1
|
Huang YH, Lu YL, Cao ZM, Zhang XD, Liu CH, Xu HS, Su CY. Multipocket Cage Enables the Binding of High-Order Bulky and Drug Guests Uncovered by MS Methodology. J Am Chem Soc 2024; 146:21677-21688. [PMID: 39042557 DOI: 10.1021/jacs.4c05758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Achieving high guest loading and multiguest-binding capacity holds crucial significance for advancement in separation, catalysis, and drug delivery with synthetic receptors; however, it remains a challenging bottleneck in characterization of high-stoichiometry guest-binding events. Herein, we describe a large-sized coordination cage (MOC-70-Zn8Pd6) possessing 12 peripheral pockets capable of accommodating multiple guests and a high-resolution electrospray ionization mass spectrometry (HR-ESI-MS)-based method to understand the solution host-guest chemistry. A diverse range of bulky guests, varying from drug molecules to rigid fullerenes as well as flexible host molecules of crown ethers and calixarenes, could be loaded into open pockets with high capacities. Notably, these hollow cage pockets provide multisites to capture different guests, showing heteroguest coloading behavior to capture binary, ternary, or even quaternary guests. Moreover, a pair of commercially applied drugs for the combination therapy of chronic lymphocytic leukemia (CLL) has been tested, highlighting its potential in multidrug delivery for combined treatment.
Collapse
Affiliation(s)
- Yin-Hui Huang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, GBRCE for Functional Molecular Engineering, LIFM, IGCME, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu-Lin Lu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, GBRCE for Functional Molecular Engineering, LIFM, IGCME, Sun Yat-Sen University, Guangzhou 510275, China
| | - Zhong-Min Cao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, GBRCE for Functional Molecular Engineering, LIFM, IGCME, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xiao-Dong Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, GBRCE for Functional Molecular Engineering, LIFM, IGCME, Sun Yat-Sen University, Guangzhou 510275, China
| | - Chen-Hui Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, GBRCE for Functional Molecular Engineering, LIFM, IGCME, Sun Yat-Sen University, Guangzhou 510275, China
| | - Hai-Sen Xu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, GBRCE for Functional Molecular Engineering, LIFM, IGCME, Sun Yat-Sen University, Guangzhou 510275, China
| | - Cheng-Yong Su
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, GBRCE for Functional Molecular Engineering, LIFM, IGCME, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
2
|
Jogdeo CM, Panja S, Kanvinde S, Kapoor E, Siddhanta K, Oupický D. Advances in Lipid-Based Codelivery Systems for Cancer and Inflammatory Diseases. Adv Healthc Mater 2023; 12:e2202400. [PMID: 36453542 PMCID: PMC10023350 DOI: 10.1002/adhm.202202400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/13/2022] [Indexed: 12/03/2022]
Abstract
Combination therapy targeting multiple therapeutic targets is a favorable strategy to achieve better therapeutic outcomes in cancer and inflammatory diseases. Codelivery is a subfield of drug delivery that aims to achieve combined delivery of diverse therapeutic cargoes within the same delivery system, thereby ensuring delivery to the same site and providing an opportunity to tailor the release kinetics as desired. Among the wide range of materials being investigated in the design of codelivery systems, lipids have stood out on account of their low toxicity, biocompatibility, and ease of formulation scale-up. This review highlights the advances of the last decade in lipid-based codelivery systems focusing on the codelivery of drug-drug, drug-nucleic acid, nucleic acid-nucleic acid, and protein therapeutic-based combinations for targeted therapy in cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Chinmay M. Jogdeo
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Sudipta Panja
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Shrey Kanvinde
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Ekta Kapoor
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - David Oupický
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| |
Collapse
|
3
|
Mallick AM, Tripathi A, Mishra S, Mukherjee A, Dutta C, Chatterjee A, Sinha Roy R. Emerging Approaches for Enabling RNAi Therapeutics. Chem Asian J 2022; 17:e202200451. [PMID: 35689534 DOI: 10.1002/asia.202200451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/04/2022] [Indexed: 11/07/2022]
Abstract
RNA interference (RNAi) is a primitive evolutionary mechanism developed to escape incorporation of foreign genetic material. siRNA has been instrumental in achieving the therapeutic potential of RNAi by theoretically silencing any gene of interest in a reversible and sequence-specific manner. Extrinsically administered siRNA generally needs a delivery vehicle to span across different physiological barriers and load into the RISC complex in the cytoplasm in its functional form to show its efficacy. This review discusses the designing principles and examples of different classes of delivery vehicles that have proved to be efficient in RNAi therapeutics. We also briefly discuss the role of RNAi therapeutics in genetic and rare diseases, epigenetic modifications, immunomodulation and combination modality to inch closer in creating a personalized therapy for metastatic cancer. At the end, we present, strategies and look into the opportunities to develop efficient delivery vehicles for RNAi which can be translated into clinics.
Collapse
Affiliation(s)
- Argha M Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Archana Tripathi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Sukumar Mishra
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Asmita Mukherjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Chiranjit Dutta
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.,Present address:Department of Biological Sciences, NUS Environmental Research Institute (NERI), National University of Singapore (NUS), Block S2 #05-01, 16 Science Drive 4, Singapore, 117558, Singapore
| | - Ananya Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Rituparna Sinha Roy
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.,Centre for Advanced Functional Materials, Indian Institute of Science Education and Research Kolkata, 741246, Mohanpur, India.,Centre for Climate and Environmental Studies, Indian Institute of Science Education and Research Kolkata, 741246, Mohanpur, India
| |
Collapse
|
4
|
Yu Y, Yang Q, Wang Z, Ding Q, Li M, Fang Y, He Q, Zhu YZ. The Anti-Inflammation and Anti-Nociception Effect of Ketoprofen in Rats Could Be Strengthened Through Co-Delivery of a H 2S Donor, S-Propargyl-Cysteine. J Inflamm Res 2021; 14:5863-5875. [PMID: 34785926 PMCID: PMC8590460 DOI: 10.2147/jir.s333326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/11/2021] [Indexed: 01/20/2023] Open
Abstract
PURPOSE Ketoprofen (KETO) is a traditional non-steroidal anti-inflammatory drug (NSAIDs) with good analgesic and antipyretic effects. However, as NASIDs, the toxicity of KETO towards gastrointestinal (GI) system might limit its clinical use. S-propargyl-cysteine (SPRC) is an excellent endogenous H2S donor showed wide application in the field of anti-inflammation, anti-oxidative stress, or even the protection of cardiovascular system through the elevation of endogenous H2S concentration. As recently studies reported, co-administration of H2S donor might potentially mitigate the GI toxicity and relevant side effects induced by series of NSAIDs. METHODS In this study, we established a SPRC and KETO co-encapsulated poly (lactic-co-glycolic acid) microsphere (SK@MS), and its particle size, morphology, storage stability and in vitro release profile were firstly investigated. The elevation of endogenous H2S level of SK@MS was then calculated, and the pharmacodynamic study (anti-inflammation and analgesic effects) of SK@MS, SPRC, and KETO towards adjuvant induced arthritis (AIA) in rats were also studied. Finally, to test the potential side effect, the heart, liver, spleen, lung, kidney, stomach, small intestine, and large intestine were resected from rats and examined by H&E staining. RESULTS A monodispersed SK@MS could be observed under the SEM, and particle size was calculated around 25.12 μm. The loading efficiency (LE) for SPRC and KETO were 6.67% and 2.64%, respectively, while the encapsulation efficiency (EE) for SPRC and KETO were 37.20% and 68.28%, respectively. SK@MS showed a sustained release of SPRC and KETO in vitro, which was up-to 15 days. SK@MS could achieve a long-term elevation of the H2S concentration in vivo, while SPRC showed an instant H2S elevation and metabolize within 6 h. Interestingly, the KETO did not show any influence on the H2S concentration in vivo. After establishment of AIA model, neither SPRC nor KETO showed scarcely anti-inflammation and anti-nociception effect, while conversely, SK@MS showed an obvious mitigation towards paw edema and pain in AIA rats, which indicated an improved anti-inflammation and anti-nociception effect when co-delivery of SRC and KETO. Besides, low stimulation towards major organs in rats observed in any experimental group. CONCLUSION A monodispersed was successfully prepared in this study, and SK@MS showed a sustained SPRC and KETO release in vitro and H2S release in vivo. In the pharmacodynamics study, SK@MS not only exhibited an excellent anti-inflammation and analgesic effects in AIA rats but also showed low stimulation towards rats.
Collapse
Affiliation(s)
- Yue Yu
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Qinyan Yang
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Zhou Wang
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Qian Ding
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Meng Li
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Yudong Fang
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Qida He
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Taipa, Macau SAR, People's Republic of China
- Shanghai Key Laboratory of Bioactive Small Molecules & School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
5
|
Multi-functional silica-based mesoporous materials for simultaneous delivery of biologically active ions and therapeutic biomolecules. Acta Biomater 2021; 129:1-17. [PMID: 34010692 DOI: 10.1016/j.actbio.2021.05.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
Abstract
Mesoporous silica-based materials, especially mesoporous bioactive glasses (MBGs), are being highly considered for biomedical applications, including drug delivery and tissue engineering, not only because of their bioactivity and biocompatibility but also due to their tunable composition and potential use as drug delivery carriers owing to their controllable nanoporous structure. Numerous researches have reported that MBGs can be doped with various therapeutic ions (strontium, copper, magnesium, zinc, lithium, silver, etc.) and loaded with specific biomolecules (e.g., therapeutic drugs, antibiotics, growth factors) achieving controllable loading and release kinetics. Therefore, co-delivery of ions and biomolecules using a single MBG carrier is highly interesting as this approach provides synergistic effects toward improved therapeutic outcomes in comparison to the strategy of sole drug or ion delivery. In this review, we discuss the state-of-the-art in the field of mesoporous silica-based materials used for co-delivery of ions and therapeutic drugs with osteogenesis/cementogenesis, angiogenesis, antibacterial and anticancer properties. The analysis of the literature reveals that specially designed mesoporous nanocarriers can release multiple ions and drugs at therapeutically safe and relevant levels, achieving the desired biological effects (in vivo, in vitro) for specific biomedical applications. It is expected that this review on the ion/drug co-delivery concept using MBG carriers will shed light on the advantages of such co-delivery systems for clinical use. Areas for future research directions are identified and discussed. STATEMENT OF SIGNIFICANCE: Many studies in literature focus on the potential of single drug or ion delivery by mesoporous silica-based materials, exploiting the bioactivity, biocompatibility, tunable composition and controllable nanoporosity of these materials. Recenlty, studies have adopted the "dual-delivery" concept, by designing multi-functional mesoporous silica-based systems which are capable to deliver both biologically active ions and biomolecules (growth factors, drugs) simultaneously in order to achieve synergy of their complementary therapeutic activities. This review summarizes the state of the art in the field, with focus on osteogenesis/cementogenesis, angiogenesis, antibacterial and anticancer properties, and discusses the challenges and prospects for further progress in this area, expecting to generate broader interest in the technology for applications in disease treatment and regenerative medicine.
Collapse
|
6
|
Biswas G, Jena BC, Samanta P, Mandal M, Dhara D. Synthesis, self-assembly and drug release study of a new dual-responsive biocompatible block copolymer containing phenylalanine derivative. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2021. [DOI: 10.1080/10601325.2021.1947748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Gargi Biswas
- Department of Chemistry, Indian Institute of Technology, Kharagpur, West Bengal, India
| | - Bikash Chandra Jena
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, West Bengal, India
| | - Pousali Samanta
- Department of Chemistry, Indian Institute of Technology, Kharagpur, West Bengal, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, West Bengal, India
| | - Dibakar Dhara
- Department of Chemistry, Indian Institute of Technology, Kharagpur, West Bengal, India
| |
Collapse
|
7
|
Aghamiri S, Raee P, Talaei S, Mohammadi-Yeganeh S, Bayat S, Rezaee D, Ghavidel AA, Teymouri A, Roshanzamiri S, Farhadi S, Ghanbarian H. Nonviral siRNA delivery systems for pancreatic cancer therapy. Biotechnol Bioeng 2021; 118:3669-3690. [PMID: 34170520 DOI: 10.1002/bit.27869] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022]
Abstract
The serious drawbacks of the conventional treatment of pancreatic ductal adenocarcinoma (PDAC) such as nonspecific toxicity and high resistance to chemo and radiation therapy, have prompted the development and application of countless small interfering RNA (siRNA)-based therapeutics. Recent advances in drug delivery systems hold great promise for improving siRNA-based therapeutics and developing a new class of drugs, known as nano-siRNA drugs. However, many fundamental questions, regarding toxicity, immunostimulation, and poor knowledge of nano-bio interactions, need to be addressed before clinical translation. In this review, we provide recent achievements in the design and development of various nonviral delivery vehicles for pancreatic cancer therapy. More importantly, codelivery of conventional anticancer drugs with siRNA as a new revolutionary pancreatic cancer combinational therapy is completely discussed.
Collapse
Affiliation(s)
- Shahin Aghamiri
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pourya Raee
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sam Talaei
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shiva Bayat
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Delsuz Rezaee
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin A Ghavidel
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Teymouri
- Department of Infectious Disease, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheil Roshanzamiri
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shohreh Farhadi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell SciencesSchool of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
DuRoss AN, Landry MR, Thomas CR, Neufeld MJ, Sun C. Fucoidan-coated nanoparticles target radiation-induced P-selectin to enhance chemoradiotherapy in murine colorectal cancer. Cancer Lett 2020; 500:208-219. [PMID: 33232787 DOI: 10.1016/j.canlet.2020.11.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death for both men and women, highlighting the need for new treatment strategies. Advanced disease is often treated with a combination of radiation and cytotoxic agents, such as DNA damage repair inhibitors and DNA damaging agents. To optimize the therapeutic window of these multimodal therapies, advanced nanomaterials have been investigated to deliver sensitizing agents or enhance local radiation dose deposition. In this study, we demonstrate the feasibility of employing an inflammation targeting nanoscale metal-organic framework (nMOF) platform to enhance CRC treatment. This novel formulation incorporates a fucoidan surface coating to preferentially target P-selectin, which is over-expressed or translocated in irradiated tumors. Using this radiation stimulated delivery strategy, a combination PARP inhibitor (talazoparib) and chemotherapeutic (temozolomide) drug-loaded hafnium and 1,4-dicarboxybenzene (Hf-BDC) nMOF was evaluated both in vitro and in vivo. Significantly, these drug-loaded P-selectin targeted nMOFs (TT@Hf-BDC-Fuco) show improved tumoral accumulation over multiple controls and subsequently enhanced therapeutic effects. The integrated radiation and nanoformulation treatment demonstrated improved tumor control (reduced volume, density, and growth rate) and increased survival in a syngeneic CRC mouse model. Overall, the data from this study support the continued investigation of radiation-priming for targeted drug delivery and further consideration of nanomedicine strategies in the clinical management of advanced CRC.
Collapse
Affiliation(s)
- Allison N DuRoss
- Department of Pharmaceutical Sciences, Oregon State University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Madeleine R Landry
- Department of Pharmaceutical Sciences, Oregon State University, 2730 S Moody Ave, Portland, OR, 97201, USA
| | - Charles R Thomas
- Department of Radiation Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Megan J Neufeld
- Department of Pharmaceutical Sciences, Oregon State University, 2730 S Moody Ave, Portland, OR, 97201, USA.
| | - Conroy Sun
- Department of Pharmaceutical Sciences, Oregon State University, 2730 S Moody Ave, Portland, OR, 97201, USA; Department of Radiation Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| |
Collapse
|
9
|
Mozafari N, Farjadian F, Mohammadi Samani S, Azadi S, Azadi A. Simvastatin-chitosan-citicoline conjugates nanoparticles as the co-delivery system in Alzheimer susceptible patients. Int J Biol Macromol 2019; 156:1396-1407. [PMID: 31760027 DOI: 10.1016/j.ijbiomac.2019.11.180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 12/17/2022]
Abstract
The main goal of this study was the preparation and characterization of a chitosan-based system for co-delivery of simvastatin and citicoline to overcome simvastatin unwanted side effects in Alzheimer's disease. This conjugated complex was synthesized in three steps, and 1HNMR, FTIR, and UV-Vis spectroscopy confirmed its success. The simvastatin conjugation rate to chitosan was 1.67 times more than citicoline. X-ray diffraction results showed that the crystalline property of both drugs converted to an amorphous state during the synthesis of the conjugated form. Further, SEM images revealed that the developed nanoparticles have a spherical shape with a size between 100 and 300 nm. Another characterization test was RBC hemolysis, with the lowest value at 6.04% and the highest value at 89.56% and became much lower after preparing nanoparticles using the ionotropic technique. TEM characterized the nanoparticles and showed that the gelation technique stabilized the particles.
Collapse
Affiliation(s)
- Negin Mozafari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soliman Mohammadi Samani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soha Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Azadi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
10
|
Assessment of unloaded polymeric nanocapsules with different coatings in female rats: Influence on toxicological and behavioral parameters. Biomed Pharmacother 2019; 121:109575. [PMID: 31689599 DOI: 10.1016/j.biopha.2019.109575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/09/2019] [Accepted: 10/21/2019] [Indexed: 11/20/2022] Open
Abstract
The unloaded polymeric nanocapsules (NCs) present incredible characteristics as drug carriers. However, the toxicity caused by NCs with different coatings is still a challenge for contemporary toxicology. Allied to this, preclinical studies are performed in males, disregarding possible gender-dependent toxicity. Thus, the aim of present study was to evaluate the influence of different NCs coatings on toxicological and behavioral parameters in female rats. The physicochemical characterization of NCs with different surface coatings: NC1 (Polysorbate 80), NC2 (PEG), NC3 (Eudragit®RS 100) and NC4 (Chitosan) were performed. Female rats were treated with saline, NC1, NC2, NC3 or NC4 daily for 14 days, p.o. After 24 h of last treatment, animals were submitted to behavioral tests. Only after behavioral tests, female rats were euthanized, organs were removed and weighted. After, histopathological, biochemical and oxidative stress analysis were performed. All NCs-coatings did not cause alterations in behavioral tests. For markers of hepatic, renal damage and lipid profile, the different coatings showed a low toxicity. NCs did not alter weight of organs and histopathological analysis. Also, all NCs-coatings did not modify redox balance in organs studied, only NC2 induced a increase of FRAP levels in intestine. This study demonstrated that the different NCs-coatings did not cause behavioral changes and showed a low toxicity in female rats.
Collapse
|
11
|
Arroyo N, Herlem G, Picaud F. Ligand nanovectorization using graphene to target cellular death receptors of cancer cell. Proteins 2019; 88:94-105. [DOI: 10.1002/prot.25772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/11/2019] [Accepted: 07/06/2019] [Indexed: 02/04/2023]
Affiliation(s)
- Nicolas Arroyo
- Laboratoire de Nanomédecine, Imagerie et Thérapeutique, EA4662, Université Bourgogne‐Franche‐Comté (UFR Sciences et Techniques, UFR Sciences Médicales et Pharmaceutiques), Centre Hospitalier Universitaire de Besançon Besançon France
| | - Guillaume Herlem
- Laboratoire de Nanomédecine, Imagerie et Thérapeutique, EA4662, Université Bourgogne‐Franche‐Comté (UFR Sciences et Techniques, UFR Sciences Médicales et Pharmaceutiques), Centre Hospitalier Universitaire de Besançon Besançon France
| | - Fabien Picaud
- Laboratoire de Nanomédecine, Imagerie et Thérapeutique, EA4662, Université Bourgogne‐Franche‐Comté (UFR Sciences et Techniques, UFR Sciences Médicales et Pharmaceutiques), Centre Hospitalier Universitaire de Besançon Besançon France
| |
Collapse
|
12
|
Allen R, Chizari S, Ma JA, Raychaudhuri S, Lewis JS. Combinatorial, Microparticle-Based Delivery of Immune Modulators Reprograms the Dendritic Cell Phenotype and Promotes Remission of Collagen-Induced Arthritis in Mice. ACS APPLIED BIO MATERIALS 2019; 2:2388-2404. [DOI: 10.1021/acsabm.9b00092] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Riley Allen
- Department of Biomedical Engineering, University of California, Davis, Sacramento, California 95616, United States
| | - Shahab Chizari
- Department of Biomedical Engineering, University of California, Davis, Sacramento, California 95616, United States
| | - Jeffrey A. Ma
- Department of Biomedical Engineering, University of California, Davis, Sacramento, California 95616, United States
| | - Siba Raychaudhuri
- Department of Rheumatology, Allergy, and Clinical Immunology, School of Medicine, University of California, Davis, Sacramento, California 95817, United States
- VA Hospital, Northern California Health Care System, Sacramento, California 95817, United States
| | - Jamal S. Lewis
- Department of Biomedical Engineering, University of California, Davis, Sacramento, California 95616, United States
| |
Collapse
|
13
|
Li Y, Chen S, Chang X, He F, Zhuo R. Efficient Co-delivery of Doxorubicin and Methotrexate by pH-Sensitive Dual-Functional Nanomicelles for Enhanced Synergistic Antitumor Efficacy. ACS APPLIED BIO MATERIALS 2019; 2:2271-2279. [DOI: 10.1021/acsabm.9b00230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Youmei Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Shu Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Xiupeng Chang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Feng He
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Renxi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
14
|
Yarian F, Alibakhshi A, Eyvazi S, Arezumand R, Ahangarzadeh S. Antibody-drug therapeutic conjugates: Potential of antibody-siRNAs in cancer therapy. J Cell Physiol 2019; 234:16724-16738. [PMID: 30908646 DOI: 10.1002/jcp.28490] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/11/2019] [Accepted: 02/19/2019] [Indexed: 12/22/2022]
Abstract
Codelivery is a promising strategy of targeted delivery of cytotoxic drugs for eradicating tumor cells. This rapidly growing method of drug delivery uses a conjugate containing drug linked to a smart carrier. Both two parts usually have therapeutic properties on the tumor cells. Monoclonal antibodies and their derivatives, such as Fab, scFv, and bsAb due to targeting high potent have now been attractive candidates as drug targeting carrier systems. The success of some therapeutic agents like small interfering RNA (siRNA), a small noncoding RNAs, with having problems such as enzymatic degradation and rapid renal filtration need to an appropriate carrier. Therefore, the aim of this study is to review the recent enhancements in development of antibody drug conjugates (ADCs), especially antibody-siRNA conjugates (SRCs), its characterizations and mechanisms in innovative cancer therapy approaches.
Collapse
Affiliation(s)
- Fatemeh Yarian
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Alibakhshi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Eyvazi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roghaye Arezumand
- Department of Medical Biotechnology and Molecular Science, North Khorasan University of Medical Science, Bojnurd, Iran
| | - Shahrzad Ahangarzadeh
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
15
|
Novel amphiphilic folic acid-cholesterol-chitosan micelles for paclitaxel delivery. Oncotarget 2018; 8:3315-3326. [PMID: 27926514 PMCID: PMC5356884 DOI: 10.18632/oncotarget.13757] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/21/2016] [Indexed: 01/26/2023] Open
Abstract
In order to decrease the toxicity of paclitaxel (PTX) and increase the efficiency, we developed an amphiphilic PTX injection system using a biodegradable and biocompatible polymer synthesized by folic acid, cholesterol, and chitosan (FACC). This FACC-based polymer had a low critical concentration (64.13μg/ml) and could self-assemble in aqueous condition to form nanoscale micelles. The particle sizes of FACC-PTX micelles were 253.2±0.56 nm, the encapsulation efficiency and loading capacity of these FACC-PTX micelles were 65.1±0.23% and 9.1±0.16%, respectively. The cumulative release rate was about 85% at pH 5.0 which was higher than that at pH 7.4 (76%). This pH-dependent release behavior was highly suggesting that PTX release from FACC-PTX micelles might be higher in a weak acidic tumor microenvironment and lower toxic for normal cells. The anti-cancer effectiveness of FACC-PTX micelles was investigated by in vitro cytotoxicity and targeting study. The results revealed that FACC micelles have non-toxic on cells as evidenced by high cell viability found (86% to 100%) in the cells cultured with various concentrations of FACC micelles (1 to 500 μg/ml). Targeting study indicated that the cytotoxic efficacy of FACC-PTX micelles was significantly higher than that with Taxol® in the Hela cells (folate receptor-positive cells). These findings indicated that the anticancer efficiency of PTX can be enhanced by adding some cancer cell positive receptor into drug carrier and the FACC micelle was a potential tumor targeting carrier for PXT delivery.
Collapse
|
16
|
Qu D, Wang L, Qin Y, Guo M, Guo J, Huang M, Liu Y, Liu C, Li H, Chen Y. Non-triggered sequential-release liposomes enhance anti-breast cancer efficacy of STS and celastrol-based microemulsion. Biomater Sci 2018; 6:3284-3299. [PMID: 30346001 DOI: 10.1039/c8bm00796a] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A codelivery system that sequentially releases its contents is an effective strategy to enhance anticancer efficacy.
Collapse
Affiliation(s)
- Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| | - Lixiang Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
| | - Yue Qin
- Jiangsu Provincial Academy of Traditional Chinese Medicine
- Nanjing 210028
- China
| | - Mengfei Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
| | - Jian Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
| | - Mengmeng Huang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| | - Congyan Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| | - Hui Li
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Sciences
- Beijing 100010
- China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| |
Collapse
|
17
|
Wu X, Wang S, Li M, Wang A, Zhou Y, Li P, Wang Y. Nanocarriers for TRAIL delivery: driving TRAIL back on track for cancer therapy. NANOSCALE 2017; 9:13879-13904. [PMID: 28914952 DOI: 10.1039/c7nr04959e] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Since its initial identification, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has been shown to be capable of selectively inducing apoptosis in cancer cells. However, translation of the encouraging preclinical studies of this cytokine into the clinic has been restricted by its extremely short half-life, the presence of resistant cancer cell populations, and its inefficient in vivo delivery. Recently, there has been exceptional progress in developing novel formulations to increase the circulatory half-life of TRAIL and new combinations to treat cancers that are resistant to TRAIL. In particular, TRAIL-based nanotherapies offer the potential to improve the stability of TRAIL and prolong its half-life in plasma, to specifically deliver TRAIL to a particular target site, and to overcome resistance to TRAIL. The aim of this review is to provide an overview of the state-of-the art drug delivery systems that are currently being tested or developed to improve the biological attributes of TRAIL-based therapies.
Collapse
Affiliation(s)
- Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Soni KS, Lei F, Desale SS, Marky LA, Cohen SM, Bronich TK. Tuning polypeptide-based micellar carrier for efficient combination therapy of ErbB2-positive breast cancer. J Control Release 2017; 264:276-287. [PMID: 28870832 DOI: 10.1016/j.jconrel.2017.08.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/19/2017] [Accepted: 08/29/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Kruti S Soni
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Fan Lei
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Swapnil S Desale
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Luis A Marky
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, NE 68198-5950, USA
| | - Samuel M Cohen
- Department of Pathology and Microbiology, University of Nebraska Medical Center/Fred and Pamela Buffet Cancer Center, Omaha, NE 68198-3135, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, NE 68198-5950, USA.
| |
Collapse
|
19
|
Babu A, Munshi A, Ramesh R. Combinatorial therapeutic approaches with RNAi and anticancer drugs using nanodrug delivery systems. Drug Dev Ind Pharm 2017; 43:1391-1401. [PMID: 28523942 PMCID: PMC6101010 DOI: 10.1080/03639045.2017.1313861] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
RNA interference (RNAi) is emerging as a powerful approach in cancer treatment. siRNA is an important RNAi tool that can be designed to specifically silence the expression of genes involved in drug resistance and chemotherapeutic inactivity. Combining siRNA and other therapeutic agents can overcome the multidrug resistance (MDR) phenomenon by simultaneously silencing genes and enhancing chemotherapeutic activity. Moreover, the therapeutic efficiency of anticancer drugs can be significantly improved by additive or synergistic effects induced by siRNA and combined therapies. Co-delivery of these diverse anticancer agents, however, requires specially designed nanocarriers. This review highlights the recent trends in siRNA/anticancer drug co-delivery systems under the major categories of liposomes/lipid, polymeric and inorganic nanoplatforms. The objective is to discuss the strategies for nanocarrier-based co-delivery systems using siRNA/anticancer drug combinations, emphasizing various siRNA targets that help overcome MDR and enhance therapeutic efficiency.
Collapse
Affiliation(s)
- Anish Babu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
- Department of Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
| | - Anupama Munshi
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
- Department of Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
- Department of Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
- Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104; USA
| |
Collapse
|
20
|
Pindiprolu SKSS, Krishnamurthy PT, Chintamaneni PK, Karri VVSR. Nanocarrier based approaches for targeting breast cancer stem cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:885-898. [PMID: 28826237 DOI: 10.1080/21691401.2017.1366337] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Breast cancer stem cells (BCSCs) are heterogeneous subpopulation of tumour initiating cells within breast tumours. They are spared even after chemotherapy and responsible for tumour relapse. Targeting BCSCs is, therefore, necessary to achieve radical cure in breast cancer. Despite the availability of agents targeting BCSCs, their clinical application is limited due to their off-target effects and bioavailability issues. Nanotechnology based drug carriers (nanocarriers) offer various advantages to deliver anti-BCSCs agents specifically to their target sites by overcoming their bioavailability issues. In this review, we describe various strategies for targeting BCSCs using nanocarriers.
Collapse
Affiliation(s)
- Sai Kiran S S Pindiprolu
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Praveen T Krishnamurthy
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Pavan Kumar Chintamaneni
- a Department of Pharmacology , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| | - Veera Venkata Satyanarayana Reddy Karri
- b Department of Pharmaceutics , JSS College of Pharmacy (A Constituent College of Jagadguru Sri Shivarathreeshwara University) , Ootacamund , Tamil Nadu , India
| |
Collapse
|
21
|
Zhang J, Wang L, Fai Chan H, Xie W, Chen S, He C, Wang Y, Chen M. Co-delivery of paclitaxel and tetrandrine via iRGD peptide conjugated lipid-polymer hybrid nanoparticles overcome multidrug resistance in cancer cells. Sci Rep 2017; 7:46057. [PMID: 28470171 PMCID: PMC5415764 DOI: 10.1038/srep46057] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 03/06/2017] [Indexed: 01/13/2023] Open
Abstract
One of the promising strategies to overcome tumor multidrug resistance (MDR) is to deliver anticancer drug along with P-glycoprotein (P-gp) inhibitor simultaneously. To enhance the cancer cellular internalization and implement the controlled drug release, herein an iRGD peptide-modified lipid-polymer hybrid nanosystem (LPN) was fabricated to coload paclitaxel (PTX) and tetrandrine (TET) at a precise combination ratio. In this co-delivery system, PTX was covalently conjugated to poly (D,L-lactide-co-glycolide) polymeric core by redox-sensitive disulfide bond, while TET was physically capsulated spontaneously for the aim to suppress P-gp in advance by the earlier released TET in cancer cells. As a result, the PTX+TET/iRGD LPNs with a core-shell structure possessed high drug loading efficiency, stability and redox-sensitive drug release profiles. Owing to the enhanced cellular uptake and P-gp suppression mediated by TET, significantly more PTX accumulated in A2780/PTX cells treated with PTX+TET/iRGD LPNs than either free drugs or non-iRGD modified LPNs. As expected, PTX+TET/iRGD LPNs presented the highest cytotoxicity against A2780/PTX cells and effectively promoted ROS production, enhanced apoptosis and cell cycle arrests particularly. Taken together, the co-delivery system demonstrated great promise as potential treatment for MDR-related tumors based on the synergistic effects of P-gp inhibition, enhanced endocytosis and intracellular sequentially drug release.
Collapse
Affiliation(s)
- Jinming Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lu Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hon Fai Chan
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Xie
- Department of Respiratory Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Sheng Chen
- Department of Respiratory Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
22
|
He YJ, Xing L, Cui PF, Zhang JL, Qiao JB, Luo CQ, Jiang G, Jiang HL. Branched polymeric prodrug/programmed cell death 4 complexes for combinational cancer therapy. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-016-0301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Li J, Mao J, Tang J, Li G, Fang F, Tang Y, Ding J. Surface spermidine functionalized PEGylated poly(lactide-co-glycolide) nanoparticles for tumor-targeted drug delivery. RSC Adv 2017. [DOI: 10.1039/c7ra02447a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
SPD functionalized nanoparticles could target the delivery of a drug into tumor cells by binding specifically with PTS.
Collapse
Affiliation(s)
- Jianming Li
- Xiangya School of Pharmaceutical Sciences
- Central South University
- Changsha 410013
- China
| | - Juan Mao
- Xiangya School of Pharmaceutical Sciences
- Central South University
- Changsha 410013
- China
| | - Jing Tang
- Department of Pharmaceutics
- Changsha Medical University
- Changsha 410219
- China
| | - Guo Li
- Xiangya School of Pharmaceutical Sciences
- Central South University
- Changsha 410013
- China
| | - Fengling Fang
- Xiangya School of Pharmaceutical Sciences
- Central South University
- Changsha 410013
- China
| | - Yana Tang
- Xiangya School of Pharmaceutical Sciences
- Central South University
- Changsha 410013
- China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences
- Central South University
- Changsha 410013
- China
| |
Collapse
|
24
|
Xu P, Zuo H, Chen D, Peng M, Jiang Y, Liu X, Ouyang J, Chen B. Anti-CD22-conjugated CdTe QDs co-loaded with doxorubicin and gambogic acid: a novel platform for lymphoma treatment. RSC Adv 2017. [DOI: 10.1039/c7ra04056c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
DOX/GA–CdTe–CD22 can precisely target lymphoma and deliver DOX and GA to lymphoma cells to improve their therapeutic effects.
Collapse
Affiliation(s)
- Peipei Xu
- Department of Hematology
- Drum Tower Hospital
- School of Medicine
- Nanjing University
- Nanjing
| | - Huaqin Zuo
- Department of Hematology
- Drum Tower Hospital
- School of Medicine
- Nanjing University
- Nanjing
| | - Dangui Chen
- Department of Hematology
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University
- Nanjing 210036
- P. R. China
| | - Miaoxin Peng
- Department of Hematology
- Drum Tower Hospital
- School of Medicine
- Nanjing University
- Nanjing
| | - Ying Jiang
- Department of Hematology
- Drum Tower Hospital
- School of Medicine
- Nanjing University
- Nanjing
| | - Xu Liu
- Department of Hematology
- Drum Tower Hospital
- School of Medicine
- Nanjing University
- Nanjing
| | - Jian Ouyang
- Department of Hematology
- Drum Tower Hospital
- School of Medicine
- Nanjing University
- Nanjing
| | - Bing Chen
- Department of Hematology
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University
- Nanjing 210036
- P. R. China
| |
Collapse
|
25
|
Han HD, Byeon Y, Kang TH, Jung ID, Lee JW, Shin BC, Lee YJ, Sood AK, Park YM. Toll-like receptor 3-induced immune response by poly(d,l-lactide-co-glycolide) nanoparticles for dendritic cell-based cancer immunotherapy. Int J Nanomedicine 2016; 11:5729-5742. [PMID: 27843314 PMCID: PMC5098754 DOI: 10.2147/ijn.s109001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Dendritic cells (DCs) are potent professional antigen-presenting cells that are capable of initiating a primary immune response and activating T cells, and they play a pivotal role in the immune responses of the host to cancer. Prior to antigen presentation, efficient antigen and adjuvant uptake by DCs is necessary to induce their maturation and cytokine generation. Nanoparticles (NPs) are capable of intracellular delivery of both antigen and adjuvant to DCs. Here, we developed an advanced poly(d,l-lactide-co-glycolide) (PLGA)-NP encapsulating both ovalbumin (OVA) as a model antigen and polyinosinic-polycytidylic acid sodium salt (Toll-like receptor 3 ligand) as an adjuvant to increase intracellular delivery and promote DC maturation. The PLGA-NPs were taken up by DCs, and their uptake greatly facilitated major histocompatibility class I antigen presentation in vitro. Moreover, vaccination with PLGA-NP-treated DCs led to the generation of ovalbumin-specific CD8+ T cells, and the resulting antitumor efficacy was significantly increased in EG.7 and TC-1 tumor-bearing mice compared to control mice (P<0.01). Taken together, these findings demonstrated that the PLGA-NP platform may be an effective method for delivering tumor-specific antigens or adjuvants to DCs.
Collapse
Affiliation(s)
- Hee Dong Han
- Department of Immunology, School of Medicine, Konkuk University, Chungwondaero, Chungju-Si, Chungcheongbuk-Do
| | - Yeongseon Byeon
- Department of Immunology, School of Medicine, Konkuk University, Chungwondaero, Chungju-Si, Chungcheongbuk-Do
| | - Tae Heung Kang
- Department of Immunology, School of Medicine, Konkuk University, Chungwondaero, Chungju-Si, Chungcheongbuk-Do
| | - In Duk Jung
- Department of Immunology, School of Medicine, Konkuk University, Chungwondaero, Chungju-Si, Chungcheongbuk-Do
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
| | - Byung Cheol Shin
- Bio/Drug Discovery Division, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon
| | - Young Joo Lee
- Department of Bioscience and Biotechnology, Sejong University, Kwang-Jin-Gu, Seoul, South Korea
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine; Department of Cancer Biology; Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Yeong-Min Park
- Department of Immunology, School of Medicine, Konkuk University, Chungwondaero, Chungju-Si, Chungcheongbuk-Do
| |
Collapse
|
26
|
Wang S, Xu Y, Chan HF, Kim HW, Wang Y, Leong KW, Chen M. Nanoparticle-mediated inhibition of survivin to overcome drug resistance in cancer therapy. J Control Release 2016; 240:454-464. [DOI: 10.1016/j.jconrel.2016.04.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 04/09/2016] [Accepted: 04/11/2016] [Indexed: 02/08/2023]
|
27
|
One-step assembly of polymeric demethylcantharate prodrug/Akt1 shRNA complexes for enhanced cancer therapy. Int J Pharm 2016; 513:612-627. [PMID: 27682215 DOI: 10.1016/j.ijpharm.2016.09.070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/22/2016] [Accepted: 09/24/2016] [Indexed: 02/06/2023]
Abstract
This report demonstrated a one-step assembly for co-delivering chemotherapeutics and therapeutic nucleic acids, constructed by integrating drug molecules into a nucleic acid condensing polymeric prodrug through degradable linkages. Demethylcantharate was selected as the model drug and pre-modified by esterifying its two carboxylic groups with 2-hydroxyethyl acrylate. The synthesized demethylcantharate diacrylate was then used to polymerize with linear polyethyleneimine (PEI 423) through a one-step Michael-addition reaction. The obtained cationic polymeric demethylcantharate prodrug was used to pack Akt1 shRNA into complexes through a one-step assembly. The formed complexes could release the parent drug demethylcantharate and Akt1 shRNA through the hydrolysis of ester bonds. Cellular assays involving cell uptake, cytotoxicity, and cell migration indicated that demethylcantharate and Akt1 shRNA co-delivered in the present form significantly and synergistically suppress the growth and metastasis of three human cancer cells. This work suggests that incorporating drug molecules into a nucleic acid-packing cationic polymer as a polymeric prodrug in a degradable form is a highly convenient and efficient way to co-deliver drugs and nucleic acids for cancer therapy.
Collapse
|
28
|
Bobbala S, Hook S. Is There an Optimal Formulation and Delivery Strategy for Subunit Vaccines? Pharm Res 2016; 33:2078-97. [DOI: 10.1007/s11095-016-1979-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/21/2016] [Indexed: 12/16/2022]
|
29
|
Co-delivery of chemotherapeutics and proteins for synergistic therapy. Adv Drug Deliv Rev 2016; 98:64-76. [PMID: 26546464 DOI: 10.1016/j.addr.2015.10.021] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/15/2023]
Abstract
Combination therapy with chemotherapeutics and protein therapeutics, typically cytokines and antibodies, has been a type of crucial approaches for synergistic cancer treatment. However, conventional approaches by simultaneous administration of free chemotherapeutic drugs and proteins lead to limitations for further optimizing the synergistic effects, due to the distinct in vivo pharmacokinetics and distribution of small drugs and proteins, insufficient tumor selectivity and tumor accumulation, unpredictable drug/protein ratios at tumor sites, short half-lives, and serious systemic adverse effects. Consequently, to obtain optimal synergistic anti-tumor efficacy, considerable efforts have been devoted to develop the co-delivery systems for co-incorporating chemotherapeutics and proteins into a single carrier system and subsequently releasing the dual or multiple payloads at desired target sites in a more controllable manner. The co-delivery systems result in markedly enhanced blood stability and in vivo half-lives of the small drugs and proteins, elevated tumor accumulation, as well as the capability of delivering the multiple agents to the same target sites with rational drug/protein ratios, which may facilitate maximizing the synergistic effects and therefore lead to optimal antitumor efficacy. This review emphasizes the recent advances in the co-delivery systems for chemotherapeutics and proteins, typically cytokines and antibodies, for systemic or localized synergistic cancer treatment. Moreover, the proposed mechanisms responsible for the synergy of chemotherapeutic drugs and proteins are discussed.
Collapse
|
30
|
Liang Y, Kiick KL. Liposome-Cross-Linked Hybrid Hydrogels for Glutathione-Triggered Delivery of Multiple Cargo Molecules. Biomacromolecules 2016; 17:601-14. [PMID: 26751084 PMCID: PMC4992983 DOI: 10.1021/acs.biomac.5b01541] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Novel, liposome-cross-linked hybrid hydrogels cross-linked by the Michael-type addition of thiols with maleimides were prepared via the use of maleimide-functionalized liposome cross-linkers and thiolated polyethylene glycol (PEG) polymers. Gelation of the materials was confirmed by oscillatory rheology experiments. These hybrid hydrogels are rendered degradable upon exposure to thiol-containing molecules such as glutathione (GSH), via the incorporation of selected thioether succinimide cross-links between the PEG polymers and liposome nanoparticles. Dynamic light scattering (DLS) characterization confirmed that intact liposomes were released upon network degradation. Owing to the hierarchical structure of the network, multiple cargo molecules relevant for chemotherapies, namely doxorubicin (DOX) and cytochrome c, were encapsulated and simultaneously released from the hybrid hydrogels, with differential release profiles that were driven by degradation-mediated release and Fickian diffusion, respectively. This work introduces a facile approach for the development of advanced, hybrid drug delivery vehicles that exhibit novel chemical degradation.
Collapse
Affiliation(s)
- Yingkai Liang
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Kristi L. Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, Delaware 19716, United States
| |
Collapse
|
31
|
Ma Y, Fan X, Li L. pH-sensitive polymeric micelles formed by doxorubicin conjugated prodrugs for co-delivery of doxorubicin and paclitaxel. Carbohydr Polym 2016; 137:19-29. [DOI: 10.1016/j.carbpol.2015.10.050] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/12/2015] [Accepted: 10/14/2015] [Indexed: 01/23/2023]
|
32
|
Immune cell impact of three differently coated lipid nanocapsules: pluronic, chitosan and polyethylene glycol. Sci Rep 2016; 6:18423. [PMID: 26728491 PMCID: PMC4700454 DOI: 10.1038/srep18423] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/09/2015] [Indexed: 12/23/2022] Open
Abstract
Lipid nanocapsules (NCs) represent promising tools in clinical practice for diagnosis and therapy applications. However, the NC appropriate functionalization is essential to guarantee high biocompatibility and molecule loading ability. In any medical application, the immune system-impact of differently functionalized NCs still remains to be fully understood. A comprehensive study on the action exerted on human peripheral blood mononuclear cells (PBMCs) and major immune subpopulations by three different NC coatings: pluronic, chitosan and polyethylene glycol-polylactic acid (PEG) is reported. After a deep particle characterization, the uptake was assessed by flow-cytometry and confocal microscopy, focusing then on apoptosis, necrosis and proliferation impact in T cells and monocytes. Cell functionality by cell diameter variations, different activation marker analysis and cytokine assays were performed. We demonstrated that the NCs impact on the immune cell response is strongly correlated to their coating. Pluronic-NCs were able to induce immunomodulation of innate immunity inducing monocyte activations. Immunomodulation was observed in monocytes and T lymphocytes treated with Chitosan-NCs. Conversely, PEG-NCs were completely inert. These findings are of particular value towards a pre-selection of specific NC coatings depending on biomedical purposes for pre-clinical investigations; i.e. the immune-specific action of particular NC coating can be excellent for immunotherapy applications.
Collapse
|
33
|
Song Y, Xie Y, Yang J, Li R, Jin X, Yang J. A poly(ascorbyl acrylate)-containing nanoplatform with anticancer activity and the sequential combination therapy with its loaded paclitaxel. J Mater Chem B 2016; 4:6588-6596. [DOI: 10.1039/c6tb01818a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The complex nanocarriers combined with the loaded therapeutic agents to achieve synergistic tumor inhibition.
Collapse
Affiliation(s)
- Yufeng Song
- State Key Laboratory of Chemical Resource
- Beijing Key Laboratory of Bioprocess
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Yanqi Xie
- State Key Laboratory of Chemical Resource
- Beijing Key Laboratory of Bioprocess
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Junjiao Yang
- College of Science
- Beijing University of Chemical Technology
- Beijing 100029
- China
| | - Ruiqiong Li
- State Key Laboratory of Chemical Resource
- Beijing Key Laboratory of Bioprocess
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Xu Jin
- Department of Anesthesiology and Pain Therapy
- Beijing Tiantan Hospital Affiliated to Capital Medical University
- Beijing 100050
- China
| | - Jing Yang
- State Key Laboratory of Chemical Resource
- Beijing Key Laboratory of Bioprocess
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| |
Collapse
|
34
|
Zeng J, Du P, Liu L, Li J, Tian K, Jia X, Zhao X, Liu P. Superparamagnetic Reduction/pH/Temperature Multistimuli-Responsive Nanoparticles for Targeted and Controlled Antitumor Drug Delivery. Mol Pharm 2015; 12:4188-99. [DOI: 10.1021/acs.molpharmaceut.5b00342] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jin Zeng
- State Key Laboratory
of Applied
Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry
and Resources Utilization of Gansu Province, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Pengcheng Du
- State Key Laboratory
of Applied
Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry
and Resources Utilization of Gansu Province, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Lei Liu
- State Key Laboratory
of Applied
Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry
and Resources Utilization of Gansu Province, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jiagen Li
- State Key Laboratory
of Applied
Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry
and Resources Utilization of Gansu Province, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Kun Tian
- State Key Laboratory
of Applied
Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry
and Resources Utilization of Gansu Province, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xu Jia
- State Key Laboratory
of Applied
Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry
and Resources Utilization of Gansu Province, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xubo Zhao
- State Key Laboratory
of Applied
Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry
and Resources Utilization of Gansu Province, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Peng Liu
- State Key Laboratory
of Applied
Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry
and Resources Utilization of Gansu Province, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
35
|
Bertucci A, Prasetyanto EA, Septiadi D, Manicardi A, Brognara E, Gambari R, Corradini R, De Cola L. Combined Delivery of Temozolomide and Anti-miR221 PNA Using Mesoporous Silica Nanoparticles Induces Apoptosis in Resistant Glioma Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:5687-95. [PMID: 26395266 DOI: 10.1002/smll.201500540] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 07/22/2015] [Indexed: 05/14/2023]
Abstract
Mesoporous silica nanoparticles (MSNPs), 100 nm in size, incorporating a Cy5 fluorophore within the silica framework, are synthesized and loaded with the anti-cancer drug temozolomide (TMZ), used in the treatment of gliomas. The surface of the particles is then decorated, using electrostatic interactions, with a polyarginine-peptide nucleic acid (R8-PNA) conjugate targeting the miR221 microRNA. The multi-functional nanosystem thus obtained is rapidly internalized into glioma C6 or T98G cells. The anti-miR activity of the PNA is retained, as confirmed by reverse transcription polymerase chain reaction (RT-PCR) measurements and induction of apoptosis is observed in temozolomide-resistant cell lines. The TMZ-loaded MSNPs show an enhanced pro-apoptotic effect, and the combined effect of TMZ and R8-PNA in the MSNPs shows the most effective induction of apoptosis (70.9% of apoptotic cells) thus far achieved in the temozolomide-resistant T98G cell line.
Collapse
Affiliation(s)
- Alessandro Bertucci
- Institut de science et d'ingénierie supramoléculaire (ISIS) & icFRC, Université de Strasbourg & CNRS, 8 Rue Gaspard Monge, Strasbourg, 67000, France
- Dipartimento di Chimica, Università di Parma, Parco Area delle Scienze 17/A, Parma, 43124, Italy
| | - Eko Adi Prasetyanto
- Institut de science et d'ingénierie supramoléculaire (ISIS) & icFRC, Université de Strasbourg & CNRS, 8 Rue Gaspard Monge, Strasbourg, 67000, France
| | - Dedy Septiadi
- Institut de science et d'ingénierie supramoléculaire (ISIS) & icFRC, Université de Strasbourg & CNRS, 8 Rue Gaspard Monge, Strasbourg, 67000, France
| | - Alex Manicardi
- Dipartimento di Chimica, Università di Parma, Parco Area delle Scienze 17/A, Parma, 43124, Italy
| | - Eleonora Brognara
- Dipartimento di Scienze della Vita e Biotecnologie, Università di Ferrara, Via Luigi Borsari 46, Ferrara, 44121, Italy
| | - Roberto Gambari
- Dipartimento di Scienze della Vita e Biotecnologie, Università di Ferrara, Via Luigi Borsari 46, Ferrara, 44121, Italy
| | - Roberto Corradini
- Dipartimento di Chimica, Università di Parma, Parco Area delle Scienze 17/A, Parma, 43124, Italy
| | - Luisa De Cola
- Institut de science et d'ingénierie supramoléculaire (ISIS) & icFRC, Université de Strasbourg & CNRS, 8 Rue Gaspard Monge, Strasbourg, 67000, France
| |
Collapse
|
36
|
Chen CY, Syu CK, Lin HC. A Stimulated Mixed Micelle System for In Vitro Study on Chemo-Photodynamic Therapy. Macromol Biosci 2015; 16:188-97. [DOI: 10.1002/mabi.201500269] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/10/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Ching-Yi Chen
- Department of Chemical Engineering; National Chung Cheng University; Chia-Yi County 621 Taiwan
| | - Chao-Kai Syu
- Department of Chemical Engineering; National Chung Cheng University; Chia-Yi County 621 Taiwan
| | - Hsin Chang Lin
- Department of Chemical Engineering; National Chung Cheng University; Chia-Yi County 621 Taiwan
| |
Collapse
|
37
|
Lao YH, Phua KKL, Leong KW. Aptamer nanomedicine for cancer therapeutics: barriers and potential for translation. ACS NANO 2015; 9:2235-54. [PMID: 25731717 DOI: 10.1021/nn507494p] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Aptamer nanomedicine, including therapeutic aptamers and aptamer nanocomplexes, is beginning to fulfill its potential in both clinical trials and preclinical studies. Especially in oncology, aptamer nanomedicine may perform better than conventional or antibody-based chemotherapeutics due to specificity compared to the former and stability compared to the latter. Many proof-of-concept studies on applying aptamers to drug delivery, gene therapy, and cancer imaging have shown promising efficacy and impressive safety in vivo toward translation. Yet, there remains ample room for improvement and critical barriers to be addressed. In this review, we will first introduce the recent progress in clinical trials of aptamer nanomedicine, followed by a discussion of the barriers at the design and in vivo application stages. We will then highlight recent advances and engineering strategies proposed to tackle these barriers. Aptamer cancer nanomedicine has the potential to address one of the most important healthcare issues of the society.
Collapse
Affiliation(s)
- Yeh-Hsing Lao
- †Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States
| | | | - Kam W Leong
- †Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States
| |
Collapse
|
38
|
Desale SS, Raja SM, Kim JO, Mohapatra B, Soni KS, Luan H, Williams SH, Bielecki TA, Feng D, Storck M, Band V, Cohen SM, Band H, Bronich TK. Polypeptide-based nanogels co-encapsulating a synergistic combination of doxorubicin with 17-AAG show potent anti-tumor activity in ErbB2-driven breast cancer models. J Control Release 2015; 208:59-66. [PMID: 25660204 DOI: 10.1016/j.jconrel.2015.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/02/2015] [Indexed: 12/29/2022]
Abstract
ErbB2-driven breast cancers constitute 20-25% of the cases diagnosed within the USA. The humanized anti-ErbB2 monoclonal antibody, Trastuzumab (Herceptin™; Genentech), with chemotherapy is the current standard of treatment. Novel agents and strategies continue to be explored, given the challenges posed by Trastuzumab-resistance development in most patients. The HSP90 inhibitor, 17-allylaminodemethoxygeldanamycin (17-AAG), which induces ErbB2 degradation and attenuates downstream oncogenic signaling, is one such agent that showed significant promise in early phase I and II clinical trials. Its low water solubility, potential toxicities and undesirable side effects observed in patients, partly due to the Cremophor-based formulation, have been discouraging factors in the advancement of this promising drug into clinical use. Encapsulation of 17-AAG into polymeric nanoparticle formulations, particularly in synergistic combination with conventional chemotherapeutics, represents an alternative approach to overcome these problems. Herein, we report an efficient co-encapsulation of 17-AAG and doxorubicin, a clinically well-established and effective modality in breast cancer treatment, into biodegradable and biocompatible polypeptide-based nanogels. Dual drug-loaded nanogels displayed potent cytotoxicity in a breast cancer cell panel and exerted selective synergistic anticancer activity against ErbB2-overexpressing breast cancer cell lines. Analysis of ErbB2 degradation confirmed efficient 17-AAG release from nanogels with activity comparable to free 17-AAG. Furthermore, nanogels containing both 17-AAG and doxorubicin exhibited superior antitumor efficacy in vivo in an ErbB2-driven xenograft model compared to the combination of free drugs. These studies demonstrate that polypeptide-based nanogels can serve as novel nanocarriers for encapsulating 17-AAG along with other chemotherapeutics, providing an opportunity to overcome solubility issues and thereby exploit its full potential as an anti-cancer agent.
Collapse
Affiliation(s)
- Swapnil S Desale
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, United States
| | - Srikumar M Raja
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC, United States.
| | - Jong Oh Kim
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, United States; College of Pharmacy, Yeungnam University, Gyeongsan 712-749, South Korea
| | - Bhopal Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC, United States
| | - Kruti S Soni
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, United States
| | - Haitao Luan
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC, United States
| | - Stetson H Williams
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC, United States
| | - Timothy A Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC, United States
| | - Dan Feng
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC, United States
| | - Matthew Storck
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC, United States
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, UNMC, United States
| | - Samuel M Cohen
- Department of Pathology and Microbiology, UNMC, United States
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC, United States.
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, United States.
| |
Collapse
|
39
|
Liu X, Jiang H, Ge W, Wu C, Chen D, Li Q, Chen Y, Wang X. Green and facile synthesis of highly biocompatible carbon nanospheres and their pH-responsive delivery of doxorubicin to cancer cells. RSC Adv 2015. [DOI: 10.1039/c4ra16359a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Carbon nanospheres with size below 71 nm are synthesized from bacterial cellulose nanofibers using a one-pot hydrothermal synthesis method.
Collapse
Affiliation(s)
- Xiaoli Liu
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab)
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing
- PR China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab)
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing
- PR China
| | - Wei Ge
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab)
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing
- PR China
| | - Changyu Wu
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab)
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing
- PR China
| | - Donghua Chen
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab)
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing
- PR China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab)
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing
- PR China
| | - Yun Chen
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab)
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing
- PR China
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab)
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing
- PR China
| |
Collapse
|
40
|
Enhanced antitumor efficacy of folate modified amphiphilic nanoparticles through co-delivery of chemotherapeutic drugs and genes. Biomaterials 2014; 35:6369-78. [DOI: 10.1016/j.biomaterials.2014.04.095] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 04/21/2014] [Indexed: 11/19/2022]
|