1
|
Gao Y, Shelling AN, Nolan E, Porter D, Leung E, Wu Z. Liposome-enabled bufalin and doxorubicin combination therapy for trastuzumab-resistant breast cancer with a focus on cancer stem cells. J Liposome Res 2024; 34:489-506. [PMID: 38269490 DOI: 10.1080/08982104.2024.2305866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024]
Abstract
Breast cancer stem cells (BCSCs) play a key role in therapeutic resistance in breast cancer treatments and disease recurrence. This study aimed to develop a combination therapy loaded with pH-sensitive liposomes to kill both BCSCs and the okbulk cancer cells using trastuzumab-sensitive and resistant human epidermal growth factor receptor 2 positive (HER2+) breast cancer cell models. The anti-BCSCs effect and cytotoxicity of all-trans retinoic acid, salinomycin, and bufalin alone or in combination with doxorubicin were compared in HER2+ cell line BT-474 and a validated trastuzumab-resistant cell line, BT-474R. The most potent anti-BCSC agent was selected and loaded into a pH-sensitive liposome system. The effects of the liposomal combination on BCSCs and bulk cancer cells were assessed. Compared with BT-474, the aldehyde dehydrogenase positive BCSC population was elevated in BT-474R (3.9 vs. 23.1%). Bufalin was the most potent agent and suppressed tumorigenesis of BCSCs by ∼50%, and showed strong synergism with doxorubicin in both BT-474 and BT-474R cell lines. The liposomal combination of bufalin and doxorubicin significantly reduced the BCSC population size by 85%, and inhibited both tumorigenesis and self-renewal, although it had little effect on the migration and invasiveness. The cytotoxicity against the bulk cancer cells was also enhanced by the liposomal combination than either formulation alone in both cell lines (p < 0.001). The liposomal bufalin and doxorubicin combination therapy may effectively target both BCSCs and bulk cancer cells for a better outcome in trastuzumab-resistant HER2+ breast cancer.
Collapse
Affiliation(s)
- Yu Gao
- Faculty of Medical and Health Sciences, School of Pharmacy, The University of Auckland, Auckland, New Zealand
| | - Andrew N Shelling
- Faculty of Medical and Health Sciences, School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Emma Nolan
- Faculty of Medical and Health Sciences, Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - David Porter
- Auckland Regional Cancer and Blood Service, Auckland City Hospital, Auckland, New Zealand
| | - Euphemia Leung
- Faculty of Medical and Health Sciences, Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Zimei Wu
- Faculty of Medical and Health Sciences, School of Pharmacy, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
2
|
Biscaia-Caleiras M, Fonseca NA, Lourenço AS, Moreira JN, Simões S. Rational formulation and industrial manufacturing of lipid-based complex injectables: Landmarks and trends. J Control Release 2024; 373:617-639. [PMID: 39002799 DOI: 10.1016/j.jconrel.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Lipid-based complex injectables are renowned for their effectiveness in delivering drugs, with many approved products. While significant strides have been made in formulating nanosystems for small molecular weight drugs, a pivotal breakthrough emerged with the recognition of lipid nanoparticles as a promising platform for delivering nucleic acids. This finding has paved the way for tackling long-standing challenges in molecular and delivery aspects (e.g., mRNA stability, intracellular delivery) that have impeded the clinical translation of gene therapy, especially in the realm of immunotherapy. Nonetheless, developing and implementing new lipid-based delivery systems pose significant challenges, as industrial manufacturing of these formulations often involves complex, multi-batch processes, giving rise to issues related to scalability, stability, sterility, and regulatory compliance. To overcome these obstacles, embracing the principles of quality-by-design (QbD) is imperative. Furthermore, adopting cutting-edge manufacturing and process analytical tools (PAT) that facilitate the transition from batch to continuous production is essential. Herein, the key milestones and insights derived from the development of currently approved lipid- nanosystems will be explored. Additionally, a comprehensive and critical overview of the latest technologies and regulatory guidelines that underpin the creation of more efficient, scalable, and flexible manufacturing processes for complex lipid-based nanoformulations will be provided.
Collapse
Affiliation(s)
- Mariana Biscaia-Caleiras
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Nuno A Fonseca
- Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal
| | - Ana Sofia Lourenço
- Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Sérgio Simões
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
3
|
Vatankhah M, Dadashzadeh S, Mahboubi A, Haeri A, Jandaghi Alaee K, Mostafavi Naeini SB, Abbasian Z. Preparation of multivesicular liposomes for the loco-regional delivery of Vancomycin hydrochloride using active loading method: drug release and antimicrobial properties. J Liposome Res 2024; 34:77-87. [PMID: 37287348 DOI: 10.1080/08982104.2023.2220805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Over the last few years, among controlled-release delivery systems, multivesicular liposomes (MVLs) have attracted attention due to their unique benefits as a loco-regional drug delivery system. Considering the clinical limitations of the current treatment strategies for osteomyelitis, MVLs can be a suitable carrier for the local delivery of effective antibiotics. This study aimed to prepare vancomycin hydrochloride (VAN HL) loaded MVLs using the active loading method which to the best of our knowledge has not been previously reported. Empty MVLS were prepared by the double emulsion (w/o/w) method and VAN HL was loaded into the prepared liposomes by the ammonium gradient method. After full characterization, the release profile of VAN HL from MVLs was assessed at two different pH values (5.5 and 7.4), and compared with the release profile of the free drug and also passively loaded MVLs. In vitro antimicrobial activities were evaluated using the disc diffusion method. Our results demonstrated that the encapsulation efficiency was higher than 90% in the optimum actively loaded MVL. The free VAN HL was released within 6-8 h, while the passively loaded MVLs and the optimum actively loaded MVL formulation released the drug in 6 days and up to 19 days, respectively. The released drug showed effective antibacterial activity against osteomyelitis-causing pathogens. In conclusion, the prepared formulation offered the advantages of sustained-release properties, appropriate particle size as well as being composed of biocompatible materials, and thus could be a promising candidate for the loco-regional delivery of VAN HL and the management of osteomyelitis.
Collapse
Affiliation(s)
- Melody Vatankhah
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simin Dadashzadeh
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Mahboubi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences
| | - Kimia Jandaghi Alaee
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Baubak Mostafavi Naeini
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Abbasian
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Rastegar G, Salman MM, Sirsi SR. Remote Loading: The Missing Piece for Achieving High Drug Payload and Rapid Release in Polymeric Microbubbles. Pharmaceutics 2023; 15:2550. [PMID: 38004529 PMCID: PMC10675060 DOI: 10.3390/pharmaceutics15112550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
The use of drug-loaded microbubbles for targeted drug delivery, particularly in cancer treatment, has been extensively studied in recent years. However, the loading capacity of microbubbles has been limited due to their surface area. Typically, drug molecules are loaded on or within the shell, or drug-loaded nanoparticles are coated on the surfaces of microbubbles. To address this significant limitation, we have introduced a novel approach. For the first time, we employed a transmembrane ammonium sulfate and pH gradient to load doxorubicin in a crystallized form in the core of polymeric microcapsules. Subsequently, we created remotely loaded microbubbles (RLMBs) through the sublimation of the liquid core of the microcapsules. Remotely loaded microcapsules exhibited an 18-fold increase in drug payload compared with physically loaded microcapsules. Furthermore, we investigated the drug release of RLMBs when exposed to an ultrasound field. After 120 s, an impressive 82.4 ± 5.5% of the loaded doxorubicin was released, demonstrating the remarkable capability of remotely loaded microbubbles for on-demand drug release. This study is the first to report such microbubbles that enable rapid drug release from the core. This innovative technique holds great promise in enhancing drug loading capacity and advancing targeted drug delivery.
Collapse
Affiliation(s)
| | | | - Shashank R. Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA; (G.R.); (M.M.S.)
| |
Collapse
|
5
|
Ahmed ETM, Hassan M, Shamma RN, Makky A, Hassan DH. Controlling the Evolution of Selective Vancomycin Resistance through Successful Ophthalmic Eye-Drop Preparation of Vancomycin-Loaded Nanoliposomes Using the Active-Loading Method. Pharmaceutics 2023; 15:1636. [PMID: 37376084 DOI: 10.3390/pharmaceutics15061636] [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: 04/28/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Vancomycin is the front-line defense and drug of choice for the most serious and life-threatening methicillin-resistant Staphylococcus aureus (MRSA) infections. However, poor vancomycin therapeutic practice limits its use, and there is a consequent rise of the threat of vancomycin resistance by complete loss of its antibacterial activity. Nanovesicles as a drug-delivery platform, with their featured capabilities of targeted delivery and cell penetration, are a promising strategy to resolve the shortcomings of vancomycin therapy. However, vancomycin's physicochemical properties challenge its effective loading. In this study, we used the ammonium sulfate gradient method to enhance vancomycin loading into liposomes. Depending on the pH difference between the extraliposomal vancomycin-Tris buffer solution (pH 9) and the intraliposomal ammonium sulfate solution (pH 5-6), vancomycin was actively and successfully loaded into liposomes (up to 65% entrapment efficiency), while the liposomal size was maintained at 155 nm. Vancomycin-loaded nanoliposomes effectively enhanced the bactericidal effect of vancomycin; the minimum inhibitory concentration (MIC) value for MRSA decreased 4.6-fold. Furthermore, they effectively inhibited and killed heteroresistant vancomycin-intermediate S.aureous (h-VISA) with an MIC of 0.338 μg mL-1. Moreover, MRSA could not develop resistance against vancomycin that was loaded into and delivered by liposomes. Vancomycin-loaded nanoliposomes could be a feasible solution for enhancing vancomycin's therapeutic use and controlling the emerging vancomycin resistance.
Collapse
Affiliation(s)
- El Tahra M Ahmed
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Giza 12585, Egypt
| | - Mariam Hassan
- Department of Microbiology and Immunology, Faculty of Pharmacy Cairo University, Cairo 12613, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, New Galala City, Suez 43511, Egypt
| | - Rehab Nabil Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy Cairo University, Cairo 12613, Egypt
| | - Amna Makky
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy Cairo University, Cairo 12613, Egypt
| | - Doaa H Hassan
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Giza 12585, Egypt
| |
Collapse
|
6
|
Fan N, Li Q, Liu Y, Ma B, Li M, Yin D. Preparation of an HI-6-loaded brain-targeted liposomes based on the nasal delivery route and the evaluation of its reactivation of central toxic acetylcholinesterase. Eur J Pharm Sci 2023; 184:106406. [PMID: 36805055 DOI: 10.1016/j.ejps.2023.106406] [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: 01/04/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
PURPOSE Organophosphorus compounds (OPs) is a serious threat to human health and life safety, but because of the existence of blood-brain barrier, most of the therapeutic drugs cannot enter the center, reactivate centrally located toxic acetylcholinesterase (AChE), it is urgent to find an efficient treatment method. METHODS The c(RGDyK) cyclic peptide modified HI-6-loaded brain targeting liposomes [c(RGDyK)-PEG2000HI-6-lipo] were prepared by ammonium sulfate gradient method. The in vitro blood-brain barrier (BBB) model was established, and the function of the liposomes was evaluated. The animal model of DDVP poisoning was established, and the central toxic enzyme reactivation ability of c(RGDyK)-PEG2000HI-6-lipo by both the intravenous and nasal administration route was verified. RESULTS The HI-6-loaded liposomes with brain targeting function were successfully synthesized and prepared with high encapsulation efficiency (70.23 ± 2.18%), drug loading (2.86 ± 0.07)%, average particle size 242.9 nm (polydispersion index 0.149), and ζ potential -16.2 mV. Combined with the in vitro and in vivo studies, the c(RGDyK)-PEG2000HI-6-lipo has better ability to cross the BBB. In addition, compared with intravenous injection, nasal administration was proved to be more effective against organophosphorus poisoning, and the reactivation rate of brain acetylcholinesterase reached (26.19 ± 7.70)%. CONCLUSION The prepared c(RGDyK)-PEG2000HI-6-lipo has a better ability to cross BBB. Nasal administration, as a way to bypass the BBB and directly deliver drugs into the brain, effectively improves the bioavailability of HI-6 in the brain. This study holds promise by providing a non-invasive approach to deliver water-soluble oxime antidote into the brain and reactivate central acetylcholinesterase via the naso-brain route.
Collapse
Affiliation(s)
- Ning Fan
- General Hospital of Xinjiang Military Command of the Chinese People's Liberation Army, Shaybak district, Urumqi, Xinjiang, China
| | - Qian Li
- General Hospital of Xinjiang Military Command of the Chinese People's Liberation Army, Shaybak district, Urumqi, Xinjiang, China
| | - Yuan Liu
- Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Bohua Ma
- Department of Pharmacy, Shihezi University, Shihezi, Xinjiang, China
| | - Meng Li
- Department of Pharmacy, Shihezi University, Shihezi, Xinjiang, China
| | - Dongfeng Yin
- General Hospital of Xinjiang Military Command of the Chinese People's Liberation Army, Shaybak district, Urumqi, Xinjiang, China.
| |
Collapse
|
7
|
Amini SM, Rezayat SM, Dinarvand R, Kharrazi S, Jaafari MR. Gold cluster encapsulated liposomes: theranostic agent with stimulus triggered release capability. Med Oncol 2023; 40:126. [PMID: 36961614 DOI: 10.1007/s12032-023-01991-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 03/07/2023] [Indexed: 03/25/2023]
Abstract
Cancer is a major cause of death worldwide. Cancer-resistant to chemo or radiotherapy treatment is a challenge that could be overcome by a nanotechnology approach. Providing a theranostic nano-platform for different cancer treatment strategies could be revolutionary. Here we introduce a multifunctional theranostic nanostructure which has the capacity for improving cancer diagnosis and treatment through better chemo and radiotherapy and current x-ray imaging systems through co-encapsulation of a small gold cluster and anticancer drug doxorubicin. 2 nm gold clusters represent good heating under radio frequency electric field (RF-EF) exposure and have been used for in vitro hyperthermia treatment of cancerous cells. Liposomal doxorubicin (169 ± 19.8 nm) with gold clusters encapsulation efficiency of 13.2 ± 3.0% and doxorubicin encapsulation efficiency of 64.7 ± 0.7% were prepared and studied as a theranostic agent with a high potential in different cancer treatment modalities. Exposure to a radiofrequency electric field on prepared formulation caused 20.2 ± 2.1% drug release and twice decreasing of IC50 on colorectal carcinoma cells. X-ray attenuation efficiency of the liposomal gold cluster was better than commercial iohexol and free gold clusters in different concentrations. Finally, treatment of gold clusters on cancerous cells results in a significant decrease in the viability of irradiated cells to cobalt-60 beam. Based on these experiments, we concluded that the conventional liposomal formulation of doxorubicin that has been co-encapsulated with small gold clusters could be a suitable theranostic nanostructure for cancer treatment and merits further investigation.
Collapse
Affiliation(s)
- Seyed Mohammad Amini
- Radiation Biology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Centre, Novel Drug Delivery Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sharmin Kharrazi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Mahmoud Reza Jaafari
- Pharmaceutical Technology Institute, School of Pharmacy, Nanotechnology Research Center, Mashhad University of Medical Sciences, 91775-1365, Mashhad, Iran.
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, 91775-1365, Mashhad, Iran.
| |
Collapse
|
8
|
Chen J. Preparation of Doxorubicin Liposomes by Remote Loading Method. Methods Mol Biol 2023; 2622:95-101. [PMID: 36781753 DOI: 10.1007/978-1-0716-2954-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Doxorubicin liposome is one of the most important nano-drug formulations. DOXIL, the first FDA-approved doxorubicin liposomes, is also the first nano-drug product in market. Since it was approved in 1995, DOXIL have been widely used in the treatment of various tumors. Several important technologies used in the development of doxorubicin liposomes, especially the remote loading technology, have an extremely important impact on the later liposome research and development. This article describes a protocol to prepare doxorubicin liposomes by remote loading in a laboratory.
Collapse
Affiliation(s)
- Jian Chen
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
9
|
Tentori P, Signore G, Camposeo A, Carretta A, Ferri G, Pingue P, Luin S, Pozzi D, Gratton E, Beltram F, Caracciolo G, Cardarelli F. Fluorescence lifetime microscopy unveils the supramolecular organization of liposomal Doxorubicin. NANOSCALE 2022; 14:8901-8905. [PMID: 35719059 DOI: 10.1039/d2nr00311b] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The supramolecular organization of Doxorubicin (DOX) within the standard Doxoves® liposomal formulation (DOX®) is investigated using visible light and phasor approach to fluorescence lifetime imaging (phasor-FLIM). First, the phasor-FLIM signature of DOX® is resolved into the contribution of three co-existing fluorescent species, each with its characteristic mono-exponential lifetime, namely: crystallized DOX (DOXc, 0.2 ns), free DOX (DOXf, 1.0 ns), and DOX bound to the liposomal membrane (DOXb, 4.5 ns). Then, the exact molar fractions of the three species are determined by combining phasor-FLIM with quantitative absorption/fluorescence spectroscopy on DOXc, DOXf, and DOXb pure standards. The final picture on DOX® comprises most of the drug in the crystallized form (∼98%), with the remaining fractions divided between free (∼1.4%) and membrane-bound drug (∼0.7%). Finally, phasor-FLIM in the presence of a DOX dynamic quencher allows us to suggest that DOXf is both encapsulated and non-encapsulated, and that DOXb is present on both liposome-membrane leaflets. We argue that the present experimental protocol can be applied to the investigation of the supramolecular organization of encapsulated luminescent drugs/molecules all the way from the production phase to their state within living matter.
Collapse
Affiliation(s)
- Paolo Tentori
- Laboratorio NEST, Scuola Normale Superiore, Pisa, Italy. E-mail:.
- Center for Nanotechnology Innovation @NEST, Pisa, Italy
| | | | - Andrea Camposeo
- NEST, Istituto Nanoscienze-CNR, Piazza S. Silvestro, 12, I-56127, Pisa, Italy
| | | | - Gianmarco Ferri
- Laboratorio NEST, Scuola Normale Superiore, Pisa, Italy. E-mail:.
| | - Pasqualantonio Pingue
- Laboratorio NEST, Scuola Normale Superiore, Pisa, Italy. E-mail:.
- NEST, Istituto Nanoscienze-CNR, Piazza S. Silvestro, 12, I-56127, Pisa, Italy
| | - Stefano Luin
- Laboratorio NEST, Scuola Normale Superiore, Pisa, Italy. E-mail:.
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California at Irvine, Irvine, California, USA
| | - Fabio Beltram
- Laboratorio NEST, Scuola Normale Superiore, Pisa, Italy. E-mail:.
- Center for Nanotechnology Innovation @NEST, Pisa, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesco Cardarelli
- Laboratorio NEST, Scuola Normale Superiore, Pisa, Italy. E-mail:.
- NEST, Istituto Nanoscienze-CNR, Piazza S. Silvestro, 12, I-56127, Pisa, Italy
| |
Collapse
|
10
|
Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon 2022; 8:e09394. [PMID: 35600452 PMCID: PMC9118483 DOI: 10.1016/j.heliyon.2022.e09394] [Citation(s) in RCA: 288] [Impact Index Per Article: 144.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/19/2022] [Accepted: 05/06/2022] [Indexed: 12/18/2022] Open
Abstract
Liposomes are now considered the most commonly used nanocarriers for various potentially active hydrophobic and hydrophilic molecules due to their high biocompatibility, biodegradability, and low immunogenicity. Liposomes also proved to enhance drug solubility and controlled distribution, as well as their capacity for surface modifications for targeted, prolonged, and sustained release. Based on the composition, liposomes can be considered to have evolved from conventional, long-circulating, targeted, and immune-liposomes to stimuli-responsive and actively targeted liposomes. Many liposomal-based drug delivery systems are currently clinically approved to treat several diseases, such as cancer, fungal and viral infections; more liposomes have reached advanced phases in clinical trials. This review describes liposomes structure, composition, preparation methods, and clinical applications.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Dima Khater
- Department of Chemistry, Faculty of Arts and Science, Applied Science Private University, Amman, Jordan
| | - Usama Sayed
- Department of Biology, The University of Jordan, Amman, 11942, Jordan
| | - Fadwa Odeh
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.,Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
11
|
Kaushik N, Borkar SB, Nandanwar SK, Panda PK, Choi EH, Kaushik NK. Nanocarrier cancer therapeutics with functional stimuli-responsive mechanisms. J Nanobiotechnology 2022; 20:152. [PMID: 35331246 PMCID: PMC8944113 DOI: 10.1186/s12951-022-01364-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Presently, nanocarriers (NCs) have gained huge attention for their structural ability, good biocompatibility, and biodegradability. The development of effective NCs with stimuli-responsive properties has acquired a huge interest among scientists. When developing drug delivery NCs, the fundamental goal is to tackle the delivery-related problems associated with standard chemotherapy and to carry medicines to the intended sites of action while avoiding undesirable side effects. These nanocarriers were able of delivering drugs to tumors through regulating their pH, temperature, enzyme responsiveness. With the use of nanocarriers, chemotherapeutic drugs could be supplied to tumors more accurately that can equally encapsulate and deliver them. Material carriers for chemotherapeutic medicines are discussed in this review keeping in viewpoint of the structural properties and targeting methods that make these carriers more therapeutically effective, in addition to metabolic pathways triggered by drug-loaded NCs. Largely, the development of NCs countering to endogenous and exogenous stimuli in tumor regions and understanding of mechanisms would encourage the progress for tumor therapy and precision diagnosis in future.
Collapse
Affiliation(s)
- Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong, 18323, Republic of Korea.
| | - Shweta B Borkar
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Sondavid K Nandanwar
- Department of Basic Science Research Institute, Pukyong National University, Busan, 48513, Korea
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Department of Physics and Astronomy, Uppsala University, Box 516, S-75120, Uppsala, Sweden
| | - Eun Ha Choi
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Nagendra Kumar Kaushik
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Republic of Korea.
| |
Collapse
|
12
|
Zhang C, Ma Y, Zhang J, Kuo JCT, Zhang Z, Xie H, Zhu J, Liu T. Modification of Lipid-Based Nanoparticles: An Efficient Delivery System for Nucleic Acid-Based Immunotherapy. Molecules 2022; 27:molecules27061943. [PMID: 35335310 PMCID: PMC8949521 DOI: 10.3390/molecules27061943] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Lipid-based nanoparticles (LBNPs) are biocompatible and biodegradable vesicles that are considered to be one of the most efficient drug delivery platforms. Due to the prominent advantages, such as long circulation time, slow drug release, reduced toxicity, high transfection efficiency, and endosomal escape capacity, such synthetic nanoparticles have been widely used for carrying genetic therapeutics, particularly nucleic acids that can be applied in the treatment for various diseases, including congenital diseases, cancers, virus infections, and chronic inflammations. Despite great merits and multiple successful applications, many extracellular and intracellular barriers remain and greatly impair delivery efficacy and therapeutic outcomes. As such, the current state of knowledge and pitfalls regarding the gene delivery and construction of LBNPs will be initially summarized. In order to develop a new generation of LBNPs for improved delivery profiles and therapeutic effects, the modification strategies of LBNPs will be reviewed. On the basis of these developed modifications, the performance of LBNPs as therapeutic nanoplatforms have been greatly improved and extensively applied in immunotherapies, including infectious diseases and cancers. However, the therapeutic applications of LBNPs systems are still limited due to the undesirable endosomal escape, potential aggregation, and the inefficient encapsulation of therapeutics. Herein, we will review and discuss recent advances and remaining challenges in the development of LBNPs for nucleic acid-based immunotherapy.
Collapse
Affiliation(s)
- Chi Zhang
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Z.Z.)
| | - Yifan Ma
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; (Y.M.); (J.Z.)
| | - Jingjing Zhang
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; (Y.M.); (J.Z.)
| | - Jimmy Chun-Tien Kuo
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Z.Z.)
| | - Zhongkun Zhang
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Z.Z.)
| | - Haotian Xie
- Department of Statistics, The Ohio State University, Columbus, OH 43210, USA;
| | - Jing Zhu
- College of Nursing and Health Innovation, The University of Texas Arlington, Arlington, TX 76010, USA
- Correspondence: (J.Z.); (T.L.); Tel.: +1-614-570-1164 (J.Z.); +86-186-6501-3854 (T.L.)
| | - Tongzheng Liu
- College of Pharmacy, Jinan University, Guangzhou 511443, China
- Correspondence: (J.Z.); (T.L.); Tel.: +1-614-570-1164 (J.Z.); +86-186-6501-3854 (T.L.)
| |
Collapse
|
13
|
Amin M, Mansourian M, Burgers PC, Amin B, Jaafari MR, ten Hagen TLM. Increased Targeting Area in Tumors by Dual-Ligand Modification of Liposomes with RGD and TAT Peptides. Pharmaceutics 2022; 14:pharmaceutics14020458. [PMID: 35214190 PMCID: PMC8878433 DOI: 10.3390/pharmaceutics14020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Modification with polyethylene glycol (PEGylation) and the use of rigid phospholipids drastically improve the pharmacokinetics of chemotherapeutics and result in more manageable or reduced side-effects. A major drawback is retarded cellular delivery of content, which, along with tumor heterogeneity, are the two main obstacles against tumor targeting. To enhance cellular delivery and reach a bigger area of a tumor, we designed liposomes decorated with two ligands: one for targeting tumor vasculature via a cyclic-pentapeptide containing arginine-glycine-aspartic acid (RGD), which impacts tumor independent of passive accumulation inside tumors, and one for extravascular targeting of tumor cells via a cell-penetrating peptide derived from human immunodeficiency virus type 1 transactivator of transcription (TAT). Liposomes with different ligand combinations were prepared and compared with respect to performance in targeting. Intravital imaging illustrates the heterogeneous behavior of RGD-liposomes in both intravascular and extravascular distribution, whereas TAT-liposomes exhibit a predictable extravascular localization but no intravascular targeting. Dual-ligand modification results in enhanced vascular targeting and a predictable extravascular behavior that improves the therapeutic efficacy of doxorubicin-loaded liposomes but also an augmented clearance rate of liposomes. However, the dual-modified liposome could be a great candidate for targeted delivery of non-toxic payloads or contrast agents for therapeutic or diagnostic purposes. Here we show that the combination of vascular-specific and tumor cell-specific ligands in a liposomal system is beneficial in bypassing the heterogeneous expression of tumor-specific markers.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Correspondence:
| | - Mercedeh Mansourian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9196773117, Iran; (M.M.); (M.R.J.)
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Peter C. Burgers
- Laboratory of Neuro-Oncology, Department of Neurology, Erasmus MC University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands;
| | - Bahareh Amin
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar 9613873136, Iran;
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9196773117, Iran; (M.M.); (M.R.J.)
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Timo L. M. ten Hagen
- Laboratory Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| |
Collapse
|
14
|
Barbălată CI, Porfire AS, Sesarman A, Rauca VF, Banciu M, Muntean D, Știufiuc R, Moldovan A, Moldovan C, Tomuță I. A Screening Study for the Development of Simvastatin-Doxorubicin Liposomes, a Co-Formulation with Future Perspectives in Colon Cancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13101526. [PMID: 34683821 PMCID: PMC8537800 DOI: 10.3390/pharmaceutics13101526] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 01/25/2023] Open
Abstract
An increasing number of studies published so far have evidenced the benefits of Simvastatin (SIM) and Doxorubicin (DOX) co-treatment in colorectal cancer. In view of this, the current study aimed to investigate the pharmaceutical development of liposomes co-encapsulating SIM and DOX, by implementing the Quality by Design (QbD) concept, as a means to enhance the antiproliferative effect of the co-formulation on C26 murine colon cancer cells co-cultured with macrophages. It is known that the quality profile of liposomes is dependent on the critical quality attributes (CQAs) of liposomes (drug entrapped concentration, encapsulation efficiency, size, zeta potential, and drug release profile), which are, in turn, directly influenced by various formulation factors and processing parameters. By using the design of experiments, it was possible to outline the increased variability of CQAs in relation to formulation factors and identify by means of statistical analysis the material attributes that are critical (phospholipids, DOX and SIM concentration) for the quality of the co-formulation. The in vitro studies performed on a murine colon cancer cell line highlighted the importance of delivering the optimal drug ratio at the target site, since the balance antiproliferative vs. pro-proliferative effects can easily be shifted when the molar ratio between DOX and SIM changes.
Collapse
Affiliation(s)
- Cristina Ioana Barbălată
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| | - Alina Silvia Porfire
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
- Correspondence:
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Valentin-Florian Rauca
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Dana Muntean
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| | - Rareș Știufiuc
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Alin Moldovan
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Cristian Moldovan
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Ioan Tomuță
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| |
Collapse
|
15
|
Kousani S, Karimi M, Teymouri M, Navashenaq JG, Darban SA, Jaafari MR. Antennapedia-derived positively-charged peptide faces multiple problems upon their usage as targeting ligand for liposomal doxorubicin. Biotechnol Prog 2021; 37:e3202. [PMID: 34405574 DOI: 10.1002/btpr.3202] [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: 03/15/2021] [Revised: 07/18/2021] [Accepted: 08/11/2021] [Indexed: 11/09/2022]
Abstract
The positively-charged peptide antp derived from Antennapedia transcription protein is demonstrated to mediate the liposome translocation across the cell membrane. In the current investigation, we prepared a stable liposomal doxorubicin (Dox) formulation and targeted it with the antp peptide from 0 to 200 ligand/liposome. These antp-containing liposomes were investigated in terms of physical stability on storage in the refrigerator and upon incubation in blood. Also, other features like cell binding, uptake, biodistribution, and treatment efficiency were evaluated in C26 colon carcinoma BALB/c mice. The Antp in liposomes resulted in enhanced particle growth with the development of the enormously large liposomes from 2000 to 6000 nm. Upon incubation in blood, these large liposomes were removed. The antp also enhanced the cell binding affinity and cell uptake rate of the liposomes and resulted in the restriction of the cancer cell proliferation, but it failed to improve the chemotherapeutic property of the Dox-liposome. The i.v. injection of antp-liposomes (15 mg Dox/kg) caused severe body weight loss and early death incidence due to probably increased toxicity. The antp targeting offered no advantage to the Dox-liposome in the delivery of Dox to the tumor, and failed to enhance the treatment efficiency of the liposomes.
Collapse
Affiliation(s)
- Sima Kousani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Karimi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Manouchehr Teymouri
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnord, Iran
| | - Jamshid Gholizadeh Navashenaq
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Noncommunicable Disease Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Shahrzad Amiri Darban
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
García MC, Naitlho N, Calderón-Montaño JM, Drago E, Rueda M, Longhi M, Rabasco AM, López-Lázaro M, Prieto-Dapena F, González-Rodríguez ML. Cholesterol Levels Affect the Performance of AuNPs-Decorated Thermo-Sensitive Liposomes as Nanocarriers for Controlled Doxorubicin Delivery. Pharmaceutics 2021; 13:pharmaceutics13070973. [PMID: 34199018 PMCID: PMC8309145 DOI: 10.3390/pharmaceutics13070973] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Stimulus-responsive liposomes (L) for triggering drug release to the target site are particularly useful in cancer therapy. This research was focused on the evaluation of the effects of cholesterol levels in the performance of gold nanoparticles (AuNPs)-functionalized L for controlled doxorubicin (D) delivery. Their interfacial and morphological properties, drug release behavior against temperature changes and cytotoxic activity against breast and ovarian cancer cells were studied. Langmuir isotherms were performed to identify the most stable combination of lipid components. Two mole fractions of cholesterol (3.35 mol% and 40 mol%, L1 and L2 series, respectively) were evaluated. Thin-film hydration and transmembrane pH-gradient methods were used for preparing the L and for D loading, respectively. The cationic surface of L allowed the anchoring of negatively charged AuNPs by electrostatic interactions, even inducing a shift in the zeta potential of the L2 series. L exhibited nanometric sizes and spherical shape. The higher the proportion of cholesterol, the higher the drug loading. D was released in a controlled manner by diffusion-controlled mechanisms, and the proportions of cholesterol and temperature of release media influenced its release profiles. D-encapsulated L preserved its antiproliferative activity against cancer cells. The developed liposomal formulations exhibit promising properties for cancer treatment and potential for hyperthermia therapy.
Collapse
Affiliation(s)
- Mónica C. García
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Haya de la Torre and Medina Allende, Science Building 2, Córdoba X5000HUA, Argentina;
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, UNITEFA, Córdoba X5000HUA, Argentina
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain; (N.N.); (A.M.R.)
- Correspondence: (M.C.G.); (M.L.G.-R.); Tel./Fax: +54-351-5353865 (M.C.G.); +34-954556397 (M.L.G.-R.)
| | - Nabila Naitlho
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain; (N.N.); (A.M.R.)
| | - José Manuel Calderón-Montaño
- Department of Pharmacology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain; (J.M.C.-M.); (M.L.-L.)
| | - Estrella Drago
- Department of Physical Chemistry, Faculty of Chemistry, Universidad de Sevilla, C/Prof. García González s/n, 41012 Seville, Spain; (E.D.); (M.R.); (F.P.-D.)
| | - Manuela Rueda
- Department of Physical Chemistry, Faculty of Chemistry, Universidad de Sevilla, C/Prof. García González s/n, 41012 Seville, Spain; (E.D.); (M.R.); (F.P.-D.)
| | - Marcela Longhi
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Haya de la Torre and Medina Allende, Science Building 2, Córdoba X5000HUA, Argentina;
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, UNITEFA, Córdoba X5000HUA, Argentina
| | - Antonio M. Rabasco
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain; (N.N.); (A.M.R.)
| | - Miguel López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain; (J.M.C.-M.); (M.L.-L.)
| | - Francisco Prieto-Dapena
- Department of Physical Chemistry, Faculty of Chemistry, Universidad de Sevilla, C/Prof. García González s/n, 41012 Seville, Spain; (E.D.); (M.R.); (F.P.-D.)
| | - María Luisa González-Rodríguez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain; (N.N.); (A.M.R.)
- Correspondence: (M.C.G.); (M.L.G.-R.); Tel./Fax: +54-351-5353865 (M.C.G.); +34-954556397 (M.L.G.-R.)
| |
Collapse
|
17
|
Omabe K, Paris C, Lannes F, Taïeb D, Rocchi P. Nanovectorization of Prostate Cancer Treatment Strategies: A New Approach to Improved Outcomes. Pharmaceutics 2021; 13:591. [PMID: 33919150 PMCID: PMC8143094 DOI: 10.3390/pharmaceutics13050591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PC) is the most frequent male cancer in the Western world. Progression to Castration Resistant Prostate Cancer (CRPC) is a known consequence of androgen withdrawal therapy, making CRPC an end-stage disease. Combination of cytotoxic drugs and hormonal therapy/or genotherapy is a recognized modality for the treatment of advanced PC. However, this strategy is limited by poor bio-accessibility of the chemotherapy to tumor sites, resulting in an increased rate of collateral toxicity and incidence of multidrug resistance (MDR). Nanovectorization of these strategies has evolved to an effective approach to efficacious therapeutic outcomes. It offers the possibility to consolidate their antitumor activity through enhanced specific and less toxic active or passive targeting mechanisms, as well as enabling diagnostic imaging through theranostics. While studies on nanomedicine are common in other cancer types, only a few have focused on prostate cancer. This review provides an in-depth knowledge of the principles of nanotherapeutics and nanotheranostics, and how the application of this rapidly evolving technology can clinically impact CRPC treatment. With particular reference to respective nanovectors, we draw clinical and preclinical evidence, demonstrating the potentials and prospects of homing nanovectorization into CRPC treatment strategies.
Collapse
Affiliation(s)
- Kenneth Omabe
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
- Department of Biochemistry & Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki 84001, Nigeria
| | - Clément Paris
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| | - François Lannes
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| | - David Taïeb
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
- Biophysics and Nuclear Medicine, La Timone University Hospital, European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Palma Rocchi
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| |
Collapse
|
18
|
Ferreira M, Pinto SN, Aires-da-Silva F, Bettencourt A, Aguiar SI, Gaspar MM. Liposomes as a Nanoplatform to Improve the Delivery of Antibiotics into Staphylococcus aureus Biofilms. Pharmaceutics 2021; 13:pharmaceutics13030321. [PMID: 33801281 PMCID: PMC7999762 DOI: 10.3390/pharmaceutics13030321] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus biofilm-associated infections are a major public health concern. Current therapies are hampered by reduced penetration of antibiotics through biofilm and low accumulation levels at infected sites, requiring prolonged usage. To overcome these, repurposing antibiotics in combination with nanotechnological platforms is one of the most appealing fast-track and cost-effective approaches. In the present work, we assessed the potential therapeutic benefit of three antibiotics, vancomycin, levofloxacin and rifabutin (RFB), through their incorporation in liposomes. Free RFB displayed the utmost antibacterial effect with MIC and MBIC50 below 0.006 µg/mL towards a methicillin susceptible S. aureus (MSSA). RFB was selected for further in vitro studies and the influence of different lipid compositions on bacterial biofilm interactions was evaluated. Although positively charged RFB liposomes displayed the highest interaction with MSSA biofilms, RFB incorporated in negatively charged liposomes displayed lower MBIC50 values in comparison to the antibiotic in the free form. Preliminary safety assessment on all RFB formulations towards osteoblast and fibroblast cell lines demonstrated that a reduction on cell viability was only observed for the positively charged liposomes. Overall, negatively charged RFB liposomes are a promising approach against biofilm S. aureus infections and further in vivo studies should be performed.
Collapse
Affiliation(s)
- Magda Ferreira
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.F.); (F.A.-d.-S.)
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Sandra N. Pinto
- Department of Bioengineering, iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal;
| | - Frederico Aires-da-Silva
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.F.); (F.A.-d.-S.)
| | - Ana Bettencourt
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Sandra I. Aguiar
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.F.); (F.A.-d.-S.)
- Correspondence: (S.I.A.); (M.M.G.)
| | - Maria Manuela Gaspar
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisboa, Portugal;
- Correspondence: (S.I.A.); (M.M.G.)
| |
Collapse
|
19
|
Allen SD, Liu X, Jiang J, Liao YP, Chang CH, Nel AE, Meng H. Immune checkpoint inhibition in syngeneic mouse cancer models by a silicasome nanocarrier delivering a GSK3 inhibitor. Biomaterials 2020; 269:120635. [PMID: 33422940 DOI: 10.1016/j.biomaterials.2020.120635] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 12/21/2020] [Accepted: 12/27/2020] [Indexed: 12/18/2022]
Abstract
Checkpoint blocking antibodies that interfere in the PD-1/PD-L1 axis provide effective cancer immunotherapy for tumors that are immune inflamed or induced to become "hot". It has also been demonstrated that a small molecule inhibitor of the signaling hub kinase GSK3 can interfere in the PD-1/PD-L1 axis in T-cells by suppressing PD-1 expression. This provides an alternative approach to intervening in the PD-1/PD-L1 axis to provide cancer immunotherapy. In this communication, we demonstrate the remote loading of GSK3 inhibitor AZD1080 into the porous interior of mesoporous silica nanoparticles coated with a lipid bilayer (a.k.a. silicasomes). In a MC38 colon cancer model, intravenous injection (IV) of silicasome-encapsulated AZD1080 significantly improved biodistribution and drug delivery to the tumor site. The improved drug delivery was accompanied by cytotoxic MC38 tumor cell killing by perforin-releasing CD8+ T-cells, exhibiting reduced PD-1 expression. IV injection of encapsulated AZD1080 also resulted in significant tumor shrinkage in other syngeneic mouse tumor models, including another colorectal tumor (CT26), as well as pancreas (KPC) and lung (LLC) cancer models. Not only was the therapeutic efficacy of encapsulated AZD1080 similar or better than anti-PD-1 antibody, but the treatment was devoid of treatment toxicity. These results provide proof-of-principal demonstration of the feasibility of using encapsulated delivery of a GSK3 inhibitor to provide cancer immunotherapy, with the possibility to be used as a monotherapy or in combination with chemotherapy or other immunomodulatory agents.
Collapse
Affiliation(s)
- Sean D Allen
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Xiangsheng Liu
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA; California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Jinhong Jiang
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Yu-Pei Liao
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Chong Hyun Chang
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA
| | - Andre E Nel
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA; California NanoSystems Institute, University of California, Los Angeles, CA, USA.
| | - Huan Meng
- Department of Medicine, Division of NanoMedicine, University of California, Los Angeles, CA, USA; California NanoSystems Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Almeida B, Nag OK, Rogers KE, Delehanty JB. Recent Progress in Bioconjugation Strategies for Liposome-Mediated Drug Delivery. Molecules 2020; 25:E5672. [PMID: 33271886 PMCID: PMC7730700 DOI: 10.3390/molecules25235672] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
In nanoparticle (NP)-mediated drug delivery, liposomes are the most widely used drug carrier, and the only NP system currently approved by the FDA for clinical use, owing to their advantageous physicochemical properties and excellent biocompatibility. Recent advances in liposome technology have been focused on bioconjugation strategies to improve drug loading, targeting, and overall efficacy. In this review, we highlight recent literature reports (covering the last five years) focused on bioconjugation strategies for the enhancement of liposome-mediated drug delivery. These advances encompass the improvement of drug loading/incorporation and the specific targeting of liposomes to the site of interest/drug action. We conclude with a section highlighting the role of bioconjugation strategies in liposome systems currently being evaluated for clinical use and a forward-looking discussion of the field of liposomal drug delivery.
Collapse
Affiliation(s)
- Bethany Almeida
- American Society for Engineering Education, Washington, DC 20036, USA;
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| | - Okhil K. Nag
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| | - Katherine E. Rogers
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
- Fischell Department of Bioengineering, 2330 Kim Engineering Building, University of Maryland, College Park, MD 20742, USA
| | - James B. Delehanty
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| |
Collapse
|
21
|
Li B, London E. Preparation and Drug Entrapment Properties of Asymmetric Liposomes Containing Cationic and Anionic Lipids. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:12521-12531. [PMID: 33070610 DOI: 10.1021/acs.langmuir.0c01968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We have developed cyclodextrin-catalyzed lipid exchange methods to prepare large unilamellar vesicles (LUVs) with asymmetric charge distributions, i.e., with different net charges on the lipids in the inner and outer leaflets. LUVs contained a mixture of a zwitterionic lipid (phosphatidylcholine), cholesterol, and various cationic lipids (O-ethyl phosphatidylcholine or dioleoyl-3-trimethylammonium propane) or anionic lipids (phosphatidylglycerol, phosphatidylserine, or phosphatidic acid). Symmetric and asymmetric LUVs with a wide variety of lipid combinations were prepared. The asymmetric LUVs contained cationic or anionic outer leaflets and inner leaflets that had either the opposite charge or were uncharged. The behavior of symmetric LUVs prepared with zwitterionic, anionic, or cationic leaflets was compared to those of asymmetric LUVs. Lipid exchange was confirmed by quantitative thin-layer chromatography, and lipid asymmetry by a novel assay measuring binding of a cationic fluorescent probe to the LUV outer leaflet. For both symmetric and asymmetric LUVs, the level of entrapment of the cationic drug doxorubicin was controlled by the charge on the inner leaflet, with the greatest entrapment and slowest leakage in vesicles with an anionic inner leaflet. This shows that it is possible to choose inner leaflet lipids to maximize liposomal loading of charged drugs independently of the identity of outer-leaflet lipids. This implies that it should also be possible to independently vary outer-leaflet lipids to, for example, impart favorable bioavailability and biodistribution properties to lipid vesicles.
Collapse
Affiliation(s)
- Bingchen Li
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| | - Erwin London
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| |
Collapse
|
22
|
Saraf S, Jain A, Tiwari A, Verma A, Jain SK. Engineered liposomes bearing camptothecin analogue for tumour targeting: in vitro and ex-vivo studies. J Liposome Res 2020; 31:326-341. [PMID: 32718195 DOI: 10.1080/08982104.2020.1801725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Shivani Saraf
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, India
| | - Ankit Jain
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, India
- Department of Materials Engineering, Indian Institute of Science, Bangalore, India
| | - Ankita Tiwari
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, India
| | - Amit Verma
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, India
| | - Sanjay K. Jain
- Pharmaceutics Research Projects Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, India
| |
Collapse
|
23
|
Chowdhury N, Chaudhry S, Hall N, Olverson G, Zhang QJ, Mandal T, Dash S, Kundu A. Targeted Delivery of Doxorubicin Liposomes for Her-2+ Breast Cancer Treatment. AAPS PharmSciTech 2020; 21:202. [PMID: 32696338 PMCID: PMC7995642 DOI: 10.1208/s12249-020-01743-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/25/2020] [Indexed: 01/06/2023] Open
Abstract
The adverse side effects and toxicity caused by the non-targeted delivery of doxorubicin has emphasized the demand of emerging a targeted delivery system. The goal of this study is to enhance the delivery of doxorubicin by formulating an aptamer-labeled liposomal nanoparticle delivery system that will carry and deliver doxorubicin specifically into Her-2+ breast cancer cells. Twelve liposomal batches were prepared using different saturated (HSPC and DPPC) and unsaturated (POPC and DOPC) lipids by thin film hydration. The liposomes were characterized for their particle size, zeta potential, and drug encapsulation efficiency. The particles were also assessed for in vitro toxicity and DOX delivery into the breast cancer cells. The formulations, F1 through F12, had a small particle size of less than 200 nm and a high entrapment efficiency of about 88 ± 5%. The best formulation, F5, had a particle size of 101 ± 14nm, zeta potential of + 5.63 ± 0.46 mV, and entrapment efficiency of ≈ 93%. The cytotoxicity studies show that the DOX-loaded liposomal formulations are more effective in killing cancer cells than the free DOX in both MCF-7 and SKBR-3 cells. The uptake studies show a significant increase in the uptake of the aptamer-labeled liposomes (i.e., F5) by more than 60% into Her-2+ MCF-7 and SKBR-3 breast cancer cells compare to non-aptamer-labeled nanoparticles. F5 also shows ≈ 1.79-fold increase in uptake of DOX in the Her-2+ cells compared to the Her-2- cells. This preliminary study indicates that aptamer-labeled F5 nanoparticles among several batches showed the highest uptake as well as the targeted delivery of doxorubicin into Her-2+ breast cancer cells. Thus, aptamer targeted approach results in substantial reduction in the dose of DOX and improves the therapeutic benefits by promoting the target specificity.
Collapse
Affiliation(s)
- Nusrat Chowdhury
- Department of Biology, Xavier University of Louisiana, 1 Drexel Dr, New Orleans, Louisiana, 70125-1098, USA
| | - Shanzay Chaudhry
- Department of Biology, Xavier University of Louisiana, 1 Drexel Dr, New Orleans, Louisiana, 70125-1098, USA
| | - Nicholas Hall
- Department of Biology, Xavier University of Louisiana, 1 Drexel Dr, New Orleans, Louisiana, 70125-1098, USA
| | - George Olverson
- Department of Biology, Xavier University of Louisiana, 1 Drexel Dr, New Orleans, Louisiana, 70125-1098, USA
| | - Qian-Jin Zhang
- Department of Biology, Xavier University of Louisiana, 1 Drexel Dr, New Orleans, Louisiana, 70125-1098, USA
| | - Tarun Mandal
- Center for Nanomedicine and Drug Delivery, Xavier University College of Pharmacy, New Orleans, Louisiana, 70125, USA
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, 70112, USA
| | - Anup Kundu
- Department of Biology, Xavier University of Louisiana, 1 Drexel Dr, New Orleans, Louisiana, 70125-1098, USA.
| |
Collapse
|
24
|
Licarete E, Rauca VF, Luput L, Drotar D, Stejerean I, Patras L, Dume B, Toma VA, Porfire A, Gherman C, Sesarman A, Banciu M. Overcoming Intrinsic Doxorubicin Resistance in Melanoma by Anti-Angiogenic and Anti-Metastatic Effects of Liposomal Prednisolone Phosphate on Tumor Microenvironment. Int J Mol Sci 2020; 21:ijms21082968. [PMID: 32340166 PMCID: PMC7215436 DOI: 10.3390/ijms21082968] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 12/31/2022] Open
Abstract
Regardless of recent progress, melanoma is very difficult to treat, mainly due to the drug resistance modulated by tumor cells as well as by the tumor microenvironment (TME). Among the immune cells recruited at the tumor site, tumor associated macrophages (TAMs) are the most abundant, promoting important tumorigenic processes: angiogenesis, inflammation and invasiveness. Furthermore, it has been shown that TAMs are involved in mediating the drug resistance of melanoma cells. Thus, in the present study, we used liposomal formulation of prednisolone disodium phosphate (LCL-PLP) to inhibit the protumor function of TAMs with the aim to sensitize the melanoma cells to the cytotoxic drug doxorubicin (DOX) to which human melanoma has intrinsic resistance. Consequently, we evaluated the in vivo effects of the concomitant administration of LCL-PLP and liposomal formulation of DOX (LCL-DOX) on B16.F10 melanoma growth and on the production of key molecular markers for tumor development. Our results demonstrated that the concomitant administration of LCL-PLP and LCL-DOX induced a strong inhibition of tumor growth, primarily by inhibiting TAMs-mediated angiogenesis as well as the tumor production of MMP-2 and AP-1. Moreover, our data suggested that the combined therapy also affected TME as the number of infiltrated macrophages in melanoma microenvironment was reduced significantly.
Collapse
Affiliation(s)
- Emilia Licarete
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400271 Cluj-Napoca, Romania
| | - Valentin Florian Rauca
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Lavinia Luput
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Denise Drotar
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
| | - Ioana Stejerean
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Bogdan Dume
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
| | - Vlad Alexandru Toma
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Institute of Biological Research, 400015 Cluj-Napoca, Romania
- National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Claudia Gherman
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
- Correspondence: ; Tel.: +40-264-431-691; Fax: +40-264-431-858
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| |
Collapse
|
25
|
Karimi Zarchi AA, Amini SM, Salimi A, Kharazi S. Synthesis and characterisation of liposomal doxorubicin with loaded gold nanoparticles. IET Nanobiotechnol 2019; 12:846-849. [PMID: 30104461 DOI: 10.1049/iet-nbt.2017.0321] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Developing nanostructures for cancer treatment is growing significantly. Liposomal doxorubicin is a drug that is used in the clinic and represents a lot of benefits over doxorubicin. The development of multifunctional liposomes with different cancer treatment capability enables broader applications of doxorubicin chemotherapy. Many efforts were carried to prepare more effective liposomal formulation through loading gold nanoparticles (GNPs) in the formulation. Here, GNPs with an average size of 6 nm were loaded in liposomal formulation alongside doxorubicin. The hydrodynamic diameter of final formulation was 177.3 ± 33.9 nm that in comparison with liposomes without GNPs (112.5 ± 10.3 nm), GNPs-loaded liposomes showed the bigger hydrodynamic diameter. GNPs-loaded liposomes are slightly positively charged (4.4 ± 1.1 mV), while liposomes without loading the GNPs were negatively charged (-18.5 ± 1.6 mV). Doxorubicin was loaded in this formulation through active loading technique. Doxorubicin loading efficiency in gold-loaded liposomes is slightly lesser than liposomes without GNPs, but still considerably high in comparison to passive loading techniques.
Collapse
Affiliation(s)
- Ali Akbar Karimi Zarchi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Mohamad Amini
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sharmin Kharazi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
26
|
Cao D, Zhang X, Akabar MD, Luo Y, Wu H, Ke X, Ci T. Liposomal doxorubicin loaded PLGA-PEG-PLGA based thermogel for sustained local drug delivery for the treatment of breast cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:181-191. [DOI: 10.1080/21691401.2018.1548470] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Dinglingge Cao
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P. R. China
| | - Xingxian Zhang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P. R. China
| | - MD. Akabar
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P. R. China
| | - Yuan Luo
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P. R. China
| | - Hao Wu
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P. R. China
| | - Xue Ke
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P. R. China
| | - Tianyuan Ci
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P. R. China
| |
Collapse
|
27
|
Zheng H, Li J, Wang M, Luo X, Qiu Q, Hu L, Li C, Song Y, Deng Y. Exhausting tumor associated macrophages with sialic acid-polyethyleneimine-cholesterol modified liposomal doxorubicin for enhancing sarcoma chemotherapy. Int J Pharm 2019; 558:187-200. [PMID: 30654062 DOI: 10.1016/j.ijpharm.2019.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/13/2018] [Accepted: 01/01/2019] [Indexed: 02/03/2023]
Abstract
To overstep the dilemma of chemical drug degradation within powerful lysosomes of tumor associated macrophages (TAMs), a sialic acid-polyethylenimine-cholesterol (SA-PEI-CH) modified liposomal doxorubicin (DOX-SPCL) was designed with both TAMs targeting and smart lysosomal trafficking. The modified liposome DOX-SPCL performed particle size as 103.2 ± 3.1 nm and zeta potential as -4.5 ± 0.9 mV with encapsulation efficiency as 95.8 ± 0.5%. In in vitro cell experiments, compared with conventional liposomal doxorubicin (DOX-CL) and PEGylated liposomal doxorubicin (DOX-PL), DOX-SPCL showed a selective binding on TAMs and a mere lysosomal concentration. In pharmacokinetic study, DOX-SPCL effectively impeded/delayed the disposition of mononuclear phagocyte system (MPS) with a value of AUC0-t as 796.03 ± 66.93 mg L-1 h. In S180 sarcomas bearing mice, DOX-SPCL showed the greatest tumor inhibition rate (92.7% ± 3.6%) compared with DOX-CL (46.4% ± 2.0%) or DOX-PL (58.8% ± 7.6%). The <0.5% positive region of TAMs in tumor section indicated a super TAMs exhaustion for DOX-SPCL treatment. Conclusively, DOX-SPCL was supposed as a safe and effective liposomal preparation for clinical sarcoma treatment via TAMs targeting/deletion delivery strategy.
Collapse
Affiliation(s)
- Huangliang Zheng
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Jiaqi Li
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Mengjing Wang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Xiang Luo
- College of Chemistry and Chemical Engineering, Shaoxing University, No. 508 Huancheng West Road, Shaoxing, Zhejiang 312000, China
| | - Qiujun Qiu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Ling Hu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Cong Li
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Yanzhi Song
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning 110016, China.
| | - Yihui Deng
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning 110016, China.
| |
Collapse
|
28
|
Development of doxorubicin hydrochloride loaded pH-sensitive liposomes: Investigation on the impact of chemical nature of lipids and liposome composition on pH-sensitivity. Eur J Pharm Biopharm 2018; 133:331-338. [DOI: 10.1016/j.ejpb.2018.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/20/2018] [Accepted: 11/02/2018] [Indexed: 12/11/2022]
|
29
|
Farzaneh H, Ebrahimi Nik M, Mashreghi M, Saberi Z, Jaafari MR, Teymouri M. A study on the role of cholesterol and phosphatidylcholine in various features of liposomal doxorubicin: From liposomal preparation to therapy. Int J Pharm 2018; 551:300-308. [DOI: 10.1016/j.ijpharm.2018.09.047] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 10/28/2022]
|
30
|
Oliveira JD, Ribeiro LNDM, Rodrigues da Silva GH, Casadei BR, Couto VM, Martinez EF, de Paula E. Sustained Release from Ionic-Gradient Liposomes Significantly Decreases ETIDOCAINE Cytotoxicity. Pharm Res 2018; 35:229. [DOI: 10.1007/s11095-018-2512-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022]
|
31
|
Khorasani S, Danaei M, Mozafari M. Nanoliposome technology for the food and nutraceutical industries. Trends Food Sci Technol 2018. [DOI: 10.1016/j.tifs.2018.07.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
32
|
Mai J, Li X, Zhang G, Huang Y, Xu R, Shen Q, Lokesh GL, Thiviyanathan V, Chen L, Liu H, Zu Y, Ma X, Volk DE, Gorenstein DG, Ferrari M, Shen H. DNA Thioaptamer with Homing Specificity to Lymphoma Bone Marrow Involvement. Mol Pharm 2018; 15:1814-1825. [PMID: 29537266 PMCID: PMC6311132 DOI: 10.1021/acs.molpharmaceut.7b01169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Selective drug accumulation in the malignant tissue is a prerequisite for effective cancer treatment. However, most drug molecules and their formulated particles are blocked en route to the destiny tissue due to the existence of multiple biological and physical barriers including the tumor microvessel endothelium. Since the endothelial cells on the surface of the microvessel wall can be modulated by inflammatory cytokines and chemokines secreted by the tumor or stromal cells, an effective drug delivery approach is to enhance interaction between the drug particles and the unique spectrum of surface proteins on the tumor endothelium. In this study, we performed in vivo screening for thioaptamers that bind to the bone marrow endothelium with specificity in a murine model of lymphoma with bone marrow involvement (BMI). The R1 thioaptamer was isolated based on its high homing potency to bones with BMI, and 40-60% less efficiency in accumulation to healthy bones. In cell culture, R1 binds to human umbilical vein endothelial cells (HUVEC) with a high affinity ( Kd ≈ 3 nM), and the binding affinity can be further enhanced when cells were treated with a mixture of lymphoma cell and bone marrow cell conditioned media. Cellular uptake of R1 is through clathrin-mediated endocytosis. Conjugating R1 on to the surface of liposomal doxorubicin nanoparticles resulted in 2-3-fold increase in drug accumulation in lymphoma BMI. Taking together, we have successfully identified a thioaptamer that preferentially binds to the endothelium of lymphoma BMI. It can serve as an affinity moiety for targeted delivery of drug particles to the disease organ.
Collapse
Affiliation(s)
- Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Xin Li
- Institute of Molecular Medicine and the Department of Nanomedicine and Biomedical Engineering, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston 77030, USA
| | - Guodong Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Yi Huang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Rong Xu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Qi Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Ganesh L. Lokesh
- Institute of Molecular Medicine and the Department of Nanomedicine and Biomedical Engineering, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston 77030, USA
| | - Varatharasa Thiviyanathan
- Institute of Molecular Medicine and the Department of Nanomedicine and Biomedical Engineering, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston 77030, USA
| | - Lingxiao Chen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Haoran Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas 77030, USA
| | - Xiaojing Ma
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York 10065, USA
| | - David E. Volk
- Institute of Molecular Medicine and the Department of Nanomedicine and Biomedical Engineering, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston 77030, USA
| | - David G. Gorenstein
- Institute of Molecular Medicine and the Department of Nanomedicine and Biomedical Engineering, McGovern Medical School, The University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York 10065, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York 10065, USA
| |
Collapse
|
33
|
Ma J, Deng H, Zhao F, Deng L, Wang W, Dong A, Zhang J. Liposomes-Camouflaged Redox-Responsive Nanogels to Resolve the Dilemma between Extracellular Stability and Intracellular Drug Release. Macromol Biosci 2018; 18:e1800049. [DOI: 10.1002/mabi.201800049] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/29/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Jinfeng Ma
- Department of Polymer Science and Engineering; Key Laboratory of Systems Bioengineering (Ministry of Education); School of Chemical Engineering and Technology; Tianjin University; Tianjin 300072 China
| | - Hongzhang Deng
- Department of Polymer Science and Engineering; Key Laboratory of Systems Bioengineering (Ministry of Education); School of Chemical Engineering and Technology; Tianjin University; Tianjin 300072 China
| | - Fuli Zhao
- Department of Polymer Science and Engineering; Key Laboratory of Systems Bioengineering (Ministry of Education); School of Chemical Engineering and Technology; Tianjin University; Tianjin 300072 China
| | - Liandong Deng
- Department of Polymer Science and Engineering; Key Laboratory of Systems Bioengineering (Ministry of Education); School of Chemical Engineering and Technology; Tianjin University; Tianjin 300072 China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin); Tianjin 300072 China
| | - Weiwei Wang
- Institute of Biomedical Engineering; Chinese Academy of Medical Science and Peking Union Medical College; Tianjin 300192 China
| | - Anjie Dong
- Department of Polymer Science and Engineering; Key Laboratory of Systems Bioengineering (Ministry of Education); School of Chemical Engineering and Technology; Tianjin University; Tianjin 300072 China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin); Tianjin 300072 China
| | - Jianhua Zhang
- Department of Polymer Science and Engineering; Key Laboratory of Systems Bioengineering (Ministry of Education); School of Chemical Engineering and Technology; Tianjin University; Tianjin 300072 China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology; Tianjin University; Tianjin 300072 China
| |
Collapse
|
34
|
Guo P, Wang B, Liu D, Yang J, Subramanyam K, McCarthy CR, Hebert J, Moses MA, Auguste DT. Using Atomic Force Microscopy to Predict Tumor Specificity of ICAM1 Antibody-Directed Nanomedicines. NANO LETTERS 2018; 18:2254-2262. [PMID: 29505261 DOI: 10.1021/acs.nanolett.7b04801] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Atomic force microscopy (AFM) is a powerful tool to detect in vitro antibody-antigen interactions. To date, however, AFM-measured antibody-antigen interactions have yet to be exploited to predict in vivo tumor specificity of antibody-directed nanomedicines. In this study, we have utilized AFM to directly measure the biomechanical interaction between live triple negative breast cancer (TNBC) cells and an antibody against ICAM1, a recently identified TNBC target. For the first time, we provide proof-of-principle evidence that in vitro TNBC cell-ICAM1 antibody binding force measured by AFM on live cells more precisely correlates with in vivo tumor accumulation and therapeutic efficacy of ICAM1 antibody-directed liposomes than ICAM1 gene and surface protein overexpression levels. These studies demonstrate that live cell-antibody binding force measurements may be used as a novel in vitro metric for predicting the in vivo tumor recognition of antibody-directed nanomedicines.
Collapse
Affiliation(s)
- P Guo
- Department of Biomedical Engineering , The City College of New York , 160 Convent Avenue , New York , New York 10031 , United States
- Vascular Biology Program , Boston Children's Hospital , 300 Longwood Avenue , Boston , Massachusetts 02115 , United States
- Department of Surgery , Harvard Medical School and Boston Children's Hospital , 300 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - B Wang
- Department of Biomedical Engineering , The City College of New York , 160 Convent Avenue , New York , New York 10031 , United States
| | - D Liu
- Department of Biomedical Engineering , The City College of New York , 160 Convent Avenue , New York , New York 10031 , United States
- Department of Chemical Engineering , Northeastern University , 360 Huntington Avenue , Boston , Massachusetts 02115 , United States
| | - J Yang
- Vascular Biology Program , Boston Children's Hospital , 300 Longwood Avenue , Boston , Massachusetts 02115 , United States
- Department of Surgery , Harvard Medical School and Boston Children's Hospital , 300 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - K Subramanyam
- School of Engineering and Applied Sciences , Harvard University , 29 Oxford Street , Cambridge , Massachusetts 02115 , United States
| | - C R McCarthy
- Department of Biomedical Engineering , The City College of New York , 160 Convent Avenue , New York , New York 10031 , United States
| | - J Hebert
- Department of Chemical Engineering , Northeastern University , 360 Huntington Avenue , Boston , Massachusetts 02115 , United States
| | - M A Moses
- Vascular Biology Program , Boston Children's Hospital , 300 Longwood Avenue , Boston , Massachusetts 02115 , United States
- Department of Surgery , Harvard Medical School and Boston Children's Hospital , 300 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - D T Auguste
- Department of Biomedical Engineering , The City College of New York , 160 Convent Avenue , New York , New York 10031 , United States
- Department of Chemical Engineering , Northeastern University , 360 Huntington Avenue , Boston , Massachusetts 02115 , United States
| |
Collapse
|
35
|
Senapati S, Mahanta AK, Kumar S, Maiti P. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduct Target Ther 2018; 3:7. [PMID: 29560283 PMCID: PMC5854578 DOI: 10.1038/s41392-017-0004-3] [Citation(s) in RCA: 1096] [Impact Index Per Article: 182.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/16/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
Although conventional chemotherapy has been successful to some extent, the main drawbacks of chemotherapy are its poor bioavailability, high-dose requirements, adverse side effects, low therapeutic indices, development of multiple drug resistance, and non-specific targeting. The main aim in the development of drug delivery vehicles is to successfully address these delivery-related problems and carry drugs to the desired sites of therapeutic action while reducing adverse side effects. In this review, we will discuss the different types of materials used as delivery vehicles for chemotherapeutic agents and their structural characteristics that improve the therapeutic efficacy of their drugs and will describe recent scientific advances in the area of chemotherapy, emphasizing challenges in cancer treatments. Improving the delivery of cancer therapies to tumor sites is crucial to reduce unwanted side effects and patient mortality rates. Pralay Maiti and colleagues at the Indian Institute of Technology in Varanasi, India, review the latest developments in drug delivery vehicles and treatment approaches designed to enhance the effectiveness of current cancer therapies. New nanoparticle-based carriers, hydrogels and hybrid materials that offer controlled and sustained drug release are showing great promise in animal models. Furthermore, materials that respond to stimuli such as heat, light, magnetic or electric fields are also being tested to aid target-specific drug delivery and, thus, avoid damage to healthy tissues. Although there are some challenges in translating these findings to the clinic, there is no doubt that technological advances are shaping better and safer treatment options.
Collapse
Affiliation(s)
- Sudipta Senapati
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Arun Kumar Mahanta
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sunil Kumar
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pralay Maiti
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
36
|
Pugliese E, Coentro JQ, Zeugolis DI. Advancements and Challenges in Multidomain Multicargo Delivery Vehicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1704324. [PMID: 29446161 DOI: 10.1002/adma.201704324] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/05/2017] [Indexed: 06/08/2023]
Abstract
Reparative and regenerative processes are well-orchestrated temporal and spatial events that are governed by multiple cells, molecules, signaling pathways, and interactions thereof. Yet again, currently available implantable devices fail largely to recapitulate nature's complexity and sophistication in this regard. Herein, success stories and challenges in the field of layer-by-layer, composite, self-assembly, and core-shell technologies are discussed for the development of multidomain/multicargo delivery vehicles.
Collapse
Affiliation(s)
- Eugenia Pugliese
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| | - João Q Coentro
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| |
Collapse
|
37
|
Sreekanth V, Medatwal N, Pal S, Kumar S, Sengupta S, Bajaj A. Molecular Self-Assembly of Bile Acid-Phospholipids Controls the Delivery of Doxorubicin and Mice Survivability. Mol Pharm 2017; 14:2649-2659. [DOI: 10.1021/acs.molpharmaceut.7b00105] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Vedagopuram Sreekanth
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India
- Manipal University, Manipal-576104, Karnataka, India
| | - Nihal Medatwal
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India
- Manipal University, Manipal-576104, Karnataka, India
| | - Sanjay Pal
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India
- KIIT University, Bhubaneswar-751024, Odisha, India
| | - Sandeep Kumar
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India
- Manipal University, Manipal-576104, Karnataka, India
| | - Sagar Sengupta
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi-110067, India
| | - Avinash Bajaj
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India
| |
Collapse
|
38
|
Wang W, Shao A, Zhang N, Fang J, Ruan JJ, Ruan BH. Cationic Polymethacrylate-Modified Liposomes Significantly Enhanced Doxorubicin Delivery and Antitumor Activity. Sci Rep 2017; 7:43036. [PMID: 28225062 PMCID: PMC5320526 DOI: 10.1038/srep43036] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/16/2017] [Indexed: 01/03/2023] Open
Abstract
Liposome (LP) encapsulation of doxorubicin (DOX) is a clinically validated method for cancer drug delivery, but its cellular uptake is actually lower than the free DOX. Therefore, we modified DOX-LP with a cationic polymer (Eudragit RL100; ER) to improve its cellular uptake and antitumor activity. The resulting DOX-ERLP was a 190 nm nanoparticle that was absorbed efficiently and caused cancer cell death in 5 hrs. Growth as measured by the MTT assay or microscopic imaging demonstrated that DOX-ERLP has at least a two-fold greater potency than the free DOX in inhibiting the growth of a DOX resistant (MCF7/adr) cell and an aggressive liver cancer H22 cell. Further, its in vivo efficacy was tested in H22-bearing mice, where four injections of DOX-ERLP reduced the tumor growth by more than 60% and caused an average of 60% tumor necrosis, which was significantly better than the DOX and DOX-LP treated groups. Our work represents the first use of polymethacrylate derivatives for DOX liposomal delivery, demonstrating the great potential of cationic polymethacrylate modified liposomes for improving cancer drug delivery.
Collapse
|
39
|
Deng C, Zhang Q, Fu Y, Sun X, Gong T, Zhang Z. Coadministration of Oligomeric Hyaluronic Acid-Modified Liposomes with Tumor-Penetrating Peptide-iRGD Enhances the Antitumor Efficacy of Doxorubicin against Melanoma. ACS APPLIED MATERIALS & INTERFACES 2017; 9:1280-1292. [PMID: 28009503 DOI: 10.1021/acsami.6b13738] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A safe and efficient tumor-targeting strategy based on oligomeric hyaluronic acid (HA) modification and coadministration of tumor-penetrating peptide-iRGD was successfully developed. In this study, common liposomes (cLip) were modified by oligomeric HA to obtain HA-Lip. After injection into rats, HA-Lip showed good stealth in the bloodstream and lower liver distribution compared with cLip. Moreover, our HA-Lip could be internalized into B16F10 cells (CD44-overexpressing tumor cells) through HA-CD44 interaction. After systemic administration to B16F10 melanoma-bearing mice, HA-Lip showed an increased distribution in tumor due to the prolonged blood circulation time and the enhanced penetration and retention effect. When coadministered with iRGD, the tumor penetration of HA-Lip was significantly enhanced, which could promote HA-Lip internalization by tumors cells located in deep tumor regions through receptor-mediated endocytosis. Furthermore, doxorubicin (DOX)-loaded HA-Lip coadministering with iRGD showed much better antitumor effect compared to DOX-loaded cLip and DOX-loaded cLip in combination with iRGD. In systemic toxicity test, DOX-loaded HA-Lip could significantly decrease the cardiotoxicity and myelosuppression of DOX. Taken together, our results demonstrated that coadministration of oligomeric HA-modified liposomes with iRGD could be a promising treatment strategy for targeted therapy of melanoma.
Collapse
Affiliation(s)
- Caifeng Deng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| | - Quan Zhang
- School of Pharmacy, Chengdu Medical College , Chengdu 610083, China
| | - Yao Fu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| |
Collapse
|
40
|
Tat peptide and hexadecylphosphocholine introduction into pegylated liposomal doxorubicin: An in vitro and in vivo study on drug cellular delivery, release, biodistribution and antitumor activity. Int J Pharm 2016; 511:236-244. [DOI: 10.1016/j.ijpharm.2016.06.117] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/21/2016] [Accepted: 06/26/2016] [Indexed: 11/20/2022]
|
41
|
Chuang EY, Lin CC, Chen KJ, Wan DH, Lin KJ, Ho YC, Lin PY, Sung HW. A FRET-guided, NIR-responsive bubble-generating liposomal system for in vivo targeted therapy with spatially and temporally precise controlled release. Biomaterials 2016; 93:48-59. [DOI: 10.1016/j.biomaterials.2016.03.040] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/07/2016] [Accepted: 03/28/2016] [Indexed: 11/26/2022]
|
42
|
Yingchoncharoen P, Kalinowski DS, Richardson DR. Lipid-Based Drug Delivery Systems in Cancer Therapy: What Is Available and What Is Yet to Come. Pharmacol Rev 2016; 68:701-87. [PMID: 27363439 PMCID: PMC4931871 DOI: 10.1124/pr.115.012070] [Citation(s) in RCA: 436] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer is a leading cause of death in many countries around the world. However, the efficacy of current standard treatments for a variety of cancers is suboptimal. First, most cancer treatments lack specificity, meaning that these treatments affect both cancer cells and their normal counterparts. Second, many anticancer agents are highly toxic, and thus, limit their use in treatment. Third, a number of cytotoxic chemotherapeutics are highly hydrophobic, which limits their utility in cancer therapy. Finally, many chemotherapeutic agents exhibit short half-lives that curtail their efficacy. As a result of these deficiencies, many current treatments lead to side effects, noncompliance, and patient inconvenience due to difficulties in administration. However, the application of nanotechnology has led to the development of effective nanosized drug delivery systems known commonly as nanoparticles. Among these delivery systems, lipid-based nanoparticles, particularly liposomes, have shown to be quite effective at exhibiting the ability to: 1) improve the selectivity of cancer chemotherapeutic agents; 2) lower the cytotoxicity of anticancer drugs to normal tissues, and thus, reduce their toxic side effects; 3) increase the solubility of hydrophobic drugs; and 4) offer a prolonged and controlled release of agents. This review will discuss the current state of lipid-based nanoparticle research, including the development of liposomes for cancer therapy, different strategies for tumor targeting, liposomal formulation of various anticancer drugs that are commercially available, recent progress in liposome technology for the treatment of cancer, and the next generation of lipid-based nanoparticles.
Collapse
Affiliation(s)
- Phatsapong Yingchoncharoen
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
43
|
Huang Y, Hemmer E, Rosei F, Vetrone F. Multifunctional Liposome Nanocarriers Combining Upconverting Nanoparticles and Anticancer Drugs. J Phys Chem B 2016; 120:4992-5001. [DOI: 10.1021/acs.jpcb.6b02013] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yue Huang
- Institut
National de la Recherche Scientifique - Énergie, Matériaux
et Télécommunications, Université du Québec, Varennes, Québec J3X 1S2, Canada
| | - Eva Hemmer
- Institut
National de la Recherche Scientifique - Énergie, Matériaux
et Télécommunications, Université du Québec, Varennes, Québec J3X 1S2, Canada
| | - Federico Rosei
- Institut
National de la Recherche Scientifique - Énergie, Matériaux
et Télécommunications, Université du Québec, Varennes, Québec J3X 1S2, Canada
- Institute
for Fundamental and Frontier Science, University of Electronic Science and Technology of China, Chengdu, PR China
- Centre
for Self-Assembled Chemical Structures, McGill University, Montreal, Québec H3A 2K6, Canada
| | - Fiorenzo Vetrone
- Institut
National de la Recherche Scientifique - Énergie, Matériaux
et Télécommunications, Université du Québec, Varennes, Québec J3X 1S2, Canada
- Institute
for Fundamental and Frontier Science, University of Electronic Science and Technology of China, Chengdu, PR China
- Centre
for Self-Assembled Chemical Structures, McGill University, Montreal, Québec H3A 2K6, Canada
| |
Collapse
|
44
|
Improving drug retention in liposomes by aging with the aid of glucose. Int J Pharm 2016; 505:194-203. [DOI: 10.1016/j.ijpharm.2016.03.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 03/20/2016] [Accepted: 03/23/2016] [Indexed: 01/24/2023]
|
45
|
Zahmatkeshan M, Gheybi F, Rezayat SM, Jaafari MR. Improved drug delivery and therapeutic efficacy of PEgylated liposomal doxorubicin by targeting anti-HER2 peptide in murine breast tumor model. Eur J Pharm Sci 2016; 86:125-35. [PMID: 26972276 DOI: 10.1016/j.ejps.2016.03.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 02/29/2016] [Accepted: 03/09/2016] [Indexed: 12/14/2022]
Abstract
Targeted cancer therapy is a powerful therapeutic strategy to management of cancer. HER2 as an anticancer target has long been studied. Its overexpression plays an important role in the pathogenesis and progressiveness of breast and other cancers. To establish efficient and reliable drug delivery to HER2-overexpressing cells, the authors of this study have developed anti-HER2 (ErbB2) peptide-liposomal formulations of doxorubicin (DOX) by an engineered breast tumor-targeting peptide ligand, AHNP, Anti-HER2/neu peptide, (FCDGFYACYADV) with three glycine amino acids as spacer before its original sequencing. Towards this goal, PEGylated liposome doxorubicin (PLD) bearing different ligand densities of AHNP was prepared and characterized for their size, zeta potential and peptide conjugation. The AHNP functionalization and density effects on breast tumor cell uptake, selective cytotoxicity, prevention of tumor growth and the tissue biodistribution of encapsulated DOX were studied in mice bearing TUBO breast cancer tumor model. The findings demonstrated that increasing the ligand density of AHNP increases cytotoxicity and cell-uptake in SKBR3 and TUBO cells which overexpress HER2 but not in MDA-MB-231with low HER2 expression profile. The anticancer activity was also superior for targeted liposomal DOX with more AHNP densities. Overall, the results showed that optimum AHNP density functionalization of PLD can significantly improve selectivity and the therapeutic index of liposomal DOX in the treatment of HER2 positive breast cancer and merits further investigation.
Collapse
Affiliation(s)
- Masoumeh Zahmatkeshan
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1417755469, Iran
| | - Fatemeh Gheybi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1417755469, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1417755469, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91775-1365, Iran.
| |
Collapse
|
46
|
Wu CH, Liu IJ, Lu RM, Wu HC. Advancement and applications of peptide phage display technology in biomedical science. J Biomed Sci 2016; 23:8. [PMID: 26786672 PMCID: PMC4717660 DOI: 10.1186/s12929-016-0223-x] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/11/2016] [Indexed: 12/25/2022] Open
Abstract
Combinatorial phage library is a powerful research tool for high-throughput screening of protein interactions. Of all available molecular display techniques, phage display has proven to be the most popular approach. Screening phage-displayed random peptide libraries is an effective means of identifying peptides that can bind target molecules and regulate their function. Phage-displayed peptide libraries can be used for (i) B-cell and T-cell epitope mapping, (ii) selection of bioactive peptides bound to receptors or proteins, disease-specific antigen mimics, peptides bound to non-protein targets, cell-specific peptides, or organ-specific peptides, and (iii) development of peptide-mediated drug delivery systems and other applications. Targeting peptides identified using phage display technology may be useful for basic research and translational medicine. In this review article, we summarize the latest technological advancements in the application of phage-displayed peptide libraries to applied biomedical sciences.
Collapse
Affiliation(s)
- Chien-Hsun Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - I-Ju Liu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Ruei-Min Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.
| |
Collapse
|
47
|
Patra S, Roy E, Madhuri R, Sharma PK. Retracted Article: Creation of ultrasound and temperature-triggered bubble liposomes from economical precursors to enhance the therapeutic efficacy of curcumin in cancer cells. RSC Adv 2016. [DOI: 10.1039/c6ra14584a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An ultrasound and temperature responsive bubble liposome has been designed with high physiological stability, targeted, rapid and tunable drug release profile.
Collapse
Affiliation(s)
- Santanu Patra
- Department of Applied Chemistry
- Indian School of Mines
- Dhanbad
- India
| | - Ekta Roy
- Department of Applied Chemistry
- Indian School of Mines
- Dhanbad
- India
| | - Rashmi Madhuri
- Department of Applied Chemistry
- Indian School of Mines
- Dhanbad
- India
| | - Prashant K. Sharma
- Functional Nanomaterials Research Laboratory
- Department of Applied Physics
- Indian School of Mines
- Dhanbad
- India
| |
Collapse
|
48
|
Teymouri M, Farzaneh H, Badiee A, Golmohammadzadeh S, Sadri K, Jaafari MR. Investigation of Hexadecylphosphocholine (miltefosine) usage in Pegylated liposomal doxorubicin as a synergistic ingredient: In vitro and in vivo evaluation in mice bearing C26 colon carcinoma and B16F0 melanoma. Eur J Pharm Sci 2015; 80:66-73. [DOI: 10.1016/j.ejps.2015.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 08/10/2015] [Accepted: 08/17/2015] [Indexed: 12/27/2022]
|
49
|
Abstract
The discovery of RNA interference (RNAi) in mammalian cells has created a new class of therapeutics based on the reversible silencing of specific disease-causing genes. This therapeutic potential depends on the ability to deliver inducers of RNAi, such as short-interfering RNA (siRNA) and micro-RNA (miRNA), to cells of target tissues. This chapter reviews various challenges and delivery strategies for siRNA, with a particular focus on the development of lipid nanoparticle (LNP) delivery technologies. Currently, LNP delivery systems are the most advanced technology for systemic delivery of siRNA, with numerous formulations under various stages of clinical trials. We also discuss methods to improve gene silencing potency of LNP-siRNA, as well as application of LNP technologies beyond siRNA to the encapsulation of other nucleic acids such as mRNA and clustered regularly interspaced short palindromic repeats (CRISPR).
Collapse
Affiliation(s)
- Alex K K Leung
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Yuen Yi C Tam
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
50
|
Yuan M, Qiu Y, Zhang L, Gao H, He Q. Targeted delivery of transferrin and TAT co-modified liposomes encapsulating both paclitaxel and doxorubicin for melanoma. Drug Deliv 2015; 23:1171-83. [PMID: 26036724 DOI: 10.3109/10717544.2015.1040527] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The purpose of this study was to develop an efficient dual-ligand based liposomal drug delivery system with targeting specificity as well as properties that would kill melanoma cells. Liposomes modified with transferrin (Tf) and cell-penetrating peptide TAT was prepared, which encapsulated two kinds of chemotherapy drugs, paclitaxel and doxorubicin (Tf/TAT-PTX/DOX-LP). The Tf ligands specifically bind to the overexpressed Tf receptors on the surface of melanoma cells, while the TAT ligands functioned as a classical cell penetrating peptide, helping dual-ligand liposomes be internalized by melanoma cells. The effect of dual-targeting system and "double-drug" combination therapy were evaluated both in vitro and in vivo. In vitro, cellular uptake, intracellular distribution and tumor spheroids penetration studies demonstrated that the system could not only be selectively and efficiently penetrate melanoma cells. Besides, apoptosis staining assay and cytotoxicity showed effective anti-tumor capability and obvious synergistic effect of combination therapy of PTX and DOX. In vivo imaging and fluorescent images of tumor section further demonstrated that Tf/TAT-PTX/DOX-LP had the highest tumor distribution. The results of these experiments demonstrated that double-drug liposomal drug delivery systems (DDS) had both enhanced targeting efficiency and increased therapeutic efficacy.
Collapse
Affiliation(s)
- Mingqing Yuan
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , P. R. China and
| | - Yue Qiu
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , P. R. China and
| | - Li Zhang
- b Elderly Digestive Department , Sichuan Provincial People's Hospital, Sichuan Academy of Medical Sciences , Chengdu , China
| | - Huile Gao
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , P. R. China and
| | - Qin He
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , P. R. China and
| |
Collapse
|