inspired Engineering, Harvard UniversityBostonMA 02115
| | - Li W. Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityCambridgeMA 02138
- Wyss Institute of Biologically Inspired Engineering, Harvard UniversityBostonMA 02115
- Harvard–MIT Health Sciences and Technology ProgramInstitute for Medical Engineering and Science, Massachusetts Institute of TechnologyCambridgeMA 02139
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard UniversityCambridgeMA 02138
- Wyss Institute of Biologically Inspired Engineering, Harvard UniversityBostonMA 02115
| |
Collapse
|
58
|
Ibric A, Eckerstorfer S, Eder M, Louko I, Tunjic L, Heffeter P, Schueffl HH, Marian B, Haider N. Position-Selective Synthesis and Biological Evaluation of Four Isomeric A-Ring Amino Derivatives of the Alkaloid Luotonin A. Molecules 2019; 24:molecules24040716. [PMID: 30781470 PMCID: PMC6412769 DOI: 10.3390/molecules24040716] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/11/2019] [Accepted: 02/14/2019] [Indexed: 12/18/2022] Open
Abstract
Following two orthogonal synthetic routes, a series of all four possible A-ring amino derivatives of the natural product Luotonin A (a known Topoisomerase I inhibitor) was synthesized. In both strategies, intramolecular cycloaddition reactions are the key step. The target compounds were obtained in good yields by mild catalytic transfer hydrogenation of the corresponding nitro precursors. In-vitro evaluation of the antiproliferative activity towards human tumor cell lines revealed the 4-amino compound (5b) to be the most effective agent, showing an interesting profile of cytotoxic activity. Among other effects, a significant G2/M cell cycle arrest was observed for this compound, suggesting that either Topoisomerase I is not the only biological target, or that some atypical mechanism is responsible for inhibition of this enzyme.
Collapse
Affiliation(s)
- Amra Ibric
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| | - Stefan Eckerstorfer
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| | - Martin Eder
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| | - Ivan Louko
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| | - Leopold Tunjic
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| | - Hemma Henrike Schueffl
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| | - Brigitte Marian
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| | - Norbert Haider
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| |
Collapse
|
59
|
Nandi A, Ghosh C, Bajpai A, Basu S. Graphene oxide nanocells for impairing topoisomerase and DNA in cancer cells. J Mater Chem B 2019. [DOI: 10.1039/c9tb00336c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We have engineered graphene oxide based nanocell to target DNA topoisomerases and nuclear DNA in cancer cells to induce apoptosis.
Collapse
Affiliation(s)
- Aditi Nandi
- Department of Chemistry
- Indian Institute of Science Education and Research (IISER)-Pune
- Pune, 411008
- India
| | - Chandramouli Ghosh
- Department of Chemistry
- Indian Institute of Science Education and Research (IISER)-Pune
- Pune, 411008
- India
| | - Aman Bajpai
- Discipline of Chemistry, Indian Institute of Technology (IIT)-Gandhinagar, Palaj
- Gandhinagar
- India
| | - Sudipta Basu
- Discipline of Chemistry, Indian Institute of Technology (IIT)-Gandhinagar, Palaj
- Gandhinagar
- India
| |
Collapse
|
60
|
Yang Q, Tan T, Zhao J, Zhou C, Guo C, Wan Z, Song X, Gong T. A reversible decomposition approach for the formation of injectable, excipient-free, self-assembling nanocrystals. Chem Commun (Camb) 2019; 55:3144-3147. [PMID: 30801073 DOI: 10.1039/c8cc10191d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A chemical procedure to generate excipient-free SN38 nanocrystals with greater efficacy and less toxicity than CPT-11.
Collapse
Affiliation(s)
- Qin Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Tiantian Tan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Juan Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Chuchu Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Chenqi Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Zhuoya Wan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Xu Song
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| |
Collapse
|
61
|
Giarma E, Mpampali Z, Lialiaris T, Mourelatos D. Cytoprotective and genotoxic effects of vitamins K1 and B1 on irinotecan in vitro. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 837:60-64. [PMID: 30595211 DOI: 10.1016/j.mrgentox.2018.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 11/26/2022]
Abstract
Cultured human lymphocytes were treated with vitamins K1 and B1, potential anticancer agents, either alone or in combination with irinotecan, a semisynthetic analogue of camptothecin. The frequency of sister chromatid exchanges (SCEs) was measured as an indicator of genotoxicity and the proliferation rate index (PRI) and mitotic index (MI) was measured as indicators of cytostatic effect. Vitamin K1 alone did not induce SCEs at the concentrations tested and combined with irinotecan does not increase SCE rates induced by irinotecan alone. Vitamin B1 significantly increased SCEs and, in combination with irinotecan, increased rates further (p < 0.05). Vitamin K1 decreased PRI and MI in combination with irinotecan, there were further increases in MI. At a low concentration, vitamin B1 reduced the levels of SCE and increased PRI induced by irinotecan. The use of these vitamins in combination with antitumor agents might reduce clinical side effects of the antineoplastics.
Collapse
Affiliation(s)
- Eleni Giarma
- Laboratory of Genetics, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Zoi Mpampali
- Laboratory of Genetics, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Th Lialiaris
- Laboratory of Genetics, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - D Mourelatos
- Lab. of Med. Biol. and Genetics, Medical Faculty, Aristotle University of Thessaloniki, 54124, Greece.
| |
Collapse
|
62
|
Liu Y, Li Y, Li J, Xie Z, Wang Y, Chen Z. Ethyl violet-bovine serum albumin fluorescent protein nanovessels target to lysosomes and mitochondria. Nanomedicine (Lond) 2018; 14:19-31. [PMID: 30547703 DOI: 10.2217/nnm-2018-0281] [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: 11/21/2022] Open
Abstract
AIM Organelles are essential in maintaining homeostasis of mammalian cells. Monitoring the morphology and dynamics of organelles is of significance in cell state determination and disease diagnosis. MATERIALS & METHODS We describe here a new material called ethyl violet-bovine serum albumin fluorescent protein nanovessel (EV-BSA FPN). Upon heating, BSA was denatured to form higher polyhedral structures, which was prone to EV binding. These dye-protein hybrid materials were red fluorescence emissive upon excitation. RESULTS EV-BSA FPNs can be readily internalized by mammalian cells and dual localized in lysosomes and mitochondria. Besides, EV-BSA FPN can serve as carriers and efficiently deliver drug into cells. CONCLUSION EV-BSA FPNs can be dual function fluorescent vessels for both dual-organelle imaging and drug delivery.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Supramolecular Structure & Materials & Institute of Theoretical Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, PR China
| | - Yuanyuan Li
- State Key Laboratory of Polymer Physics & Chemistry, The First Hospital of Jilin University, Xinmin Street, Changchun, Jilin 130021, PR China
| | - Jia Li
- Beijing Key Laboratory of Gene Resource & Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics & Chemistry, The First Hospital of Jilin University, Xinmin Street, Changchun, Jilin 130021, PR China
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource & Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Zhijun Chen
- State Key Laboratory of Supramolecular Structure & Materials & Institute of Theoretical Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, PR China
| |
Collapse
|
63
|
Lai CH, Chang CC, Weng YL, Chuang TH. Synthesis, Experimental and Density Functional Theory (DFT) Studies on Solubility of Camptothecin Derivatives. Molecules 2018; 23:molecules23123170. [PMID: 30513778 PMCID: PMC6320962 DOI: 10.3390/molecules23123170] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 11/16/2022] Open
Abstract
Two camptothecin derivatives, 10-cyclohexyl-7-methyl-20(S)-camptothecin and 7-methyl-10-morpholino-20(S)-camptothecin, were synthesized and their differences in solubility were investigated using four chosen solvent systems. Based on our results, 10-cyclohexyl-7-methyl-20(S)-camptothecin exhibited higher solubilities than 7-methyl-10-morpholino-20(S)-camptothecin in polar aprotic solvents. However, these two camptothecin derivatives did not exhibit apparent differences in solubility between 5% dimethyl sulfoxide (DMSO)/95% normal saline co-solvent system and 5% dimethylacetamide (DMAC)/95% normal saline co-solvent system. To rationalize their differences in solubility, we also tried to perform a DFT-B3LYP study to investigate their interaction with one water molecule.
Collapse
Affiliation(s)
- Chin-Hung Lai
- Department of Medical Applied Chemistry, Chung Shan Medical University, Taichung 40201, Taiwan.
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Chia-Chin Chang
- Master Program for Pharmaceutical Manufacture, China Medical University, Taichung 40402, Taiwan.
| | - Yi-Lin Weng
- Master Program for Pharmaceutical Manufacture, China Medical University, Taichung 40402, Taiwan.
| | - Ta-Hsien Chuang
- Master Program for Pharmaceutical Manufacture, China Medical University, Taichung 40402, Taiwan.
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
64
|
Mukwaya V, Wang C, Dou H. Saccharide-based nanocarriers for targeted therapeutic and diagnostic applications. POLYM INT 2018. [DOI: 10.1002/pi.5702] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Vincent Mukwaya
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering; Shanghai Jiao Tong University; Shanghai PR China
| | - Chenglong Wang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering; Shanghai Jiao Tong University; Shanghai PR China
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering; Shanghai Jiao Tong University; Shanghai PR China
| |
Collapse
|
65
|
Abstract
Glioblastoma multiforme (GBM), a grade IV astrocytoma as defined by the World Health Organization (WHO) criteria, is the most common primary central nervous system tumor in adults. After treatment with the current standard of care consisting of surgical resection, concurrent temozolomide (TMZ), and radiation, the median survival is only 15 months. The limited and less-effective treatment options for these highly aggressive GBMs call for the development of new techniques and the improvement of existing technologies. Nanotechnology has shown promise in treating this disease, and some nanomaterials have demonstrated the ability to cross the blood–brain barrier (BBB) and remain in GBM tissues. Although the retention of nanoparticles (NPs) in GBM tissue is necessary to elicit an antitumor response, the delivery of the NP needs to be enhanced. Current research in nanotechnology is directed at increasing the active targeting of GBM tissue not only for the aid of chemotherapeutic drug delivery but also for imaging studies. This review is aimed at describing advancements in increasing nanotechnology specificity to GBM tissue.
Collapse
|
66
|
Bhori M, Singh K, Marar T, Chilakapati MK. Exploring the effect of vitamin E in cancer chemotherapy-A biochemical and biophysical insight. JOURNAL OF BIOPHOTONICS 2018; 11:e201800104. [PMID: 29770585 DOI: 10.1002/jbio.201800104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/15/2018] [Indexed: 06/08/2023]
Abstract
Many oncologists contend that patient undergoing chemotherapy must avoid antioxidant supplementation as it may interfere with the activity of the drug. In the present investigation, we have explored the influence of vitamin E, a well-known antioxidant on Camptothecin (CPT), a potent anti-cancer drug induced cell apoptosis and death of cervical cancer cells. HeLa cells were treated with different concentrations of CPT in presence and absence of 100 μm vitamin E. Treated cells were subjected to cytotoxicity studies, catalase assay, DNA fragmentation assay, clonogenic assay and flow cytometry based apoptosis detection. Also, Raman spectroscopy a label free technique which provides global information, in conjunction with multivariate tools like PCA, PCLDA and FDA, was investigated to explore vitamin E supplementation induced alterations. Our data based on biochemical and biophysical experimental analysis reveals that CPT causes DNA damage along with protein and lipid alteration culminating in cell death. Importantly, Raman spectroscopic analysis could uniquely differentiate the cluster of control and vitamin E control from CPT and CPT + Vit E treated cells. We conclusively prove that presence of vitamin E at 100 μM concentration shows promising antioxidant activity and displays no modulatory role on CPT induced effect, thereby causing no possible hindrance with the efficacy of the drug. Vitamin E may prove beneficial to alleviate chemotherapy associated side effects in patients during clinical settings which may open the doors further for subsequent exploration in in vivo preclinical studies.
Collapse
Affiliation(s)
- Mustansir Bhori
- School of Biotechnology and Bioinformatics, D. Y. Patil Deemed to be University, Navi Mumbai, India
| | - Kanchanlata Singh
- School of Biotechnology and Bioinformatics, D. Y. Patil Deemed to be University, Navi Mumbai, India
| | - Thankamani Marar
- School of Biotechnology and Bioinformatics, D. Y. Patil Deemed to be University, Navi Mumbai, India
| | - Murali Krishna Chilakapati
- Chilakapati Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
67
|
Liang D, Wu X, Hasinoff BB, Herbert DE, Tranmer GK. Evaluation of Nitrobenzyl Derivatives of Camptothecin as Anti-Cancer Agents and Potential Hypoxia Targeting Prodrugs. Molecules 2018; 23:molecules23082041. [PMID: 30111719 PMCID: PMC6222813 DOI: 10.3390/molecules23082041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/14/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
As part of our initial efforts into developing a tumor-targeting therapy, C-10 substituted derivatives of a camptothecin analog (SN-38) have been synthesized (2-, 3- and 4-nitrobenzyl) for use as potential hypoxia-activated prodrugs and evaluated for their cytotoxicity, topoisomerase I inhibition and electrochemical (reductive) properties. All three derivatives were found to possess reduced toxicity towards human leukemia K562 cells compared to SN-38, validating a condition for prodrug action. Using an MTS assay, IC50's were found to be 3.0, 25.9, 12.2 and 58.0 nM for SN-38, 2-nitro-, 3-nitro- and 4-nitrobenzyl-C10-substituted-SN-38, respectively, representing an 8-, 4- and 19-fold decrease in cytotoxicity. Using a topoisomerase I assay, one of the analogs (4-nitrobenzyl) was shown to inhibit the ability of this enzyme to relax supercoiled pBR322 DNA, at a similar concentration to the clinically-approved active metabolite SN-38. Cyclic voltammetry detailed the reductive nature of the analogs, and was used to infer the potential of these compounds to serve as hypoxia-targeting prodrugs. The electrochemical results also validated the quasi-reversible nature of the first reduction step, and served as a proof-of-principle that hypoxia-targeting prodrugs of SN-38 can participate in a redox-futile cycle, the proposed mechanism of activation and targeting. Chemical reduction of the 4-nitrobenzyl analog led to the formation/release of SN-38 and validated the prodrug ability of the C-10 substituted derivative.
Collapse
Affiliation(s)
- Dinghua Liang
- College of Pharmacy, Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
| | - Xing Wu
- College of Pharmacy, Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
| | - Brian B Hasinoff
- College of Pharmacy, Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
| | - David E Herbert
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - Geoffrey K Tranmer
- College of Pharmacy, Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| |
Collapse
|
68
|
Cheng QQ, Massey LA, Willett BS, Deng Y, Arman H, Doyle MP. Copper-Catalyzed Formal [4+2] Cycloaddition of Enoldiazoimides with Sulfur Ylides. Angew Chem Int Ed Engl 2018; 57:10343-10346. [PMID: 29863775 PMCID: PMC6421837 DOI: 10.1002/anie.201805323] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/29/2018] [Indexed: 12/29/2022]
Abstract
Enoldiazoimides, a new subclass of enoldiazo compounds, generate enol-substituted carbonyl ylides whose reactions with sulfur ylides enable an unprecedented formal [4+2] cycloaddition. The resulting multifunctionalized indolizidinones, which incorporate sulfur, are formed in good yields under mild reaction conditions. The uniqueness of this transformation stems from the role of the silyl-protected enol, since the corresponding acetyldiazoimide failed to provide any cross-products in metal-catalyzed reactions with sulfur ylides. This copper-catalyzed cycloaddition is initiated with the generation of enol-substituted carbonyl ylides and sulfur ylides from enoldiazoimides and sulfonium salts, respectively, and proceeds through stepwise six-membered ring formation, C-O and C-S bond cleavage, and silyl and acetyl group migration.
Collapse
Affiliation(s)
- Qing-Qing Cheng
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Lynée A Massey
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Brook S Willett
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Yongming Deng
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
- Current address: Department of Chemistry, Western Kentucky University, Bowling Green, KY, 42101, USA
| | - Hadi Arman
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Michael P Doyle
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| |
Collapse
|
69
|
Cheng Q, Massey LA, Willett BS, Deng Y, Arman H, Doyle MP. Copper‐Catalyzed Formal [4+2] Cycloaddition of Enoldiazoimides with Sulfur Ylides. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201805323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Qing‐Qing Cheng
- Department of Chemistry The University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| | - Lynée A. Massey
- Department of Chemistry The University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| | - Brook S. Willett
- Department of Chemistry The University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| | - Yongming Deng
- Department of Chemistry The University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
- Current address: Department of Chemistry Western Kentucky University Bowling Green KY 42101 USA
| | - Hadi Arman
- Department of Chemistry The University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| | - Michael P. Doyle
- Department of Chemistry The University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| |
Collapse
|
70
|
Arthur DE, Uzairu A. Molecular docking study and structure-based design of novel camptothecin analogues used as topoisomerase I inhibitor. J CHIN CHEM SOC-TAIP 2018. [DOI: 10.1002/jccs.201700314] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- David Ebuka Arthur
- Department of Chemistry; Ahmadu Bello University; Zaria Kaduna State Nigeria
| | - Adamu Uzairu
- Department of Chemistry; Ahmadu Bello University; Zaria Nigeria
| |
Collapse
|
71
|
Park IH, Im SA, Jung KH, Sohn JH, Park YH, Lee KS, Sim SH, Park KH, Kim JH, Nam BH, Kim HJ, Kim TY, Lee KH, Kim SB, Ahn JH, Lee S, Ro J. Randomized Open Label Phase III Trial of Irinotecan Plus Capecitabine versus Capecitabine Monotherapy in Patients with Metastatic Breast Cancer Previously Treated with Anthracycline and Taxane: PROCEED Trial (KCSG BR 11-01). Cancer Res Treat 2018; 51:43-52. [PMID: 29458237 PMCID: PMC6333992 DOI: 10.4143/crt.2017.562] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/12/2018] [Indexed: 01/22/2023] Open
Abstract
Purpose We investigated whether irinotecan plus capecitabine improved progression-free survival (PFS) compared with capecitabine alone in patients with human epidermal growth factor 2 (HER2) negative and anthracycline and taxane pretreated metastatic breast cancer (MBC). Materials and Methods A total of 221 patients were randomly assigned to irinotecan (80 mg/m2, days 1 and 8) and capecitabine (1,000 mg/m2 twice a day, days 1-14) or capecitabine alone (1,250 mg/m2 twice a day, days 1-14) every 3 weeks. The primary endpoint was PFS. Results There was no significant difference in PFS between the combination and monotherapy arm (median, 6.4 months vs. 4.7 months; hazard ratio [HR], 0.84; 95% confidence interval [CI], 0.63 to 1.11; p=0.84). In patients with triple-negative breast cancer (TNBC, n=90), the combination significantly improved PFS (median, 4.7 months vs. 2.5 months; HR, 0.58; 95% CI, 0.37 to 0.91; p=0.02). Objective response rate was numerically higher in the combination arm, though it failed to reach statistical significance (44.4% vs. 33.3%, p=0.30). Overall survival did not differ between arms (median, 20.4 months vs. 24.0 months; p=0.63). While grade 3 or 4 neutropenia was more common in the combination arm (39.6% vs. 9.0%), hand-foot syndrome was more often observed in capecitabine arm. Quality of life measurements in global health status was similar. However, patients in the combination arm showed significantly worse symptom scales especially in nausea/vomiting and diarrhea. Conclusion Irinotecan plus capecitabine did not prove clinically superior to single-agent capecitabine in anthracycline- and taxane-pretreated HER2 negative MBC patients. Toxicity profiles of the two groups differed but were manageable. The role of added irinotecan in patients with TNBC remains to be elucidated.
Collapse
Affiliation(s)
- In Hae Park
- Division of Internal Medicine, Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, Ulsan University College of Medicine, Seoul, Korea
| | - Joo Hyuk Sohn
- Department of Oncology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - Keun Seok Lee
- Division of Internal Medicine, Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - Sung Hoon Sim
- Division of Internal Medicine, Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - Kyong-Hwa Park
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jee Hyun Kim
- Division of Oncology/Hematology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Byung Ho Nam
- Center for Clinical Trials, National Cancer Center, Goyang, Korea
| | - Hee-Jun Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, Ulsan University College of Medicine, Seoul, Korea
| | - Jin-Hee Ahn
- Department of Oncology, Asan Medical Center, Ulsan University College of Medicine, Seoul, Korea
| | - Suee Lee
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Jungsil Ro
- Division of Internal Medicine, Center for Breast Cancer, National Cancer Center, Goyang, Korea
| |
Collapse
|
72
|
Toyota M, Kagawa N, Nishimura K, Abe S. Concise Approach to Mono- and Disubstituted Luotonin A Analogs and Their Cytotoxicity Test. HETEROCYCLES 2018. [DOI: 10.3987/com-17-s(t)6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
73
|
Structure-activity relationships of flavonoids as natural inhibitors against E. coli β-glucuronidase. Food Chem Toxicol 2017; 109:975-983. [DOI: 10.1016/j.fct.2017.03.042] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 11/23/2022]
|
74
|
Mucaji P, Atanasov AG, Bak A, Kozik V, Sieron K, Olsen M, Pan W, Liu Y, Hu S, Lan J, Haider N, Musiol R, Vanco J, Diederich M, Ji S, Zitko J, Wang D, Agbaba D, Nikolic K, Oljacic S, Vucicevic J, Jezova D, Tsantili-Kakoulidou A, Tsopelas F, Giaginis C, Kowalska T, Sajewicz M, Silberring J, Mielczarek P, Smoluch M, Jendrzejewska I, Polanski J, Jampilek J. The Forty-Sixth Euro Congress on Drug Synthesis and Analysis: Snapshot †. Molecules 2017; 22:molecules22111848. [PMID: 29143778 PMCID: PMC6150335 DOI: 10.3390/molecules22111848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/26/2017] [Accepted: 10/26/2017] [Indexed: 01/08/2023] Open
Abstract
The 46th EuroCongress on Drug Synthesis and Analysis (ECDSA-2017) was arranged within the celebration of the 65th Anniversary of the Faculty of Pharmacy at Comenius University in Bratislava, Slovakia from 5-8 September 2017 to get together specialists in medicinal chemistry, organic synthesis, pharmaceutical analysis, screening of bioactive compounds, pharmacology and drug formulations; promote the exchange of scientific results, methods and ideas; and encourage cooperation between researchers from all over the world. The topic of the conference, "Drug Synthesis and Analysis," meant that the symposium welcomed all pharmacists and/or researchers (chemists, analysts, biologists) and students interested in scientific work dealing with investigations of biologically active compounds as potential drugs. The authors of this manuscript were plenary speakers and other participants of the symposium and members of their research teams. The following summary highlights the major points/topics of the meeting.
Collapse
Affiliation(s)
- Pavel Mucaji
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia.
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland.
- Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| | - Andrzej Bak
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Violetta Kozik
- Department of Synthesis Chemistry, Faculty of Mathematics, Physics and Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Karolina Sieron
- Department of Physical Medicine, Medical University of Silesia, Medykow 18, 40752 Katowice, Poland.
| | - Mark Olsen
- Department of Pharmaceutical Sciences, College of Pharmacy Glendale, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA.
| | - Weidong Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang, 550014, China.
| | - Yazhou Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang, 550014, China.
| | - Shengchao Hu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang, 550014, China.
| | - Junjie Lan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang, 550014, China.
| | - Norbert Haider
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| | - Robert Musiol
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Jan Vanco
- Department of Inorganic Chemistry & Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, 17. listopadu 12, 77146 Olomouc, Czech Republic.
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Seoul 08826, Korea.
| | - Seungwon Ji
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Seoul 08826, Korea.
| | - Jan Zitko
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic.
| | - Dongdong Wang
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland.
- Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| | - Danica Agbaba
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Slavica Oljacic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Jelica Vucicevic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Daniela Jezova
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia.
| | - Anna Tsantili-Kakoulidou
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece.
| | - Fotios Tsopelas
- Laboratory of Inorganic and Analytical Chemistry, School of Chemical Engineering, National Technical University of Athens, Iroon Polytechniou 9, 15780 Athens, Greece.
| | - Constantinos Giaginis
- Department of Food Science and Nutrition, School of Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece.
| | - Teresa Kowalska
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Mieczyslaw Sajewicz
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Jerzy Silberring
- Department of Biochemistry and Neurobiology, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza 30, 30059 Krakow, Poland.
| | - Przemyslaw Mielczarek
- Department of Biochemistry and Neurobiology, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza 30, 30059 Krakow, Poland.
| | - Marek Smoluch
- Department of Biochemistry and Neurobiology, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza 30, 30059 Krakow, Poland.
| | - Izabela Jendrzejewska
- Department of Crystallography, Faculty of Mathematics, Physics and Chemistry, University of Silesia, Bankowa 12, 40006 Katowice, Poland.
| | - Jaroslaw Polanski
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Josef Jampilek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia.
| |
Collapse
|
75
|
Khageh Hosseini S, Kolterer S, Steiner M, von Manstein V, Gerlach K, Trojan J, Waidmann O, Zeuzem S, Schulze JO, Hahn S, Steinhilber D, Gatterdam V, Tampé R, Biondi RM, Proschak E, Zörnig M. Camptothecin and its analog SN-38, the active metabolite of irinotecan, inhibit binding of the transcriptional regulator and oncoprotein FUBP1 to its DNA target sequence FUSE. Biochem Pharmacol 2017; 146:53-62. [PMID: 29031818 DOI: 10.1016/j.bcp.2017.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/11/2017] [Indexed: 01/03/2023]
Abstract
The transcriptional regulator FUSE Binding Protein 1 (FUBP1) is overexpressed in more than 80% of all human hepatocellular carcinomas (HCCs) and other solid tumor entities including prostate and colorectal carcinoma. FUBP1 expression is required for HCC tumor cell expansion, and it functions as an important pro-proliferative and anti-apoptotic oncoprotein that binds to the single-stranded DNA sequence FUSE to regulate the transcription of a variety of target genes. In this study, we screened an FDA-approved drug library and discovered that the Topoisomerase I (TOP1) inhibitor camptothecin (CPT) and its derivative 7-ethyl-10-hydroxycamptothecin (SN-38), the active irinotecan metabolite that is used in the clinics in combination with other chemotherapeutics to treat carcinoma, inhibit FUBP1 activity. Both molecules prevent in vitro the binding of FUBP1 to its single-stranded target DNA FUSE, and they induce deregulation of FUBP1 target genes in HCC cells. Our results suggest the interference with the FUBP1/FUSE interaction as a further molecular mechanism that, in addition to the inactivation of TOP1, may contribute to the therapeutic potential of CPT/SN-38. Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression.
Collapse
Affiliation(s)
- Sabrina Khageh Hosseini
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Stefanie Kolterer
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Marlene Steiner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Viktoria von Manstein
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Katharina Gerlach
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany
| | - Jörg Trojan
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany; Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Oliver Waidmann
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany
| | - Stefan Zeuzem
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany
| | - Jörg O Schulze
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany
| | - Steffen Hahn
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany
| | - Volker Gatterdam
- Institute of Biochemistry, Biocenter/Cluster of Excellence-Macromolecular Complexes, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter/Cluster of Excellence-Macromolecular Complexes, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany
| | - Ricardo M Biondi
- Department of Internal Medicine I, University Hospital Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany; Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Martin Zörnig
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, D-60596 Frankfurt/Main, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany.
| |
Collapse
|
76
|
Sugumaran A, Ponnusamy C, Kandasamy P, Krishnaswami V, Palanichamy R, Kandasamy R, Lakshmanan M, Natesan S. Development and evaluation of camptothecin loaded polymer stabilized nanoemulsion: Targeting potential in 4T1-breast tumour xenograft model. Eur J Pharm Sci 2017; 116:15-25. [PMID: 28987538 DOI: 10.1016/j.ejps.2017.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 09/06/2017] [Accepted: 10/03/2017] [Indexed: 11/18/2022]
Abstract
Targeted delivery of anticancer agents is poised to improve cancer therapy, for which polymers can serve as targeting ligands or nanocarriers for chemotherapeutic agents. In this study, we have developed and evaluated the efficacy of a camptothecin (CPT)-loaded polymer stabilized nanoemulsion (PSNE) for the passive targeted delivery to breast cancer. Based on the pseudo-ternary phase diagrams, PSNEs were developed using capmul MCM:poloxamer 407 (4:1), solutol HS 15:simulsol P23 (1:2) and water. CPT polymer mixture was developed by solvent evaporation technique. The PSNEs were characterized for droplet size distribution, plasma protein adsorption, drug release, in-vivo targeting potential, hemolytic potential, cytotoxicity, genotoxicity, in-vivo biodistribution and CPT lactone ring stability. The developed PSNEs showed uniform droplet distribution, extended drug release (76.59±6.12% at 24h), acceptable hemolytic potential, significant cytotoxicity (IC50=176±4.3ng/mL) and genotoxicity against MCF-7 cancer cells but low DNA damage potential in human peripheral blood lymphocytes. The efficiency of PSNEs for the targeted delivery of CPT into the tumour regions was documented in 4T1-breast tumour xenografted BALB/c mice. In-vivo biodistribution study shows that 7105.84±568.46ng/g of CPT was passively targeted from PSNE to breast cancer tissue. About 80% of the lactone form was stable for 24h. Taken together, our study provides a promising strategy for developing PSNE-targeted drug delivery system for the breast cancer therapy.
Collapse
Affiliation(s)
- Abimanyu Sugumaran
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, India; Department of Pharmaceutics, SRM College of Pharmacy, SRM University, Kattankulathur, India
| | - Chandrasekar Ponnusamy
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, India
| | - Palanivel Kandasamy
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| | - Venkateshwaran Krishnaswami
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, India
| | - Rajaguru Palanichamy
- Department of Biotechnology, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, India
| | - Ruckmani Kandasamy
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, India
| | | | - Subramanian Natesan
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, India.
| |
Collapse
|
77
|
A Facile Oxidative Opening of the C-Ring in Luotonin A and Derivatives. Molecules 2017; 22:molecules22091540. [PMID: 28895937 PMCID: PMC6151605 DOI: 10.3390/molecules22091540] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/11/2017] [Accepted: 09/11/2017] [Indexed: 11/17/2022] Open
Abstract
An oxidative ring opening reaction of the central ring C in the alkaloid Luotonin A and two of its derivatives was found to occur upon heating with an excess amine and potassium carbonate in dimethylsulfoxide (DMSO) solution in the presence of air oxygen. The structure of the novel amide-type products was elucidated and a possible mechanism for this reaction is proposed. Four of the new compounds show moderate in vitro anticancer activity towards human colon adenocarcinoma cells.
Collapse
|
78
|
Chen C, Tao R, Ding D, Kong D, Fan A, Wang Z, Zhao Y. Ratiometric co-delivery of multiple chemodrugs in a single nanocarrier. Eur J Pharm Sci 2017. [DOI: 10.1016/j.ejps.2017.06.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
79
|
Kulhari H, Telukutla SR, Pooja D, Shukla R, Sistla R, Bansal V, Adams DJ. Peptide grafted and self-assembled poly(γ-glutamic acid)-phenylalanine nanoparticles targeting camptothecin to glioma. Nanomedicine (Lond) 2017. [DOI: 10.2217/nnm-2017-0067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To synthesize cRGDfK peptide conjugated poly(γ-glutamic acid)-phenylalanine nanoparticles to improve the therapeutic efficacy of camptothecin (CPT) against glioblastoma multiforme. Methods: Peptide-conjugated, drug-loaded nanoparticles (cRGDfK-conjugated camptothecin-loaded PGA–PA nanoparticles [RCPN]) were prepared and physico-chemically characterized using different techniques. Nanoparticles were evaluated for in vitro anticancer activity, cellular uptake, induction of apoptosis and wound healing cell migration against U87MG human glioblastoma cells. Results: RCPN, with a particle size of <100 nm and 65% CPT encapsulation efficiency, exhibited a dose- and time-dependent cytotoxicity to glioblastoma cells. Compared with native CPT or unconjugated nanoparticles, RCPN induced apoptosis, increased reactive oxygen species generation and inhibited U87MG cell migration. Conclusion: cRGDfK-mediated and amphiphilic copolymer-based nanomedicines represent a new approach for improved delivery of anticancer drugs to and treatment of glioblastoma multiforme.
Collapse
Affiliation(s)
- Hitesh Kulhari
- Ian Potter NanoBioSensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Health Innovations Research Institute, RMIT University, Melbourne, VIC 3083, Australia
- IICT-RMIT Research Centre, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500607, India
- Medicinal Chemistry & Pharmacology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500607, India
| | - Srinivasa R Telukutla
- Ian Potter NanoBioSensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- IICT-RMIT Research Centre, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500607, India
- Medicinal Chemistry & Pharmacology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500607, India
| | - Deep Pooja
- Medicinal Chemistry & Pharmacology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500607, India
| | - Ravi Shukla
- Ian Potter NanoBioSensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Health Innovations Research Institute, RMIT University, Melbourne, VIC 3083, Australia
| | - Ramakrishna Sistla
- Medicinal Chemistry & Pharmacology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500607, India
| | - Vipul Bansal
- Ian Potter NanoBioSensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Health Innovations Research Institute, RMIT University, Melbourne, VIC 3083, Australia
| | - David J Adams
- Health Innovations Research Institute, RMIT University, Melbourne, VIC 3083, Australia
- Illawarra Health & Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
80
|
Atia M, Bogdán D, Brügger M, Haider N, Mátyus P. Remarkable regioselectivities in the course of the synthesis of two new Luotonin A derivatives. Tetrahedron 2017. [DOI: 10.1016/j.tet.2017.04.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
81
|
Chernov L, Deyell RJ, Anantha M, Dos Santos N, Gilabert‐Oriol R, Bally MB. Optimization of liposomal topotecan for use in treating neuroblastoma. Cancer Med 2017; 6:1240-1254. [PMID: 28544814 PMCID: PMC5463073 DOI: 10.1002/cam4.1083] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/29/2017] [Accepted: 04/03/2017] [Indexed: 12/20/2022] Open
Abstract
The purpose of this work was to develop an optimized liposomal formulation of topotecan for use in the treatment of patients with neuroblastoma. Drug exposure time studies were used to determine that topotecan (Hycamtin) exhibited great cytotoxic activity against SK-N-SH, IMR-32 and LAN-1 neuroblastoma human cell lines. Sphingomyelin (SM)/cholesterol (Chol) and 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC)/Chol liposomes were prepared using extrusion methods and then loaded with topotecan by pH gradient and copper-drug complexation. In vitro studies showed that SM/Chol liposomes retained topotecan significantly better than DSPC/Chol liposomes. Decreasing the drug-to-lipid ratio engendered significant increases in drug retention. Dose-range finding studies on NRG mice indicated that an optimized SM/Chol liposomal formulation of topotecan prepared with a final drug-to-lipid ratio of 0.025 (mol: mol) was better tolerated than the previously described DSPC/Chol topotecan formulation. Pharmacokinetic studies showed that the optimized SM/Chol liposomal topotecan exhibited a 10-fold increase in plasma half-life and a 1000-fold increase in AUC0-24 h when compared with Hycamtin administered at equivalent doses (5 mg/kg). In contrast to the great extension in exposure time, SM/Chol liposomal topotecan increased the life span of mice with established LAN-1 neuroblastoma tumors only modestly in a subcutaneous and systemic model. The extension in exposure time may still not be sufficient and the formulation may require further optimization. In the future, liposomal topotecan will be assessed in combination with high-dose radiotherapy such as 131 I-metaiodobenzylguanidine, and immunotherapy treatment modalities currently used in neuroblastoma therapy.
Collapse
Affiliation(s)
- Lina Chernov
- Experimental TherapeuticsBC Cancer Agency675 West 10 AvenueVancouverBritish ColumbiaV5Z 1L3Canada
- Department of Pathology and Laboratory MedicineUniversity of British Columbia2211 Wesbrook MallVancouverBritish ColumbiaV6T 2B5Canada
| | - Rebecca J. Deyell
- Division of Pediatric Hematology/OncologyBritish Columbia Children's Hospital and the University of British Columbia4480 Oak StreetVancouverBritish ColumbiaV6H 3V4Canada
- Michael Cuccione Childhood Cancer Research ProgramBritish Columbia Children's Hospital Research Institute950 West 28 AvenueVancouverBritish ColumbiaV5Z 4H4Canada
| | - Malathi Anantha
- Experimental TherapeuticsBC Cancer Agency675 West 10 AvenueVancouverBritish ColumbiaV5Z 1L3Canada
| | - Nancy Dos Santos
- Experimental TherapeuticsBC Cancer Agency675 West 10 AvenueVancouverBritish ColumbiaV5Z 1L3Canada
| | - Roger Gilabert‐Oriol
- Experimental TherapeuticsBC Cancer Agency675 West 10 AvenueVancouverBritish ColumbiaV5Z 1L3Canada
| | - Marcel B. Bally
- Experimental TherapeuticsBC Cancer Agency675 West 10 AvenueVancouverBritish ColumbiaV5Z 1L3Canada
- Department of Pathology and Laboratory MedicineUniversity of British Columbia2211 Wesbrook MallVancouverBritish ColumbiaV6T 2B5Canada
- Faculty of Pharmaceutical SciencesUniversity of British Columbia2405 Wesbrook MallVancouverBritish ColumbiaV6T 1Z3Canada
- Centre for Drug Research and Development4‐2405 Wesbrook MallVancouverBritish ColumbiaV6T 1Z3Canada
| |
Collapse
|
82
|
In vitro assay for measuring real time topotecan release from liposomes: release kinetics and cellular internalization. Drug Deliv Transl Res 2017; 7:544-557. [DOI: 10.1007/s13346-017-0380-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
83
|
Shah V, Bellantone RA, Taft DR. Evaluating the Potential for Delivery of Irinotecan via the Buccal Route: Physicochemical Characterization and In Vitro Permeation Assessment Across Porcine Buccal Mucosa. AAPS PharmSciTech 2017; 18:867-874. [PMID: 27363416 DOI: 10.1208/s12249-016-0578-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/19/2016] [Indexed: 11/30/2022] Open
Abstract
Irinotecan (CPT-11) is used to treat advanced colorectal cancer as an intravenous therapy. Depending on pH, CPT-11 exists in either a lactone (active) or carboxylate (inactive) form, or both. In this investigation, the feasibility for systemic delivery of CPT-11 through the buccal route was evaluated. Permeation of CPT-11 across porcine buccal mucosa was studied in vitro using side-by-side flow through diffusion cells at 37°C. Experiments were performed over a pH range from 4 to 9, and the permeability of both the lactone and carboxylate forms of CPT-11 was measured. CPT-11 steady state flux was determined over a range of donor concentrations at pH 4 (0.5, 1, 5, 10, 15, 20 mg/ml) and pH 6.8 (0.5, 5, 10 mg/ml). Steady state flux increased linearly with increasing donor concentration of CPT-11 at pH 4 (r 2 = 0.9935) and at pH 6.8 (r 2 = 0.9886). CPT-11 permeability was independent of pH, although the distribution coefficient increased with increasing pH. Estimates of permeability for the lactone and carboxylate forms were 4.16 × 10-5 cm/s and 2.6 × 10-5 cm/s, respectively. These calculated permeability values were in agreement with the in vitro experimental data. Overall, CPT-11 was found to permeate through porcine buccal mucosa via passive diffusion. CPT-11 permeability was independent of pH, suggesting that the compound was transported mainly via a paracellular route. Overall, the results of this research suggest that the buccal route is a potential extravascular mode of delivery for CPT-11.
Collapse
|
84
|
Martino E, Della Volpe S, Terribile E, Benetti E, Sakaj M, Centamore A, Sala A, Collina S. The long story of camptothecin: From traditional medicine to drugs. Bioorg Med Chem Lett 2016; 27:701-707. [PMID: 28073672 DOI: 10.1016/j.bmcl.2016.12.085] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022]
Abstract
20-(S)-Camptothecin (CPT) is a natural alkaloid extracted from the bark of Camptotheca acuminata (Chinese happy tree). It acts as a DNA topoisomerase 1 poison with an interesting antitumor activity and its use is limited by low stability and solubility and unpredictable drug-drug interactions. Since the late 20th century, it has been widely used in cancer therapy and, since extraction yields from plant tissues are very low, various synthetic routes have been developed to satisfy the increase in demand for CPT. Moreover, SAR studies have allowed for the development of more potent CPT analogues topotecan and irinotecan. Unfortunately, resistance has already occurred in several tumour lines. Additional studies are needed to better understand the relationship between substituents and resistance, its clinical relevance and the impact of related gene polymorphism. One of the latest research approaches focuses on modifying the delivery mode to improve tumour cell uptake and reduce toxicity.
Collapse
Affiliation(s)
- Emanuela Martino
- Department of Earth and Environmental Sciences, University of Pavia, Via S. Epifanio 14, 27100 Pavia, Italy
| | - Serena Della Volpe
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Elisa Terribile
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Emanuele Benetti
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Mirena Sakaj
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Adriana Centamore
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Andrea Sala
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Simona Collina
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| |
Collapse
|
85
|
Zhao L, Shao Z, Shanks JV. Anticancer Drugs. Ind Biotechnol (New Rochelle N Y) 2016. [DOI: 10.1002/9783527807833.ch8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Le Zhao
- Iowa State University; Department of Chemical and Biological Engineering; 4140 Biorenewables Research Laboratory, 617 Bissell Road Ames 50011 IA USA
| | - Zengyi Shao
- Iowa State University; Department of Chemical and Biological Engineering; 4140 Biorenewables Research Laboratory, 617 Bissell Road Ames 50011 IA USA
| | - Jacqueline V Shanks
- Iowa State University; Department of Chemical and Biological Engineering; 4140 Biorenewables Research Laboratory, 617 Bissell Road Ames 50011 IA USA
| |
Collapse
|
86
|
Integrating a novel SN38 prodrug into the PEGylated liposomal system as a robust platform for efficient cancer therapy in solid tumors. Int J Pharm 2016; 512:39-48. [PMID: 27544846 DOI: 10.1016/j.ijpharm.2016.08.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/26/2016] [Accepted: 08/17/2016] [Indexed: 12/20/2022]
Abstract
Liposomal nanoassemblies have been used extensively as carriers for the delivery of both lipophilic and hydrophilic drugs. They represent a mature, versatile technology with considerable potential for improving the pharmacokinetics of drugs. However, the formulation of many chemotherapeutics into liposome systems has posed a significant challenge due to their incompatible physicochemical properties, as was the case with camptothecin-based chemotherapeutics. Here, we present a rational paradigm of potent chemotherapeutics that were reconstructed and subsequently integrated into liposomal nanoassemblies. Using SN38 (7-ethyl-10-hydroxy camptothecin) as a model drug, a lipophilic prodrug 1 (designated as LA-SN38) was constructed by tethering the linoleic acid (LA) moiety via esterification, which was further facilitated to form liposomal nanoparticles (LipoNP) through supramolecular nanoassembly. The resulting 1-loaded LipoNP exhibited sustained drug release kinetics and decreased cellular uptake by macrophage cells. Uptake by tumor cells was enhanced relative to our previous supramolecular nanoparticles (SNP 1), which were derived from the self-assembling prodrug 1. Notably, LipoNP outperformed SNP 1 in terms of pharmacokinetics and in vivo therapeutic efficacy in both human BEL-7402 hepatocellular carcinoma (HCC) and HCT-116 colorectal cancer-derived xenograft mouse models. These results were likely due to the improved systemic circulation and preferential accumulation of nanodrugs in tumors. Hence, our results suggest that the combination of liposomal delivery platforms with rational prodrug engineering may emerge as a promising approach for the effective and safe delivery of anticancer chemotherapeutics.
Collapse
|
87
|
Vutey V, Castelli S, D'Annessa I, Sâmia LBP, Souza-Fagundes EM, Beraldo H, Desideri A. Human topoisomerase IB is a target of a thiosemicarbazone copper(II) complex. Arch Biochem Biophys 2016; 606:34-40. [PMID: 27431056 DOI: 10.1016/j.abb.2016.07.009] [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] [Received: 03/14/2016] [Revised: 06/30/2016] [Accepted: 07/12/2016] [Indexed: 02/03/2023]
Abstract
The human topoisomerase IB inhibition and the antiproliferative activity of 3-(4-bromophenyl)-1-pyridin-2-ylprop-2-en-1-one thiosemicarbazone HPyCT4BrPh alone and its copper(II) complex [Cu(PyCT4BrPh)Cl] was investigated. [Cu(PyCT4BrPh)Cl] inhibits both the DNA cleavage and religation step of the enzyme, whilst the ligand alone does not display any effect. In addition we show that coordination to copper(II) improves the cytotoxicity of HPyCT4BrPh against THP-1 leukemia and MCF-7 breast cancer cells. The data indicate that the copper(II) thiosemicarbazone complex may hit human topoisomerase IB and that metal coordination can be useful to improve cytotoxicity of this versatile class of compounds.
Collapse
Affiliation(s)
- Venn Vutey
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Silvia Castelli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Ilda D'Annessa
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Luciana B P Sâmia
- Departamento de Química, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine M Souza-Fagundes
- Departamento de Química, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | - Heloisa Beraldo
- Departamento de Química, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
88
|
Wang L, Fu P, Zhao Y, Wang G, Yu R, Wang X, Tang Z, Imperato-McGinley J, Zhu YS. Dissociation of NSC606985 induces atypical ER-stress and cell death in prostate cancer cells. Int J Oncol 2016; 49:529-38. [PMID: 27277821 DOI: 10.3892/ijo.2016.3555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/17/2016] [Indexed: 11/05/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) is a major cause of prostate cancer (Pca) death. Chemotherapy is able to improve the survival of CRPC patients. We previously found that NSC606985 (NSC), a highly water-soluble camptothecin analog, induced cell death in Pca cells via interaction with topoisomerase 1 and activation of the mitochondrial apoptotic pathway. To further elucidate the role of NSC, we studied the effect of NSC on ER-stress and its association with NSC-induced cell death in Pca cells. NSC produced a time- and dose-dependent induction of GRP78, CHOP and XBP1s mRNA, and CHOP protein expression in Pca cells including DU145, indicating an activation of ER-stress. However, unlike conventional ER-stress in which GRP78 protein is increased, NSC produced a time- and dose-dependent U-shape change in GRP78 protein in DU145 cells. The NSC-induced decrease in GRP78 protein was blocked by protease inhibitors, N-acetyl-L-leucyl-L-leucylnorleucinal (ALLN), a lysosomal protease inhibitor, and epoxomicin (EPO), a ubiquitin-protease inhibitor. ALLN, but not EPO, also partially inhibited NSC-induced cell death. However, both 4-PBA and TUDCA, two chemical chaperons that effectively reduced tunicamycin-induced ER-stress, failed to attenuate NSC-induced GRP78, CHOP and XBP1s mRNA expression and cell death. Moreover, knockdown of NSC induction of CHOP expression using a specific siRNA had no effect on NSC-induced cytochrome c release and NSC-induced cell death. These results suggest that NSC produced an atypical ER-stress that is dissociated from NSC-induced activation of the mitochondrial apoptotic pathway and NSC-induced cell death in DU145 prostate cancer cells.
Collapse
Affiliation(s)
- Liping Wang
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Pengcheng Fu
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yuan Zhao
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Guo Wang
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Richard Yu
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xin Wang
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Zehai Tang
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Yuan-Shan Zhu
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
89
|
Jensen NF, Agama K, Roy A, Smith DH, Pfister TD, Rømer MU, Zhang HL, Doroshow JH, Knudsen BR, Stenvang J, Brünner N, Pommier Y. Characterization of DNA topoisomerase I in three SN-38 resistant human colon cancer cell lines reveals a new pair of resistance-associated mutations. J Exp Clin Cancer Res 2016; 35:56. [PMID: 27029323 PMCID: PMC4815242 DOI: 10.1186/s13046-016-0335-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/23/2016] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND DNA topoisomerase I (Top1) is a DNA unwinding protein and the specific target of the camptothecin class of chemotherapeutic drugs. One of these, irinotecan, acting through its active metabolite SN-38, is used in the treatment of metastatic colorectal cancer. However, resistance to irinotecan represents a major clinical problem. Since molecular alterations in Top1 may result in resistance to irinotecan, we characterized Top1 in three human colon cancer cell lines with acquired resistance to SN-38. METHODS Three SN-38 resistant (20-67 fold increased resistance) cell lines were generated and compared to wild-type parental cells with regards to: TOP1 gene copy number and gene sequence, Top1 expression (mRNA and protein), Top1 enzymatic activity in the absence and presence of drug, and Top1-DNA cleavage complexes in drug treated cells. TOP1 mutations were validated by PCR using mutant specific primers. Furthermore, cross-resistance to two indenoisoquinoline Top1-targeting drugs (NSC 725776 and NSC 743400) and two Top2-targeting drugs (epirubicin and etoposide) was investigated. RESULTS Two of three SN-38 resistant cell lines carried TOP1 gene copy number aberrations: A TOP1 gene copy gain and a loss of chromosome 20, respectively. One resistant cell line harbored a pair of yet unreported TOP1 mutations (R364K and G717R) in close proximity to the drug binding site. Mutant TOP1 was expressed at a markedly higher level than wild-type TOP1. None or very small reductions were observed in Top1 expression or Top1 activity in the absence of drug. In all three SN-38 resistant cell lines Top1 activity was maintained in the presence of high concentrations of SN-38. None or only partial cross-resistance were observed for etoposide and epirubicin, respectively. SN-38 resistant cells with wild-type TOP1 remained sensitive to NSC 743400, while cells with mutant TOP1 was fully cross-resistant to both indenoisoquinolines. Top1-DNA cleavage complex formation following drug treatment supported the other findings. CONCLUSIONS This study adds to the growing knowledge about resistance mechanisms for Top1-targeting chemotherapeutic drugs. Importantly, two yet unreported TOP1 mutations were identified, and it was underlined that cross-resistance to the new indenoisoquinoline drugs depends on the specific underlying molecular mechanism of resistance to SN-38.
Collapse
Affiliation(s)
- Niels Frank Jensen
- />Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Keli Agama
- />National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Molecular Pharmacology, 37 Convent Drive, Building 37, Room 5068, Bethesda, MD 20892-4255 USA
| | - Amit Roy
- />Department of Molecular Biology and Genetics, Aarhus University, C.F. Møllers Allé 3, Building 1130, DK-8000 Aarhus C, Denmark
- />Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Vaishali 844102 India
| | - David Hersi Smith
- />Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
- />Dako Denmark A/S, R&D, Produktionsvej 42, DK-2600 Glostrup, Denmark
| | - Thomas D. Pfister
- />Laboratory of Human Toxicology and Pharmacology, Applied/Developmental Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Maria Unni Rømer
- />Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
- />Department for Clinical Physiology and Nuclear Medicine, Frederiksberg Hospital, Nordre Fasanvej 57, DK-2000 Frederiksberg C, Denmark
| | - Hong-Liang Zhang
- />National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Molecular Pharmacology, 37 Convent Drive, Building 37, Room 5068, Bethesda, MD 20892-4255 USA
| | - James H. Doroshow
- />National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Molecular Pharmacology, 37 Convent Drive, Building 37, Room 5068, Bethesda, MD 20892-4255 USA
- />Laboratory of Human Toxicology and Pharmacology, Applied/Developmental Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Birgitta R. Knudsen
- />Department of Molecular Biology and Genetics, Aarhus University, C.F. Møllers Allé 3, Building 1130, DK-8000 Aarhus C, Denmark
| | - Jan Stenvang
- />Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Nils Brünner
- />Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Yves Pommier
- />National Institutes of Health, National Cancer Institute, Center for Cancer Research, Laboratory of Molecular Pharmacology, 37 Convent Drive, Building 37, Room 5068, Bethesda, MD 20892-4255 USA
| |
Collapse
|
90
|
Nicolay NH, Rühle A, Perez RL, Trinh T, Sisombath S, Weber KJ, Schmezer P, Ho AD, Debus J, Saffrich R, Huber PE. Mesenchymal stem cells exhibit resistance to topoisomerase inhibition. Cancer Lett 2016; 374:75-84. [PMID: 26876302 DOI: 10.1016/j.canlet.2016.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/29/2016] [Accepted: 02/02/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND Inhibition of cellular topoisomerases has been established as an effective way of treating certain cancers, albeit with often high levels of toxicity to the bone marrow. While the involvement of mesenchymal stem cells (MSCs) in bone marrow homeostasis and regeneration has been well established, the effects of topoisomerase-inhibiting anticancer agents remain largely unknown. MATERIALS AND METHODS Human bone marrow MSCs were treated with topoisomerase I inhibitor irinotecan or topoisomerase II inhibitor etoposide, and survival and apoptosis levels were measured. The influence of topoisomerase inhibition on cellular morphology, adhesion and migration potential and the ability to differentiate was assessed. Additionally, the role of individual DNA double-strand break repair pathways in MSCs was investigated as a potential cellular mechanism of resistance to topoisomerase inhibitors. RESULTS Human bone marrow MSCs were found relatively resistant to topoisomerase I and II inhibitors and show survival levels comparable to these of differentiated fibroblasts. Treatment with irinotecan or etoposide did not significantly influence cellular adhesion, migratory ability, surface marker expression or induction of apoptosis in human MSCs. The ability to differentiate was found preserved in MSCs after exposure to high doses of irinotecan or etoposide. MSCs were able to efficiently repair DNA double-strand breaks induced by topoisomerase inhibitors both by non-homologous end joining and homologous recombination pathways. CONCLUSION Our data demonstrate a topoisomerase-resistant phenotype of human MSCs that may at least in part be due to the stem cells' ability to efficiently remove DNA damage caused by these anticancer agents. The observed resistance of MSCs warrants further investigation of these cells as a potential therapeutic option for treating topoisomerase inhibitor-induced bone marrow damage.
Collapse
Affiliation(s)
- Nils H Nicolay
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Alexander Rühle
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Thuy Trinh
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Sonevisay Sisombath
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Klaus-Josef Weber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Peter Schmezer
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Anthony D Ho
- Department of Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Rainer Saffrich
- Department of Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
91
|
Synergistic Effects of Simvastatin and Irinotecan against Colon Cancer Cells with or without Irinotecan Resistance. Gastroenterol Res Pract 2016; 2016:7891374. [PMID: 26966430 PMCID: PMC4757714 DOI: 10.1155/2016/7891374] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/30/2015] [Accepted: 01/11/2016] [Indexed: 01/15/2023] Open
Abstract
Aims. We here investigated whether the combination of simvastatin and irinotecan could induce the synergistic effect on colon cancer cells with or without resistance to irinotecan. Methods. We investigated cell proliferation assay and assessed cell death detection ELISA and caspase-3 activity assay of various concentrations of simvastatin and irinotecan to evaluate the efficacy of drug combination on colon cancer cells with or without irinotecan resistance. Results. The IC50 values of simvastatin alone and irinotecan alone were 115.4 ± 0.14 μM (r = 0.98) and 62.5 ± 0.18 μM (r = 0.98) in HT-29 cells without resistance to irinotecan. The IC50 values of these two drugs were 221.9 ± 0.22 μM (r = 0.98) and 195.9 ± 0.16 μM (r = 0.99), respectively, in HT-29 cell with resistance to irinotecan. The results of combinations of the various concentrations of two drugs showed that combined treatment with irinotecan and simvastatin more efficiently suppressed cell proliferation of HT-29 cells even with resistance to irinotecan as well as without resistance. Furthermore, the combination of simvastatin and irinotecan at 2 : 1 molar ratio showed the best synergistic interaction. Conclusion. Simvastatin could act synergistically with irinotecan to overcome irinotecan resistance of colon cancer.
Collapse
|
92
|
Dosio F, Arpicco S, Stella B, Fattal E. Hyaluronic acid for anticancer drug and nucleic acid delivery. Adv Drug Deliv Rev 2016; 97:204-36. [PMID: 26592477 DOI: 10.1016/j.addr.2015.11.011] [Citation(s) in RCA: 403] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 01/06/2023]
Abstract
Hyaluronic acid (HA) is widely used in anticancer drug delivery, since it is biocompatible, biodegradable, non-toxic, and non-immunogenic; moreover, HA receptors are overexpressed on many tumor cells. Exploiting this ligand-receptor interaction, the use of HA is now a rapidly-growing platform for targeting CD44-overexpressing cells, to improve anticancer therapies. The rationale underlying approaches, chemical strategies, and recent advances in the use of HA to design drug carriers for delivering anticancer agents, are reviewed. Comprehensive descriptions are given of HA-based drug conjugates, particulate carriers (micelles, liposomes, nanoparticles, microparticles), inorganic nanostructures, and hydrogels, with particular emphasis on reports of preclinical/clinical results.
Collapse
|
93
|
Yi L, Lin G, Zhang K, Wang L, Zhang R, Xie J, Li J. Molecular Genetics External Quality Assessment Pilot Scheme for Irinotecan-Related UGT1A1 Genotyping in China. PLoS One 2016; 11:e0148081. [PMID: 26820647 PMCID: PMC4731084 DOI: 10.1371/journal.pone.0148081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 01/12/2016] [Indexed: 12/27/2022] Open
Abstract
Irinotecan is widely used in the treatment of solid tumors, especially in colorectal cancer and lung cancer. Molecular testing for UGT1A1 genotyping is increasingly required in China for optimum irinotecan administration. In order to determine the performance of laboratories with regard to the whole testing process for UGT1A1 to ensure the consistency and accuracy of the test results, the National Center for Clinical Laboratories conducted an external quality assessment program for UGT1A1*28 genotyping in 2015. The panel, which comprised of four known mutational samples and six wild-type samples, was distributed to 45 laboratories that test for the presence of UGT1A1*28 polymorphisms. Participating laboratories were allowed to perform polymorphism analysis by using their routine methods. The accuracy of the genotyping and reporting of results was analyzed. Other information from the individual laboratories, including the number of samples tested each month, accreditation/certification status, and test methodology, was reviewed. Forty-four of the 45 participants reported the correct results for all samples. There was only one genotyping error, with a corresponding analytical sensitivity of 99.44% (179/180 challenges; 95% confidence interval: 96.94-99.99%) and an analytical specificity of 100% (270/270 challenges; 95% confidence interval: 98.64-100%). Both commercial kits and laboratory development tests were commonly used by the laboratories, and pyrosequencing was the main methodology used (n = 26, 57.8%). The style of the written reports showed large variation, and many reports showed a shortage of information. In summary, the first UGT1A1 genotyping external quality assessment result demonstrated that UGT1A1 genotype analysis of good quality was performed in the majority of pharmacogenetic testing centers that were investigated. However, greater education on the reporting of UGT1A1 genetic testing results is needed.
Collapse
Affiliation(s)
- Lang Yi
- National Center for Clinical Laboratories, Beijing Hospital, Beijing 100730, P R China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P R China
| | - Guigao Lin
- National Center for Clinical Laboratories, Beijing Hospital, Beijing 100730, P R China
| | - Kuo Zhang
- National Center for Clinical Laboratories, Beijing Hospital, Beijing 100730, P R China
| | - Lunan Wang
- National Center for Clinical Laboratories, Beijing Hospital, Beijing 100730, P R China
| | - Rui Zhang
- National Center for Clinical Laboratories, Beijing Hospital, Beijing 100730, P R China
| | - Jiehong Xie
- National Center for Clinical Laboratories, Beijing Hospital, Beijing 100730, P R China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, Beijing 100730, P R China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P R China
| |
Collapse
|
94
|
Zheng J, Chan T, Zhu L, Yan X, Cao Z, Wang Y, Zhou F. The inhibitory effects of camptothecin (CPT) and its derivatives on the substrate uptakes mediated by human solute carrier transporters (SLCs). Xenobiotica 2016; 46:831-40. [DOI: 10.3109/00498254.2015.1129080] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jian Zheng
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration in Oil Field, Ministry of Education, Harbin, P.R. China,
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia,
| | - Ting Chan
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia,
| | - Ling Zhu
- Save Sight Institute, University of Sydney, Sydney, NSW, Australia, and
| | - Xiufeng Yan
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration in Oil Field, Ministry of Education, Harbin, P.R. China,
| | - Zhisong Cao
- The CHRISTUS Stehlin Foundation for Cancer Research, Houston, TX, USA
| | - Yang Wang
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration in Oil Field, Ministry of Education, Harbin, P.R. China,
| | - Fanfan Zhou
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia,
| |
Collapse
|
95
|
Martínez-Carmona M, Lozano D, Colilla M, Vallet-Regí M. Selective topotecan delivery to cancer cells by targeted pH-sensitive mesoporous silica nanoparticles. RSC Adv 2016. [DOI: 10.1039/c6ra07763c] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Topotecan targeted pH-sensitive delivery system based in mesoporous silica nanoparticles coated with a multifunctional biopolymer coating for cancer therapy.
Collapse
Affiliation(s)
- M. Martínez-Carmona
- Departamento de Química Inorgánica y Bioinorgánica
- Facultad de Farmacia
- Universidad Complutense de Madrid
- Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12
- 28040 Madrid
| | - D. Lozano
- Departamento de Química Inorgánica y Bioinorgánica
- Facultad de Farmacia
- Universidad Complutense de Madrid
- Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12
- 28040 Madrid
| | - M. Colilla
- Departamento de Química Inorgánica y Bioinorgánica
- Facultad de Farmacia
- Universidad Complutense de Madrid
- Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12
- 28040 Madrid
| | - M. Vallet-Regí
- Departamento de Química Inorgánica y Bioinorgánica
- Facultad de Farmacia
- Universidad Complutense de Madrid
- Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12
- 28040 Madrid
| |
Collapse
|
96
|
Sakasai R, Iwabuchi K. The distinctive cellular responses to DNA strand breaks caused by a DNA topoisomerase I poison in conjunction with DNA replication and RNA transcription. Genes Genet Syst 2015; 90:187-94. [PMID: 26616758 DOI: 10.1266/ggs.15-00023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Camptothecin (CPT) inhibits DNA topoisomerase I (Top1) through a non-catalytic mechanism that stabilizes the Top1-DNA cleavage complex (Top1cc) and blocks the DNA re-ligation step, resulting in the accumulation in the genome of DNA single-strand breaks (SSBs), which are converted to secondary strand breaks when they collide with the DNA replication and RNA transcription machinery. DNA strand breaks mediated by replication, which have one DNA end, are distinct in repair from the DNA double-strand breaks (DSBs) that have two ends and are caused by ionizing radiation and other agents. In contrast to two-ended DSBs, such one-ended DSBs are preferentially repaired through the homologous recombination pathway. Conversely, the repair of one-ended DSBs by the non-homologous end-joining pathway is harmful for cells and leads to cell death. The choice of repair pathway has a crucial impact on cell fate and influences the efficacy of anticancer drugs such as CPT derivatives. In addition to replication-mediated one-ended DSBs, transcription also generates DNA strand breaks upon collision with the Top1cc. Some reports suggest that transcription-mediated DNA strand breaks correlate with neurodegenerative diseases. However, the details of the repair mechanisms of, and cellular responses to, transcription-mediated DNA strand breaks still remain unclear. In this review, combining our recent results and those of previous reports, we introduce and discuss the responses to CPT-induced DNA damage mediated by DNA replication and RNA transcription.
Collapse
Affiliation(s)
- Ryo Sakasai
- Department of Biochemistry I, Kanazawa Medical University
| | | |
Collapse
|
97
|
Lee H, Uhm S, Shin JW, Jeon HM, Dongbang S, Jung HS, Na YC, Kang C, Kim JS. HepG2 Cell Resistance against Camptothecin from a Lysosomal Drug Delivery. Chem Asian J 2015; 10:2695-700. [DOI: 10.1002/asia.201500913] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Hoyeon Lee
- The School of East-West Medical Science; Kyung Hee University; Yongin 446-701 Korea
| | - Soojin Uhm
- Department of Chemistry; Korea University; Seoul 136-701 Korea
| | - Jung-Won Shin
- The School of East-West Medical Science; Kyung Hee University; Yongin 446-701 Korea
| | - Hyun Mi Jeon
- The School of East-West Medical Science; Kyung Hee University; Yongin 446-701 Korea
| | - Sun Dongbang
- Department of Chemistry; Korea University; Seoul 136-701 Korea
| | - Hyo Sung Jung
- Department of Chemistry; Korea University; Seoul 136-701 Korea
| | - Yun-Cheol Na
- Omics System Research Team; Korea Basic Science Institute; 150 Bugahyeon-ro, Seodaemun-gu Seoul 120-140 Republic of Korea
| | - Chulhun Kang
- The School of East-West Medical Science; Kyung Hee University; Yongin 446-701 Korea
| | - Jong Seung Kim
- Department of Chemistry; Korea University; Seoul 136-701 Korea
| |
Collapse
|
98
|
Camptothecin and topotecan inhibit adipocyte differentiation by inducing degradation of PPARγ. Biochem Biophys Res Commun 2015; 463:1122-8. [DOI: 10.1016/j.bbrc.2015.06.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/09/2015] [Indexed: 12/21/2022]
|
99
|
Naumczuk B, Hyz K, Kawęcki R, Bocian W, Bednarek E, Sitkowski J, Wielgus E, Kozerski L. DOSY NMR and MALDI-TOF evidence of covalent binding the DNA duplex by trimethylammonium salts of topotecan upon near UV irradiation. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2015; 53:565-571. [PMID: 26017759 DOI: 10.1002/mrc.4255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/25/2015] [Accepted: 04/01/2015] [Indexed: 06/04/2023]
Abstract
Using DOSY NMR and MALDI-TOF MS techniques, we present evidence that quaternary trimethylammonium salts of topotecan, [TPT-NMe3 ](+) X(-) (X = CF3SO3, HCOO), bind covalently the natural DNA oligomer upon near UV irradiation in water under physiological conditions. It is shown that formate salt is very reactive at pH 7 and requires short irradiation time. This weak irradiation at 365 nm paves the way for a new application of TPT derivatives in clinical use, which can dramatically increase the therapeutic effects of a medicine.
Collapse
Affiliation(s)
- Beata Naumczuk
- Institute of Organic Chemistry, Polish Academy of Sciences, 01-224, Warsaw, Kasprzaka 44, Poland
| | - Karolina Hyz
- Institute of Organic Chemistry, Polish Academy of Sciences, 01-224, Warsaw, Kasprzaka 44, Poland
| | - Robert Kawęcki
- Institute of Organic Chemistry, Polish Academy of Sciences, 01-224, Warsaw, Kasprzaka 44, Poland
- University of Natural Sciences and Humanities, 80-110, Siedlce, Poland
| | - Wojciech Bocian
- Institute of Organic Chemistry, Polish Academy of Sciences, 01-224, Warsaw, Kasprzaka 44, Poland
- National Medicines Institute, 00-725, Warsaw, Chełmska 30/34, Poland
| | - Elżbieta Bednarek
- National Medicines Institute, 00-725, Warsaw, Chełmska 30/34, Poland
| | - Jerzy Sitkowski
- Institute of Organic Chemistry, Polish Academy of Sciences, 01-224, Warsaw, Kasprzaka 44, Poland
- National Medicines Institute, 00-725, Warsaw, Chełmska 30/34, Poland
| | - Ewelina Wielgus
- Center of Molecular and Macromolecular Studies, Polish Academy of Sciences, Łódź, Poland
| | - Lech Kozerski
- Institute of Organic Chemistry, Polish Academy of Sciences, 01-224, Warsaw, Kasprzaka 44, Poland
- National Medicines Institute, 00-725, Warsaw, Chełmska 30/34, Poland
| |
Collapse
|
100
|
Xu G, Shi C, Guo D, Wang L, Ling Y, Han X, Luo J. Functional-segregated coumarin-containing telodendrimer nanocarriers for efficient delivery of SN-38 for colon cancer treatment. Acta Biomater 2015; 21:85-98. [PMID: 25910639 DOI: 10.1016/j.actbio.2015.04.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/25/2015] [Accepted: 04/16/2015] [Indexed: 12/17/2022]
Abstract
Four coumarin-containing telodendrimers (denoted as P-I, P-II, P-III and P-IV) were designed and synthesized to self-assemble into the corresponding nanoparticles. Of those, two nanoparticles (P-II and P-IV micelles) were screened and selected for targeted drug delivery of 7-ethyl-10-hydroxy camptothecin (SN-38), a prominent and efficacious anticancer agent, for the treatment of colon cancers. The nanoparticle encapsulation significantly increased the solubility of SN-38 in aqueous solution. Dynamic light scattering (DLS) showed the size of these SN-38 nanoparticles to be around 50 nm, and rod-shaped micelles were observed using transmission electron microscopy (TEM). These two novel nanoformulations of SN-38/P-II and SN-38/P-IV were found to exhibit similar in vitro cytotoxic activity against colon cancer cells as the free drug (SN-38 in DMSO) and were 500-fold more potent than irinotecan (a prodrug of SN-38). In addition, near infrared fluorescent (NIRF) optical imaging was utilized to monitor the tumor targeted delivery of SN-38/NPs via co-loading a NIRF dye. It was demonstrated that these NPs preferentially accumulated in tumors when compared to healthy tissue. A pharmacokinetics study showed that SN-38 micelle formulations had a longer circulating time in blood than irinotecan. Furthermore, SN-38 loaded nanoformulations exhibit superior anti-tumor efficacy when compared with irinotecan at equivalent SN-38 dose in HT-29 human colon cancer xenograft models.
Collapse
Affiliation(s)
- Gaofei Xu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States; Department of Applied Chemistry, Science College, China Agricultural University, Beijing 100193, China
| | - Changying Shi
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Dandan Guo
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Lili Wang
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Yun Ling
- Department of Applied Chemistry, Science College, China Agricultural University, Beijing 100193, China
| | - Xiaobing Han
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Juntao Luo
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States.
| |
Collapse
|