1
|
Wang A, Wang X, Li D, Li A, He M, Yuan Y, Ye L, Liu J. A superior method for antitumor therapy and application: dual-ligand nanomedicines. J Mater Chem B 2025. [PMID: 40396464 DOI: 10.1039/d5tb00044k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Currently, nanomedicines have been widely applied in the treatment of various types of tumors. However, due to the complexity of the tumor microenvironment, conventional nanomedicines often exhibit poor efficacy, insufficient site specificity, and susceptibility to off-target effects. In contrast, dual-ligand nanomedicines demonstrate superior targeting ability and drug penetration in tumor therapy. These nanomedicines are equipped with two ligands on their surface, enabling targeting of specific receptors on the same or different cells. The specific binding between ligands and receptors significantly enhances the selectivity and targeting of dual-ligand nanomedicines towards tumors. This review systematically describes the preparation of dual-ligand nanomedicines, the influencing factors, and the types of delivered drugs, focusing on the application of dual-ligand nanomedicines in targeting the treatment of various tumors. We highlight the comprehensiveness of dual-ligand nanomedicines for the treatment of tumors, including glioblastoma, lung cancer, breast cancer, gastric cancer, and many other types of tumors. Finally, the possible challenges for the future development of dual-ligand nanomedicines in terms of preparation, clinic, and safety are further analyzed. We look forward to exploring dual-ligand nanomedicines in greater depth to provide references for their future development and clinical applications.
Collapse
Affiliation(s)
- Ailing Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Xuejun Wang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Aixue Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Mengyuan He
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Yingying Yuan
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
- College of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Ye
- School of Pharmacy and (R & D Center) Lab. for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau SAR, 999078, China.
| | - Jiyong Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| |
Collapse
|
2
|
Liu Y, Chen X, Fischer KS, Fu S, Yuan L, Hu X. Keloids revisited: Current concepts in treatment and differential diagnosis. Cancer Lett 2025; 625:217802. [PMID: 40374155 DOI: 10.1016/j.canlet.2025.217802] [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: 01/21/2025] [Revised: 05/08/2025] [Accepted: 05/13/2025] [Indexed: 05/17/2025]
Abstract
Keloid is a special type of scar considered prototypic of skin fibrosis. Unlike hypertrophic scars, keloids exceed the margins of the original wound, and exist over time without a quiescent or regressive phase. Although keloids do not metastasize, they exhibit tumor-like characteristics, and share many similarities. Large epidemiological study demonstrates that patients with keloids have a 1.49-fold higher risk for cancers. Keloids can lead to severe functional impairments and diminish quality of life which increases hidden costs for patients and medical systems. The main goals of treatments are to improve scar appearance, symptoms and patient's quality of life (QoL). However, the microenvironment, pathogenesis, formation and development of the keloid are complex, the efficacy of multiple treatments were limited. Therefore, this up-to-date review aimed to target the current concepts in keloid treatment and differential diagnosis. The goal is to provide a reference for doctors and researchers to improve the accuracy of diagnosis and facilitate the selection of personalized treatment methods for patients with keloids.
Collapse
Affiliation(s)
- Yanhui Liu
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Xiao Chen
- Department of Medical Cosmetology, The First People's Hospital of Changde City, Changde, Hunan, 415003, China.
| | - Katharina S Fischer
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University, Stanford, CA, 94305, USA; Department of Surgery, University of Arizona, Tucson, AZ, 85724, USA.
| | - Siqi Fu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Li Yuan
- Department of Nuclear Medicine, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China.
| | - Xing Hu
- Department of Medical Cosmetology, Changsha Aier Eye Hospital, Changsha, Hunan, 410015, China; Aier Eye Institute, Central South University, Changsha, Hunan, 410083, China.
| |
Collapse
|
3
|
Saberian E, Jenčová J, Jenča A, Jenča A, Salehipoor F, Zare-Zardini H, Petrášová A, Džupa P, Ebrahimifar M, Allahyartorkaman M, Jenča J. Bleomycin-loaded folic acid-conjugated nanoliposomes: a novel formulation for targeted treatment of oral cancer. Front Bioeng Biotechnol 2025; 13:1535793. [PMID: 40297282 PMCID: PMC12034650 DOI: 10.3389/fbioe.2025.1535793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Targeted delivery of anticancer drugs holds great promise for enhancing therapeutic efficacy while minimizing adverse effects. The folate receptor (FR)-mediated approach offers a selective strategy to target cancer cells overexpressing FR. Bleomycin, an established antitumor antibiotic, suffers from limited efficacy due to poor diffusion into tumor cells. This study examined the anti-cancer potential of folate-targeted liposomal Bleomycin (FL-BLEOMYCIN) in comparison to non-targeted L-BLEOMYCIN on oral cavity cancer (CAL27). The study also investigated FL-Bleomycin's capacity to halt the cell cycle in the G2/M phase using flow cytometry. Methods FL-Bleomycin was produced using thin-layer hydration, followed by incorporation of folic acid into nanoliposomes. To evaluate the release profile, drug release tests were carried out. Cytotoxicity of FL-Bleomycin, L-Bleomycin, and traditional Bleomycin was evaluated using cell viability assays. The cell cycle arrest caused by FL-Bleomycin was examined using flow cytometry. Finally, FL-Bleomycin uptake studies were performed to assess the internalization of FL-Bleomycin by CAL27 cells. Results Compared to L-Bleomycin and traditional Bleomycin, FL-Bleomycin showed noticeably more cytotoxicity against CAL 27 cells. The effective arrest of CAL 27 cells in the G2/M phase of the cell cycle by FL-Bleomycin was verified by flow cytometry. Uptake studies revealed increased internalization of FL-Bleomycin by CAL 27 cells compared to standard formulations. Drug release studies showed a consistent, non-explosive release profile. Cells treated with these nanoliposomes, compared to control groups, exhibited a dose-dependent decrease in the intensity of the 170-kDa EGF-R band as observed by Western blot analysis. Discussion The findings suggest that FL-Bleomycin is a potential method for delivering drugs precisely in tumors expressing folic acid receptors. Its potential for successful cancer treatment is shown by its higher internalization, improved cytotoxicity, and cell cycle prevention in CAL 27 cells. To find out how effective FL-Bleomycin is in vivo and whether it may be used to treat other FR-expressing tumors, more research is necessary.
Collapse
Affiliation(s)
- Elham Saberian
- Pavol Jozef Šafárik University, Clinic of Dentistry and Maxillofacial Surgery Academy of Košice, Kosice, Slovakia
| | - Janka Jenčová
- Pavol Jozef Šafárik University, Clinic of Dentistry and Maxillofacial Surgery Academy of Košice, Kosice, Slovakia
| | - Andrej Jenča
- Pavol Jozef Šafárik University, Clinic of Dentistry and Maxillofacial Surgery Academy of Košice, Kosice, Slovakia
| | - Andrej Jenča
- Pavol Jozef Šafárik University, Clinic of Dentistry and Maxillofacial Surgery Academy of Košice, Kosice, Slovakia
| | - Fateme Salehipoor
- Department of Medicine, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Hadi Zare-Zardini
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Adriána Petrášová
- Pavol Jozef Šafárik University, Clinic of Dentistry and Maxillofacial Surgery Academy of Košice, Kosice, Slovakia
| | - Peter Džupa
- Department of Medicine, Slovak Medical University in Bratislava, Bratislava, Slovakia
| | - Meysam Ebrahimifar
- Department of Toxicology, Faculty of Pharmacy, Islamic Azad University, Shahreza Branch, Shahreza, Iran
| | | | - Jozef Jenča
- Pavol Jozef Šafárik University, Clinic of Dentistry and Maxillofacial Surgery Academy of Košice, Kosice, Slovakia
| |
Collapse
|
4
|
Hong S, Lin C, Hu J, Piao J, Piao MG. Octa-Arginine-Conjugated Liposomal Nimodipine Incorporated in a Temperature-Responsive Gel for Nasoencephalic Delivery. Mol Pharm 2024; 21:5217-5237. [PMID: 39185610 DOI: 10.1021/acs.molpharmaceut.4c00634] [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] [Indexed: 08/27/2024]
Abstract
Nimodipine is the primary clinical drug used to treat cerebral vasospasm following subarachnoid hemorrhage. Currently, tablets have low bioavailability when taken orally, and injections contain ethanol. Therefore, we investigated a new method of nimodipine administration, namely, nasoencephalic administration. Nasal administration of nimodipine was carried out by attaching the cell-penetrating peptide octa-arginine (R8) to liposomes of nimodipine and incorporating it into a temperature-sensitive in situ gel. The prepared liposomes and gels underwent separate evaluations for in vitro characterization. In vitro release exhibited a significant slow-release effect. In vitro toad maxillary cilia model, RPMI 2650 cytotoxicity, and in vivo SD rat pathological histotoxicity experiments showed that all the dosage from the groups had no significant toxicity to toad maxillary cilia, RPMI 2650 cells, and SD rat tissues and organs, and the cilia continued to oscillate up to 694 ± 10.15 min, with the survival rate of the cells being above 85%. A transwell nasal mucosa cell model and an isolated porcine nasal mucosa model were established, and the results showed that the osmolality of the R8-modified nimodipine liposomal gel to nasal mucosal cells and isolated porcine nasal mucosa was 30.41 ± 2.14 and 65.9 ± 7.34 μg/mL, respectively, which was significantly higher than that of the NM-Solution and PEGylated nimodipine liposome gel groups. Animal fluorescence imaging studies revealed that the R8-modified nimodipine liposomal gel displayed increased brain fluorescence intensity compared to the normal liposomal gel. Pharmacokinetic results showed that after transnasal administration, the AUC(0-∞) of the R8-modified nimodipine liposomal gel was 11.662 ± 1.97 μg·mL-1, which was significantly higher than that of the plain nimodipine liposomal gel (5.499 ± 2.89 μg·mL-1). Brain-targeting experiments showed that the brain-targeting efficiencies of the PEGylated nimodipine liposome gel and R8-modified PEGylated nimodipine liposome gels were 20.44 and 33.45, respectively, suggesting that R8/PEG/Lip-NM-TSG significantly increased the brain-targeting of the drug.
Collapse
Affiliation(s)
- Shuai Hong
- College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Changxiu Lin
- Central Laboratory of the Affiliated Hospital, Yanbian University, Yanji 133000, Jilin, China
| | - Junsheng Hu
- College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Jingshu Piao
- College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Ming Guan Piao
- College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, Jilin, China
| |
Collapse
|
5
|
Functionalised penetrating peptide-chondroitin sulphate‑gold nanoparticles: Synthesis, characterization, and applications as an anti-Alzheimer's disease drug. Int J Biol Macromol 2023; 230:123125. [PMID: 36603725 DOI: 10.1016/j.ijbiomac.2022.123125] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/04/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023]
Abstract
The purpose of this study was to construct a transmembrane peptide-chondroitin sulphate‑gold nanoparticle (TAT-CS@Au) delivery system and investigate its activity as an anti-Alzheimer's disease (AD) drug. We successfully prepared TAT-CS@Au nanoparticles, investigated their anti-AD effects, and explored the possible mechanisms in in vitro models. TAT-CS@Au exhibited excellent cellular uptake and transport capacity, effectively inhibited the accumulation of Aβ1-40, and significantly reduced Aβ1-40-induced apoptosis in SH-SY5Y cells. Furthermore, TAT-CS@Au significantly reduced oxidative stress damage and cholinergic injury induced by Aβ1-40 by regulating intracellular concentrations of reactive oxygen species (ROS), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), and acetylcholine (ACh). Western blotting results demonstrated that TAT-CS@Au inhibited aberrant tau phosphorylation (Ser199, Thr205, Ser404, and Ser396) through GSK3β inactivation. TAT-CS@Au decreased the levels of inflammatory factors, specifically TNF-α, IL-6, and IL-1β, by inhibiting NF-κB nuclear translocation by activating MAPK signalling pathways. Overall, these results indicate that TAT-CS@Au exhibits excellent transmembrane ability, inhibits Aβ1-40 accumulation, antagonises oxidative stress, reduces aberrant tau phosphorylation, and suppresses the expression of inflammatory factors. TAT-CS@Au may be a multi-target anti-AD drug with good cell permeability, providing new insights into the design and research of anti-AD therapeutics.
Collapse
|
6
|
Farooq MU, Sahin YM, Naz MY, Ijaz S, Shukrullah S, Makhlouf MM. Surface engineered AuNPs for paclitaxel-loaded bleomycin delivery as a supplementation therapy. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-022-02645-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
7
|
Xie Q, Li S, Feng X, Shi J, Li Y, Yuan G, Yang C, Shen Y, Kong L, Zhang Z. All-in-one approaches for triple-negative breast cancer therapy: metal-phenolic nanoplatform for MR imaging-guided combinational therapy. J Nanobiotechnology 2022; 20:226. [PMID: 35549947 PMCID: PMC9097361 DOI: 10.1186/s12951-022-01416-7] [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: 12/19/2021] [Accepted: 04/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Conventional chemotherapy has poor efficacy in triple-negative breast cancer (TNBC) which is highly heterogeneous and aggressive. Imaging-guided therapy is usually combined with diverse treatment modalities, could realize the integration of diagnosis and treatments. Therefore, the primary challenge for combinational therapy is designing proper delivery systems to accomplish multiple synergistic effects. RESULTS Herein, a facile nanoplatform was manufactured to fulfill the all-in-one approaches for TNBC combinational therapy. Fe3+-based metal-phenolic networks (MPNs) with bovine serum albumin (BSA) modification served as drug delivery carriers to encapsulate bleomycin (BLM), forming BFE@BSA NPs. The self-assembly mechanism, pH-responsive drug release behavior, and other physicochemical properties of this system were characterized. The potential of BFE@BSA NPs as photothermal transduction agents and magnetic resonance imaging (MRI) contrast agents was explored. The synergistic anti-tumor effects consisting of BLM-induced chemotherapy, Fenton reactions-mediated chemodynamic therapy, and photothermal therapy-induced apoptosis were studied both in vitro and in vivo. Once internalized into tumor cells, released BLM could cause DNA damage, while Fenton reactions were initiated to produce highly toxic •OH. Upon laser irradiation, BFE@BSA NPs could convert light into heat to achieve synergistic effects. After intravenous administration, BFE@BSA NPs exhibited great therapeutic effects in 4T1 tumor xenograft model. Moreover, as T1-weighted MRI contrast agents, BFE@BSA NPs could provide diagnosis and treatment monitoring for individualized precise therapy. CONCLUSIONS A nano-system that integrated imaging and combinational therapy (chemotherapy, chemodynamic therapy and photothermal therapy) were developed to kill the tumor and monitor therapeutic efficacy. This strategy provided an all-in-one theranostic nanoplatform for MRI-guided combinational therapy against TNBC.
Collapse
Affiliation(s)
- Qi Xie
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shichao Li
- Department of Radiology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xingxing Feng
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingyu Shi
- Liyuan Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yang Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guanjie Yuan
- Department of Radiology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yaqi Shen
- Department of Radiology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China. .,National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Hubei Engineering Research Center for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
8
|
Rusiecka I, Gągało I, Kocić I. Cell-penetrating peptides improve pharmacokinetics and pharmacodynamics of anticancer drugs. Tissue Barriers 2022; 10:1965418. [PMID: 34402743 PMCID: PMC8794253 DOI: 10.1080/21688370.2021.1965418] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022] Open
Abstract
This review concentrates on the research concerning conjugates of anticancer drugs with versatile cell-penetrating peptides (CPPs). For a better insight into the relationship between the components of the constructs, it starts with the characteristic of the peptides and considers its following aspects: mechanisms of cellular internalization, interaction with cancer-modified membranes, selectivity against tumor tissue. Also, CPPs with anticancer activity have been distinguished and summarized with their mechanisms of action. With respect to the conjugates, the preclinical studies (in vitro, in vivo) indicated that they possess several merits in comparison to the parent drugs. They concerned not only better cellular internalization but also other improvements in pharmacokinetics (e.g. access to the brain tissue) and pharmacodynamics (e.g. overcoming drug resistance). The anticancer activity of the conjugates was usually superior to that of the unconjugated drug. Certain anticancer CPPs and conjugates entered clinical trials.
Collapse
Affiliation(s)
- Izabela Rusiecka
- Department of Pharmacology, Medical University of Gdansk, Gdansk, Poland
| | - Iwona Gągało
- Department of Pharmacology, Medical University of Gdansk, Gdansk, Poland
| | - Ivan Kocić
- Department of Pharmacology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
9
|
Shoari A, Tooyserkani R, Tahmasebi M, Löwik DWPM. Delivery of Various Cargos into Cancer Cells and Tissues via Cell-Penetrating Peptides: A Review of the Last Decade. Pharmaceutics 2021; 13:1391. [PMID: 34575464 PMCID: PMC8470549 DOI: 10.3390/pharmaceutics13091391] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 01/03/2023] Open
Abstract
Cell-penetrating peptides (CPPs), also known as protein transduction domains, are a class of diverse amino acid sequences with the ability to cross cellular membranes. CPPs can deliver several bioactive cargos, including proteins, peptides, nucleic acids and chemotherapeutics, into cells. Ever since their discovery, synthetic and natural CPPs have been utilized in therapeutics delivery, gene editing and cell imaging in fundamental research and clinical experiments. Over the years, CPPs have gained significant attention due to their low cytotoxicity and high transduction efficacy. In the last decade, multiple investigations demonstrated the potential of CPPs as carriers for the delivery of therapeutics to treat various types of cancer. Besides their remarkable efficacy owing to fast and efficient delivery, a crucial benefit of CPP-based cancer treatments is delivering anticancer agents selectively, rather than mediating toxicities toward normal tissues. To obtain a higher therapeutic index and to improve cell and tissue selectivity, CPP-cargo constructions can also be complexed with other agents such as nanocarriers and liposomes to obtain encouraging outcomes. This review summarizes various types of CPPs conjugated to anticancer cargos. Furthermore, we present a brief history of CPP utilization as delivery systems for anticancer agents in the last decade and evaluate several reports on the applications of CPPs in basic research and preclinical studies.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Raheleh Tooyserkani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Mehdi Tahmasebi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
| | - Dennis W. P. M. Löwik
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| |
Collapse
|
10
|
Joshi R, Acharya B, Bhandari K, Bhandari S, Duwal R, Ghimire R, Shakya R, Sweidan KA, Shrestha BG. Dichloroacetyl chloride conjugated peptide-based probes: design, synthesis, and in vitro evaluation in breast cancer cells. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2020.1765358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Rajendra Joshi
- Department of Chemical Science and Engineering, School of Engineering, Kathmandu University, Dhulikhel, Nepal
| | - Baku Acharya
- Department of Pharmacy, School of Science, Kathmandu University, Dhulikhel, Nepal
| | - Kritisha Bhandari
- Department of Pharmacy, School of Science, Kathmandu University, Dhulikhel, Nepal
| | - Suzeeta Bhandari
- Department of Pharmacy, School of Science, Kathmandu University, Dhulikhel, Nepal
| | - Rajina Duwal
- Department of Pharmacy, School of Science, Kathmandu University, Dhulikhel, Nepal
| | - Ranju Ghimire
- Department of Pharmacy, School of Science, Kathmandu University, Dhulikhel, Nepal
| | - Rajani Shakya
- Department of Pharmacy, School of Science, Kathmandu University, Dhulikhel, Nepal
| | - Kamal A. Sweidan
- Department of Chemistry, The University of Jordan, Amman, Jordan
| | - Bhupal G. Shrestha
- Department of Biotechnology, School of Science, Kathmandu University, Dhulikhel, Nepal
| |
Collapse
|
11
|
Serra AS, Eusébio D, Neves AR, Albuquerque T, Bhatt H, Biswas S, Costa D, Sousa Â. Synthesis and Characterization of Mannosylated Formulations to Deliver a Minicircle DNA Vaccine. Pharmaceutics 2021; 13:673. [PMID: 34067176 PMCID: PMC8150592 DOI: 10.3390/pharmaceutics13050673] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 12/24/2022] Open
Abstract
DNA vaccines still represent an emergent area of research, giving rise to continuous progress towards several biomedicine demands. The formulation of delivery systems to specifically target mannose receptors, which are overexpressed on antigen presenting cells (APCs), is considered a suitable strategy to improve the DNA vaccine immunogenicity. The present study developed binary and ternary carriers, based on polyethylenimine (PEI), octa-arginine peptide (R8), and mannose ligands, to specifically deliver a minicircle DNA (mcDNA) vaccine to APCs. Systems were prepared at various nitrogen to phosphate group (N/P) ratios and characterized in terms of their morphology, size, surface charge, and complexation capacity. In vitro studies were conducted to assess the biocompatibility, cell internalization ability, and gene expression of formulated carriers. The high charge density and condensing capacity of both PEI and R8 enhance the interaction with the mcDNA, leading to the formation of smaller particles. The addition of PEI polymer to the R8-mannose/mcDNA binary system reduces the size and increases the zeta potential and system stability. Confocal microscopy studies confirmed intracellular localization of targeting systems, resulting in sustained mcDNA uptake. Furthermore, the efficiency of in vitro transfection can be influenced by the presence of R8-mannose, with great implications for gene expression. R8-mannose/PEI/mcDNA ternary systems can be considered valuable tools to instigate further research, aiming for advances in the DNA vaccine field.
Collapse
Affiliation(s)
- Ana Sofia Serra
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.S.S.); (D.E.); (A.R.N.); (T.A.)
| | - Dalinda Eusébio
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.S.S.); (D.E.); (A.R.N.); (T.A.)
| | - Ana Raquel Neves
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.S.S.); (D.E.); (A.R.N.); (T.A.)
| | - Tânia Albuquerque
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.S.S.); (D.E.); (A.R.N.); (T.A.)
| | - Himanshu Bhatt
- Department of Pharmacy, Nanomedicine Research Laboratory, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, Telangana 500078, India; (H.B.); (S.B.)
| | - Swati Biswas
- Department of Pharmacy, Nanomedicine Research Laboratory, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, Telangana 500078, India; (H.B.); (S.B.)
| | - Diana Costa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.S.S.); (D.E.); (A.R.N.); (T.A.)
| | - Ângela Sousa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.S.S.); (D.E.); (A.R.N.); (T.A.)
| |
Collapse
|
12
|
Raber HF, Heerde T, El Din SN, Flaig C, Hilgers F, Bitzenhofer N, Jäger KE, Drepper T, Gottschalk KE, Bodenberger NE, Weil T, Kubiczek DH, Rosenau F. Azulitox—A Pseudomonas aeruginosa P28-Derived Cancer-Cell-Specific Protein Photosensitizer. Biomacromolecules 2020; 21:5067-5076. [DOI: 10.1021/acs.biomac.0c01216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Heinz Fabian Raber
- Institute for Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Thomas Heerde
- Institute for Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Suzanne Nour El Din
- Institute for Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Carolin Flaig
- Institute for Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Institute for Experimental Physics, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Fabienne Hilgers
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf at Forschungszentrum Jülich, Stetternicher Forst, 52426 Jülich, Germany
| | - Nora Bitzenhofer
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf at Forschungszentrum Jülich, Stetternicher Forst, 52426 Jülich, Germany
| | - Karl-Erich Jäger
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf at Forschungszentrum Jülich, Stetternicher Forst, 52426 Jülich, Germany
- Institute of Bio- and Geosciences (IBG-1: Biotechnology) Forschungszentrum Jülich, Stetternicher Forst, 52426 Jülich, Germany
| | - Thomas Drepper
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf at Forschungszentrum Jülich, Stetternicher Forst, 52426 Jülich, Germany
| | - Kay-Eberhard Gottschalk
- Institute for Experimental Physics, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | | | - Tanja Weil
- Max Planck Institute for Polymer Research Mainz, Ackermannweg 10, 55128 Mainz, Germany
| | - Dennis Horst Kubiczek
- Institute for Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Frank Rosenau
- Institute for Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Max Planck Institute for Polymer Research Mainz, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
13
|
El Maghraby GM, Arafa MF. Liposomes for Enhanced Cellular Uptake of Anticancer Agents. Curr Drug Deliv 2020; 17:861-873. [PMID: 32640957 DOI: 10.2174/1567201817666200708113131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/08/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
Cancers are life threatening diseases and their traditional treatment strategies have numerous limitations which include poor pharmacokinetic profiles, non-specific drug distribution in the body tissues and organs and deprived tumor cells penetration. This attracted the attention of researchers to tailor efficient drug delivery system for anticancer agents to overcome these limitations. Liposomes are one of the newly developed delivery systems for anticancer agents. They are vesicular structures, which were fabricated to enhance drug targeting to tumor tissues either via active or passive targeting. They can be tailored to penetrate tumor cells membrane which is considered the main rate limiting step in antineoplastic therapy. This resulted in enhancing drug cellular uptake and internalization and increasing drug cytotoxic effect. These modifications were achieved via various approaches which included the use of cell-penetrating peptides, the use of lipid substances that can increase liposome fusogenic properties or increase the cell membrane permeability toward amphiphilic drugs, surface modification or ligand targeted liposomes and immuno-liposomes. The modified liposomes were able to enhance anticancer agent's cellular uptake and this was reflected in their ability to destroy tumor tissues. This review outlines different approaches employed for liposomes modification for enhancing anticancer agent's cellular uptake.
Collapse
Affiliation(s)
- Gamal M El Maghraby
- Department of Pharmaceutical Technology, College of Pharmacy, University of Tanta, Tanta, Egypt
| | - Mona F Arafa
- Department of Pharmaceutical Technology, College of Pharmacy, University of Tanta, Tanta, Egypt
| |
Collapse
|
14
|
Influence of cell-penetrating peptides on the activity and stability of virus-based nanoparticles. Int J Pharm 2020; 576:119008. [DOI: 10.1016/j.ijpharm.2019.119008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/23/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023]
|
15
|
Kim B, Park JH, Sailor MJ. Rekindling RNAi Therapy: Materials Design Requirements for In Vivo siRNA Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1903637. [PMID: 31566258 PMCID: PMC6891135 DOI: 10.1002/adma.201903637] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/12/2019] [Indexed: 05/07/2023]
Abstract
With the recent FDA approval of the first siRNA-derived therapeutic, RNA interference (RNAi)-mediated gene therapy is undergoing a transition from research to the clinical space. The primary obstacle to realization of RNAi therapy has been the delivery of oligonucleotide payloads. Therefore, the main aims is to identify and describe key design features needed for nanoscale vehicles to achieve effective delivery of siRNA-mediated gene silencing agents in vivo. The problem is broken into three elements: 1) protection of siRNA from degradation and clearance; 2) selective homing to target cell types; and 3) cytoplasmic release of the siRNA payload by escaping or bypassing endocytic uptake. The in vitro and in vivo gene silencing efficiency values that have been reported in publications over the past decade are quantitatively summarized by material type (lipid, polymer, metal, mesoporous silica, and porous silicon), and the overall trends in research publication and in clinical translation are discussed to reflect on the direction of the RNAi therapeutics field.
Collapse
Affiliation(s)
- Byungji Kim
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Michael J Sailor
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| |
Collapse
|
16
|
Kardani K, Milani A, H Shabani S, Bolhassani A. Cell penetrating peptides: the potent multi-cargo intracellular carriers. Expert Opin Drug Deliv 2019; 16:1227-1258. [PMID: 31583914 DOI: 10.1080/17425247.2019.1676720] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Cell penetrating peptides (CPPs) known as protein translocation domains (PTD), membrane translocating sequences (MTS), or Trojan peptides (TP) are able to cross biological membranes without clear toxicity using different mechanisms, and facilitate the intracellular delivery of a variety of bioactive cargos. CPPs could overcome some limitations of drug delivery and combat resistant strains against a broad range of diseases. Despite delivery of different therapeutic molecules by CPPs, they lack cell specificity and have a short duration of action. These limitations led to design of combined cargo delivery systems and subsequently improvement of their clinical applications. Areas covered: This review covers all our studies and other researchers in different aspects of CPPs such as classification, uptake mechanisms, and biomedical applications. Expert opinion: Due to low cytotoxicity of CPPs as compared to other carriers and final degradation to amino acids, they are suitable for preclinical and clinical studies. Generally, the efficiency of CPPs was suitable to penetrate the cell membrane and deliver different cargos to specific intracellular sites. However, no CPP-based therapeutic approach has approved by FDA, yet; because there are some disadvantages for CPPs including short half-life in blood, and nonspecific CPP-mediated delivery to normal tissue. Thus, some methods were used to develop the functions of CPPs in vitro and in vivo including the augmentation of cell specificity by activatable CPPs, specific transport into cell organelles by insertion of corresponding localization sequences, incorporation of CPPs into multifunctional dendrimeric or liposomal nanocarriers to improve selectivity and efficiency especially in tumor cells.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Samaneh H Shabani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| |
Collapse
|
17
|
El-Sawy HS, Al-Abd AM, Ahmed TA, El-Say KM, Torchilin VP. Stimuli-Responsive Nano-Architecture Drug-Delivery Systems to Solid Tumor Micromilieu: Past, Present, and Future Perspectives. ACS NANO 2018; 12:10636-10664. [PMID: 30335963 DOI: 10.1021/acsnano.8b06104] [Citation(s) in RCA: 296] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The microenvironment characteristics of solid tumors, renowned as barriers that harshly impeded many drug-delivery approaches, were precisely studied, investigated, categorized, divided, and subdivided into a complex diverse of barriers. These categories were further studied with a particular perspective, which makes all barriers found in solid-tumor micromilieu turn into different types of stimuli, and were considered triggers that can increase and hasten drug-release targeting efficacy. This review gathers data concerning the nature of solid-tumor micromilieu. Past research focused on the treatment of such tumors, the recent efforts employed for engineering smart nanoarchitectures with the utilization of the specified stimuli categories, the possibility of combining more than one stimuli for much-greater targeting enhancement, examples of the approved nanoarchitectures that already translated clinically as well as the obstacles faced by the use of these nanostructures, and, finally, an overview of the possible future implementations of smart-chemical engineering for the design of more-efficient drug delivery and theranostic systems and for making nanosystems with a much-higher level of specificity and penetrability features.
Collapse
Affiliation(s)
- Hossam S El-Sawy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy , Egyptian Russian University , Badr City , Cairo 63514 , Egypt
| | - Ahmed M Al-Abd
- Department of Pharmaceutical Sciences, College of Pharmacy , Gulf Medical University , Ajman , United Arab Emirates
- Pharmacology Department, Medical Division , National Research Centre , Giza 12622 , Egypt
| | - Tarek A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Khalid M El-Say
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , 140 The Fenway, Room 211/214, 360 Huntington Aveue , Boston , Massachusetts 02115 , United States
| |
Collapse
|
18
|
Deshpande P, Jhaveri A, Pattni B, Biswas S, Torchilin V. Transferrin and octaarginine modified dual-functional liposomes with improved cancer cell targeting and enhanced intracellular delivery for the treatment of ovarian cancer. Drug Deliv 2018; 25:517-532. [PMID: 29433357 PMCID: PMC6058534 DOI: 10.1080/10717544.2018.1435747] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Off-target effects of drugs severely limit cancer therapy. Targeted nanocarriers are promising to enhance the delivery of therapeutics to tumors. Among many approaches for active tumor-targeting, arginine-rich cell penetrating peptides (AR-CPP) and ligands specific to target over-expressed receptors on cancer-cell surfaces, are popular. Earlier, we showed that the attachment of an AR-CPP octaarginine (R8) to the surface of DOXIL® (Doxorubicin encapsulated PEGylated liposomes) improved cytoplasmic and nuclear DOX delivery that enhanced the cytotoxic effect in vitro and improved therapeutic efficacy in vivo. Here, we report on DOX-loaded liposomes, surface-modified with, R8 and transferrin (Tf) (Dual DOX-L), to improve targeting of A2780 ovarian carcinoma cells via the over-expressed transferrin receptors (TfRs) with R8-mediated intracellular DOX delivery. Flow cytometry analysis with fluorescently labeled DualL (without DOX) showed two-fold higher cancer-cell association than other treatments after 4 h treatment. Blocking entry pathways of R8 (macropinocytosis) and Tf (receptor-mediated endocytosis, RME) resulted in a decreased cancer-cell association of DualL. Confocal microscopy confirmed involvement of both entry pathways and cytoplasmic liposome accumulation with nuclear DOX delivery for Dual DOX-L. Dual DOX-L exhibited enhanced cytotoxicity in vitro and was most effective in controlling tumor growth in vivo in an A2780 ovarian xenograft model compared to other treatments. A pilot biodistribution study showed improved DOX accumulation in tumors after Dual DOX-L treatment. All results collectively presented a clear advantage of the R8 and Tf combination to elevate the therapeutic potential of DOX-L by exploiting TfR over-expression imparting specificity followed by endosomal escape and intracellular delivery via R8.
Collapse
Affiliation(s)
- Pranali Deshpande
- a Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , Boston , MA , USA
| | - Aditi Jhaveri
- a Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , Boston , MA , USA
| | - Bhushan Pattni
- a Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , Boston , MA , USA
| | - Swati Biswas
- b Department of Pharmacy , Birla Institute of Technology & Science-Pilani, Hyderabad Campus , Hyderabad , India
| | - Vladimir Torchilin
- a Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , Boston , MA , USA
| |
Collapse
|
19
|
Intracellular delivery of colloids: Past and future contributions from microinjection. Adv Drug Deliv Rev 2018; 132:3-15. [PMID: 29935217 DOI: 10.1016/j.addr.2018.06.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/06/2018] [Accepted: 06/18/2018] [Indexed: 01/07/2023]
Abstract
The manipulation of single cells and whole tissues has been possible since the early 70's, when semi-automatic injectors were developed. Since then, microinjection has been used to introduce an ever-expanding range of colloids of up to 1000 nm in size into living cells. Besides injecting nucleic acids to study transfection mechanisms, numerous cellular pathways have been unraveled through the introduction of recombinant proteins and blocking antibodies. The injection of nanoparticles has also become popular in recent years to investigate toxicity mechanisms and intracellular transport, and to conceive semi-synthetic cells containing artificial organelles. This article reviews colloidal systems such as proteins, nucleic acids and nanoparticles that have been injected into cells for different research aims, and discusses the scientific advances achieved through them. The colloids' intracellular processing and ultimate fate are also examined from a drug delivery perspective with an emphasis on the differences observed for endocytosed versus microinjected material.
Collapse
|
20
|
Spicer CD, Jumeaux C, Gupta B, Stevens MM. Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications. Chem Soc Rev 2018; 47:3574-3620. [PMID: 29479622 PMCID: PMC6386136 DOI: 10.1039/c7cs00877e] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peptide- and protein-nanoparticle conjugates have emerged as powerful tools for biomedical applications, enabling the treatment, diagnosis, and prevention of disease. In this review, we focus on the key roles played by peptides and proteins in improving, controlling, and defining the performance of nanotechnologies. Within this framework, we provide a comprehensive overview of the key sequences and structures utilised to provide biological and physical stability to nano-constructs, direct particles to their target and influence their cellular and tissue distribution, induce and control biological responses, and form polypeptide self-assembled nanoparticles. In doing so, we highlight the great advances made by the field, as well as the challenges still faced in achieving the clinical translation of peptide- and protein-functionalised nano-drug delivery vehicles, imaging species, and active therapeutics.
Collapse
Affiliation(s)
- Christopher D Spicer
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, Stockholm, Sweden.
| | | | | | | |
Collapse
|
21
|
Cai L, Gu Z, Zhong J, Wen D, Chen G, He L, Wu J, Gu Z. Advances in glycosylation-mediated cancer-targeted drug delivery. Drug Discov Today 2018; 23:1126-1138. [DOI: 10.1016/j.drudis.2018.02.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/22/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022]
|
22
|
Chiani M, Norouzian D, Shokrgozar MA, Azadmanesh K, Najmafshar A, Mehrabi MR, Akbarzadeh A. Folic acid conjugated nanoliposomes as promising carriers for targeted delivery of bleomycin. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017. [DOI: 10.1080/21691401.2017.1337029] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Mohsen Chiani
- Nanobiotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Dariush Norouzian
- Nanobiotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Aazam Najmafshar
- Clinical Biochemistry Department, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Azim Akbarzadeh
- Nanobiotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
Chiani M, Azadmanesh K, Shokrgozar MA, Mehrabi MR, Akbarzadeh A, Norouzian D. Enhanced antitumor effect of targeted nanoliposomal bleomycin. Chem Biol Drug Des 2017; 90:953-961. [DOI: 10.1111/cbdd.13021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/24/2017] [Accepted: 05/01/2017] [Indexed: 12/01/2022]
Affiliation(s)
- Mohsen Chiani
- Pilot Nanobiotechnology Department; Pasteur Institute of Iran; Tehran Iran
| | | | | | | | - Azim Akbarzadeh
- Pilot Nanobiotechnology Department; Pasteur Institute of Iran; Tehran Iran
| | - Dariush Norouzian
- Pilot Nanobiotechnology Department; Pasteur Institute of Iran; Tehran Iran
| |
Collapse
|
24
|
Janicka M, Gubernator J. Use of nanotechnology for improved pharmacokinetics and activity of immunogenic cell death inducers used in cancer chemotherapy. Expert Opin Drug Deliv 2016; 14:1059-1075. [DOI: 10.1080/17425247.2017.1266333] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Martyna Janicka
- Faculty of Biotechnology, Department of Lipids and Liposomes, University of Wroclaw, Wroclaw, Poland
| | - Jerzy Gubernator
- Faculty of Biotechnology, Department of Lipids and Liposomes, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
25
|
Liu Y, Wu P, Jiang J, Wu J, Chen Y, Tan Y, Tan C, Jiang Y. Conjugated Polyelectrolyte Nanoparticles for Apoptotic Cell Imaging. ACS APPLIED MATERIALS & INTERFACES 2016; 8:21984-21989. [PMID: 27525500 DOI: 10.1021/acsami.6b09347] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Three anionic conjugated polyelectrolytes (CPEs) with poly(p-phenylene ethynylene thiophene) backbones were designed and synthesized, among which PPET3-CO2Na showed greater molar extinction coefficient with red-shifted bands in both absorption and emission spectra compared to the well-studied PPE-CO2Na polymer. PPET3-CO2Na was thus chosen to construct CPE-based nanoparticles (CPNs) with cationic octaarginine (R8) peptide through electrostatic-interaction-induced self-assembly. Due to plasma membrane permeabilization and mitochondrial outer membrane permeabilization (MOMP) in early apoptotic cells, PPET3/R8 CPNs demonstrated excellent colocalization with MitoTracker Red in apoptotic cells instead of normal cells, which had potential application in cell imaging for early apoptosis recognition.
Collapse
Affiliation(s)
- Yu Liu
- Department of Chemistry, Tsinghua University , Beijing 100084, P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab - Shenzhen Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University , Shenzhen 518055, P. R. China
| | - Pan Wu
- Department of Chemistry, Tsinghua University , Beijing 100084, P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab - Shenzhen Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University , Shenzhen 518055, P. R. China
| | - Jianhua Jiang
- Department of Chemistry, Tsinghua University , Beijing 100084, P. R. China
| | - Jiatao Wu
- Department of Chemistry, Tsinghua University , Beijing 100084, P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab - Shenzhen Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University , Shenzhen 518055, P. R. China
| | - Yan Chen
- The Ministry-Province Jointly Constructed Base for State Key Lab - Shenzhen Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University , Shenzhen 518055, P. R. China
- Shenzhen Technology and Engineering Laboratory for Personalized Cancer Diagnostics and Therapeutics, Shenzhen Kivita Innovative Drug Discovery Institute , Shenzhen 518055, P. R. China
| | - Ying Tan
- Department of Chemistry, Tsinghua University , Beijing 100084, P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab - Shenzhen Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University , Shenzhen 518055, P. R. China
| | - Chunyan Tan
- Department of Chemistry, Tsinghua University , Beijing 100084, P. R. China
- The Ministry-Province Jointly Constructed Base for State Key Lab - Shenzhen Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University , Shenzhen 518055, P. R. China
| | - Yuyang Jiang
- The Ministry-Province Jointly Constructed Base for State Key Lab - Shenzhen Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University , Shenzhen 518055, P. R. China
- Department of Pharmacology and Pharmaceutical Sciences, School of Medicine, Tsinghua University , Beijing 100084, P. R. China
| |
Collapse
|
26
|
Mussa Farkhani S, Asoudeh Fard A, Zakeri-Milani P, Shahbazi Mojarrad J, Valizadeh H. Enhancing antitumor activity of silver nanoparticles by modification with cell-penetrating peptides. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:1029-1035. [DOI: 10.1080/21691401.2016.1200059] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Samad Mussa Farkhani
- Research Center for Pharmaceutical Nanotechnology and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Asoudeh Fard
- Research Center for Pharmaceutical Nanotechnology and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Shahbazi Mojarrad
- Biotechnology Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
GUO ZHENGRONG, PENG HUANYAN, KANG JIWEN, SUN DIANXING. Cell-penetrating peptides: Possible transduction mechanisms and therapeutic applications. Biomed Rep 2016; 4:528-534. [PMID: 27123243 PMCID: PMC4840506 DOI: 10.3892/br.2016.639] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/09/2016] [Indexed: 01/09/2023] Open
Abstract
Cell-penetrating peptides (CPPs), also known as protein transduction domains, are a class of diverse peptides with 5-30 amino acids. CPPs are divided into cationic, amphipathic and hydrophobic CPPs. They are able to carry small molecules, plasmid DNA, small interfering RNA, proteins, viruses, imaging agents and other various nanoparticles across the cellular membrane, resulting in internalization of the intact cargos. However, the mechanisms of CPP internalization remain to be elucidated. Recently, CPPs have received considerable attention due to their high transduction efficiency and low cytotoxicity. These peptides have a significant potential for diagnostic and therapeutic applications, such as delivery of fluorescent or radioactive compounds for imaging, delivery of peptides and proteins for therapeutic application, and delivery of molecules into induced pluripotent stem cells for directing differentiation. The present study reviews the classifications and transduction mechanisms of CPPs, as well as their potential applications.
Collapse
Affiliation(s)
- ZHENGRONG GUO
- The Liver Diseases Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, Hebei 050082, P.R. China
- Department of Gastroenterology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - HUANYAN PENG
- The Liver Diseases Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, Hebei 050082, P.R. China
- Hebei Medical University Graduate School, Shijiazhuang, Hebei 050017, P.R. China
| | - JIWEN KANG
- The Liver Diseases Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, Hebei 050082, P.R. China
| | - DIANXING SUN
- The Liver Diseases Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, Hebei 050082, P.R. China
| |
Collapse
|
28
|
Jhaveri A, Torchilin V. Intracellular delivery of nanocarriers and targeting to subcellular organelles. Expert Opin Drug Deliv 2015; 13:49-70. [DOI: 10.1517/17425247.2015.1086745] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
Valizadeh H, Ghanbarzadeh S, Zakeri-Milani P. Fusogenic liposomal formulation of sirolimus: improvement of drug anti-proliferative effect on human T-cells. Drug Dev Ind Pharm 2014; 41:1558-65. [DOI: 10.3109/03639045.2014.971032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
30
|
Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat Rev Drug Discov 2014; 13:813-27. [PMID: 25287120 DOI: 10.1038/nrd4333] [Citation(s) in RCA: 1063] [Impact Index Per Article: 96.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The use of nanoparticulate pharmaceutical drug delivery systems (NDDSs) to enhance the in vivo effectiveness of drugs is now well established. The development of multifunctional and stimulus-sensitive NDDSs is an active area of current research. Such NDDSs can have long circulation times, target the site of the disease and enhance the intracellular delivery of a drug. This type of NDDS can also respond to local stimuli that are characteristic of the pathological site by, for example, releasing an entrapped drug or shedding a protective coating, thus facilitating the interaction between drug-loaded nanocarriers and target cells or tissues. In addition, imaging contrast moieties can be attached to these carriers to track their real-time biodistribution and accumulation in target cells or tissues. Here, I highlight recent developments with multifunctional and stimuli-sensitive NDDSs and their therapeutic potential for diseases including cancer, cardiovascular diseases and infectious diseases.
Collapse
|
31
|
Abstract
The use of nanoparticulate pharmaceutical drug delivery systems (NDDSs) to enhance the in vivo effectiveness of drugs is now well established. The development of multifunctional and stimulus-sensitive NDDSs is an active area of current research. Such NDDSs can have long circulation times, target the site of the disease and enhance the intracellular delivery of a drug. This type of NDDS can also respond to local stimuli that are characteristic of the pathological site by, for example, releasing an entrapped drug or shedding a protective coating, thus facilitating the interaction between drug-loaded nanocarriers and target cells or tissues. In addition, imaging contrast moieties can be attached to these carriers to track their real-time biodistribution and accumulation in target cells or tissues. Here, I highlight recent developments with multifunctional and stimuli-sensitive NDDSs and their therapeutic potential for diseases including cancer, cardiovascular diseases and infectious diseases.
Collapse
|
32
|
Mansourian M, Badiee A, Jalali SA, Shariat S, Yazdani M, Amin M, Jaafari MR. Effective induction of anti-tumor immunity using p5 HER-2/neu derived peptide encapsulated in fusogenic DOTAP cationic liposomes co-administrated with CpG-ODN. Immunol Lett 2014; 162:87-93. [PMID: 25086399 DOI: 10.1016/j.imlet.2014.07.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/06/2014] [Accepted: 07/21/2014] [Indexed: 12/29/2022]
Abstract
Cationic liposomes have been used as efficient antigen delivery systems for cancer vaccination. The current study has investigated whether the incorporation of the helper-fusogenic lipid dioleoylphosphatidylethanolamine (DOPE) in cationic liposomes composed of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP)-cholesterol enhances the cytosolic delivery of p5 HER-2/neu derived peptide (p5) and promotes cytotoxic T lymphocytes (CTL) response. The p5, which is a very hydrophobic peptide, was encapsulated into liposomes by using three different methods and characterized for their colloidal properties. A chaotropic loading method using 7 M urea provided the highest encapsulation yields. Mice were first immunized with encapsulated p5 in liposomes composed of either DOTAP-cholesterol or DOTAP-cholesterol-DOPE, alone or co-administered with CpG-ODN, as an immunoadjuvant, then, inoculated with a subcutaneous injection of TUBO tumor cells. Results obtained from enzyme-linked immunospot, cytotoxicity and intracellular cytokine assays as well as tumor sizes and animal survival analysis demonstrated that p5 encapsulated in DOTAP-cholesterol-DOPE liposomes co-administered with CpG-ODN greatly enhanced the cytotoxic T lymphocytes response and highly inhibited the tumor progression. The outperformance of DOTAP-cholesterol-DOPE liposomes+CpG-ODN was found to be attributed to its capability in induction of both CD8+ and CD4+ responses. This formulation could be a suitable vaccine candidate against Her2 positive cancers and merits further investigations.
Collapse
Affiliation(s)
- Mercedeh Mansourian
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Amir Jalali
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Immunogenetic and Cell Culture Department, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sheida Shariat
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Yazdani
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohamdreza Amin
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
33
|
Sriraman SK, Aryasomayajula B, Torchilin VP. Barriers to drug delivery in solid tumors. Tissue Barriers 2014; 2:e29528. [PMID: 25068098 PMCID: PMC4106925 DOI: 10.4161/tisb.29528] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 06/06/2014] [Accepted: 06/09/2014] [Indexed: 02/07/2023] Open
Abstract
Over the last decade, significant progress has been made in the field of drug delivery. The advent of engineered nanoparticles has allowed us to circumvent the initial limitations to drug delivery such as pharmacokinetics and solubility. However, in spite of significant advances to tumor targeting, an effective treatment strategy for malignant tumors still remains elusive. Tumors possess distinct physiological features which allow them to resist traditional treatment approaches. This combined with the complexity of the biological system presents significant hurdles to the site-specific delivery of therapeutic drugs. One of the key features of engineered nanoparticles is that these can be tailored to execute specific functions. With this review, we hope to provide the reader with a clear understanding and knowledge of biological barriers and the methods to exploit these characteristics to design multifunctional nanocarriers, effect useful dosing regimens and subsequently improve therapeutic outcomes in the clinic.
Collapse
Affiliation(s)
- Shravan Kumar Sriraman
- Center for Pharmaceutical Biotechnology and Nanomedicine; Northeastern University; Boston, MA USA
| | - Bhawani Aryasomayajula
- Center for Pharmaceutical Biotechnology and Nanomedicine; Northeastern University; Boston, MA USA
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine; Northeastern University; Boston, MA USA
| |
Collapse
|
34
|
Farkhani SM, Valizadeh A, Karami H, Mohammadi S, Sohrabi N, Badrzadeh F. Cell penetrating peptides: efficient vectors for delivery of nanoparticles, nanocarriers, therapeutic and diagnostic molecules. Peptides 2014; 57:78-94. [PMID: 24795041 DOI: 10.1016/j.peptides.2014.04.015] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/19/2014] [Accepted: 04/19/2014] [Indexed: 01/24/2023]
Abstract
Efficient delivery of therapeutic and diagnostic molecules to the cells and tissues is a difficult challenge. The cellular membrane is very effective in its role as a selectively permeable barrier. While it is essential for cell survival and function, also presents a major barrier for intracellular delivery of cargo such as therapeutic and diagnostic agents. In recent years, cell-penetrating peptides (CPPs), that are relatively short cationic and/or amphipathic peptides, received great attention as efficient cellular delivery vectors due to their intrinsic ability to enter cells and mediate uptake of a wide range of macromolecular cargo such as plasmid DNA (pDNA), small interfering RNA (siRNAs), drugs, and nanoparticulate pharmaceutical carriers. This review discusses the various uptake mechanisms of these peptides. Furthermore, we discuss recent advances in the use of CPP for the efficient delivery of nanoparticles, nanocarriers, DNA, siRNA, and anticancer drugs to the cells. In addition, we have been highlighting new results for improving endosomal escape of CPP-cargo molecules. Finally, pH-responsive and activable CPPs for tumor-targeting therapy have been described.
Collapse
Affiliation(s)
- Samad Mussa Farkhani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alireza Valizadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran.
| | - Hadi Karami
- Department of Medical Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Sardasht, 38481 Arak, Iran.
| | - Samane Mohammadi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran.
| | - Nasrin Sohrabi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran.
| | - Fariba Badrzadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, 51664 Tabriz, Iran.
| |
Collapse
|
35
|
Biswas S, Torchilin VP. Nanopreparations for organelle-specific delivery in cancer. Adv Drug Deliv Rev 2014; 66:26-41. [PMID: 24270008 DOI: 10.1016/j.addr.2013.11.004] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 10/30/2013] [Accepted: 11/13/2013] [Indexed: 01/07/2023]
Abstract
To efficiently deliver therapeutics into cancer cells, a number of strategies have been recently investigated. The toxicity associated with the administration of chemotherapeutic drugs due to their random interactions throughout the body necessitates the development of drug-encapsulating nanopreparations that significantly mask, or reduce, the toxic side effects of the drugs. In addition to reduced side effects associated with drug encapsulation, nanocarriers preferentially accumulate in tumors as a result of its abnormally leaky vasculature via the Enhanced Permeability and Retention (EPR) effect. However, simple passive nanocarrier delivery to the tumor site is unlikely to be enough to elicit a maximum therapeutic response as the drug-loaded carriers must reach the intracellular target sites. Therefore, efficient translocation of the nanocarrier through the cell membrane is necessary for cytosolic delivery of the cargo. However, crossing the cell membrane barrier and reaching cytosol might still not be enough for achieving maximum therapeutic benefit, which necessitates the delivery of drugs directly to intracellular targets, such as bringing pro-apoptotic drugs to mitochondria, nucleic acid therapeutics to nuclei, and lysosomal enzymes to defective lysosomes. In this review, we discuss the strategies developed for tumor targeting, cytosolic delivery via cell membrane translocation, and finally organelle-specific targeting, which may be applied for developing highly efficacious, truly multifunctional, cancer-targeted nanopreparations.
Collapse
Affiliation(s)
- Swati Biswas
- Center for Pharmaceutical Biotechnology and Nanomedicine, 360 Huntington Avenue, 140 The Fenway, Northeastern University, Boston, 02115, USA; Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad, Andhra Pradesh 500078, India
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, 360 Huntington Avenue, 140 The Fenway, Northeastern University, Boston, 02115, USA.
| |
Collapse
|
36
|
Deshpande PP, Biswas S, Torchilin VP. Current trends in the use of liposomes for tumor targeting. Nanomedicine (Lond) 2013; 8:1509-28. [PMID: 23914966 PMCID: PMC3842602 DOI: 10.2217/nnm.13.118] [Citation(s) in RCA: 436] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The use of liposomes for drug delivery began early in the history of pharmaceutical nanocarriers. These nanosized, lipid bilayered vesicles have become popular as drug delivery systems owing to their efficiency, biocompatibility, nonimmunogenicity, enhanced solubility of chemotherapeutic agents and their ability to encapsulate a wide array of drugs. Passive and ligand-mediated active targeting promote tumor specificity with diminished adverse off-target effects. The current field of liposomes focuses on both clinical and diagnostic applications. Recent efforts have concentrated on the development of multifunctional liposomes that target cells and cellular organelles with a single delivery system. This review discusses the recent advances in liposome research in tumor targeting.
Collapse
Affiliation(s)
- Pranali P Deshpande
- Center for Pharmaceutical Biotechnology & Nanomedicine, 360 Huntington Avenue, 140 The Fenway, Northeastern University, Boston, MA 02115, USA
| | - Swati Biswas
- Center for Pharmaceutical Biotechnology & Nanomedicine, 360 Huntington Avenue, 140 The Fenway, Northeastern University, Boston, MA 02115, USA
- Department of Pharmacy, Birla Institute of Technology & Sciences – PiIani, Hyderabad Campus, Jawahar Nagar, Hyderabad, Andhra Pradesh 500078, India
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology & Nanomedicine, 360 Huntington Avenue, 140 The Fenway, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
37
|
Impedimetric detection of in situ interaction between anti-cancer drug bleomycin and DNA. Int J Biol Macromol 2013; 61:295-301. [PMID: 23892034 DOI: 10.1016/j.ijbiomac.2013.07.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 07/21/2013] [Accepted: 07/23/2013] [Indexed: 11/21/2022]
Abstract
Surface confined interaction of anti-cancer drug bleomycin (BLM) with nucleic acids: single stranded and double stranded DNA was investigated herein by using electrochemical impedance spectroscopy (EIS) technique in combination with a graphite sensor technology. The experimental conditions were optimized: such as, dsDNA concentration, BLM concentration and interaction time. The main features of impedimetric DNA biosensor, such as its detection limit and the repeatability, were also discussed. The in situ interaction of BLM with dsDNA was also tested impedimetrically in the absence or presence of other chemotherapeutic agents, such as mitomycin C (MC) and cis-platin (cis-DDP) for testing the selectivity.
Collapse
|
38
|
Recent trends in multifunctional liposomal nanocarriers for enhanced tumor targeting. JOURNAL OF DRUG DELIVERY 2013; 2013:705265. [PMID: 23533772 PMCID: PMC3606784 DOI: 10.1155/2013/705265] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 02/06/2013] [Indexed: 12/30/2022]
Abstract
Liposomes are delivery systems that have been used to formulate a vast variety of therapeutic and imaging agents for the past several decades. They have significant advantages over their free forms in terms of pharmacokinetics, sensitivity for cancer diagnosis and therapeutic efficacy. The multifactorial nature of cancer and the complex physiology of the tumor microenvironment require the development of multifunctional nanocarriers. Multifunctional liposomal nanocarriers should combine long blood circulation to improve pharmacokinetics of the loaded agent and selective distribution to the tumor lesion relative to healthy tissues, remote-controlled or tumor stimuli-sensitive extravasation from blood at the tumor's vicinity, internalization motifs to move from tumor bounds and/or tumor intercellular space to the cytoplasm of cancer cells for effective tumor cell killing. This review will focus on current strategies used for cancer detection and therapy using liposomes with special attention to combination therapies.
Collapse
|
39
|
Biswas S, Deshpande PP, Perche F, Dodwadkar NS, Sane SD, Torchilin VP. Octa-arginine-modified pegylated liposomal doxorubicin: an effective treatment strategy for non-small cell lung cancer. Cancer Lett 2013; 335:191-200. [PMID: 23419527 DOI: 10.1016/j.canlet.2013.02.020] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/09/2013] [Accepted: 02/08/2013] [Indexed: 02/01/2023]
Abstract
The present study aims to evaluate the efficacy of octa-arginine (R8)-modified pegylated liposomal doxorubicin (R8-PLD) for the treatment of non-small cell lung cancer, for which the primary treatment modality currently consists of surgery and radiotherapy. Cell-penetrating peptide R8 modification of Doxorubicin-(Dox)-loaded liposomes was performed by post-insertion of an R8-conjugated amphiphilic PEG-PE copolymer (R8-PEG-DOPE) into the liposomal lipid bilayer. In vitro analysis with the non-small cell lung cancer cell line, A549 confirmed the efficient cellular accumulation of Dox, delivered by R8-PLD compared to PLD. It led to the early initiation of apoptosis and a 9-fold higher level of the apoptotic regulator, caspase 3/7 (9.24±0.34) compared to PLD (1.07±0.19) at Dox concentration of 100 μg/mL. The treatment of A549 monolayers with R8-PLD increased the level of cell death marker lactate dehydrogenase (LDH) secretion (1.2±0.1 for PLD and 2.3±0.1 for R8-PLD at Dox concentration of 100 μg/mL) confirming higher cytotoxicity of R8-PLD than PLD, which was ineffective under the same treatment regimen (cell viability 90±6% in PLD vs. 45±2% in R8-PLD after 24h). R8-PLD had significantly higher penetration into the hypoxic A549 tumor spheroids compared to PLD. R8-PLD induced greater level of apoptosis to A549 tumor xenograft and dramatic inhibition of tumor volume and tumor weight reduction. The R8-PLD treated tumor lysate had a elevated caspase 3/7 expression than with R8-PLD treatment. This suggested system improved the delivery efficiency of Dox in selected model of cancer which supports the potential usefulness of R8-PLD in cancer treatment, lung cancer in particular.
Collapse
Affiliation(s)
- Swati Biswas
- Center for Pharmaceutical Biotechnology and Nanomedicine, 360 Huntington Avenue, 140 The Fenway, Northeastern University, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
40
|
Surface functionalization of doxorubicin-loaded liposomes with octa-arginine for enhanced anticancer activity. Eur J Pharm Biopharm 2013; 84:517-25. [PMID: 23333899 DOI: 10.1016/j.ejpb.2012.12.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 12/26/2012] [Accepted: 12/27/2012] [Indexed: 11/21/2022]
Abstract
Doxorubicin-loaded PEGylated liposomes (commercially available as DOXIL or Lipodox) were surface functionalized with a cell-penetrating peptide, octa-arginine (R8). For this purpose, R8-peptide was conjugated to the polyethylene glycol-dioleoyl phosphatidylethanolamine (PEG-DOPE) amphiphilic co-polymer. The resultant R8-PEG-PE conjugate was introduced into the lipid bilayer of liposomes at 2 mol% of total lipid amount via spontaneous micelle-transfer technique. The liposomal modification did not alter the particle size distribution, as measured by Particle Size Analyzer and transmission electron microscopy (TEM). However, surface-associated cationic peptide increased zeta potential of the modified liposomes. R8-functionalized liposomes (R8-Dox-L) markedly increased the intracellular and intratumoral delivery of doxorubicin as measured by flow cytometry and visualizing by confocal laser scanning microscopy (CLSM) compared to unmodified Doxorubicin-loaded PEGylated liposomes (Dox-L). R8-Dox-L delivered loaded Doxorubicin to the nucleus, being released from the endosomes at higher efficiency compared to unmodified liposomes, which had marked entrapment in the endosomes at tested time point of 1h. The significantly higher accumulation of loaded drug to its site of action for R8-Dox-L resulted in improved cytotoxic activity in vitro (cell viability of 58.5 ± 7% for R8-Dox-L compared to 90.6 ± 2% for Dox-L at Dox dose of 50 μg/mL for 4h followed by 24h incubation) and enhanced suppression of tumor growth (348 ± 53 mm(3) for R8-Dox-L, compared to 504 ± 54 mm(3) for Dox-L treatment) in vivo compared to Dox-L. R8-modification has the potential for broadening the therapeutic window of pegylated liposomal doxorubicin treatment, which could lead to lower non-specific toxicity.
Collapse
|