1
|
Shrestha P, Ghanwatkar Y, Mahto S, Pramanik N, Mahato RI. Gemcitabine-Lipid Conjugate and ONC201 Combination Therapy Effectively Treats Orthotopic Pancreatic Tumor-Bearing Mice. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29686-29698. [PMID: 38813771 DOI: 10.1021/acsami.4c02626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Gemcitabine (GEM) is a nucleoside analogue approved as a first line of therapy for pancreatic ductal adenocarcinoma (PDAC). However, rapid metabolism by plasma cytidine deaminase leading to the short half-life, intricate intracellular metabolism, ineffective cell uptake, and swift development of chemoresistance downgrades the clinical efficacy of GEM. ONC201 is a small molecule that inhibits the Akt and ERK pathways and upregulates the TNF-related apoptosis-inducing ligand (TRAIL), which leads to the reversal of both intrinsic and acquired GEM resistance in PDAC treatment. Moreover, the pancreatic cancer cells that were able to bypass apoptosis after treatment of ONC201 get arrested in the G1-phase, which makes them highly sensitive to GEM. To enhance the in vivo stability of GEM, we first synthesized a disulfide bond containing stearate conjugated GEM (lipid-GEM), which makes it sensitive to the redox tumor microenvironment (TME) comprising high glutathione levels. In addition, with the help of colipids 1,2-dioleoyl-glycero-3-phosphocholine (DOPC), cholesterol, and 1,2-distearoyl-glycero-3-phosphoethanolamine-poly(ethylene glycol)-2000 (DSPE-PEG 2000), we were able to synthesize the lipid-GEM conjugate and ONC201 releasing liposomes. A cumulative drug release study confirmed that both ONC201 and GEM showed sustained release from the formulation. Since MUC1 is highly expressed in 70-90% PDAC, we conjugated a MUC1 binding peptide in the liposomes which showed higher cytotoxicity, apoptosis, and cellular internalization by MIA PaCa-2 cells. A biodistribution study further confirmed that the systemic delivery of the liposomes through the tail vein resulted in a higher accumulation of drugs in orthotopic PDAC tumors in NSG mice. The IHC of the excised tumor grafts further confirmed the higher apoptosis and lower metastasis and cell proliferation. Thus, our MUC1 targeting binary drug-releasing liposomal formulation showed higher drug payload, enhanced plasma stability, and accumulation of drugs in the pancreatic orthotopic tumor and thus is a promising therapeutic alternative for the treatment of PDAC.
Collapse
Affiliation(s)
- Prakash Shrestha
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yashwardhan Ghanwatkar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Sohan Mahto
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Nilkamal Pramanik
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
2
|
Han H, Li S, Zhong Y, Huang Y, Wang K, Jin Q, Ji J, Yao K. Emerging pro-drug and nano-drug strategies for gemcitabine-based cancer therapy. Asian J Pharm Sci 2022; 17:35-52. [PMID: 35261643 PMCID: PMC8888143 DOI: 10.1016/j.ajps.2021.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/19/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
Gemcitabine has been extensively applied in treating various solid tumors. Nonetheless, the clinical performance of gemcitabine is severely restricted by its unsatisfactory pharmacokinetic parameters and easy deactivation mainly because of its rapid deamination, deficiencies in deoxycytidine kinase (DCK), and alterations in nucleoside transporter. On this account, repeated injections with a high concentration of gemcitabine are adopted, leading to severe systemic toxicity to healthy cells. Accordingly, it is highly crucial to fabricate efficient gemcitabine delivery systems to obtain improved therapeutic efficacy of gemcitabine. A large number of gemcitabine pro-drugs were synthesized by chemical modification of gemcitabine to improve its biostability and bioavailability. Besides, gemcitabine-loaded nano-drugs were prepared to improve the delivery efficiency. In this review article, we introduced different strategies for improving the therapeutic performance of gemcitabine by the fabrication of pro-drugs and nano-drugs. We hope this review will provide new insight into the rational design of gemcitabine-based delivery strategies for enhanced cancer therapy.
Collapse
Affiliation(s)
- Haijie Han
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Su Li
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yueyang Zhong
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Kai Wang
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ke Yao
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Zhejiang Provincial Key Lab of Ophthalmology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
3
|
Han XW, Daugulis O, Brookhart M. Synthesis of End-Functionalized Poly(norbornenes) and Poly(ethylidene norbornenes) Using a Pd(II) Catalyst in Combination with Chain Transfer Agents. Organometallics 2021. [DOI: 10.1021/acs.organomet.1c00333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xing-Wang Han
- Center for Polymer Chemistry, Department of Chemistry, University of Houston, Houston, Texas 77204-5003, United States
| | - Olafs Daugulis
- Center for Polymer Chemistry, Department of Chemistry, University of Houston, Houston, Texas 77204-5003, United States
| | - Maurice Brookhart
- Center for Polymer Chemistry, Department of Chemistry, University of Houston, Houston, Texas 77204-5003, United States
| |
Collapse
|
4
|
Alzhrani RF, Xu H, Valdes SA, Naguib YW, Cui Z. Effect of surface mannosylation on the cytotoxicity and cellular uptake of stearoyl gemcitabine-incorporated, acid-sensitive micelles. COLLOID AND INTERFACE SCIENCE COMMUNICATIONS 2021; 43:100441. [PMID: 34307073 PMCID: PMC8294156 DOI: 10.1016/j.colcom.2021.100441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Elevated expression of C-type like receptors (CLRs) by tumor cells and tumor-associated macrophages (TAMs) present a unique target for the delivery of anticancer agents. Stearoyl gemcitabine (GemC18)-incorporated, acid-sensitive micelles (G-AS-M) prepared with a stearoyl polyethylene glycol (PEG2000) hydrazone were surface-mannosylated in this study for potential targeted killing of tumor cells and TAMs. The surface mannosylated micelles (i.e. G-MAS-M) were significantly more cytotoxic than the G-AS-M micelles to macrophages and tumor cells that express CLRs. Surprisingly, the uptake of GemC18 in the mannosylated G-MAS-M micelles by the macrophages and tumor cells was lower than that of GemC18 in the G-AS-M micelles. The lack of correlation between the cytoxicity and cellular uptake of GemC18 in the micelles was likely caused by a reduction in the sensitivity of the hydrazone bond linking the PEG2000 to the mannosylated G-MAS-M micelles to hydrolysis, resulting in more stable micelles.
Collapse
Affiliation(s)
- Riyad F. Alzhrani
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
| | - Haiyue Xu
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
| | - Solange A. Valdes
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
| | - Youssef W. Naguib
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
- Deparment of Pharmaceutics, Faculty of Pharmacy, Minia
University, Minia, Egypt
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
| |
Collapse
|
5
|
Catarata R, Azim N, Bhattacharya S, Zhai L. Controlled drug release from polyelectrolyte-drug conjugate nanoparticles. J Mater Chem B 2021; 8:2887-2894. [PMID: 32191246 DOI: 10.1039/d0tb00012d] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Encapsulating drugs in functional nanoparticles provides controlled and targeted release of drugs. In this study, a general approach for encapsulating hydrophobic drugs in polyelectrolyte nanoparticles was developed for a controlled drug release. Gemcitabine (GEM), an anticancer drug for pancreatic ductal adenocarcinoma (PDAC), was used as a model drug to produce poly(acrylic acid) (PAA)-GEM conjugate nanoparticles to achieve a controlled release of GEM in cells. The PAA-GEM conjugate nanoparticles were fabricated by coupling GEM onto PAA through the formation of amide bonds. The hydrophobic interactions of GEM molecules induced the formation of the nanoparticles with the GEM core and PAA shell. Fabrication conditions such as the PAA/GEM ratio and pH were optimized to achieve high structure stability and drug loading efficiency. The size and surface charge of the nanoparticles were characterized by transmission electron microscopy (TEM), dynamic light scattering (DLS) and zeta potential measurement. The optimized PAA-GEM nanoparticles had a size around 12 nm, 30 nm and 60 nm in dry state, water, and phosphate buffered saline (PBS), respectively. The encapsulation efficiency was 29.29 ± 1.7%, and the loading capacity was 9.44 ± 0.46%. Less than 7% GEM was released from the PAA-GEM nanoparticles after 96 hour incubation in phosphate buffered saline. The cytotoxic efficacy of the PAA-GEM nanoparticles in cancer cells was investigated through viability studies of PANC-1, a human pancreatic cancer cell line. It was found that the PAA-GEM nanoparticles had more than a 48 hour delay of releasing GEM and had the same cytotoxic efficacy in PANC-1 cells as free GEM. The uptake of the PAA-GEM nanoparticles by PANC-1 cells was investigated using PAA-GEM labeled by rhodamine G6. Fluorescence and bright field overlay images indicated that the PAA-GEM nanoparticles were taken up by PANC-1 cells within 2 hours. It is believed that the PAA-GEM nanoparticles were decomposed in PANC-1 cells and GEM was released from the nanoparticles.
Collapse
Affiliation(s)
- Ruginn Catarata
- NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, USA.
| | - Nilab Azim
- NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, USA. and Department of Chemistry, University of Central Florida, Orlando, Florida 32816, USA
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Jacksonville, Florida 32224, USA.
| | - Lei Zhai
- NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, USA. and Department of Chemistry, University of Central Florida, Orlando, Florida 32816, USA and Department of Material Science and Engineering, University of Central Florida, Orlando, Florida 32816, USA
| |
Collapse
|
6
|
Yin N, Yu H, Zhang X, Lv X. Enhancement of Pancreatic Cancer Therapy Efficacy by Type-1 Matrix Metalloproteinase-Functionalized Nanoparticles for the Selective Delivery of Gemcitabine and Erlotinib. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4465-4475. [PMID: 33122890 PMCID: PMC7591159 DOI: 10.2147/dddt.s270303] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/02/2020] [Indexed: 12/23/2022]
Abstract
Purpose Pancreatic cancer (PCa) is projected to become the second leading cause of cancer-related deaths by 2030. Gemcitabine (GEM) combined with erlotinib (ERL) have been approved by the FDA for locally advanced, unresectable or metastatic pancreatic cancer therapy since 2005. Type-1 matrix metalloproteinase (MT1-MMP) has been recognized as a critical mediator of several steps in PCa progression including activating TGF-β or releasing latent TGF-β from LTBP-1, resulting in increased collagen production and cleavage collagen. Methods In the present research, GEM and ERL co-loaded nanoparticles (GEM/ERL NPs) were prepared. A non-substrate MT1-MMP binding peptide was decorated onto the GEM/ERL NPs surface. Results M-M GEM/ERL NPs exhibited the highest uptake ability (67.65 ± 2.87%), longest half-life period, largest area under the curve, and the best tumor inhibition efficiency (69.81 ± 4.13%). The body weight, blood urine nitrogen (BUN), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) of the system were steady when tested in mice model. Conclusion In conclusion, M-M GEM/ERL NPs protected the drugs in the plasma, improved cellular uptake capacity, exhibited the most remarkable tumor cell inhibition ability, and showed the most efficient tumor growth inhibition capacity in vivo. M-M GEM/ERL NPs could be applied as an efficient and safe system for the synergistic combination chemotherapy of PCa.
Collapse
Affiliation(s)
- Na Yin
- Department of Pharmacy, Jinan Infectious Diseases Hospital Affiliated to Shandong University, Jinan 250000, People's Republic of China
| | - Hui Yu
- Department of Pharmacy, Jinan Infectious Diseases Hospital Affiliated to Shandong University, Jinan 250000, People's Republic of China
| | - Xiaodi Zhang
- Department of Pharmacy, Jinan Infectious Diseases Hospital Affiliated to Shandong University, Jinan 250000, People's Republic of China
| | - Xiaodan Lv
- Department of Pharmacy, Jinan Infectious Diseases Hospital Affiliated to Shandong University, Jinan 250000, People's Republic of China
| |
Collapse
|
7
|
Guo R, Long Y, Lu Z, Deng M, He P, Li M, He Q. Enhanced stability and efficacy of GEM-TOS prodrug by co-assembly with antimetastatic shell LMWH-TOS. Acta Pharm Sin B 2020; 10:1977-1988. [PMID: 33163348 PMCID: PMC7606181 DOI: 10.1016/j.apsb.2019.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/08/2019] [Accepted: 06/20/2019] [Indexed: 01/01/2023] Open
Abstract
Chemotherapy agents have been widely used for cancer treatment, while the insolubility, instability and toxicity seriously restrict their efficacy. Thus, prodrug strategy was devised. Since some prodrugs are still with poor solubility or stability, a synergy strategy is needed to enhance their efficacy. Gemcitabine (GEM) is a prescribed anticancer drug, however, the rapid clearance, growing resistance and serious side effects limit its clinical efficacy. Conjugating GEM with d-α-tocopherol succinate (TOS) is an effective solution, while the GEM-TOS (GT) is unstable in aqueous solution. d-α-Tocopherol polyethylene glycol succinate (TPGS) has been used to enhance the stability, but GT stabilized by TPGS (GTT) has limited effect on tumor metastases. Tumor metastases lead to high mortality in patients suffering from cancers. In order to further achieve antimetastatic effect, an amphiphilic polymer (LT) was synthesized by connecting low-molecular-weight heparin (LMWH) with TOS, and eventually obtained desired self-delivery micellar NPs (GLT) by co-assembly GT with LT. The GLT not only possessed excellent stability, but also inhibited the metastases by acting on different phases of the metastatic cascade. The hydrophobic TOS inhibited the secretion of matrix metalloproteinase-9 (MMP-9), the hydrophilic LMWH inhibited the interaction between tumor cells and platelets. As a result, GLT reduced tumor cells entering the blood and implanting at the distant organs, leading to a much more excellent inhibitory effect on the lung metastasis than GEM and GTT.
Collapse
|
8
|
Alzhrani RF, Xu H, Valdes SA, Cui Z. Intranasal delivery of a nicotine vaccine candidate induces antibodies in mouse blood and lung mucosal secretions that specifically neutralize nicotine. Drug Dev Ind Pharm 2020; 46:1656-1664. [PMID: 32892651 DOI: 10.1080/03639045.2020.1820033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Cigarette smoking is one of the leading causes of death in the world. The majority of the smokers have tried to quit, but only a few of them were able to achieve long-term abstinence, due to the high addictiveness of nicotine. Nicotine-specific antibodies have the potential to block the euphoric effect of nicotine by forming antibody-antigen complexes in the blood circulation. Since nicotine is taken largely by inhalation, inducing anti-nicotine antibodies in lung and nasal mucosal secretions, in addition to blood circulation, is expected to be beneficial. SIGNIFICANCE The importance of this study is to establish the feasibility of inducing nicotine-neutralizing antibodies not only in the blood, but also in the lung and nasal mucosal secretions, by intranasal administration of a nicotine vaccine candidate. METHODS Nicotine-keyhole limpet hemocyanin conjugate (Nic-KLH) was prepared and mixed with monophosphoryl lipid A (MPL) as an adjuvant. Nic-KLH/MPL was given intranasally or subcutaneously to mice, and the titers, affinity, and specificity of the nicotine-specific antibodies in nasal and lung mucosal secretions and blood samples were determined using (competitive) ELISA. RESULTS Nasal Nic-KLH/MPL immunization elicited robust nicotine-specific neutralizing IgA in mouse nasal and lung secretions, in additional to anti-nicotine IgG in blood circulation. The nicotine-specific IgG level in mice nasally immunized with Nic-KLH/MPL was lower than in mice subcutaneously immunized with the same Nic-KLH/MPL, but a heterologous prime-boost immunization strategy helped to increase it. CONCLUSION Intranasal immunization with a nicotine vaccine candidate can induce systemic and mucosal antibodies that specifically neutralize nicotine.
Collapse
Affiliation(s)
- Riyad F Alzhrani
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA
| | - Haiyue Xu
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA
| | - Solange A Valdes
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA
| | - Zhengrong Cui
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
9
|
Fattahi N, Shahbazi MA, Maleki A, Hamidi M, Ramazani A, Santos HA. Emerging insights on drug delivery by fatty acid mediated synthesis of lipophilic prodrugs as novel nanomedicines. J Control Release 2020; 326:556-598. [PMID: 32726650 DOI: 10.1016/j.jconrel.2020.07.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/25/2022]
Abstract
Many drug molecules that are currently in the market suffer from short half-life, poor absorption, low specificity, rapid degradation, and resistance development. The design and development of lipophilic prodrugs can provide numerous benefits to overcome these challenges. Fatty acids (FAs), which are lipophilic biomolecules constituted of essential components of the living cells, carry out many necessary functions required for the development of efficient prodrugs. Chemical conjugation of FAs to drug molecules may change their pharmacodynamics/pharmacokinetics in vivo and even their toxicity profile. Well-designed FA-based prodrugs can also present other benefits, such as improved oral bioavailability, promoted tumor targeting efficiency, controlled drug release, and enhanced cellular penetration, leading to improved therapeutic efficacy. In this review, we discuss diverse drug molecules conjugated to various unsaturated FAs. Furthermore, various drug-FA conjugates loaded into various nanostructure delivery systems, including liposomes, solid lipid nanoparticles, emulsions, nano-assemblies, micelles, and polymeric nanoparticles, are reviewed. The present review aims to inspire readers to explore new avenues in prodrug design based on the various FAs with or without nanostructured delivery systems.
Collapse
Affiliation(s)
- Nadia Fattahi
- Department of Chemistry, Faculty of Science, University of Zanjan, P.O. Box 45195-313, Zanjan, Iran; Trita Nanomedicine Research Center (TNRC), Trita Third Millennium Pharmaceuticals, 45331-55681 Zanjan, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Aziz Maleki
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehrdad Hamidi
- Trita Nanomedicine Research Center (TNRC), Trita Third Millennium Pharmaceuticals, 45331-55681 Zanjan, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Ali Ramazani
- Department of Chemistry, Faculty of Science, University of Zanjan, P.O. Box 45195-313, Zanjan, Iran; Research Institute of Modern Biological Techniques (RIMBT), University of Zanjan, P.O. Box 45195-313, Zanjan, Iran
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Helsinki Institute of Life Science (HiLIFE), Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.
| |
Collapse
|
10
|
Valdes SA, Alzhrani RF, Lansakara-P DSP, Cui Z. Effect of a Solid Lipid Nanoparticle Formulation on the Bioavailability of 4-(N)-Docosahexaenoyl 2', 2'-Difluorodeoxycytidine After Oral Administration. AAPS PharmSciTech 2020; 21:77. [PMID: 31970527 DOI: 10.1208/s12249-020-1617-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/26/2019] [Indexed: 11/30/2022] Open
Abstract
Previously, we developed a solid lipid nanoparticle (SLN) formulation of 4-(N)-docosahexaenoyl 2', 2'-difluorodeoxycytidine (DHA-dFdC), a compound with promising antitumor activity. Herein, we studied the feasibility of administering the DHA-dFdC by the oral route using the solid lipid nanoparticles (i.e., DHA-dFdC-SLNs). In simulated gastrointestinal fluids, the DHA-dFdC-SLNs did not aggregate. The release of the DHA-dFdC from the solid lipid nanoparticles in simulated gastrointestinal fluid was slow, but was slightly faster in simulated intestinal fluid than in simulated gastric fluid. In mice orally administered with DHA-dFdC-SLNs, plasma DHA-dFdC concentration vs. time curve has a Tmax of ~ 1.7 h and a Cmax of 17.01 μg/mL. The absolute oral bioavailability of DHA-dFdC when given as DHA-dFdC-SLNs was ~ 68% (based on AUC0-24 h values), while the relative oral bioavailability DHA-dFdC (compared with DHA-dFdC in a Tween 80/ethanol-in-water solution) was 126%. Finally, in mice with pre-establish B16-F10 murine melanoma, oral DHA-dFdC-SLNs increased their survival significantly, as compared with oral administration of the DHA-dFdC solution. It is concluded that the solid lipid nanoparticle formulation increased the bioavailability of the DHA-dFdC upon oral administration, as compared with the DHA-dFdC solution.
Collapse
|
11
|
Samanta K, Setua S, Kumari S, Jaggi M, Yallapu MM, Chauhan SC. Gemcitabine Combination Nano Therapies for Pancreatic Cancer. Pharmaceutics 2019; 11:E574. [PMID: 31689930 PMCID: PMC6920852 DOI: 10.3390/pharmaceutics11110574] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is one of the deadliest causes of cancer-related death in the United States, with a 5-year overall survival rate of 6 to 8%. These statistics suggest that immediate medical attention is needed. Gemcitabine (GEM) is the gold standard first-line single chemotherapy agent for pancreatic cancer but, after a few months, cells develop chemoresistance. Multiple clinical and experimental investigations have demonstrated that a combination or co-administration of other drugs as chemotherapies with GEM lead to superior therapeutic benefits. However, such combination therapies often induce severe systemic toxicities. Thus, developing strategies to deliver a combination of chemotherapeutic agents more securely to patients is needed. Nanoparticle-mediated delivery can offer to load a cocktail of drugs, increase stability and availability, on-demand and tumor-specific delivery while minimizing chemotherapy-associated adverse effects. This review discusses the available drugs being co-administered with GEM and the limitations associated during the process of co-administration. This review also helps in providing knowledge of the significant number of delivery platforms being used to overcome problems related to gemcitabine-based co-delivery of other chemotherapeutic drugs, thereby focusing on how nanocarriers have been fabricated, considering the modes of action, targeting receptors, pharmacology of chemo drugs incorporated with GEM, and the differences in the physiological environment where the targeting is to be done. This review also documents the focus on novel mucin-targeted nanotechnology which is under development for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Kamalika Samanta
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Saini Setua
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sonam Kumari
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Meena Jaggi
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| | - Murali M Yallapu
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| |
Collapse
|
12
|
O'Mary HL, Hanafy MS, Aldayel AM, Valdes SA, Alzhrani RF, Hufnagel S, Koleng JJ, Cui Z. Effect of the Ratio of Betamethasone to TNF-α siRNA Coencapsulated in Solid Lipid Nanoparticles on the Acute Proinflammatory Activity of the Nanoparticles. Mol Pharm 2019; 16:4496-4506. [PMID: 31525980 DOI: 10.1021/acs.molpharmaceut.9b00629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is evidence that encapsulating glucocorticoids into nucleic acid-containing nanoparticles reduces the inflammatory toxicities of the nanoparticles. Herein, using betamethasone acetate (BA), a glucocorticoid, and a solid lipid nanoparticle formulation of siRNA, we confirmed that coencapsulating BA into the siRNA solid lipid nanoparticles significantly reduced the proinflammatory activity of the siRNA nanoparticles in a mouse model. Using TNF-α siRNA, we then showed that the BA and TNF-α siRNA coencapsulated into the solid lipid nanoparticles acted as a dual anti-inflammatory and synergistically reduced TNF-α release by mouse macrophages in culture following stimulation with lipopolysaccharide, as compared to solid lipid nanoparticles encapsulated with TNF-α siRNA or BA alone. Importantly, upon studying the effect of the ratio of BA and TNF-α siRNA on the proinflammatory activity of the resultant nanoparticles, we identified that BA and TNF-α siRNA coencapsulated solid lipid nanoparticles prepared with a BA to TNF-α siRNA weight ratio of 2:1 induced the lowest proinflammatory cytokine production by macrophages in culture. This result was in comparison to nanoparticles prepared with BA to TNF-α siRNA ratios both higher and lower than 2:1 (i.e., 4:1, 1:1, and 0.5:1) and is likely due to differences in molecular interactions among the various components in the BA and TNF-α-siRNA coencapsulated solid lipid nanoparticles at these ratios. Encapsulating glucocorticoids into siRNA-nanoparticles represents a viable strategy to reduce the proinflammatory activity of the nanoparticles; however, the ratio of the glucocorticoid to siRNA in the nanoparticles requires optimization.
Collapse
Affiliation(s)
- Hannah L O'Mary
- The University of Texas at Austin , College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery , Austin , Texas 78712 , United States
| | - Mahmoud S Hanafy
- The University of Texas at Austin , College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery , Austin , Texas 78712 , United States
| | - Abdulaziz M Aldayel
- The University of Texas at Austin , College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery , Austin , Texas 78712 , United States.,Medical Research Core Facility and Platforms , King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS) , King Abdulaziz Medical City (KAMC) NGHA , Riyadh 11426 , Saudi Arabia
| | - Solange A Valdes
- The University of Texas at Austin , College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery , Austin , Texas 78712 , United States
| | - Riyad F Alzhrani
- The University of Texas at Austin , College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery , Austin , Texas 78712 , United States
| | - Stephanie Hufnagel
- The University of Texas at Austin , College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery , Austin , Texas 78712 , United States
| | - John J Koleng
- Via Therapeutics, LLC , Austin , Texas 78750 , United States
| | - Zhengrong Cui
- The University of Texas at Austin , College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery , Austin , Texas 78712 , United States
| |
Collapse
|
13
|
Valdes SA, Alzhrani RF, Rodriguez A, Lansakara-P DSP, Thakkar SG, Cui Z. A solid lipid nanoparticle formulation of 4-(N)-docosahexaenoyl 2', 2'-difluorodeoxycytidine with increased solubility, stability, and antitumor activity. Int J Pharm 2019; 570:118609. [PMID: 31415878 DOI: 10.1016/j.ijpharm.2019.118609] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/03/2019] [Accepted: 08/09/2019] [Indexed: 01/27/2023]
Abstract
Previously, we synthesized 4-(N)-docosahexaenoyl 2', 2'-difluorodeoxycytidine (DHA-dFdC), a novel lipophilic compound with a potent, broad-spectrum antitumor activity. Herein, we report a solid lipid nanoparticle (SLN) formulation of DHA-dFdC with improved apparent aqueous solubility, chemical stability, as well as efficacy in a mouse model. The SLNs were prepared from lecithin/glycerol monostearate-in-water emulsions emulsified with D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) and Tween 20. The resultant DHA-dFdC-SLNs were 102.2 ± 7.3 nm in diameter and increased the apparent solubility of DHA-dFdC in water to at least 5.2 mg/mL, more than 200-fold higher than its intrinsic water solubility. DHA-dFdC in a lyophilized powder of DHA-dFdC-SLNs was significantly more stable than the waxy solid of pure DHA-dFdC. DHA-dFdC-SLNs also showed an increased cytotoxicity against certain tumor cells than DHA-dFdC. The plasma concentration of DHA-dFdC in mice intravenously injected with DHA-dFdC-SLNs in dispersion followed a bi-exponential model, with a half-life of ~44 h. In mice bearing B16-F10 murine melanoma, DHA-dFdC-SLNs were significantly more effective than DHA-dFdC in controlling the tumor growth. In addition, histology evaluation revealed a high level of apoptosis and tumor encapsulation in tumors in mice treated with DHA-dFdC-SLNs. DHA-dFdC-SLNs represents a new DHA-dFdC formulation with improved antitumor activity.
Collapse
Affiliation(s)
- Solange A Valdes
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Riyad F Alzhrani
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | | | - Dharmika S P Lansakara-P
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Sachin G Thakkar
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Zhengrong Cui
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
14
|
Raghupathi K, Kumar V, Sridhar U, Ribbe AE, He H, Thayumanavan S. Role of Oligoethylene Glycol Side Chain Length in Responsive Polymeric Nanoassemblies. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:7929-7936. [PMID: 31095400 DOI: 10.1021/acs.langmuir.9b00676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
An oft-desired feature of a responsive nanomaterial is that it should undergo disassembly or morphological change upon application of a specific stimulus. The extent of response has been found to depend on factors such as the nature and the number of responsive functionalities incorporated into these particles. In this work, the length of oligoethylene glycol (OEG) side chains associated with the polymers has been shown to greatly influence the responsive behavior of polymeric nanoparticles. The integrity of these OEG-based polymeric assemblies was found to depend not only on the chemical cross-links but also on the physical cross-links in these aggregates in cases where the polymer chains bear long OEG side chains. The physical cross-linking in longer OEG side chain containing polymeric nanogels is present in the form of crystalline domains. Our results here highlight that these ethylene glycol-based hydrophilic units are not to be ignored as spectator units with water-solubilization characteristics but must be analyzed in the context of assembly stabilization and triggerability with the targeted stimulus.
Collapse
|
15
|
Ren Q, Liang Z, Jiang X, Gong P, Zhou L, Sun Z, Xiang J, Xu Z, Peng X, Li S, Li W, Cai L, Tang J. Enzyme and pH dual-responsive hyaluronic acid nanoparticles mediated combination of photodynamic therapy and chemotherapy. Int J Biol Macromol 2019; 130:845-852. [DOI: 10.1016/j.ijbiomac.2019.03.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 10/27/2022]
|
16
|
Zhang H, Zhou T, Yu Q, Yang Z, Sun Y, Cai Z, Cang H. pH-Sensitive betulinic acid polymer prodrug nanoparticles for efficient and targeted cancer cells treatment. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2019.1596916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Huaihong Zhang
- School of Chemistry and Biology, Yancheng Institute of Technology, Yancheng, China
| | - Tao Zhou
- School of Chemistry and Biology, Yancheng Institute of Technology, Yancheng, China
| | - Qing Yu
- School of Chemistry and Biology, Yancheng Institute of Technology, Yancheng, China
| | - Zhenqing Yang
- School of Chemistry and Biology, Yancheng Institute of Technology, Yancheng, China
| | - Yu Sun
- College of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Zhaosheng Cai
- School of Chemistry and Biology, Yancheng Institute of Technology, Yancheng, China
| | - Hui Cang
- School of Chemistry and Biology, Yancheng Institute of Technology, Yancheng, China
| |
Collapse
|
17
|
Li X, Valdes SA, Alzhrani RF, Hufnagel S, Hursting SD, Cui Z. Zoledronic Acid-containing Nanoparticles With Minimum Premature Release Show Enhanced Activity Against Extraskeletal Tumor. ACS APPLIED MATERIALS & INTERFACES 2019; 11:7311-7319. [PMID: 30689348 PMCID: PMC6582365 DOI: 10.1021/acsami.8b16588] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Bisphosphonates are generally used to treat bone diseases, such as bone metastasis from cancer. There is evidence that, through the modification of the pharmacokinetics and biodistribution of bisphosphonates by formulating them into nanoparticles, they may be able to treat extraskeletal tumors. However, many previously reported bisphosphonate nanoparticle formulations show extensive premature release of bisphosphonates. Herein, using zoledronate (Zol), a third-generation bisphosphonate, we developed a new Zol nanoparticle formulation (denoted as Zol-NPs) by encapsulating anionic lipid-coated Zol-calcium nanocomplexes into poly(lactic- co-glycolic) acid nanoparticles emulsified with octadecanoic acid-hydrazone-polyethylene glycol (2000), an acid-sensitive cleavable emulsifying agent. The resultant Zol-NPs, about 180 nm in hydrodynamic diameter, show very limited premature release of Zol (i.e., <5% in 48 h in a simulated physiological condition) and enhanced cytotoxicity to both murine cancer cells and macrophages. In a mouse model with orthotopically transplanted mammary tumors, Zol-NPs significantly reduced the distribution of Zol in bones, but increased its distribution in tumors. Importantly, Zol-NPs also significantly inhibited tumor growth, whereas the equivalent dose of free Zol did not. This platform technology may be exploited to treat extraskeletal tumors with bisphosphonates.
Collapse
Affiliation(s)
- Xu Li
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, 78723 USA
| | - Solange A. Valdes
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, 78723 USA
| | - Riyad F. Alzhrani
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, 78723 USA
| | - Stephanie Hufnagel
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, 78723 USA
| | - Stephen D. Hursting
- University of North Carolina at Chapel Hill, Department of
Nutrition, Chapel Hill, NC, 27599 USA
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy,
Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, 78723 USA
- Northwest University, College of Life Sciences,
Xi’an, Shaanxi, 710069 China
| |
Collapse
|
18
|
Ohara Y, Nakai K, Ahmed S, Matsumura K, Ishihara K, Yusa SI. pH-Responsive Polyion Complex Vesicle with Polyphosphobetaine Shells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:1249-1256. [PMID: 29940726 DOI: 10.1021/acs.langmuir.8b00632] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
When a bioactive molecule is taken into cells by endocytosis, it is sometimes unable to escape from the lysosomes, resulting in inefficient drug release. We prepared pH-responsive polyion complex (PIC) vesicles that collapse under acidic conditions such as those inside a lysosome. Furthermore, under acidic conditions, cationic polymer was released from the PIC vesicles to break the lysosome membranes. Diblock copolymers (P20M167 and P20A190) consisting of water-soluble zwitterionic poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC) block and cationic or anionic blocks were synthesized via reversible addition-fragmentation chain transfer (RAFT) radical polymerization. Poly(3-(methacrylamidopropyl) trimethylammonium chloride) (PMAPTAC) and poly(sodium 6-acrylamidohexanoate) (PAaH) were used as the cationic and anionic blocks, respectively. The pendant hexanoate groups in the PAaH block are ionized in basic water and in phosphate buffered saline (PBS), while the hexanoate groups are protonated in acidic water. In basic water, PIC vesicles were formed from a charge neutralized mixture of oppositely charged diblock copolymers. At the interface of PIC vesicle and water exists biocompatible PMPC shells. Under acidic conditions, the PIC vesicles collapsed, because the charge balance shifted due to protonation of the PAaH block. After collapse of the PIC vesicles, P20A190 formed micelles composed of protonated PAaH core and PMPC shells, while P20M167 was released as unimers. PIC vesicles can encapsulate hydrophilic nonionic guest molecules into their hollow core. Under acidic conditions, the PIC vesicles can release the guest molecules and P20M167. The cationic P20M167 can break the lysosome membrane to efficiently release the guest molecules from the lysosomes to the cytoplasm.
Collapse
Affiliation(s)
- Yuki Ohara
- Department of Applied Chemistry, Graduate School of Engineering , University of Hyogo , 2167 Shosha , Himeji , Hyogo 671-2280 , Japan
| | - Keita Nakai
- Department of Applied Chemistry, Graduate School of Engineering , University of Hyogo , 2167 Shosha , Himeji , Hyogo 671-2280 , Japan
| | - Sana Ahmed
- School of Materials Science , Japan Advanced Institute of Science and Technology , 1-1 Asahidai , Nomi , Ishikawa 923-1292 , Japan
| | - Kazuaki Matsumura
- School of Materials Science , Japan Advanced Institute of Science and Technology , 1-1 Asahidai , Nomi , Ishikawa 923-1292 , Japan
| | - Kazuhiko Ishihara
- Department of Materials Engineering, School of Engineering , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku , Tokyo 113-8656 , Japan
| | - Shin-Ichi Yusa
- Department of Applied Chemistry, Graduate School of Engineering , University of Hyogo , 2167 Shosha , Himeji , Hyogo 671-2280 , Japan
| |
Collapse
|
19
|
Brega V, Scaletti F, Zhang X, Wang LS, Li P, Xu Q, Rotello VM, Thomas SW. Polymer Amphiphiles for Photoregulated Anticancer Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2019; 11:2814-2820. [PMID: 30582802 PMCID: PMC6623983 DOI: 10.1021/acsami.8b18099] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
We report the synthesis of amphiphilic polymers featuring lipophilic stearyl chains and hydrophilic poly(ethylene glycol) polymers that are connected through singlet oxygen-cleavable alkoxyanthracene linkers. These amphiphilic polymers assembled in water to form micelles with diameters of ∼20 nm. Reaction of the alkoxyanthracene linkers with light and O2 cleaved the ether C-O bonds, resulting in formation of the corresponding 9,10-anthraquinone derivatives and concomitant disruption of the micelles. These micelles were loaded with the chemotherapeutic agent doxorubicin, which was efficiently released upon photo-oxidation. The drug-loaded reactive micelles were effective at killing cancer cells in vitro upon irradiation at 365 nm, functioning through both doxorubicin release and photodynamic mechanisms.
Collapse
Affiliation(s)
- Valentina Brega
- Department of Chemistry, Tufts University, 62 Talbot Avenue, Medford MA 02155, United States
| | - Federica Scaletti
- Department of Chemistry, University of Massachusetts Amherst, 710 Nt. Pleasant Street, Amherst MA 01003, United States
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 Nt. Pleasant Street, Amherst MA 01003, United States
| | - Li-Sheng Wang
- Department of Chemistry, University of Massachusetts Amherst, 710 Nt. Pleasant Street, Amherst MA 01003, United States
| | - Prudence Li
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford MA 02155, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford MA 02155, United States
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 Nt. Pleasant Street, Amherst MA 01003, United States
| | - Samuel W. Thomas
- Department of Chemistry, Tufts University, 62 Talbot Avenue, Medford MA 02155, United States
- Corresponding Author: (S.W.T.)
| |
Collapse
|
20
|
Seo J, Lee J, Lee CB, Bae SK, Na K. Nonpolymeric pH-Sensitive Carbon Dots for Treatment of Tumor. Bioconjug Chem 2019; 30:621-632. [DOI: 10.1021/acs.bioconjchem.8b00813] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jeongdeok Seo
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Jonghwan Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Chae Bin Lee
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Soo Kyung Bae
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| |
Collapse
|
21
|
Li X, Zhao H, Ji Y, Yin C, Li J, Yang Z, Tang Y, Zhang Q, Fan Q, Huang W. Lysosome-Assisted Mitochondrial Targeting Nanoprobe Based on Dye-Modified Upconversion Nanophosphors for Ratiometric Imaging of Mitochondrial Hydrogen Sulfide. ACS APPLIED MATERIALS & INTERFACES 2018; 10:39544-39556. [PMID: 30387597 DOI: 10.1021/acsami.8b16818] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Hydrogen sulfide (H2S) is a versatile modulator in mitochondria and involved in numerous diseases caused by mitochondrial dysfunction. Therefore, many efforts have been made to develop fluorescent probes for mitochondrial H2S detection. However, these cationic small molecule probes are inapplicable for in vivo imaging because of the shallow tissue penetration and poor biostability. Herein, a ratiometric upconversion luminescence nanoprobe with an acid-activated targeting strategy is developed for detecting and bioimaging of mitochondrial H2S. The merocyanine triphenylamine-merocyanine (TPAMC)-modified upconversion nanophosphors, acting as the targeting and response component, are encapsulated into a pH-sensitive husk, composed of 1,2-distearoyl- sn-glycero-3-phosphoethanolamine- N-[methoxy-(poly(ethylene glycol))-2000] (DSPE-PEG) and poly(l-histidine)- b-PEG, which improved the nanoprobe's stability during transport in vivo. Under lysosomal pH, the PEG shell is interrupted and the targeting sites are exposed to further attach to mitochondria. Taking advantage of the luminescence resonance energy transfer process between TPAMC and upconversion nanophosphors, the ratiometric detection of mitochondrial H2S can be achieved with high selectivity and sensitivity. Cellular testing reveals the precise targeting to mitochondria via a lysosome delivery process. Importantly, the nanoprobe can be used for monitoring mitochondrial H2S levels in living cells and colon cancer mouse models.
Collapse
Affiliation(s)
- Xiang Li
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
| | - Hui Zhao
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
| | - Yu Ji
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
| | - Chao Yin
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
| | - Jie Li
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
| | - Zhen Yang
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
| | - Yufu Tang
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
| | - Qichun Zhang
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
- School of Materials Science and Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
| | - Wei Huang
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , China
- Shaanxi Institute of Flexible Electronics (SIFE) , Northwestern Polytechnical University (NPU) , Xi'an 710072 , China
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing Tech University (NanjingTech) , Nanjing 211816 , China
| |
Collapse
|
22
|
Markovic M, Ben‐Shabat S, Keinan S, Aponick A, Zimmermann EM, Dahan A. Lipidic prodrug approach for improved oral drug delivery and therapy. Med Res Rev 2018; 39:579-607. [DOI: 10.1002/med.21533] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Milica Markovic
- Department of Clinical PharmacologySchool of Pharmacy, Faculty of Health Sciences, Ben‐Gurion University of the NegevBeer‐Sheva Israel
| | - Shimon Ben‐Shabat
- Department of Clinical PharmacologySchool of Pharmacy, Faculty of Health Sciences, Ben‐Gurion University of the NegevBeer‐Sheva Israel
| | | | - Aaron Aponick
- Department of ChemistryUniversity of FloridaGainesville Florida
| | - Ellen M. Zimmermann
- Department of MedicineDivision of Gastroenterology, University of FloridaGainesville Florida
| | - Arik Dahan
- Department of Clinical PharmacologySchool of Pharmacy, Faculty of Health Sciences, Ben‐Gurion University of the NegevBeer‐Sheva Israel
| |
Collapse
|
23
|
Improving Plasma Stability and Bioavailability In Vivo of Gemcitabine Via Nanoparticles of mPEG-PLG-GEM Complexed with Calcium Phosphate. Pharm Res 2018; 35:230. [DOI: 10.1007/s11095-018-2506-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/18/2018] [Indexed: 02/05/2023]
|
24
|
Uthaman S, Huh KM, Park IK. Tumor microenvironment-responsive nanoparticles for cancer theragnostic applications. Biomater Res 2018; 22:22. [PMID: 30155269 PMCID: PMC6108142 DOI: 10.1186/s40824-018-0132-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cancer is one of the deadliest threats to human health. Abnormal physiochemical conditions and dysregulated biosynthetic intermediates in the tumor microenvironment (TME) play a significant role in modulating cancer cells to evade or defend conventional anti-cancer therapy such as surgery, chemotherapy and radiotherapy. One of the most important challenges in the development of anti-tumor therapy is the successful delivery of therapeutic and imaging agents specifically to solid tumors. MAIN BODY The recent progresses in development of TME responsive nanoparticles offers promising strategies for combating cancer by making use of the common attributes of tumor such as acidic and hypoxic microenvironments. In this review, we discussed the prominent strategies utilized in the development of tumor microenvironment-responsive nanoparticles and mode of release of therapeutic cargo. CONCLUSION Tumor microenvironment-responsive nanoparticles offers a universal approach for anti-cancer therapy.
Collapse
Affiliation(s)
- Saji Uthaman
- Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 Republic of Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju, 61469 Republic of Korea
| |
Collapse
|
25
|
Lipid nanoparticles with minimum burst release of TNF-α siRNA show strong activity against rheumatoid arthritis unresponsive to methotrexate. J Control Release 2018; 283:280-289. [PMID: 29859232 DOI: 10.1016/j.jconrel.2018.05.035] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/29/2018] [Indexed: 12/18/2022]
Abstract
TNF-α siRNA has shown promising therapeutic benefits in animal models of rheumatoid arthritis. However, there continues to be a need for siRNA delivery systems that have high siRNA encapsulation efficiency and minimum burst release of TNF-α siRNA, and can target inflamed tissues after intravenous administration. Herein we report a novel acid-sensitive sheddable PEGylated solid-lipid nanoparticle formulation of TNF-α-siRNA, AS-TNF-α-siRNA-SLNs, prepared by incorporating lipophilized TNF-α-siRNA into solid-lipid nanoparticles composed of biocompatible lipids such as lecithin and cholesterol. The nanoparticles are approximately 120 nm in diameter, have a high siRNA encapsulation efficiency (>90%) and a minimum burst release of siRNA (<5%), and increase the deilvery of the siRNA in chronic inflammation sites in mouse models, including in a mouse model with collagen-induced arthritis. Importantly, in a mouse model of collagen antibody-induced arthritis that does not respond to methotrexate therapy, intravenous injection of the AS-TNF-α-siRNA-SLNs significantly reduced paw thickness, bone loss, and histopathological scores. These findings highlight the potential of using this novel siRNA nanoparticle formulation to effectively treat arthritis, potentially in patients who do not respond adequately to methotrexate.
Collapse
|
26
|
Abstract
The incidence of malignant melanoma is increasing rapidly on a global scale. Although some types of melanoma, for example primary cutaneous melanoma, can be managed by surgery, metastatic melanoma cannot and it has a high mortality rate. Both oncogene and immune-targeted strategies have shown marked efficacy in some patients, but their effect on overall survival is still variable. Therefore, newer therapeutic approaches are needed. Fortunately, new advances in molecular medicine have led to an understanding of an individual patient's cancer at the genomic level. This information is now being used in all stages of cancer treatment including diagnosis, treatment selection, and treatment monitoring. This new strategy of personalized medicine may lead to marked shifts in immunotherapeutic treatment approaches such as individualized cancer vaccines and adoptive transfer of genetically modified T cells. This review provides an overview of recent approaches in cancer research and expected impact on the future of treatment for metastatic melanoma.
Collapse
|
27
|
Morey M, Pandit A. Responsive triggering systems for delivery in chronic wound healing. Adv Drug Deliv Rev 2018; 129:169-193. [PMID: 29501700 DOI: 10.1016/j.addr.2018.02.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/27/2018] [Accepted: 02/26/2018] [Indexed: 12/31/2022]
Abstract
Non-communicable diseases including cancer, cardiovascular disease, diabetes, and neuropathy are chronic in nature. Treatment of these diseases with traditional delivery systems is limited due to lack of site-specificity, non-spatiotemporal release and insufficient doses. Numerous responsive delivery systems which respond to both physiological and external stimuli have been reported in the literature. However, effective strategies incorporating a multifactorial approach are required to control these complex wounds. This can be achieved by fabricating spatiotemporal release systems, multimodal systems or dual/multi-stimuli responsive delivery systems loaded with one or more bioactive components. Critically, these next generation stimuli responsive delivery systems that are at present not feasible are required to treat chronic wounds. This review provides a critical assessment of recent developments in the field of responsive delivery systems, highlighting their limitations and providing a perspective on how these challenges can be overcome.
Collapse
Affiliation(s)
- Mangesh Morey
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
28
|
Wang C, Zheng Y, Sand oval MA, Valdes SA, Chen Z, Lansakara-P DS, Du M, Shi Y, Cui Z. Oral 4-( N)-stearoyl gemcitabine nanoparticles inhibit tumor growth in mouse models. Oncotarget 2017; 8:89876-89886. [PMID: 29163795 PMCID: PMC5685716 DOI: 10.18632/oncotarget.21264] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/08/2017] [Indexed: 01/14/2023] Open
Abstract
In spite of recent advances in targeted tumor therapy, systemic chemotherapy with cytotoxic agents remains a vital cancer treatment modality. Gemcitabine is a nucleoside analog commonly used in the treatment of various solid tumors, but an oral gemcitabine dosage form remain unavailable. Previously, we developed the 4-(N)-stearoyl gemcitabine solid lipid nanoparticles (GemC18-SLNs) by incorporating 4-(N)-stearoyl gemcitabine (GemC18), an amide prodrug of gemcitabine, into solid lipid nanoparticles. GemC18-SLNs, when administered intravenously, showed strong antitumor activity against various human and mouse tumors in mouse models. In the present study, we defined the plasma pharmacokinetics of gemcitabine when GemC18-SLNs were given orally to healthy mice and evaluated the antitumor activity of GemC18-SLNs when given orally in mouse models of lung cancer. In mice orally gavaged with GemC18-SLNs, plasma gemcitabine concentration followed an absorption phase and then clearance phase, with a Tmax of ~2 h. The absolute oral bioavailability of gemcitabine in the GemC18-SLNs was ~70% (based on AUC0-24 h values). In mice with pre-established tumors (i.e. mouse TC-1 or LLC lung cancer cells), oral GemC18-SLNs significantly inhibited the tumor growth and increased mouse survival time, as compared to the molar equivalent dose of gemcitabine hydrochloride or GemC18 in vegetable oil or in Tween 20. Immunohistostaining revealed that oral GemC18-SLNs also have significant antiproliferative, antiangiogenic, and proapoptotic activity in LLC tumors. Formulating a lipophilic amide prodrug of gemcitabine into solid lipid nanoparticles may represent a viable approach toward developing a safe and efficacious gemcitabine oral dosage form.
Collapse
Affiliation(s)
- Caixia Wang
- Inner Mongolia Medical University, School of Basic Sciences, Inner Mongolia Key Laboratory of Molecular Biology, Hohhot, Inner Mongolia, China
| | - Yuanqiang Zheng
- Inner Mongolia University, Research Center for Laboratory Animal Sciences, Hohhot, Inner Mongolia, China
| | - Michael A. Sand oval
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
| | - Solange A. Valdes
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
| | - Zhe Chen
- Inner Mongolia Medical University, School of Basic Sciences, Inner Mongolia Key Laboratory of Molecular Biology, Hohhot, Inner Mongolia, China
| | - Dharmika S. Lansakara-P
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
| | - Maolin Du
- Inner Mongolia Medical University, School of Public Health, Hohhot, Inner Mongolia, China
| | - Yanchun Shi
- Inner Mongolia Medical University, School of Basic Sciences, Inner Mongolia Key Laboratory of Molecular Biology, Hohhot, Inner Mongolia, China
| | - Zhengrong Cui
- Inner Mongolia Medical University, School of Basic Sciences, Inner Mongolia Key Laboratory of Molecular Biology, Hohhot, Inner Mongolia, China
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, USA
| |
Collapse
|
29
|
Imam ZI, Kenyon LE, Ashby G, Nagib F, Mendicino M, Zhao C, Gadok AK, Stachowiak JC. Phase-Separated Liposomes Enhance the Efficiency of Macromolecular Delivery to the Cellular Cytoplasm. Cell Mol Bioeng 2017; 10:387-403. [PMID: 29104698 PMCID: PMC5665383 DOI: 10.1007/s12195-017-0489-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/11/2017] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION From viruses to organelles, fusion of biological membranes is used by diverse biological systems to deliver macromolecules across membrane barriers. Membrane fusion is also a potentially efficient mechanism for the delivery of macromolecular therapeutics to the cellular cytoplasm. However, a key shortcoming of existing fusogenic liposomal systems is that they are inefficient, requiring a high concentration of fusion-promoting lipids in order to cross cellular membrane barriers. OBJECTIVES Toward addressing this limitation, our experiments explore the extent to which membrane fusion can be amplified by using the process of lipid membrane phase separation to concentrate fusion-promoting lipids within distinct regions of the membrane surface. METHODS We used confocal fluorescence microscopy to investigate the integration of fusion-promoting lipids into a ternary lipid membrane system that separated into liquid-ordered and liquid-disordered membrane phases. Additionally, we quantified the impact of membrane phase separation on the efficiency with which liposomes transferred lipids and encapsulated macromolecules to cells, using a combination of confocal fluorescence imaging and flow cytometry. RESULTS Here we report that concentrating fusion-promoting lipids within phase-separated lipid domains on the surfaces of liposomes significantly increases the efficiency of liposome fusion with model membranes and cells. In particular, membrane phase separation enhanced the delivery of lipids and model macromolecules to the cytoplasm of tumor cells by at least 4-fold in comparison to homogenous liposomes. CONCLUSIONS Our findings demonstrate that phase separation can enhance membrane fusion by locally concentrating fusion-promoting lipids on the surface of liposomes. This work represents the first application of lipid membrane phase separation in the design of biomaterials-based delivery systems. Additionally, these results lay the ground work for developing fusogenic liposomes that are triggered by physical and molecular cues associated with target cells.
Collapse
Affiliation(s)
- Zachary I. Imam
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| | - Laura E. Kenyon
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| | - Grant Ashby
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| | - Fatema Nagib
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| | - Morgan Mendicino
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| | - Chi Zhao
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| | - Avinash K. Gadok
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| | - Jeanne C. Stachowiak
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| |
Collapse
|
30
|
Mondal G, Almawash S, Chaudhary AK, Mahato RI. EGFR-Targeted Cationic Polymeric Mixed Micelles for Codelivery of Gemcitabine and miR-205 for Treating Advanced Pancreatic Cancer. Mol Pharm 2017; 14:3121-3133. [PMID: 28719220 DOI: 10.1021/acs.molpharmaceut.7b00355] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gemcitabine (GEM), a first-line chemotherapy for pancreatic cancer undergoes rapid metabolism and develops chemoresistance after repeated administration. We previously demonstrated that the combination of GEM and miR-205 provides an effective therapeutic strategy to sensitize GEM-resistant pancreatic cancer cells. Since epidermal growth factor receptor (EGFR) is overexpressed in pancreatic cancer cells, in this study, we aimed to deliver mixed micelles containing GEM and miR-205 decorated with EGFR-targeting cetuximab (C225) monoclonal antibody for targeted therapy. Cetuximab C225 was conjugated to malemido-poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate-graft-dodecanol (C225-PEG-PCD) to prepare mixed micelles with mPEG-b-PCC-g-GEM-g-DC-g-TEPA for targeted codelivery of GEM and miR-205. This mixed micelle formulation showed a significant enhancement in EGFR-mediated cellular uptake in GEM-resistant MIA PaCa-2R cells. Further, an enhanced tumor accumulation of C225-micelles conjugated with near-infrared fluorescent Cy7.5 dye and Dy677-labeled miR-205 in orthotopic pancreatic tumor bearing NSG mice was evident after systemic administration. In addition, inhibition of tumor growth was also observed with increased apoptosis and reduced EMT after treatment with C225-micelles containing GEM and miR-205. Therefore, we believe that the targeted delivery of GEM and miR-205 in combination could be a novel strategy for treating advanced pancreatic cancer.
Collapse
Affiliation(s)
- Goutam Mondal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Saud Almawash
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Amit Kumar Chaudhary
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| |
Collapse
|
31
|
Abu-Fayyad A, Nazzal S. Synthesis, physiochemical characterization, and in vitro antitumor activity of the amide and pH cleavable hydrazone conjugates of γ-tocotrienol isomer of vitamin E with methoxy-poly(ethylene) glycol. Int J Pharm 2017. [PMID: 28627454 DOI: 10.1016/j.ijpharm.2017.06.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The anticancer activity of water soluble methoxy polyethylene glycol (mPEG) derivatives of tocotrienol (T3) isomers of vitamin E was previously found to be reduced when compared to the parent free isomers. This could be due to the ester bond formation between the mPEG and the 6-OH group on the chroman moiety of the T3 isomer. To further investigate, the objectives of the current study were to (1) synthesize and characterize stable amide and cleavable hydrazone conjugates between mPEG and carbon-5 on the chroman moiety of T3, and (2) examine the cytotoxicity of the newly synthesized mPEG conjugates against breast (MCF-7 and MDA-MB-231) and pancreatic (BxPC-3 and PANC-1) cancer cells. Conjugates were synthesized by direct conjugation of succinyl chloride derivatives of mPEG to the α-tocopherol and γ-tocotrienol isomers of vitamin E, and were characterized by 1H NMR, FT-IR, and mass spectrometry. The micelles of the amide and hydrazone self-assembled conjugates were characterized for size, zeta, CMC, and stability at different pH media. The hydrolysis of the hydrazone conjugate was pH dependent with highest release at acidic (pH 5.5) conditions, whereas the amide conjugate was stable in all tested media. The amide conjugate nonetheless showed greater cytotoxicity than the hydrazone conjugate, which suggested that maintaining solubility and the presence of free 6-OH group are important for γ-T3 to exert anticancer activity in vitro. The results from the current study demonstrated the importance of considering the nature of the chemical bond between T3 and mPEG when designing functional ingredients for use in drug delivery.
Collapse
Affiliation(s)
- Ahmed Abu-Fayyad
- College of Health and Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Sami Nazzal
- College of Health and Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA; College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
32
|
Abu-Fayyad A, Nazzal S. Gemcitabine-vitamin E conjugates: Synthesis, characterization, entrapment into nanoemulsions, and in-vitro deamination and antitumor activity. Int J Pharm 2017. [PMID: 28627455 DOI: 10.1016/j.ijpharm.2017.06.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Gemcitabine is the first line therapy for pancreatic cancer. It is, however, extensively metabolized to the inactive form by deamination enzymatic reaction. Conjugation of gemcitabine with fatty acids on its 4-amino group was found to protect it from deamination deactivation reaction. The objective of the present study was to test the in-vitro anticancer activity of gemcitabine conjugated to the γ-tocotrienol isomer of vitamin E against pancreatic tumor cells. This objective was based on reported studies in which it was demonstrated that free tocotrienol isomers of vitamin E can potentiate the anticancer activity of gemcitabine. To accomplish this objective, a full synthesis scheme for gemcitabine conjugation to fatty acids (stearic and linoleic) and the tocopherol and tocotrienol isomers of vitamin E (α-T and γ-T3) was presented. The conjugates were characterized by 1H NMR and mass spectrometry analysis and tested for their susceptibility to deamination. Also discussed is the impact of entrapping the conjugates into nanoemulsions on the physiochemical properties of the delivery system and the in vitro anticancer activity of gemcitabine against Bx-PC-3 and PNAC-1 pancreatic cancer cells. In-vitro enzymatic deamination study showed that the γ-T3 conjugate of gemcitabine was least affected by deamination deactivation reaction when compared with the free and conjugated gemcitabine in solution. Furthermore, in-vitro cytotoxicity study demonstrated that entrapment of gemcitabine-lipid conjugates into nanoemulsions significantly enhanced their anticancer activity when compared to the free drug. It was concluded that conjugation to the γ-T3 isomer is a viable option for gemcitabine delivery and is worthy of further investigation.
Collapse
Affiliation(s)
- Ahmed Abu-Fayyad
- College of Health and Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Sami Nazzal
- College of Health and Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA; College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
33
|
O'Mary HL, Aldayel AM, Valdes SA, Naguib YW, Li X, Salvady K, Cui Z. Acid-Sensitive Sheddable PEGylated, Mannose-Modified Nanoparticles Increase the Delivery of Betamethasone to Chronic Inflammation Sites in a Mouse Model. Mol Pharm 2017; 14:1929-1937. [PMID: 28463518 DOI: 10.1021/acs.molpharmaceut.7b00024] [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] [Indexed: 12/15/2022]
Abstract
Inflammation is implicated in a host of chronic illnesses. Within these inflamed tissues, the pH of the microenvironment is decreased and immune cells, particularly macrophages, infiltrate the area. Additionally, the vascular integrity of these sites is altered with increased fenestrations between endothelial cells. These distinctive properties may be exploited to enhance targeted delivery of anti-inflammatory therapies. Using a mouse model of chronic inflammation, we previously showed that acid-sensitive sheddable PEGylation increases the distribution and retention of nanoparticles in chronic inflammation sites. Here we demonstrated that surface modification of the acid-sensitive sheddable PEGylated nanoparticles with mannose, a ligand to mannose receptors present in chronic inflammation sites, significantly increases the targeted delivery of the nanoparticles to these areas. Furthermore, we showed that the acid-sensitive sheddable PEGylated, mannose-modified nanoparticles are able to significantly increase the delivery of betamethasone-21-acetate (BA), a model anti-inflammatory compound, to chronic inflammation sites as compared to free BA. These results highlight the ability to engineer formulations to target chronic inflammation sites by exploiting the microenvironment of these regions.
Collapse
Affiliation(s)
- Hannah L O'Mary
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin , Austin, Texas 78712, United States
| | - Abdulaziz M Aldayel
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin , Austin, Texas 78712, United States
| | - Solange A Valdes
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin , Austin, Texas 78712, United States
| | - Youssef W Naguib
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin , Austin, Texas 78712, United States
| | - Xu Li
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin , Austin, Texas 78712, United States
| | - Karun Salvady
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin , Austin, Texas 78712, United States
| | - Zhengrong Cui
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin , Austin, Texas 78712, United States.,Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University , Hohhot, Inner Mongolia, China
| |
Collapse
|
34
|
Birhanu G, Javar HA, Seyedjafari E, Zandi-Karimi A. Nanotechnology for delivery of gemcitabine to treat pancreatic cancer. Biomed Pharmacother 2017; 88:635-643. [PMID: 28142120 DOI: 10.1016/j.biopha.2017.01.071] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/02/2017] [Accepted: 01/12/2017] [Indexed: 12/21/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most deadly and quickly fatal human cancers with a 5-year mortality rate close to 100%. Its prognosis is very poor, mainly because of its hostile biological behavior and late onset of symptoms for clinical diagnosis; these bring limitations on therapeutic interventions. Factors contributing for the difficulties in treating PC include: high rate of drug resistance, fast metastasis to different organs, poor prognosis and relapse of the tumor after therapy. After being approved by US FDA 1997, Gemcitabine (Gem) is the first line and the gold standard drug for all stages of advanced PC till now. However, its efficacy is unsatisfactory, mainly due to; its chemical instability and poor cellular uptake, resulting in an extremely short half-life and low bioavailability. To solve this drawbacks and increase the therapeutic outcome important progress has been achieved in the field of nanotechnology and offers a promising and effective alternative. This review mainly focus on the most commonly investigated nanoparticle (NP) delivery systems of Gem for PC treatment and the latest progresses achieved. Novel nanocarriers with better tumor targeting efficiencies and maximum treatment outcome to treat this deadly due are given much attention.
Collapse
Affiliation(s)
- Gebremariam Birhanu
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, International Campus (TUMS-IC), Tehran, Iran; School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Hamid Akbari Javar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | - Ali Zandi-Karimi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
35
|
Abstract
Lipid-drug conjugates (LDCs) are drug molecules that have been covalently modified with lipids. The conjugation of lipids to drug molecules increases lipophilicity and also changes other properties of drugs. The conjugates demonstrate several advantages including improved oral bioavailability, improved targeting to the lymphatic system, enhanced tumor targeting, and reduced toxicity. Based on the chemical nature of drugs and lipids, various conjugation strategies and chemical linkers can be utilized to synthesize LDCs. Linkers and/or conjugation methods determine how drugs are released from LDCs and are critical for the optimal performance of LDCs. In this review, different lipids used for preparing LDCs and various conjugation strategies are summarized. Although LDCs can be administered without a delivery carrier, most of them are loaded into appropriate delivery systems. The lipid moiety in the conjugates can significantly enhance drug loading into hydrophobic components of delivery carriers and thus generate formulations with high drug loading and superior stability. Different delivery carriers such as emulsions, liposomes, micelles, lipid nanoparticles, and polymer nanoparticles are also discussed in this review.
Collapse
Affiliation(s)
- Danielle Irby
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University , Hampton, Virginia 23668, United States
| | - Chengan Du
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University , Hampton, Virginia 23668, United States
| | - Feng Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University , Hampton, Virginia 23668, United States
| |
Collapse
|
36
|
Liu J, Xu H, Tang X, Xu J, Jin Z, Li H, Wang S, Gou J, Jin X. Simple and tunable surface coatings via polydopamine for modulating pharmacokinetics, cell uptake and biodistribution of polymeric nanoparticles. RSC Adv 2017. [DOI: 10.1039/c7ra01354j] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A strategy that can modulate biological response such as pharmacokinetics, cell uptake and biodistribution of NPs simply by tunable coatings was established.
Collapse
Affiliation(s)
- Jingshuo Liu
- Department of Pharmaceutics
- College of Pharmacy Sciences
- Jilin University
- Changchun 130021
- China
| | - Hui Xu
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Xing Tang
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Jinghua Xu
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Zeng Jin
- Department of Pharmacology
- University of Alberta
- Edmonton
- Canada
| | - Hui Li
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Shihan Wang
- Department of Pharmaceutics
- College of Pharmacy Sciences
- Jilin University
- Changchun 130021
- China
| | - Jingxin Gou
- Department of Pharmaceutics
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
| | - Xiangqun Jin
- Department of Pharmaceutics
- College of Pharmacy Sciences
- Jilin University
- Changchun 130021
- China
| |
Collapse
|
37
|
Villamil Giraldo AM, Fyrner T, Wennmalm S, Parikh AN, Öllinger K, Ederth T. Spontaneous Vesiculation and pH-Induced Disassembly of a Lysosomotropic Detergent: Impacts on Lysosomotropism and Lysosomal Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:13566-13575. [PMID: 27936755 DOI: 10.1021/acs.langmuir.6b03458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Lysosomotropic detergents (LDs) selectively rupture lysosomal membranes through mechanisms that have yet to be characterized. A consensus view, currently, holds that LDs, which are weakly basic, diffuse across cellular membranes as monomers in an uncharged state, and via protonation in the acidic lysosomal compartment, they become trapped, accumulate, and subsequently solubilize the membrane and induce lysosomal membrane permeabilization. Here we demonstrate that the lysosomotropic detergent O-methyl-serine dodecylamide hydrochloride (MSDH) spontaneously assembles into vesicles at, and above, cytosolic pH, and that the vesicles disassemble as the pH reaches 6.4 or lower. The aggregation commences at concentrations below the range of those used in cell studies. Assembly and disassembly of the vesicles was studied via dynamic light scattering, zeta potential measurements, cryo-TEM, and fluorescence correlation spectroscopy and was found to be reversible via control of the pH. Aggregation of MSDH into closed vesicles under cytosolic conditions is at variance with the commonly held view of LD behavior, and we propose that endocytotic pathways should be considered as possible routes of LD entry into lysosomes. We further demonstrate that MSDH vesicles can be loaded with fluorophores via a solution transition from low to high pH, for subsequent release when the pH is lowered again. The ability to encapsulate molecular cargo into MSDH vesicles together with its ability to disaggregate at low pH and to permeabilize the lysosomal membrane presents an intriguing possibility to use MSDH as a delivery system.
Collapse
Affiliation(s)
- Ana M Villamil Giraldo
- Experimental Pathology, Department of Clinical and Experimental Medicine, Linköping, University , SE-581 85 Linköping, Sweden
| | | | - Stefan Wennmalm
- Royal Institute of Technology, Department of Applied Physics, Experimental Biomolecular Physics, Scilifelab , 171 65 Solna, Sweden
| | - Atul N Parikh
- Departments of Biomedical Engineering and Materials Science & Engineering, University of California , Davis, California 95616, United States
| | - Karin Öllinger
- Experimental Pathology, Department of Clinical and Experimental Medicine, Linköping, University , SE-581 85 Linköping, Sweden
| | | |
Collapse
|
38
|
Naguib YW, Lansakara-P D, Lashinger LM, Rodriguez BL, Valdes S, Niu M, Aldayel AM, Peng L, Hursting SD, Cui Z. Synthesis, Characterization, and In Vitro and In Vivo Evaluations of 4-(N)-Docosahexaenoyl 2', 2'-Difluorodeoxycytidine with Potent and Broad-Spectrum Antitumor Activity. Neoplasia 2016; 18:33-48. [PMID: 26806350 PMCID: PMC5965255 DOI: 10.1016/j.neo.2015.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 12/28/2022] Open
Abstract
In this study, a new compound, 4-(N)-docosahexaenoyl 2′, 2′-difluorodeoxycytidine (DHA-dFdC), was synthesized and characterized. Its antitumor activity was evaluated in cell culture and in mouse models of pancreatic cancer. DHA-dFdC is a poorly soluble, pale yellow waxy solid, with a molecular mass of 573.3 Da and a melting point of about 96°C. The activation energy for the degradation of DHA-dFdC in an aqueous Tween 80–based solution is 12.86 kcal/mol, whereas its stability is significantly higher in the presence of vitamin E. NCI-60 DTP Human Tumor Cell Line Screening revealed that DHA-dFdC has potent and broad-spectrum antitumor activity, especially in leukemia, renal, and central nervous system cancer cell lines. In human and murine pancreatic cancer cell lines, the IC50 value of DHA-dFdC was up to 105-fold lower than that of dFdC. The elimination of DHA-dFdC in mouse plasma appeared to follow a biexponential model, with a terminal phase t1/2 of about 58 minutes. DHA-dFdC significantly extended the survival of genetically engineered mice that spontaneously develop pancreatic ductal adenocarcinoma. In nude mice with subcutaneously implanted human Panc-1 pancreatic tumors, the antitumor activity of DHA-dFdC was significantly stronger than the molar equivalent of dFdC alone, DHA alone, or the physical mixture of them (1:1, molar ratio). DHA-dFdC also significantly inhibited the growth of Panc-1 tumors orthotopically implanted in the pancreas of nude mice, whereas the molar equivalent dose of dFdC alone did not show any significant activity. DHA-dFdC is a promising compound for the potential treatment of cancers in organs such as the pancreas.
Collapse
Affiliation(s)
- Youssef W Naguib
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Dharmika Lansakara-P
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Laura M Lashinger
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712
| | - B Leticia Rodriguez
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Solange Valdes
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Mengmeng Niu
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Abdulaziz M Aldayel
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Lan Peng
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Stephen D Hursting
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599
| | - Zhengrong Cui
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712.
| |
Collapse
|
39
|
Giannotti MI, Abasolo I, Oliva M, Andrade F, García-Aranda N, Melgarejo M, Pulido D, Corchero JL, Fernández Y, Villaverde A, Royo M, García-Parajo MF, Sanz F, Schwartz S. Highly Versatile Polyelectrolyte Complexes for Improving the Enzyme Replacement Therapy of Lysosomal Storage Disorders. ACS APPLIED MATERIALS & INTERFACES 2016; 8:25741-25752. [PMID: 27610822 DOI: 10.1021/acsami.6b08356] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Lysosomal storage disorders are currently treated by enzyme replacement therapy (ERT) through the direct administration of the unprotected recombinant protein to the patients. Herein we present an ionically cross-linked polyelectrolyte complex (PEC) composed of trimethyl chitosan (TMC) and α-galactosidase A (GLA), the defective enzyme in Fabry disease, with the capability of directly targeting endothelial cells by incorporating peptide ligands containing the RGD sequence. We assessed the physicochemical properties, cytotoxicity, and hemocompatibility of RGD-targeted and untargeted PECs, the uptake by endothelial cells and the intracellular activity of PECs in cell culture models of Fabry disease. Moreover, we also explored the effect of different freeze-drying procedures in the overall activity of the PECs. Our results indicate that the use of integrin-binding RGD moiety within the PEC increases their uptake and the efficacy of the GLA enzyme, while the freeze-drying allows the activity of the therapeutic protein to remain intact. Overall, these results highlight the potential of TMC-based PECs as a highly versatile and feasible drug delivery system for improving the ERT of lysosomal storage disorders.
Collapse
Affiliation(s)
- Marina I Giannotti
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Physical Chemistry Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Ibane Abasolo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Mireia Oliva
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Pharmacy and Pharmaceutical Technology Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Fernanda Andrade
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Pharmacy and Pharmaceutical Technology Department, Universitat de Barcelona , 08028 Barcelona, Spain
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto , 4050-313 Porto, Portugal
| | - Natalia García-Aranda
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Marta Melgarejo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Daniel Pulido
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - José L Corchero
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - Yolanda Fernández
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| | - Antonio Villaverde
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona , Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - Miriam Royo
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Combinatorial Chemistry Unit, Barcelona Science Park , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - María F García-Parajo
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology , ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Fausto Sanz
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- Nanoprobes & Nanoswitches, Institute for Bioengineering of Catalonia (IBEC) , Baldiri Reixac 10, 08028 Barcelona, Spain
- Physical Chemistry Department, Universitat de Barcelona , 08028 Barcelona, Spain
| | - Simó Schwartz
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , 28029 Madrid, Spain
- CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona , 08035 Barcelona, Spain
| |
Collapse
|
40
|
Dubey RD, Saneja A, Gupta PK, Gupta PN. Recent advances in drug delivery strategies for improved therapeutic efficacy of gemcitabine. Eur J Pharm Sci 2016; 93:147-62. [PMID: 27531553 DOI: 10.1016/j.ejps.2016.08.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 02/07/2023]
Abstract
Gemcitabine (2',2'-difluoro-2'-deoxycytidine; dFdC) is an efficacious anticancer agent acting against a wide range of solid tumors, including pancreatic, non-small cell lung, bladder, breast, ovarian, thyroid and multiple myelomas. However, short plasma half-life due to metabolism by cytidine deaminase necessitates administration of high dose, which limits its medical applicability. Further, due to its hydrophilic nature, it cannot traverse cell membranes by passive diffusion and, therefore, enters via nucleoside transporters that may lead to drug resistance. To circumvent these limitations, macromolecular prodrugs and nanocarrier-based formulations of Gemcitabine are gaining wide recognition. The nanoformulations based approaches by virtue of their controlled release and targeted delivery have proved to improve bioavailability, increase therapeutic efficacy and reduce adverse effects of the drug. Furthermore, the combination of Gemcitabine with other anticancer agents as well as siRNAs using nanocarriers has also been investigated in order to enhance its therapeutic potential. This review deals with challenges and recent advances in the delivery of Gemcitabine with particular emphasis on macromolecular prodrugs and nanomedicines.
Collapse
Affiliation(s)
- Ravindra Dhar Dubey
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India
| | - Ankit Saneja
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India
| | - Prasoon K Gupta
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India.
| | - Prem N Gupta
- Formulation & Drug Delivery Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi 180001, India.
| |
Collapse
|
41
|
Acid-Sensitive Sheddable PEGylated PLGA Nanoparticles Increase the Delivery of TNF-α siRNA in Chronic Inflammation Sites. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e340. [PMID: 27434685 PMCID: PMC5330937 DOI: 10.1038/mtna.2016.39] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/09/2016] [Indexed: 02/04/2023]
Abstract
There has been growing interest in utilizing small interfering RNA (siRNA) specific to pro-inflammatory cytokines, such as tumor necrosis factor-α ( TNF-α), in chronic inflammation therapy. However, delivery systems that can increase the distribution of the siRNA in chronic inflammation sites after intravenous administration are needed. Herein we report that innovative functionalization of the surface of siRNA-incorporated poly (lactic-co-glycolic) acid (PLGA) nanoparticles significantly increases the delivery of the siRNA in the chronic inflammation sites in a mouse model. The TNF-α siRNA incorporated PLGA nanoparticles were prepared by the standard double emulsion method, but using stearoyl-hydrazone-polyethylene glycol 2000, a unique acid-sensitive surface active agent, as the emulsifying agent, which renders (i) the nanoparticles PEGylated and (ii) the PEGylation sheddable in low pH environment such as that in chronic inflammation sites. In a mouse model of lipopolysaccharide-induced chronic inflammation, the acid-sensitive sheddable PEGylated PLGA nanoparticles showed significantly higher accumulation or distribution in chronic inflammation sites than PLGA nanoparticles prepared with an acid-insensitive emulsifying agent (i.e., stearoyl-amide-polyethylene glycol 2000) and significantly increased the distribution of the TNF-α siRNA incorporated into the nanoparticles in inflamed mouse foot.
Collapse
|
42
|
Chi X, Zhang H, Vargas-Zúñiga GI, Peters GM, Sessler JL. A Dual-Responsive Bola-Type Supra-amphiphile Constructed from a Water-Soluble Calix[4]pyrrole and a Tetraphenylethene-Containing Pyridine Bis-N-oxide. J Am Chem Soc 2016; 138:5829-32. [PMID: 27123813 DOI: 10.1021/jacs.6b03159] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Complexation between a water-soluble calix[4]pyrrole and a ditopic pyridine N-oxide derivative in aqueous media produces a bola-type supra-amphiphile that self-assembles to produce higher order morphologies, including multilamellar vesicles and micelles depending on the pH. The present bola-type supra-amphiphile exhibits strong fluorescence due to structural changes and aggregation induced by host-guest complexation. The resulting structures may be used to recognize, encapsulate, and release non-fluorescent, water-soluble small molecules.
Collapse
Affiliation(s)
- Xiaodong Chi
- Department of Chemistry, The University of Texas at Austin , 105 East 24th Street, Stop A5300, Austin, Texas 78712-1224, United States
| | - Huacheng Zhang
- Department of Chemistry, The University of Texas at Austin , 105 East 24th Street, Stop A5300, Austin, Texas 78712-1224, United States
| | - Gabriela I Vargas-Zúñiga
- Department of Chemistry, The University of Texas at Austin , 105 East 24th Street, Stop A5300, Austin, Texas 78712-1224, United States
| | - Gretchen M Peters
- Department of Chemistry, The University of Texas at Austin , 105 East 24th Street, Stop A5300, Austin, Texas 78712-1224, United States
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin , 105 East 24th Street, Stop A5300, Austin, Texas 78712-1224, United States
| |
Collapse
|
43
|
Palao-Suay R, Gómez-Mascaraque L, Aguilar M, Vázquez-Lasa B, Román JS. Self-assembling polymer systems for advanced treatment of cancer and inflammation. Prog Polym Sci 2016. [DOI: 10.1016/j.progpolymsci.2015.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
44
|
cRGDyK modified pH responsive nanoparticles for specific intracellular delivery of doxorubicin. Acta Biomater 2016; 30:285-298. [PMID: 26602824 DOI: 10.1016/j.actbio.2015.11.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 12/16/2022]
Abstract
Stimuli-responsive nanocarriers attract wide attention because of the unique differences in microenvironment between solid tumors and normal tissues. Herein, we reported a novel cRGDyK peptide modified pH-sensitive nanoparticle system based on poly(ethylene glycol)-poly(2,4,6-trimethoxy benzylidene-pentaerythritol carbonate) (PEG-PTMBPEC) diblock copolymer, which was expected to destroy tumor angiogenesis and kill tumor cells simultaneously. Doxorubicin (DOX)-loaded nanoparticles (NPs) were characterized to have a uniform size distribution, high entrapment efficiency, good stability in plasma as well as a pH dependent drug release pattern. Blank NPs were non-toxic to both tumor cells and normal cells, while DOX-loaded cRGDyK peptide modified NPs (cRGDyK-NPs) exhibited the pronounced cytotoxicity against B16 cells and human umbilical vein endothelial cells (HUVEC) overexpressing αvβ3 integrin receptors. Cellular uptake studies revealed that the highly efficient uptake of cRGDyK-NPs was attributed to the receptor-mediated endocytosis and acidic-triggered drug release. Importantly, cRGDyK-NPs could dramatically reduce the systemic toxicity of DOX and exert excellent tumor killing activity in vivo. The cRGDyK modified pH-sensitive nanocarrier is a promising vehicle for intracellular drug delivery to αvβ3 integrin receptor overexpressed tumor cells and neovascular cells. STATEMENT OF SIGNIFICANCE Slow intracellular drug release and poor tumor targeting capacity are still the critical barriers of polymeric nanoparticles (NPs) for the treatment efficiency of chemotherapy. In the present study, we designed cRGDyK peptide modified poly(ethylene glycol)-poly(2,4,6-trimethoxybenzylidene-pentaerythritol carbonate) (cRGDyK-PEG-PTMBPEC) NPs with active targeting and fast pH-triggered drug release. Doxorubicin (DOX)-loaded cRGDyK-PEG-PTMBPEC NPs exhibited pronounced cytotoxicity and enhanced cellular uptake against B16 cells and human umbilical vein endothelial cells overexpressing αvβ3 integrin receptors. Moreover, the active targeted pH-sensitive NPs can enhance the antitumor activity and reduce the systematic toxicity of DOX in vivo.
Collapse
|
45
|
Hung BY, Kuthati Y, Kankala RK, Kankala S, Deng JP, Liu CL, Lee CH. Utilization of Enzyme-Immobilized Mesoporous Silica Nanocontainers (IBN-4) in Prodrug-Activated Cancer Theranostics. NANOMATERIALS (BASEL, SWITZERLAND) 2015; 5:2169-2191. [PMID: 28347114 PMCID: PMC5304787 DOI: 10.3390/nano5042169] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/26/2015] [Indexed: 12/12/2022]
Abstract
To develop a carrier for use in enzyme prodrug therapy, Horseradish peroxidase (HRP) was immobilized onto mesoporous silica nanoparticles (IBN-4: Institute of Bioengineering and Nanotechnology), where the nanoparticle surfaces were functionalized with 3-aminopropyltrimethoxysilane and further conjugated with glutaraldehyde. Consequently, the enzymes could be stabilized in nanochannels through the formation of covalent imine bonds. This strategy was used to protect HRP from immune exclusion, degradation and denaturation under biological conditions. Furthermore, immobilization of HRP in the nanochannels of IBN-4 nanomaterials exhibited good functional stability upon repetitive use and long-term storage (60 days) at 4 °C. The generation of functionalized and HRP-immobilized nanomaterials was further verified using various characterization techniques. The possibility of using HRP-encapsulated IBN-4 materials in prodrug cancer therapy was also demonstrated by evaluating their ability to convert a prodrug (indole-3- acetic acid (IAA)) into cytotoxic radicals, which triggered tumor cell apoptosis in human colon carcinoma (HT-29 cell line) cells. A lactate dehydrogenase (LDH) assay revealed that cells could be exposed to the IBN-4 nanocomposites without damaging their membranes, confirming apoptotic cell death. In summary, we demonstrated the potential of utilizing large porous mesoporous silica nanomaterials (IBN-4) as enzyme carriers for prodrug therapy.
Collapse
Affiliation(s)
- Bau-Yen Hung
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien-974, Taiwan.
| | - Yaswanth Kuthati
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien-974, Taiwan.
| | - Ranjith Kumar Kankala
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien-974, Taiwan.
| | | | - Jin-Pei Deng
- Department of Chemistry, Tamkang University, New Taipei City 251, Taiwan.
| | - Chen-Lun Liu
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien-974, Taiwan.
| | - Chia-Hung Lee
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien-974, Taiwan.
| |
Collapse
|
46
|
Utama RH, Jiang Y, Zetterlund PB, Stenzel MH. Biocompatible Glycopolymer Nanocapsules via Inverse Miniemulsion Periphery RAFT Polymerization for the Delivery of Gemcitabine. Biomacromolecules 2015; 16:2144-56. [PMID: 26027950 DOI: 10.1021/acs.biomac.5b00545] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Encapsulation of hydrophilic cancer drugs in polymeric nanocapsules was achieved in a one-pot process via the inverse miniemulsion periphery RAFT polymerization (IMEPP) approach. The chosen guest molecule was gemcitabine hydrochloride, which is used as the first-line treatment of pancreatic cancer. The resulting nanocapsules were confirmed to be ∼200 nm, with excellent encapsulation (∼96%) and loading (∼12%) efficiency. Postpolymerization reaction was successfully conducted to create glyocopolymer nanocapsules without any impact on the loads as well as the nanocapsules size or morphology. The loaded nanocapsules were specifically designed to be responsive in a reductive environment. This was confirmed by the successful disintegration of the nanocapsules in the presence of glutathione. The gemcitabine-loaded nanocapsules were tested in vitro against pancreatic cancer cells (AsPC-1), with the results showing an enhancement in the cytotoxicity by two fold due to selective accumulation and release of the nanocapsules within the cells. The results demonstrated the versatility of IMEPP as a tool to synthesize functionalized, loaded-polymeric nanocapsules suitable for drug-delivery application.
Collapse
Affiliation(s)
- Robert H Utama
- ‡Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney 2052, Australia
| | - Yanyan Jiang
- †Centre for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney 2052, Australia.,‡Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney 2052, Australia
| | - Per B Zetterlund
- †Centre for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney 2052, Australia
| | - Martina H Stenzel
- †Centre for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney 2052, Australia.,‡Centre for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
47
|
Wexselblatt E, Esko JD, Tor Y. GNeosomes: Highly Lysosomotropic Nanoassemblies for Lysosomal Delivery. ACS NANO 2015; 9:3961-3968. [PMID: 25831231 DOI: 10.1021/nn507382n] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
GNeosomes, lysosomotropic lipid vesicles decorated with guanidinoneomycin, can encapsulate and facilitate the cellular internalization and lysosomal delivery of cargo ranging from small molecules to high molecular weight proteins, in a process that is exclusively dependent on cell surface glycosaminoglycans. Their cellular uptake mechanism and co-localization with lysosomes, as well as the delivery, release, and activity of internalized cargo, are quantified. GNeosomes are proposed as a universal platform for lysosomal delivery with potential as a basic research tool and a therapeutic vehicle.
Collapse
Affiliation(s)
- Ezequiel Wexselblatt
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jeffrey D Esko
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yitzhak Tor
- †Chemistry and Biochemistry and ‡Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
48
|
Mura S, Bui DT, Couvreur P, Nicolas J. Lipid prodrug nanocarriers in cancer therapy. J Control Release 2015; 208:25-41. [PMID: 25617724 DOI: 10.1016/j.jconrel.2015.01.021] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/12/2015] [Accepted: 01/20/2015] [Indexed: 10/24/2022]
Abstract
Application of nanotechnology in the medical field (i.e., nanomedicine) plays an important role in the development of novel drug delivery methods. Nanoscale drug delivery systems can indeed be customized with specific functionalities in order to improve the efficacy of the treatments. However, despite the progresses of the last decades, nanomedicines still face important obstacles related to: (i) the physico-chemical properties of the drug moieties which may reduce the total amount of loaded drug; (ii) the rapid and uncontrolled release (i.e., burst release) of the encapsulated drug after administration and (iii) the instability of the drug in biological media where a fast transformation into inactive metabolites can occur. As an alternative strategy to alleviate these drawbacks, the prodrug approach has found wide application. The covalent modification of a drug molecule into an inactive precursor from which the drug will be freed after administration offers several benefits such as: (i) a sustained drug release (mediated by chemical or enzymatic hydrolysis of the linkage between the drug-moiety and its promoiety); (ii) an increase of the drug chemical stability and solubility and, (iii) a reduced toxicity before the metabolization occurs. Lipids have been widely used as building blocks for the design of various prodrugs. Interestingly enough, these lipid-derivatized drugs can be delivered through a nanoparticulate form due to their ability to self-assemble and/or to be incorporated into lipid/polymer matrices. Among the several prodrugs developed so far, this review will focus on the main achievements in the field of lipid-based prodrug nanocarriers designed to improve the efficacy of anticancer drugs. Gemcitabine (Pubchem CID: 60750); 5-fluorouracil (Pubchem CID: 3385); Doxorubicin (Pubchem CID: 31703); Docetaxel (Pubchem CID: 148124); Methotrexate (Pubchem CID: 126941); Paclitaxel (Pubchem CID: 36314).
Collapse
Affiliation(s)
- Simona Mura
- Institut Galien Paris-Sud, UMR CNRS 8612, Univ Paris-Sud, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France.
| | - Duc Trung Bui
- Institut Galien Paris-Sud, UMR CNRS 8612, Univ Paris-Sud, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMR CNRS 8612, Univ Paris-Sud, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France
| | - Julien Nicolas
- Institut Galien Paris-Sud, UMR CNRS 8612, Univ Paris-Sud, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France.
| |
Collapse
|
49
|
Ji X, Li Y, Wang H, Zhao R, Tang G, Huang F. Facile construction of fluorescent polymeric aggregates with various morphologies by self-assembly of supramolecular amphiphilic graft copolymers. Polym Chem 2015. [DOI: 10.1039/c5py00801h] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Facile construction of fluorescent polymeric aggregates with various morphologies was realized by self-assembly of supramolecular amphiphilic graft copolymers.
Collapse
Affiliation(s)
- Xiaofan Ji
- State Key Laboratory of Chemical Engineering
- Center for Chemistry of High-Performance & Novel Materials
- Department of Chemistry
- Zhejiang University
- 310027 Hangzhou
| | - Yang Li
- Department of Chemistry
- Institute of Chemical Biology and Pharmaceutical Chemistry
- Zhejiang University
- Hangzhou 310027
- P. R. China
| | - Hu Wang
- State Key Laboratory of Chemical Engineering
- Center for Chemistry of High-Performance & Novel Materials
- Department of Chemistry
- Zhejiang University
- 310027 Hangzhou
| | - Run Zhao
- State Key Laboratory of Chemical Engineering
- Center for Chemistry of High-Performance & Novel Materials
- Department of Chemistry
- Zhejiang University
- 310027 Hangzhou
| | - Guping Tang
- Department of Chemistry
- Institute of Chemical Biology and Pharmaceutical Chemistry
- Zhejiang University
- Hangzhou 310027
- P. R. China
| | - Feihe Huang
- State Key Laboratory of Chemical Engineering
- Center for Chemistry of High-Performance & Novel Materials
- Department of Chemistry
- Zhejiang University
- 310027 Hangzhou
| |
Collapse
|
50
|
Recent advances in targeted nanoparticles drug delivery to melanoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:769-94. [PMID: 25555352 DOI: 10.1016/j.nano.2014.11.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/31/2014] [Accepted: 11/15/2014] [Indexed: 12/30/2022]
Abstract
Melanoma is one of the most aggressive skin cancers, notorious for its high multidrug resistance and low survival rate. Conventional therapies (e.g., dacarbazine, interferon-alpha-2b and interleukin-2) are limited by low response rate and demonstrate no overall survival benefit. Novel targeted therapies (e.g., vemurafenib, dabrafenib and trametinib) have higher initial response rate and clear impact on the overall survival, but relapse usually occurs within 6 to 9 months. Although immunotherapy (e.g., ipilimumab, pembrolizumab and nivolumab) can achieve long-term and durable response, rate of adverse events is extremely high. With the development of nanotechnology, the applications of nanocarriers are widely expected to change the landscape of melanoma therapy for foreseeable future. In this review, we will relate recent advances in the application of multifunctional nanocarriers for targeted drug delivery to melanoma, in melanoma nanotheranostics and combination therapy, and nanopharmaceutical associated melanoma clinical trials, followed by challenges and perspectives. From the clinical editor: The team of authors describes the current treatment regimes of malignant melanoma emphasizing the importance of achieving a better efficacy and the need to develop a better understanding of melanoma tumorigenesis.
Collapse
|