1
|
Huang X, Xu W, Li M, Zhang P, Zhang YS, Ding J, Chen X. Antiviral biomaterials. MATTER 2021; 4:1892-1918. [DOI: 10.1016/j.matt.2021.03.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
|
2
|
Piao X, Yin H, Guo S, Wang H, Guo P. RNA Nanotechnology to Solubilize Hydrophobic Antitumor Drug for Targeted Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900951. [PMID: 31763137 PMCID: PMC6864502 DOI: 10.1002/advs.201900951] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/06/2019] [Indexed: 05/15/2023]
Abstract
Small-molecule drugs are used extensively in clinics for cancer treatment; however, many antitumor chemical drugs dissolve poorly in aqueous solution. Their poor solubility and nonselective delivery in vivo often cause severe side effects. Here, the application of RNA nanotechnology to enhance the solubility of hydrophobic drugs, using camptothecin (CPT) for proof-of-concept in targeted delivery for cancer treatment is reported. Multiple CPT prodrug molecules are conjugated to RNA oligos via a click reaction, and the resulting CPT-RNA conjugates efficiently self-assemble into thermodynamically stable RNA three-way junction (3WJ) nanoparticles. The RNA 3WJ is covalently linked with seven hydrophobic CPT prodrug molecules through cleavable ester bonds and a folic acid ligand for specific tumor targeting while remaining soluble in aqueous solutions without detectable aggregation at therapeutic dose. This CPT-RNA nanoparticle exhibits efficient and specific cell binding and internalization, leading to cell apoptosis. Tumor growth is effectively inhibited by CPT-RNA nanoparticles; the targeted delivery, strengthened by tumor ligand, further enhances tumor suppression. Compared with the traditional formulation, solubilization of CPT in aqueous buffer using RNA nanoparticles as a carrier is found to be safe and efficacious, demonstrating that RNA nanoparticles are a promising platform for the solubilization and the delivery of hydrophobic antitumor drugs.
Collapse
Affiliation(s)
- Xijun Piao
- Center for RNA Nanobiotechnology and NanomedicineThe Ohio State UniversityColumbusOH43210USA
- College of PharmacyDivision of Pharmaceutics and PharmacologyThe Ohio State UniversityColumbusOH43210USA
| | - Hongran Yin
- Center for RNA Nanobiotechnology and NanomedicineThe Ohio State UniversityColumbusOH43210USA
- College of PharmacyDivision of Pharmaceutics and PharmacologyThe Ohio State UniversityColumbusOH43210USA
| | - Sijin Guo
- Center for RNA Nanobiotechnology and NanomedicineThe Ohio State UniversityColumbusOH43210USA
- College of PharmacyDivision of Pharmaceutics and PharmacologyThe Ohio State UniversityColumbusOH43210USA
| | - Hongzhi Wang
- Center for RNA Nanobiotechnology and NanomedicineThe Ohio State UniversityColumbusOH43210USA
- College of PharmacyDivision of Pharmaceutics and PharmacologyThe Ohio State UniversityColumbusOH43210USA
| | - Peixuan Guo
- Center for RNA Nanobiotechnology and NanomedicineThe Ohio State UniversityColumbusOH43210USA
- College of PharmacyDivision of Pharmaceutics and PharmacologyThe Ohio State UniversityColumbusOH43210USA
- College of MedicineDorothy M. Davis Heart and Lung Research InstituteThe Ohio State UniversityColumbusOH43210USA
- James Comprehensive Cancer CenterThe Ohio State UniversityColumbusOH43210USA
| |
Collapse
|
3
|
Zhou Z, Piao Y, Hao L, Wang G, Zhou Z, Shen Y. Acidity-responsive shell-sheddable camptothecin-based nanofibers for carrier-free cancer drug delivery. NANOSCALE 2019; 11:15907-15916. [PMID: 31414111 DOI: 10.1039/c9nr03872h] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Small molecular prodrugs that self-assemble into nanoparticles have many advantages over commonly studied nanomedicines based upon nanoscale carriers such as liposomes, micelles and polymeric nanoparticles. These carrier-free nanodrugs exhibit favorable nanoproperties without the help of a nanocarrier, and they have many unique merits, such as a simple synthetic procedure, well-defined structure and high drug loading capacity. To date, most of these carrier-free nanodrugs have been spherical and very few nonspherical nanodrugs have been synthesized and studied. Herein, we report a camptothecin (CPT) prodrug that self-assembles into nanofibers. These carrier-free CPT nanofibers have a width of approximately one hundred nanometers and a length of several micrometers. The cellular uptake and tumor penetration behaviour of these nanofibers were observed by time-lapse video microscopy. These nanofibers can rapidly enter cancer cells by penetrating the cell membrane, gradually dissolve intracellularly and efficiently release the active drug. Coating the surface of these nanofibers with a pH-responsive PEG layer improves the stability of these nanofibers and shields their positive charge to minimize nonspecific interactions. These pH-responsive nanofibers are sheddable in the acidic tumor microenvironment and deliver carried cargoes deep into tumors. Our findings demonstrate that small molecular CPT prodrugs that form nanofibers are efficient for cancer drug delivery.
Collapse
Affiliation(s)
- Zhuha Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, East Qingchun Road 3, 310016, Hangzhou, Zhejiang, China.
| | | | | | | | | | | |
Collapse
|
4
|
|
5
|
Lancelot A, Clavería-Gimeno R, Velázquez-Campoy A, Abian O, Serrano JL, Sierra T. Nanostructures based on ammonium-terminated amphiphilic Janus dendrimers as camptothecin carriers with antiviral activity. Eur Polym J 2017. [DOI: 10.1016/j.eurpolymj.2017.03.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
6
|
Liu Y, Li D, Guo X, Xu H, Li Z, Zhang Y, Song C, Fan R, Tang X, Zhang Z. A pH-responsive prodrug delivery system of 10-HCPT for controlled release and tumor targeting. Int J Nanomedicine 2017; 12:2227-2242. [PMID: 28356739 PMCID: PMC5367588 DOI: 10.2147/ijn.s125849] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We synthesized a pH-responsive conjugate of 10-hydroxycamptothecin-thiosemicarbazide-linear polyethylene glycol 2000 (PEG2000). The conjugate was confirmed by matrix-assisted laser desorption time of flight mass spectrometry, 1H NMR, and 13C NMR. The water solubility of the prodrug was increased by over 3,000 times; much longer body circulation time, higher tumor-targeting ability, and reduced toxicity were observed, compared with commercial 10-HCPT injection. The linker contains a pH-sensitive hydrazone bond, which breaks under low pH conditions in the tumor microenvironment. The conjugates showed good stability in phosphate-buffered saline (pH 7.4) and rat plasma. This amphiphilic conjugate could self-assemble into nanosized micelles of 80–100 nm. Cytotoxicity assay results indicate significantly higher efficacy of the conjugate (IC50 [half maximal inhibitory concentration] =0.117 µM on SW180 cells) than 10-HCPT solution (IC50 =0.241 µM on SW480 cells). Cellular uptake analysis suggested its rapid internalization and nuclear transport. Pharmacokinetic analysis of the conjugates demonstrated that the conjugate circulated for a longer time in the blood circulation system (T2/1 =10.516±1.158 h) than did 10-HCPT solution (T2/1 =1.859±1.385 h), and that it also enhanced the targeting and mean residence time (MRT0–inf =39.873±4.549 h) in the tumor site, compared with 10-HCPT (MRT0–inf =9.247±1.026 h). Finally, the conjugate demonstrated an increased tumor growth inhibition effect (TIR =82.66%±7.175%) in vivo and lower side effects than 10-HCPT (TIR =63.85%±5.233%). This prodrug holds great promise in improving therapeutic efficacy and overcoming multidrug resistance.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang; Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou; Department of Pharmaceutics, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan, Zhengzhou
| | - Dan Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou
| | - Xinhong Guo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou
| | - Haiwei Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou
| | - Zhi Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou
| | - Yanling Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou
| | - Chuanjun Song
- Department of Organic Chemistry, College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Ruhan Fan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang
| | - Zhenzhong Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou
| |
Collapse
|
7
|
Li B, Zhao W, Zhang X, Wang J, Luo X, Baker SD, Jordan CT, Dong Y. Design, synthesis and evaluation of anti-CD123 antibody drug conjugates. Bioorg Med Chem 2016; 24:5855-5860. [PMID: 27687970 DOI: 10.1016/j.bmc.2016.09.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 01/02/2023]
Abstract
Leukemia stem cells (LSCs) account for the development of drug resistance and increased recurrence rate in acute myeloid leukemia (AML) patients. Targeted drug delivery to leukemia stem cells remains a major challenge in AML chemotherapy. Overexpressed interleukin-3 receptor alpha chain, CD123, on the surface of leukemia stem cells was reported to be a potential target in AML treatment. Here, we designed and developed an antibody drug conjugate (CD123-CPT) by integrating anti-CD123 antibody with a chemotherapeutic agent, Camptothecin (CPT), via a disulfide linker. The linker is biodegradable in the presence of Glutathione (GSH, an endogenous component in cells), which leads to release of CPT. Anti-CD123 antibody conjugates showed significant higher cellular uptake in CD123-overexpressed tumor cells. More importantly, CD123-CPT demonstrated potent inhibitory effects on CD123-overexpressed tumor cells. Consequently, these results provide a promising targeted chemotherapeutical strategy for AML treatment.
Collapse
Affiliation(s)
- Bin Li
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Weiyu Zhao
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Xinfu Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Junfeng Wang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Xiao Luo
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Sharyn D Baker
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Craig T Jordan
- Division of Hematology, University of Colorado, Aurora, CO 80045, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
8
|
Román JV, Galán MA, del Valle EMM. Preparation and preliminary evaluation of alginate crosslinked microcapsules as potential drug delivery system (DDS) for human lung cancer therapy. Biomed Phys Eng Express 2016. [DOI: 10.1088/2057-1976/2/3/035015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
9
|
Lopes-de-Araújo J, Neves AR, Gouveia VM, Moura CC, Nunes C, Reis S. Oxaprozin-Loaded Lipid Nanoparticles towards Overcoming NSAIDs Side-Effects. Pharm Res 2015; 33:301-14. [PMID: 26350105 DOI: 10.1007/s11095-015-1788-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/31/2015] [Indexed: 01/15/2023]
Abstract
PURPOSE Nanostructured Lipid Carriers (NLCs) loading oxaprozin were developed to address an effective drug packaging and targeted delivery, improving the drug pharmacokinetics and pharmacodynamics properties and avoiding the local gastric side-effects. Macrophages actively phagocyte particles with sizes larger than 200 nm and, when activated, over-express folate beta receptors - features that in the case of this work constitute the basis for passive and active targeting strategies. METHODS Two formulations containing oxaprozin were developed: NLCs with and without folate functionalization. In order to target the macrophages folate receptors, a DSPE-PEG2000-FA conjugate was synthesized and added to the NLCs. RESULTS These formulations presented a relatively low polydispersity index (approximately 0.2) with mean diameters greater than 200 nm and zeta potential inferior to -40 mV. The encapsulation efficiency of the particles was superior to 95% and the loading capacity was of 9%, approximately. The formulations retained the oxaprozin release in simulated gastric fluid (only around 10%) promoting its release on simulated intestinal fluid. MTT and LDH assays revealed that the formulations only presented cytotoxicity in Caco-2 cells for oxaprozin concentrations superior to 100 μM. Permeability studies in Caco-2 cells shown that oxaprozin encapsulation did not interfered with oxaprozin permeability (around 0.8 × 10(-5) cm/s in simulated intestinal fluid and about 1.45 × 10(-5) cm/s in PBS). Moreover, in RAW 264.7 cells NLCs functionalization promoted an increased uptake over time mainly mediated by a caveolae uptake mechanism. CONCLUSIONS The developed nanoparticles enclose a great potential for oxaprozin oral administration with significant less gastric side-effects.
Collapse
Affiliation(s)
- José Lopes-de-Araújo
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana Rute Neves
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Virgínia M Gouveia
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Catarina C Moura
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Cláudia Nunes
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Salette Reis
- UCIBIO, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| |
Collapse
|
10
|
Zhang Y, Chen T, Yuan P, Tian R, Hu W, Tang Y, Jia Y, Zhang L. Encapsulation of honokiol into self-assembled pectin nanoparticles for drug delivery to HepG2 cells. Carbohydr Polym 2015; 133:31-8. [PMID: 26344251 DOI: 10.1016/j.carbpol.2015.06.102] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/25/2015] [Accepted: 06/26/2015] [Indexed: 12/28/2022]
Abstract
Self-assembled pectin nanoparticles was prepared and evaluated for delivering the hydrophobic drug, honokiol (HK), to HepG2 cells. These hydrophobic drug-loaded nanoparticles were developed without using any surfactant and organic solvent. Hydroxypropyl-β-cyclodextrin (HCD) was used to fabricate an inclusion complex with HK (HKHCD) to increase the solubility of the drug and thus facilitate its encapsulation and dispersion in the pectin nanoparticles. Investigation of the in vitro release indicated that the drug-loaded nanoparticles exhibited a higher drug release rate than free honokiol and an effective sustained-release. Cytotoxicity, cell apoptosis and cellular uptake studies further confirmed that the pectin nanoparticles with galactose residues generated higher cytotoxicity than free honokiol on HepG2 cells which highly expressed asialoglycoprotein receptors (ASGR). Nevertheless, these findings were not observed in ASGR-negative A549 cells under similar condition. Therefore, pectin nanoparticles demonstrated a specific active targeting ability to ASGR-positive HepG2 cells and could be used as a potential drug carrier for treatment of liver-related tumors.
Collapse
Affiliation(s)
- Yuxia Zhang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Medicine Engineering Research Center, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Tong Chen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, PR China
| | - Pei Yuan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Medicine Engineering Research Center, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Rui Tian
- The Experimental Teaching Centre, Chongqing Medical University, Chongqing 400016, PR China
| | - Wenjing Hu
- Chongqingshi Shapingba District People's Hospital, Chongqing 400030, PR China
| | - Yalan Tang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Medicine Engineering Research Center, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Yuntao Jia
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Liangke Zhang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Medicine Engineering Research Center, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
11
|
A comparative study of folate receptor-targeted doxorubicin delivery systems: Dosing regimens and therapeutic index. J Control Release 2015; 208:106-20. [DOI: 10.1016/j.jconrel.2015.04.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/03/2015] [Accepted: 04/09/2015] [Indexed: 01/07/2023]
|
12
|
Bonzi G, Salmaso S, Scomparin A, Eldar-Boock A, Satchi-Fainaro R, Caliceti P. Novel pullulan bioconjugate for selective breast cancer bone metastases treatment. Bioconjug Chem 2015; 26:489-501. [PMID: 25613006 DOI: 10.1021/bc500614b] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A novel polysaccharide bioconjugate was designed to selectively target breast cancer bone metastases using a bisphosphonate moiety (alendronate, ALN). Paclitaxel (PTX) was first covalently conjugated to pullulan (Pull) through a Cathepsin K-sensitive tetrapeptide spacer followed by a self-immolative aminobenzyl alcohol spacer to obtain Pull-(GGPNle-φ-PTX). ALN was then conjugated to the polymeric backbone of Pull-(GGPNle-φ-PTX) via a PEG spacer. The final bioconjugate Pull-(GGPNle-φ-PTX)-(PEG-ALN) was found to assemble into colloidal spherical structures, which were physically and chemically stable under physiological conditions. In vitro studies showed that Pull-(GGPNle-φ-PTX)-(PEG-ALN) had strong affinity for hydroxyapatite, which simulates the bone tissue. Paclitaxel was rapidly released from the bioconjugate by Cathepsin K cleavage under pathological conditions. All studies performed using human MDA-MB-231-BM (bone metastases-originated clone), murine 4T1 breast cancer cells, murine K7M2, and human SAOS-2 osteosarcoma cells showed that the bioconjugate exerted an enhanced antiproliferative activity compared to the conjugate without the ALN. Furthermore, the nanoconjugate inhibited the migration of cancer cells and further displayed potent anti-angiogenic activity. In conclusion, the results showed that this conjugate has an excellent potential for selective treatment of bone neoplasms such as breast cancer bone metastases and osteosarcoma.
Collapse
Affiliation(s)
- Gwénaëlle Bonzi
- †Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Stefano Salmaso
- †Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Anna Scomparin
- ‡Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anat Eldar-Boock
- ‡Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- ‡Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Paolo Caliceti
- †Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| |
Collapse
|
13
|
Gao J, Chen P, Singh Y, Zhang X, Szekely Z, Stein S, Sinko PJ. Novel monodisperse PEGtide dendrons: design, fabrication, and evaluation of mannose receptor-mediated macrophage targeting. Bioconjug Chem 2014; 24:1332-44. [PMID: 23808323 DOI: 10.1021/bc400011v] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Novel PEGtide dendrons of generations 1 through 5 (G1.0–5.0) containing alternating discrete poly(ethylene glycol) (dPEG) and amino acid/peptide moieties were designed and developed. To demonstrate their targeting utility as nanocarriers, PEGtide dendrons functionalized with mannose residues were developed and evaluated for macrophage targeting. PEGtide dendrons were synthesized using 9-fluorenylmethyloxycarbonyl (Fmoc) solid-phase peptide synthesis (SPPS) protocols. The N-α-Fmoc-N-ε-(5-carboxyfluorescein)-l-lysine (Fmoc-Lys(5-FAM)-OH) and monodisperse Fmoc-dPEG6-OH were sequentially coupled to Fmoc-β-Ala-resin to obtain the resin-bound intermediate Fmoc-dPEG6-Lys(5-FAM)-β-Ala (1). G1.0 dendrons were obtained by sequentially coupling Fmoc-Lys(Fmoc)-OH, Fmoc-β-Ala-OH, and Fmoc-dPEG6-OH to 1. Dendrons of higher generation, G2.0–5.0, were obtained by repeating the coupling cycles used for the synthesis of G1.0. Dendrons containing eight mannose residues (G3.0-mannose8) were developed for mannose receptor (MR) mediated macrophage targeting by conjugating α-d-mannopyranosylphenyl isothiocyanate to G3.0 dendrons. In the present study PEGtide dendrons up to G5.0 were synthesized. The molecular weights of the dendrons determined by MALDI-TOF were in agreement with calculated values. The hydrodynamic diameters measured using dynamic light scattering (DLS) ranged from 1 to 8 nm. Cell viability in the presence of G3.0 and G3.0-mannose8 was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and was found to be statistically indistinguishable from that of untreated cells. G3.0-mannose8 exhibited 12-fold higher uptake than unmodified G3.0 control dendrons in MR-expressing J774.E murine macrophage-like cells. Uptake was nearly completely inhibited in the presence of 10 mg/mL mannan, a mannose analogue and known MR substrate. Confocal microscopy studies demonstrated the presence of significant intracellular punctate fluorescence colocalized with a fluid endocytosis marker with little surface fluorescence in cells incubated with G3.0-mannose8. No significant cell-associated fluorescence was observed in cells incubated with G3.0 dendrons that did not contain the targeting ligand mannose. The current studies suggest that PEGtide dendrons could be useful as nanocarriers in drug delivery and imaging applications.
Collapse
|
14
|
Zhang H, Wang K, Cheng X, Lu Y, Zhang Q. Synthesis and In vitrocytotoxicity of poly(ethylene glycol)-epothilone B conjugates. J Appl Polym Sci 2014. [DOI: 10.1002/app.41123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Haiyan Zhang
- School of Pharmacy; Central South University; Changsha People's Republic of China
- Department of Experimental Hematology; Beijing Institute of Radiation Medicine; Beijing 100850 People's Republic of China
| | - Kun Wang
- Department of Experimental Hematology; Beijing Institute of Radiation Medicine; Beijing 100850 People's Republic of China
| | - Xiaochen Cheng
- Department of Experimental Hematology; Beijing Institute of Radiation Medicine; Beijing 100850 People's Republic of China
| | - Yuxin Lu
- Department of Experimental Hematology; Beijing Institute of Radiation Medicine; Beijing 100850 People's Republic of China
| | - Qinglin Zhang
- Department of Experimental Hematology; Beijing Institute of Radiation Medicine; Beijing 100850 People's Republic of China
| |
Collapse
|
15
|
Li Y, Liu R, Yang J, Ma G, Zhang Z, Zhang X. Dual sensitive and temporally controlled camptothecin prodrug liposomes codelivery of siRNA for high efficiency tumor therapy. Biomaterials 2014; 35:9731-45. [PMID: 25189519 DOI: 10.1016/j.biomaterials.2014.08.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/12/2014] [Indexed: 11/26/2022]
Abstract
The combination of chemotherapeutic drug camptothecin (CPT) and siPlk1 could prohibit cancer development with combined effects. To ensure the two drugs could be simultaneously delivered to tumor region with high loading content, and the modulator siPlk1 could be released in advance to down-regulate the Plk1 expression to improve the sensitivity of CPT to cancer cells, dual sensitive and temporally controlled CPT prodrug based cationic liposomes with siPlk1 codelivery system was constructed. The pH-sensitive zwitterionic polymer poly(carboxybetaine) (PCB) was conjugated with CPT through pH and esterase-sensitive ester bond to enhance the stability and loading content of CPT. CPT-based cationic liposomes consisted of CPT-PCB prodrug and cationic lipid DDAB were then constructed for siRNA codelivery for combination therapy. The dual sensitive CPT-PCB/siPlk1 lipoplexes simultaneously delivered the two drugs to tumor cells and enabled a temporally controlled release of two drugs, that the siRNA was quickly released after 4 h incubation due to the protonation of PCB in endosomes/lysosomes, and CPT was released in a sustained manner in response to pH and esterase and highly accumulated in nucleus after 12 h incubation. The CPT-PCB/siPlk1 lipoplexes induced significant cell apoptosis and cytotoxicity in vitro with a synergistic effect. Furthermore, the dual sensitive CPT-PCB lipoplexes enhanced the tumor accumulation of the two payloads and exhibited a synergistic tumor suppression effect in tumor-bearing mice in vivo, which proved to be a promising delivery system for codelivery of CPT and siPlk1 for cancer therapy.
Collapse
Affiliation(s)
- Yan Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ruiyuan Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Jun Yang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Guanghui Ma
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China.
| | - Xin Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
16
|
Kim SH, Kaplan JA, Sun Y, Shieh A, Sun HL, Croce CM, Grinstaff MW, Parquette JR. The self-assembly of anticancer camptothecin-dipeptide nanotubes: a minimalistic and high drug loading approach to increased efficacy. Chemistry 2014; 21:101-5. [PMID: 25384556 DOI: 10.1002/chem.201404520] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Indexed: 01/03/2023]
Abstract
20-(S)-Camptothecin (CPT)-conjugated dipeptides are reported that preassemble into nanotubes with diameters ranging from 80-120 nm. These nanoassemblies maintain a high (∼47 %) drug loading and exhibit greater drug stability (i.e., resistance to lactone hydrolysis), and consequently greater efficacy against several human cancer cells (HT-29, A549, H460, and H23) in vitro compared with the clinically used prodrug irinotecan. A key and defining feature of this system is the use of the CPT-conjugated dipeptide as both the drug and precursor to the nanostructured carrier, which simplifies the overall fabrication process.
Collapse
Affiliation(s)
- Se Hye Kim
- Department of Chemistry and Biochemistry, The Ohio State University, 100 W. 18th Ave. Columbus, OH 43210 (USA), Fax: (+1) 614-292-1685
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Liu C, Yuan J, Luo X, Chen M, Chen Z, Zhao Y, Li X. Folate-decorated and reduction-sensitive micelles assembled from amphiphilic polymer-camptothecin conjugates for intracellular drug delivery. Mol Pharm 2014; 11:4258-69. [PMID: 25238329 DOI: 10.1021/mp500468d] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
It is one of the challenges for a wide clinical application of polymer micelles to address the structure disintegration and premature drug release before reaching a pathological site. In the current study, folic acid (FA)-decorated polymer-drug conjugates (FSC) were synthesized with disulfide linkages between camptothecin (CPT) and amphiphilic poly(ethylene glycol)-b-poly(ε-caprolactone) (PECL) copolymers. FSC conjugates were proposed to assemble into micelles with a hydrophobic core of PCL segments and CPT and a hydrophilic corona of PEG segments. The addition of hexadecanol during micelle formation (FSC-16) was proposed to modulate the interactions of hydrophobic segments in micelles and enhance the reductive sensitivity. FSC-16 micelles were obtained with critical micelle concentration of around 2 μg/mL and an average size of around 200 nm, and the conjugated CPT was rapidly released out in response to glutathione. The reductive sensitivity was also demonstrated with respect to the changes of micelle size and morphologies as well as the fluorescent intensity of pyrene loaded in micelles. Benefiting from the FA receptor-mediated uptake and the reduction-sensitive release of CPT, significant cytotoxicity and cell apoptosis were identified for FSC-16 micelles against SKOV-3 cells with strong expressions of FA receptors. Flow cytometry and confocal laser scanning microscopy analyses demonstrated that CPT was distributed into nuclei after cellular uptake and intracellular release from FSC-16 micelles. Thus, the FA-decorated and reduction-sensitive micelles assembled from polymer-drug conjugates show advantages in inhibiting premature release during circulation, enhancing cellular uptake at the tumor tissues, and promoting intracellular release and nuclei location of the active moieties.
Collapse
Affiliation(s)
- Chaoyu Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P. R. China
| | | | | | | | | | | | | |
Collapse
|
18
|
Yang A, Liu Z, Yan B, Zhou M, Xiong X. Preparation of camptothecin-loaded targeting nanoparticles and their antitumor effects on hepatocellular carcinoma cell line H22. Drug Deliv 2014; 23:1699-706. [PMID: 25148540 DOI: 10.3109/10717544.2014.950767] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Camptothecin (CPT) is an effective anticancer agent against various cancers but the clinical application is limited because of its poor water solubility, low bioavailability and severe toxic side effects. The aim of the present study was to evaluate the feasibility of using targeted NPs as a high-performance CPT delivery system that targets liver cancer cells through intravenous (i.v.) administration route. CPT was incorporated into biotin-F127-PLA or F127-PLA polymeric nanoparticles (NPs) by a dialysis method. The preparation of the targeting NPs was performed by conjugating biotin-F127-PLA NPs with anti-3A5 antibody. The antitumor effect of the CPT-loaded nanoparticles against H22 cells in vitro was determined using an MTT assay. Tissue distribution and tumor inhibition in vivo were also evaluated. Survivin mRNA expression was assessed by real-time polymerase chain reaction. Results showed that the targeted CPT NPs exhibited regular spherical shapes with a mean diameter of approximately 180 nm. In vitro release of the targeted CPT NPs exhibited an initial burst (40%) within 12 h, followed by a slow release. Cytotoxicity test against H22 cells indicated that the targeted CPT NPs exerted significant antitumor effects. Compared with free CPT and non-targeted CPT NPs, the targeted CPT NPs showed superior inhibition ratio against tumor in vivo, which may be associated with reduced survivin mRNA expression. The results suggested that the new targeted CPT NPs may be a promising injectable delivery system for cancer therapy.
Collapse
Affiliation(s)
- Anshu Yang
- a State Key Laboratory of Food Science and Technology , Nanchang University , Nanchang , China .,b Sino-German Joint Research Institute, Nanchang University , Nanchang , China
| | - Zhiyong Liu
- c Laboratory Animal Center of Science and Technology , Jiangxi University of Traditional Chinese Medicine , Nanchang , China .,d Experimental Animal Center , Jiangxi Institute of Occupational Disease Prevention , Nanchang , China , and
| | - Bin Yan
- d Experimental Animal Center , Jiangxi Institute of Occupational Disease Prevention , Nanchang , China , and
| | - Ming Zhou
- d Experimental Animal Center , Jiangxi Institute of Occupational Disease Prevention , Nanchang , China , and
| | - Xiangyuan Xiong
- e School of Life Science , Jiangxi Science & Technology Normal University , Nanchang , China
| |
Collapse
|
19
|
Li G, Li Y, Tang Y, Zhang Y, Zhang Y, Yin T, Xu H, Cai C, Tang X. Hydroxyethyl starch conjugates for improving the stability, pharmacokinetic behavior and antitumor activity of 10-hydroxy camptothecin. Int J Pharm 2014; 471:234-44. [DOI: 10.1016/j.ijpharm.2014.05.038] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/15/2014] [Accepted: 05/19/2014] [Indexed: 11/15/2022]
|
20
|
Gilad Y, Firer MA, Rozovsky A, Ragozin E, Redko B, Albeck A, Gellerman G. "Switch off/switch on" regulation of drug cytotoxicity by conjugation to a cell targeting peptide. Eur J Med Chem 2014; 85:139-46. [PMID: 25084142 DOI: 10.1016/j.ejmech.2014.07.073] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/19/2014] [Accepted: 07/21/2014] [Indexed: 11/17/2022]
Abstract
Bi-nuclear amino acid platforms loaded with various drugs for conjugation to a peptide carrier were synthesized using simple and convenient orthogonally protective solid-phase organic synthesis (SPOS). Each arm of the platform carries a different anticancer agent linked through the same or different functional group, providing discrete chemo- and bio-release profiles for each drug, and also enabling "switch off/switch on" regulation of drug cytotoxicity by conjugation to the platform and to a cell targeting peptide. The versatility of this approach enables efficient production of drug-loaded platforms and determination of favorable drug combinations/modes of linkage for subsequent conjugation to a carrier moiety for targeted cancer cell therapy. The results presented here potentiate the application of amino acid platforms for targeted drug delivery (TDD).
Collapse
Affiliation(s)
- Yossi Gilad
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel; The Julius Spokojny Bioorganic Chemistry Laboratory, Department of Chemistry, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Michael A Firer
- Department of Chemical Engineering, Ariel University, Ariel, 40700, Israel
| | - Alex Rozovsky
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel; The Julius Spokojny Bioorganic Chemistry Laboratory, Department of Chemistry, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Elena Ragozin
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel
| | - Boris Redko
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel; The Julius Spokojny Bioorganic Chemistry Laboratory, Department of Chemistry, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Amnon Albeck
- The Julius Spokojny Bioorganic Chemistry Laboratory, Department of Chemistry, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Gary Gellerman
- Department of Biological Chemistry, Ariel University, Ariel, 40700, Israel.
| |
Collapse
|
21
|
Minko T. Soluble polymer conjugates for drug delivery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 2:15-20. [PMID: 24981750 DOI: 10.1016/j.ddtec.2005.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The use of water-soluble polymeric conjugates as drug carriers offers several possible advantages. These advantages include: (1) improved drug pharmacokinetics; (2) decreased toxicity to healthy organs; (3) possible facilitation of accumulation and preferential uptake by targeted cells; (4) programmed profile of drug release. In this review, we will consider the main types of useful polymeric conjugates and their role and effectiveness as carriers in drug delivery systems.:
Collapse
Affiliation(s)
- Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854-8020, USA.
| |
Collapse
|
22
|
Yu CY, Wang YM, Li NM, Liu GS, Yang S, Tang GT, He DX, Tan XW, Wei H. In Vitro and in Vivo Evaluation of Pectin-Based Nanoparticles for Hepatocellular Carcinoma Drug Chemotherapy. Mol Pharm 2014; 11:638-44. [DOI: 10.1021/mp400412c] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Cui-Yun Yu
- Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China
| | - Yan-Mei Wang
- Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China
| | - Na-Mei Li
- Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China
| | - Ge-Sha Liu
- Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China
| | - Sa Yang
- Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China
| | - Guo-Tao Tang
- Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China
| | - Dong-Xiu He
- Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China
| | - Xiang-Wen Tan
- Department
of Laboratory Animal Science, University of South China, Hengyang 421001, China
| | - Hua Wei
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
23
|
Li G, Cai C, Ren T, Tang X. Development and application of a UPLC–MS/MS method for the pharmacokinetic study of 10-hydroxy camptothecin and hydroxyethyl starch conjugate in rats. J Pharm Biomed Anal 2014; 88:345-53. [DOI: 10.1016/j.jpba.2013.08.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 08/16/2013] [Accepted: 08/18/2013] [Indexed: 10/26/2022]
|
24
|
Synthesis and activity of a folate targeted monodisperse PEG camptothecin conjugate. Bioorg Med Chem Lett 2013; 23:5810-3. [DOI: 10.1016/j.bmcl.2013.08.113] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 08/26/2013] [Accepted: 08/29/2013] [Indexed: 01/28/2023]
|
25
|
Ibrahim S, Gao D, Sinko PJ. Selective cytotoxicity and combined effects of camptothecin or paclitaxel with sodium-R-alpha lipoate on A549 human non-small cell lung cancer cells. Nutr Cancer 2013; 66:492-9. [PMID: 24063429 DOI: 10.1080/01635581.2013.749290] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Nonsmall cell lung cancer (NSCLC) is the most common type of lung cancer and remains the deadliest form of cancer in the United States and worldwide. New therapies are highly sought after to improve outcome. The effect of sodium-R-alpha lipoate on camptothecin- and paclitaxel-induced cytotoxicity was evaluated on A549 NSCLC and BEAS-2B "normal" lung epithelial cells. Combination indices (CI) and dose reduction indices (DRI) were investigated by studying the cytotoxicity of sodium-R-alpha lipoate (0-16 mM), camptothecin (0-25 nM) and paclitaxel (0-0.06 nM) alone and in combination. 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium-bromide (MTT) was used to assess cytotoxicity. The combinational cytotoxic effects of sodium-R-alpha lipoate with camptothecin or paclitaxel were analyzed using a simulation of dose effects (CompuSyn® 3.01). The effects of sodium-R-alpha lipoate on camptothecin- and paclitaxel-induced cytotoxicity varied based on concentrations and treatment times. It was found that sodium-R-alpha lipoate wasn't cytotoxic toward BEAS-2B cells at any of the concentrations tested. For A549 cells, CIs [(additive (CI = 1); synergistic (CI < 1); antagonistic (CI < 1)] were lower and DRIs were higher for the camptothecin/sodium-R-alpha-lipoate combination (CI = ∼0.17-1.5; DRI = ∼2.2-22.6) than the paclitaxel/sodium-R-alpha-lipoate combination (CI = ∼0.8-9.9; DRI = ∼0.10-5.8) suggesting that the camptothecin regimen was synergistic and that the addition of sodium-R-alpha lipoate was important for reducing the camptothecin dose and potential for adverse effects.
Collapse
Affiliation(s)
- Sherif Ibrahim
- a Department of Pharmaceutics , Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , New Brunswick , New Jersey , USA
| | | | | |
Collapse
|
26
|
Lewis DR, Kholodovych V, Tomasini MD, Abdelhamid D, Petersen LK, Welsh WJ, Uhrich KE, Moghe PV. In silico design of anti-atherogenic biomaterials. Biomaterials 2013; 34:7950-9. [PMID: 23891521 DOI: 10.1016/j.biomaterials.2013.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 07/01/2013] [Indexed: 01/10/2023]
Abstract
Atherogenesis, the uncontrolled deposition of modified lipoproteins in inflamed arteries, serves as a focal trigger of cardiovascular disease (CVD). Polymeric biomaterials have been envisioned to counteract atherogenesis based on their ability to repress scavenger mediated uptake of oxidized lipoprotein (oxLDL) in macrophages. Following the conceptualization in our laboratories of a new library of amphiphilic macromolecules (AMs), assembled from sugar backbones, aliphatic chains and poly(ethylene glycol) tails, a more rational approach is necessary to parse the diverse features such as charge, hydrophobicity, sugar composition and stereochemistry. In this study, we advance a computational biomaterials design approach to screen and elucidate anti-atherogenic biomaterials with high efficacy. AMs were quantified in terms of not only 1D (molecular formula) and 2D (molecular connectivity) descriptors, but also new 3D (molecular geometry) descriptors of AMs modeled by coarse-grained molecular dynamics (MD) followed by all-atom MD simulations. Quantitative structure-activity relationship (QSAR) models for anti-atherogenic activity were then constructed by screening a total of 1164 descriptors against the corresponding, experimentally measured potency of AM inhibition of oxLDL uptake in human monocyte-derived macrophages. Five key descriptors were identified to provide a strong linear correlation between the predicted and observed anti-atherogenic activity values, and were then used to correctly forecast the efficacy of three newly designed AMs. Thus, a new ligand-based drug design framework was successfully adapted to computationally screen and design biomaterials with cardiovascular therapeutic properties.
Collapse
Affiliation(s)
- Daniel R Lewis
- Department of Chemical and Biochemical Engineering, Rutgers University, NJ 08854, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhang Z, Yao J. Preparation of irinotecan-loaded folate-targeted liposome for tumor targeting delivery and its antitumor activity. AAPS PharmSciTech 2012; 13:802-10. [PMID: 22639238 DOI: 10.1208/s12249-012-9776-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Accepted: 03/21/2012] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study was to investigate the in vivo distribution and antitumor activity of irinotecan (camptothecin (CPT)-11)-loaded folate-targeted liposome (F-Lip) in tumor-bearing mice following i.v. administration. Folate-poly(ethylene glycol)-distearoylphosphatidylcholine (FA-PEG-DSPE) was synthesized by amide reaction of DSPE-PEG-NH(2) and FA. F-Lip modified by FA-PEG-DSPE was prepared by an ammonium sulfate gradient. The mean particle size and entrapment efficiency of F-Lip with negative charge were 197.8 ± 4.58 nm and 91.39 ± 2.34 %, respectively. The distributions of CPT-11 and SN-38 in the tumor after i.v. administration of F-Lip, CPT-11-loaded liposomes (C-Lip), and CPT-11 injection (C-Inj) were far greater with the F-Lip group in comparison to the C-Inj and C-Lip, which might contribute to folate-meditated targeting uptake by the folate receptor on the surface of the tumor cells. The uptake of CPT-11 in the liver and rectum for two liposome groups were all markedly increased as compared to the C-Inj. Moreover, F-Lip exhibited a dose-dependent tumor growth inhibition and superior anticancer activity to C-Lip and C-Inj after i.v. administration. It also showed no significant body weight loss and much lower toxicity on the center immune organs. Therefore, F-Lip may be presented as potential candidates for tumor targeting drug delivery.
Collapse
|
28
|
Li D, Diao J, Wang D, Liu J, Zhang J. Design, synthesis and biological evaluation of folate-porphyrin: a new photosensitizer for targeted photodynamic therapy. J PORPHYR PHTHALOCYA 2012. [DOI: 10.1142/s1088424610002379] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A novel folate-porphyrin conjugate 1 for targeted photodynamic therapy of tumor was designed and synthesized. The results of fluorescence spectroscopy and confocal laser scanning microscope demonstrated that the cellular uptake of conjugate 1 by HeLa cells was 35 times higher than that of precursor porphyrin 3 after 24 h incubation, and that the presence of excessive free folic acid inhibited the cellular uptake of conjugate 1. Cytotoxicity against folate-receptor positive HeLa cells in vitro measured by MTT assay demonstrated that conjugate 1 exhibited much lower dark cytotoxicity but significant photocytotoxicity, with 86.4% of cell growth inhibition ratio after irradiation. However, conjugate 1 induced lower photocytotoxicity for normal cells and folate-receptor negative cells. These results suggest that folate-porphyrin like photosensitizers could induce a potentially useful targeted photodynamic therapy modality for folate-receptor-positive cancer cells due to the folate-receptor mediated endocytosis.
Collapse
Affiliation(s)
- Donghong Li
- State Key Laboratory of Trauma, Burn and Combined Injury, the 2nd Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Junlin Diao
- State Key Laboratory of Trauma, Burn and Combined Injury, the 2nd Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Dong Wang
- Cancer Center, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jianchang Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, the 2nd Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jiaotao Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, the 2nd Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
29
|
Synthesis and activity of folate conjugated didemnin B for potential treatment of inflammatory diseases. Bioorg Med Chem Lett 2012; 22:709-12. [DOI: 10.1016/j.bmcl.2011.10.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/11/2011] [Accepted: 10/11/2011] [Indexed: 01/29/2023]
|
30
|
Tan H, Zhang Y, Wang M, Zhang Z, Zhang X, Yong AM, Wong SY, Chang AYC, Chen ZK, Li X, Choolani M, Wang J. Silica-shell cross-linked micelles encapsulating fluorescent conjugated polymers for targeted cellular imaging. Biomaterials 2011; 33:237-46. [PMID: 21963283 DOI: 10.1016/j.biomaterials.2011.09.037] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 09/15/2011] [Indexed: 12/20/2022]
Abstract
A bioinspired silification approach was successfully used to encapsulate fluorescent conjugated polymers inside silica-shell cross-linked polymeric micelles (CP-SSCL) in the highly benign synthesis environment of room temperature and near-neutral aqueous environment. Four different conjugated polymers were employed to demonstrate the versatility of the bioinspired silification, resulting in the formation of CP-SSCL with different emission wavelengths across the visible spectrum. The CP-SSCL are characterized by a large absorption coefficient and high quantum yield, indicating that they exhibit the required high fluorescence brightness for cellular imaging application. In addition, the CP-SSCL also exhibit a high colloidal stability and low cytotoxicity. The in vitro studies of using MDA-MB-231 breast cancer cells show that the CP-SSCL are successfully uptaken by the cancer cells and located at the cytoplasm of the cells. Furthermore, by conjugating folic acid on their surfaces, the uptake of CP-SSCL by MDA-MB-231 cells was enhanced significantly, suggesting their great potential for targeted imaging and early detection of cancer cells.
Collapse
Affiliation(s)
- Happy Tan
- NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, Singapore 117456, Singapore
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Improved therapeutic effect of folate-decorated PLGA–PEG nanoparticles for endometrial carcinoma. Bioorg Med Chem 2011; 19:4057-66. [DOI: 10.1016/j.bmc.2011.05.016] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Revised: 05/11/2011] [Accepted: 05/11/2011] [Indexed: 12/21/2022]
|
32
|
Feng X, Pinaud J, Chaikof EL, Taton D, Gnanou Y. Sequential functionalization of janus-type dendrimer-like poly(ethylene oxide)s with camptothecin and folic acid. ACTA ACUST UNITED AC 2011. [DOI: 10.1002/pola.24718] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
33
|
A validated stability-indicating HPLC method for the determination of PEGylated puerarin in aqueous solutions. J Chromatogr B Analyt Technol Biomed Life Sci 2010; 878:2061-6. [DOI: 10.1016/j.jchromb.2010.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 05/28/2010] [Accepted: 06/01/2010] [Indexed: 11/23/2022]
|
34
|
Pulmonary targeting microparticulate camptothecin delivery system: anticancer evaluation in a rat orthotopic lung cancer model. Anticancer Drugs 2010; 21:65-76. [PMID: 19966540 DOI: 10.1097/cad.0b013e328332a322] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Large (>6 microm) rigid microparticles (MPs) become passively entrapped within the lungs after intravenous (i.v.) injection making them an attractive and highly efficient alternative to inhalation for pulmonary delivery. In this study, PEGylated 6 microm polystyrene MPs with multiple copies of the norvaline (Nva) alpha-amino acid prodrug of camptothecin (CPT) were prepared. Surface morphology was characterized using a scanning electron microscope. CPT was released from the CPT-Nva-MPs over 24 h in rat plasma at 37 degrees C. In-vivo CPT plasma concentrations were low (approximately 1 ng/ml or less) and constant over a period of 4 days after a single i.v. injection of CPT-Nva-MPs as compared with high but short-lived systemic exposures after an i.v. injection of free CPT. This suggests that sustained local CPT concentrations were achieved in the lung after administration of the MP delivery system. Anticancer efficacy was evaluated in an orthotopic lung cancer animal model and compared with a bolus injection of CPT. Animals receiving free CPT (2 mg/kg) and CPT-Nva-MPs (0.22 mg/kg CPT and 100 mg/kg MPs) were found to have statistically significant smaller areas of lung cancer (P<0.05 and 0.01, respectively) than untreated animals. In addition, 40% of the animals receiving CPT-Nva-MPs were found to be free of cancer. The CPT dose using targeted MPs was 10 times lower than after i.v. injection of free CPT, but was more effective in reducing the amount of cancerous areas. In conclusion, CPT-Nva-MPs were able to achieve effective local lung and low systemic CPT concentrations at a dose that was 10 times lower than systemically administered CPT resulting in a significant improvement in anticancer efficacy in an orthotopic rat model of lung cancer.
Collapse
|
35
|
Deshmukh M, Chao P, Kutscher HL, Gao D, Sinko PJ. A series of alpha-amino acid ester prodrugs of camptothecin: in vitro hydrolysis and A549 human lung carcinoma cell cytotoxicity. J Med Chem 2010; 53:1038-47. [PMID: 20063889 DOI: 10.1021/jm901029n] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The objective of the present study was to identify a camptothecin (CPT) prodrug with optimal release and cytotoxicity properties for immobilization on a passively targeted microparticle delivery system. A series of alpha-amino acid ester prodrugs of CPT were synthesized, characterized, and evaluated. Four CPT prodrugs were synthesized with increasing aliphatic chain length (glycine (Gly) (2a), alanine (Ala) (2b), aminobutyric acid (Abu) (2c), and norvaline (Nva) (2d)). Prodrug reconversion was studied at pH 6.6, 7.0, and 7.4 corresponding to tumor, lung, and extracellular/physiological pH, respectively. Cytotoxicity was evaluated in A549 human lung carcinoma cells using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The hydrolytic reconversion rate to parent CPT increased with decreasing side chain length as well as increasing pH. The Hill slope of 2d was significantly less than CPT and the other prodrugs tested, indicating a higher cell death rate at lower concentrations. These results suggest that 2d is the best candidate for a passively targeted sustained release lung delivery system.
Collapse
Affiliation(s)
- Manjeet Deshmukh
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | | | |
Collapse
|
36
|
Shen Y, Jin E, Zhang B, Murphy CJ, Sui M, Zhao J, Wang J, Tang J, Fan M, Van Kirk E, Murdoch WJ. Prodrugs Forming High Drug Loading Multifunctional Nanocapsules for Intracellular Cancer Drug Delivery. J Am Chem Soc 2010; 132:4259-65. [DOI: 10.1021/ja909475m] [Citation(s) in RCA: 480] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Youqing Shen
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Erlei Jin
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Bo Zhang
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Caitlin J. Murphy
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Meihua Sui
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Jian Zhao
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Jinqiang Wang
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Jianbin Tang
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Maohong Fan
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - Edward Van Kirk
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| | - William J. Murdoch
- Center for Bionanoengineering and the State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China 310027, Department of Chemical and Petroleum Engineering and Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071
| |
Collapse
|
37
|
Abstract
Anticancer prodrugs designed to target specifically tumor cells should increase therapeutic effectiveness and decrease systemic side effects in the treatment of cancer. Over the last 20 years, significant advances have been made in the development of anticancer prodrugs through the incorporation of triggers for reductive activation. Reductively activated prodrugs have been designed to target hypoxic tumor tissues, which are known to overexpress several endogenous reductive enzymes. In addition, exogenous reductive enzymes can be delivered to tumor cells through fusion with tumor-specific antibodies or overexpressed in tumor cells through gene delivery approaches. Many anticancer prodrugs have been designed to use both the endogenous and exogenous reductive enzymes for target-specific activation and these prodrugs often contain functional groups such as quinones, nitroaromatics, N-oxides, and metal complexes. Although no new agents have been approved for clinical use, several reductively activated prodrugs are in various stages of clinical trial. This review mainly focuses on the medicinal chemistry aspects of various classes of reductively activated prodrugs including design principles, structure-activity relationships, and mechanisms of activation and release of active drug molecules.
Collapse
Affiliation(s)
- Yu Chen
- Department of Pharmaceutical Chemistry, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | |
Collapse
|
38
|
Abstract
Major advances in the use of carrier vehicles delivering pharmacologic agents and enzymes to sites of disease have occurred over the past 10 years. This review focuses on the concepts and clinical evaluation of carrier-mediated anticancer agents that are administered i.v. or orally. The primary types of carrier-mediated anticancer agents are nanoparticles, nanosomes, which are nanoparticle-sized liposomes, and conjugated agents. Nanosomes are further subdivided into stabilized and nonstabilized or conventional nanosomes. Nanospheres and dendrimers are subclasses of nanoparticles. Conjugated agents consist of polymer-linked and pegylated agents. The theoretical advantages of carrier-mediated drugs are greater solubility, longer duration of exposure, selective delivery of entrapped drug to the site of action, superior therapeutic index, and the potential to overcome resistance associated with the regular anticancer agent. The pharmacokinetic disposition of carrier-mediated agents depends on the physiochemical characteristics of the carrier, such as size, surface charge, membrane lipid packing, steric stabilization, dose, and route of administration. The primary sites of accumulation of carrier-mediated agents are the tumor, liver, and spleen, compared with noncarrier formulations. The drug that remains encapsulated in or linked to the carrier (e.g., the nanosome or nanoparticle) is an inactive prodrug, and thus the drug must be released from the carrier to be active. The factors affecting the pharmacokinetic and pharmacodynamic variability of these agents remain unclear, but most likely include the reticuloendothelial system, which has also been called the mononuclear phagocyte system. Future studies need to evaluate the mechanism of clearance of carrier-mediated agents and identify the factors associated with the pharmacokinetic and pharmacodynamic variability of carrier agents in patients and specifically in tumors.
Collapse
Affiliation(s)
- William C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina, 3308 Kerr Hall CB 7360, 311 Pharmacy Lane, Chapel Hill, NC 27599-7360, USA.
| |
Collapse
|
39
|
Minko T, Khandare JJ, Vetcher AA, Soldatenkov VA, Garbuzenko OB, Saad M, Pozharov VP. Multifunctional Nanotherapeutics for Cancer. MULTIFUNCTIONAL PHARMACEUTICAL NANOCARRIERS 2008. [DOI: 10.1007/978-0-387-76554-9_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
|
40
|
Besić E. Physical mechanisms and methods employed in drug delivery to tumors. ACTA PHARMACEUTICA 2007; 57:249-68. [PMID: 17878107 DOI: 10.2478/v10007-007-0021-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In addition to several well-known drug delivery strategies developed to facilitate effective chemotherapy with anticancer agents, some new approaches have been recently established, based on specific effects arising from the applications of ultrasound, magnetic and electric fields on drug delivery systems. This paper gives an overview of newly developed methods of drug delivery to tumors and of the related anticancer therapies based on the combined use of different physical methods and specific drug carriers. The conventional strategies and new approaches have been put into perspective to revisit the existing and to propose new directions to overcome the threatening problem of cancer diseases.
Collapse
Affiliation(s)
- Erim Besić
- Department of Biophysics, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10002 Zagreb, Croatia.
| |
Collapse
|
41
|
Sinha R, Kim GJ, Nie S, Shin DM. Nanotechnology in cancer therapeutics: bioconjugated nanoparticles for drug delivery. Mol Cancer Ther 2006; 5:1909-17. [PMID: 16928810 DOI: 10.1158/1535-7163.mct-06-0141] [Citation(s) in RCA: 451] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nanotechnology refers to the interactions of cellular and molecular components and engineered materials-typically, clusters of atoms, molecules, and molecular fragments into incredibly small particles-between 1 and 100 nm. Nanometer-sized particles have novel optical, electronic, and structural properties that are not available either in individual molecules or bulk solids. The concept of nanoscale devices has led to the development of biodegradable self-assembled nanoparticles, which are being engineered for the targeted delivery of anticancer drugs and imaging contrast agents. Nanoconstructs such as these should serve as customizable, targeted drug delivery vehicles capable of ferrying large doses of chemotherapeutic agents or therapeutic genes into malignant cells while sparing healthy cells. Such "smart" multifunctional nanodevices hold out the possibility of radically changing the practice of oncology, allowing easy detection and then followed by effective targeted therapeutics at the earliest stages of the disease. In this article, we briefly discuss the use of bioconjugated nanoparticles for the delivery and targeting of anticancer drugs.
Collapse
Affiliation(s)
- Rajni Sinha
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, Room 3090, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
42
|
Khandare JJ, Jayant S, Singh A, Chandna P, Wang Y, Vorsa N, Minko T. Dendrimer Versus Linear Conjugate: Influence of Polymeric Architecture on the Delivery and Anticancer Effect of Paclitaxel. Bioconjug Chem 2006; 17:1464-72. [PMID: 17105225 DOI: 10.1021/bc060240p] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The relative difference in polymeric architectures of dendrimer and linear bis(poly(ethylene glycol)) (PEG) polymer in conjugation with paclitaxel has been described. Paclitaxel, a poorly soluble anticancer drug, was covalently conjugated with PAMAM G4 hydroxyl-terminated dendrimer and bis(PEG) polymer for the potential enhancement of drug solubility and cytotoxicity. Both conjugates were characterized by 1NMR, HPLC, and MALDI/TOF. In addition, molecular conformations of dendrimer, bis(PEG), paclitaxel, and its polymeric conjugates were studied by molecular modeling. Hydrolysis of the ester bond in the conjugate was analyzed by HPLC using esterase hydrolyzing enzyme. In vitro cytotoxicity of dendrimer, bis(PEG), paclitaxel, and polymeric conjugates containing paclitaxel was evaluated using A2780 human ovarian carcinoma cells. Cytotoxicity increased by 10-fold with PAMAM dendrimer-succinic acid-paclitaxel conjugate when compared with free nonconjugated drug. Data obtained indicate that the nanosized dendritic polymer conjugates can be used with good success as anticancer drug carriers.
Collapse
Affiliation(s)
- Jayant J Khandare
- Department of Pharmaceutics, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Folic Acid Immobilized Ferrimagnetic DP-Bioglass to Target Tumor Cell for Cancer Hyperthermia Treatment. ACTA ACUST UNITED AC 2006. [DOI: 10.4028/www.scientific.net/ast.53.50] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
DP-bioglass is one of biodegradable glasses, which can be used as bioactive material in soft tissue and bone. It was often used in orthopedy and plastic surgery. Recently, bioglass was also used as a carrier for drug and gene delivery systems. Additionally, ferrimagnetic DP-bioglass can be potential candidates for magnetic induction hyperthermia, by using a magnetic field. The aim of this work is the preparation and characterization of surface-modified ferrimagnetic DP-bioglass. First DP-bioglass has to be surface-modified with polyethylene glycol (PEG) and folic acid (FA) to improve its intracellular uptake and ability to target specific cells. PEG-FA complex was synthesized using carbodiimide (DCC) to link PEG with FA. Then PEG-FA complex were immobilized on the surface of DP-bioglass by using amino-silane (AEAPS) as a coupling agent. Fourier transform infrared spectroscopy (FTIR), nuclear magnetic resonance (1H NMR), and thermogravimetric analysis (TGA) was used to demonstrate this immobilization process. In biological study showed that immobilized ferromagnetic DP-bioglass with PEG-FA was non-cytotoxicity and significantly enhanced the intracellular uptake of DP-bioglass by target cells.
Collapse
|
44
|
Henne WA, Doorneweerd DD, Hilgenbrink AR, Kularatne SA, Low PS. Synthesis and activity of a folate peptide camptothecin prodrug. Bioorg Med Chem Lett 2006; 16:5350-5. [PMID: 16901694 DOI: 10.1016/j.bmcl.2006.07.076] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Accepted: 07/24/2006] [Indexed: 10/24/2022]
Abstract
A folate receptor targeted camptothecin prodrug was synthesized using a hydrophilic peptide spacer linked to folate via a releasable disulfide carbonate linker. The conjugate was found to possess high affinity for folate receptor-expressing cells and inhibited cell proliferation in human KB cells with an IC(50) of 10nM. Activity of the prodrug was completely blocked by excess folic acid, demonstrating receptor-mediated uptake.
Collapse
Affiliation(s)
- Walter A Henne
- Department of Chemistry and Purdue Cancer Center, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | |
Collapse
|
45
|
Lalloo A, Chao P, Hu P, Stein S, Sinko PJ. Pharmacokinetic and pharmacodynamic evaluation of a novel in situ forming poly(ethylene glycol)-based hydrogel for the controlled delivery of the camptothecins. J Control Release 2006; 112:333-42. [PMID: 16650910 DOI: 10.1016/j.jconrel.2006.03.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2005] [Revised: 02/27/2006] [Accepted: 03/03/2006] [Indexed: 11/24/2022]
Abstract
Inadequate drug delivery, due to problems associated with achieving constant therapeutic blood levels, has hampered the use of anticancer agents of the camptothecin (CPT) class. The objective of the current studies was to develop a depot delivery system for the water-soluble analog of CPT, topotecan (TPT). In this study, a 2-phase drug depot consisting of TPT-loaded liposomes entrapped in a poly(ethylene glycol) hydrogel was designed. Physically entrapped unaltered TPT displayed a rapid release rate from the hydrogel. Controlled release was demonstrated in vitro and in vivo from the 2-phase system with constant blood levels being achieved for several days in rats. Cytotoxicity and antitumor activity were also evaluated in rats inoculated with syngeneic MAT B III breast cancer cells. Rats treated with the liposome-loaded hydrogel displayed significantly longer tumor growth suppression and did not exhibit body weight loss compared to those treated with other delivery modes. These experiments constitute a proof-of-principle of the 2-phase depot concept and its potential value for enhancing safety and efficacy in chemotherapy.
Collapse
Affiliation(s)
- Anita Lalloo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | | | | | | | |
Collapse
|
46
|
Abstract
Despite several advancements in chemotherapy, the real therapy of cancer still remains a challenge. The development of new anti-cancer drugs for the treatment of cancer has not kept pace with the progress in cancer therapy, because of the nonspecific drug distribution resulting in low tumour concentrations and systemic toxicity. The main hindrance for the distribution of anti-cancer agents to the tumour site is the highly disorganized tumour vasculature, high blood viscosity in the tumour, and high interstitial pressure within the tumour tissue. Recently, several approaches such as drug modifications and development of new carrier systems for anti-cancer agents have been attempted to enhance their tumour reach. Approaches such as drug delivery through enhanced permeability and retention (EPR) effect have resulted in a significant improvement in concentration in tumours, while approaches such as drug-carrier implants and microparticles have resulted in improvement in local chemotherapy of cancer. This review discusses different strategies employed for the delivery of anti-cancer agents to tumours, such as through EPR effect, local chemotherapeutic approaches using drug delivery systems, and special strategies such as receptor-mediated delivery, pH-based carriers, application of ultrasound and delivery to resistant tumour cells and brain using nanoparticles.
Collapse
Affiliation(s)
- L Harivardhan Reddy
- Drug Delivery Research Laboratory, Center of Relevance and Excellence in NDDS, Pharmacy Department, Fatehgunj, M.S. University of Baroda, Baroda-390002, Gujarat, India.
| |
Collapse
|
47
|
Affiliation(s)
- William C Zamboni
- Molecular Therapeutics Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15213-1863, USA
| |
Collapse
|
48
|
Hilgenbrink AR, Low PS. Folate Receptor-Mediated Drug Targeting: From Therapeutics to Diagnostics. J Pharm Sci 2005; 94:2135-46. [PMID: 16136558 DOI: 10.1002/jps.20457] [Citation(s) in RCA: 455] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Folate targeted drug delivery has emerged as an alternative therapy for the treatment and imaging of many cancers and inflammatory diseases. Due to its small molecular size and high binding affinity for cell surface folate receptors (FR), folate conjugates have the ability to deliver a variety of molecular complexes to pathologic cells without causing harm to normal tissues. Complexes that have been successfully delivered to FR expressing cells, to date, include protein toxins, immune stimulants, chemotherapeutic agents, liposomes, nanoparticles, and imaging agents. This review will summarize the applications of folic acid as a targeting ligand and highlight the various methods being developed for delivery of therapeutic and imaging agents to FR-expressing cells.
Collapse
|
49
|
Paranjpe PV, Stein S, Sinko PJ. Tumor-targeted and activated bioconjugates for improved camptothecin delivery. Anticancer Drugs 2005; 16:763-75. [PMID: 16027527 DOI: 10.1097/01.cad.0000172834.78068.7c] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Earlier reports from our laboratory described bioconjugates of camptothecin (CPT) for tumor targeting. In the current work, the rate and site of CPT release from the bioconjugates were modulated using increasingly sterically hindered amino acids and cysteine proteinase-sensitive peptide linkers, respectively. Polyethylene glycol served as a spacer/scaffold between CPT and folic acid. The folic acid receptor, overexpressed on many cancer cells, was targeted using folate. The delivery system was tested in vitro for hydrolytic stability, enzyme-mediated cleavage, cytotoxicity and targeting potential. The linkers successfully modulated the hydrolysis rate (around 1--100 h) and potential site (tumor microenvironment) of CPT release. Preliminary molecular modeling approaches were utilized to assess the influence of molecular volume on hydrolysis half-life (i.e. CPT release). There was a clear, but non-linear, relationship between in vitro CPT release and increasing steric hindrance offered by the peptide linker. The efficacy of four conjugates was studied in a syngeneic rat breast cancer model. Histopathological analysis on treated tumors was performed to evaluate disease prognosis. The results demonstrate that programmed bioconjugates may provide superior efficacy and greater control over the rate and site of CPT release, resulting in higher anti-tumor efficacy and lower toxicity.
Collapse
Affiliation(s)
- Pankaj V Paranjpe
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | |
Collapse
|
50
|
Zhang Z, Tanabe K, Hatta H, Nishimoto SI. Bioreduction activated prodrugs of camptothecin: molecular design, synthesis, activation mechanism and hypoxia selective cytotoxicity. Org Biomol Chem 2005; 3:1905-10. [PMID: 15889173 DOI: 10.1039/b502813b] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Several water-soluble derivatives (CPT3, CPT3a-d) of camptothecin (CPT) were synthesized, among which CPT3 bearing an N,N'-dimethyl-1-aminoethylcarbamate side-chain was further conjugated with reductively eliminating structural units of indolequinone, 4-nitrobenzyl alcohol and 4-nitrofuryl alcohol to produce novel prodrugs of camptothecin (CPT4-6). All CPT derivatives were of lower cytotoxicity than their parent compound of CPT. In contrast, CPT4 and CPT6 showed higher hypoxia selectivity of cytotoxicity towards tumor cells than CPT. A mechanism by which a representative prodrug CPT4 is activated in the presence of DT-diaphorase to release CPT was also discussed. The bioreduction activated CPT prodrugs including CPT4 and CPT6 are identified to be promising for application to the hypoxia targeting tumor chemotherapy.
Collapse
Affiliation(s)
- Zhouen Zhang
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura Campus, Nishikyo-ku, Kyoto, 615-8510, Japan
| | | | | | | |
Collapse
|