1
|
Chu L, Sun Y, Zhao Y, Wang A, Sun Y, Duan X, Li N, Xia H, Liu W, Sun K. Exosome-mediated delivery platform of biomacromolecules into the brain: Cetuximab in combination with doxorubicin for glioblastoma therapy. Int J Pharm 2024; 660:124262. [PMID: 38815637 DOI: 10.1016/j.ijpharm.2024.124262] [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: 01/23/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
Monoclonal antibodies (mAbs) have become the predominant treatment modality for various diseases due to their high affinity and specificity. Although antibodies also have great potential for neurological diseases, they couldn't fully meet the therapeutic requirements due to their high molecular weight and limitations in crossing the blood-brain barrier (BBB). Herein, an innovative strategy based on exosomes (Exos) platform was developed to enhance the delivery of cetuximab (CTX) into the brain, and in combination with doxorubicin (DOX) for the synergistic targeted therapy of glioblastoma (GBM). The in vitro/vivo experiments have shown that exosomes could effectively promote BBB penetration and increase the content of CTX in glioma cells and brain lesions. Cytotoxicity and wound healing experiments have shown that CTX-Exo-DOX could significantly inhibit the proliferation of tumor cells. Finally, in vivo results showed that CTX-Exo-DOX significantly prolonged the survival time of tumor-bearing rats to 28 days, which was 1.47 times that of the DOX group. In summary, exosomes could deliver more antibodies into the brain, and CTX-Exo-DOX is a promising co-delivery system for the treatment of GBM. The results of this study will also provide a prospective strategy for antibody drugs in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Liuxiang Chu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China; Yantai Laishan Fourth People's Hospital, Yantai, 264003, China
| | - Yuchen Sun
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China
| | - Yanyan Zhao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China
| | - Aiping Wang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China.
| | - Yiying Sun
- Shandong Business Institute, Yantai, 264670, China
| | - Xinliu Duan
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China
| | - Nuannuan Li
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China
| | - Hangyu Xia
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China
| | - Wanhui Liu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China
| | - Kaoxiang Sun
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Novapath Pharma (Chengdu) Co., Ltd., Chengdu, Sichuan 610200, China.
| |
Collapse
|
2
|
Dong T, Yang N, Qin J, Zhao C, Gao T, Ma H, Zhu C, Xu H. Tanshinone IIA Liposomes Treat Doxorubicin-Induced Glomerulonephritis by Modulating the Microenvironment of Fibrotic Kidneys. Mol Pharm 2024; 21:3281-3295. [PMID: 38848439 DOI: 10.1021/acs.molpharmaceut.4c00042] [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: 06/09/2024]
Abstract
Renal fibrosis plays a key role in the pathogenesis of chronic kidney disease (CKD), in which the persistent high expression of transforming growth factor β1 (TGF-β1) and α-smooth muscle actin (α-SMA) contributes to the progression of CKD to renal failure. In order to improve the solubility, bioavailability, and targeting of tanshinone IIA (Tan IIA), a novel targeting material, aminoethyl anisamide-polyethylene glycol-1,2-distearoyl-sn-glycero-3-phosphate ethanolamine (AEAA-PEG-DSPE, APD) modified Tan IIA liposomes (APD-Tan IIA-L) was constructed. An animal model of glomerulonephritis induced by doxorubicin in BALB/c mice was established. APD-Tan IIA-L significantly decreased blood urea nitrogen and serum creatinine (SCr), and the consequences of renal tissue oxidative stress indicators showed that APD-Tan IIA-L downregulated malondialdehyde, upregulated superoxide dismutase, catalase, and glutathione peroxidase. Masson's trichrome staining showed that the deposition of collagen in the APD-Tan IIA-L group decreased significantly. The pro-fibrotic factors (fibronectin, collagen I, TGF-β1, and α-SMA) and epithelial-mesenchymal transition marker (N-cadherin) were significantly inhibited by APD-Tan IIA-L. By improving the microenvironment of fibrotic kidneys, APD-Tan IIA-L attenuated TGF-β1-induced excessive proliferation of fibroblasts and alleviated oxidative stress damage to the kidney, providing a new strategy for the clinical treatment of renal fibrosis.
Collapse
Affiliation(s)
- Tingjun Dong
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Ning Yang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Jian Qin
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Chengcheng Zhao
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Tingyu Gao
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Hao Ma
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Caili Zhu
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Huan Xu
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| |
Collapse
|
3
|
Yang W, Sun Q, Zhang X, Zheng L, Yang X, He N, Pang Y, Wang X, Lai Z, Zheng W, Zheng S, Wang W. A novel doxorubicin/CTLA-4 blocker co-loaded drug delivery system improves efficacy and safety in antitumor therapy. Cell Death Dis 2024; 15:386. [PMID: 38824143 PMCID: PMC11144200 DOI: 10.1038/s41419-024-06776-6] [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: 12/04/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/03/2024]
Abstract
Doxorubicin's antitumor effectiveness may be constrained with ineffective tumor penetration, systemic adverse effects, as well as drug resistance. The co-loading of immune checkpoint inhibitors and doxorubicin into liposomes can produce synergistic benefits and address problems, including quick drug clearance, toxicity, and low drug penetration efficiency. In our previous study, we modified a nanobody targeting CTLA-4 onto liposomes (LPS-Nb36) to be an extremely potent CTLA-4 signal blocker which improve the CD8+ T-cell activity against tumors under physiological conditions. In this study, we designed a drug delivery system (LPS-RGD-Nb36-DOX) based on LPS-Nb36 that realized the doxorubicin and anti-CTLA-4 Nb co-loaded and RGD modification, and was applied to antitumor therapy. We tested whether LPS-RGD-Nb36-DOX could targets the tumor by in vivo animal photography, and more importantly, promote cytotoxic T cells proliferation, pro-inflammatory cytokine production, and cytotoxicity. Our findings demonstrated that the combination of activated CD8+ T cells with doxorubicin/anti-CTLA-4 Nb co-loaded liposomes can effectively eradicate tumor cells both in vivo and in vitro. This combination therapy is anticipated to have synergistic antitumor effects. More importantly, it has the potential to reduce the dose of chemotherapeutic drugs and improve safety.
Collapse
Affiliation(s)
- Wenli Yang
- Public Research Center, Hainan Medical University, Haikou, China
- Department of Anatomy, Zunyi Medical University, Zunyi, China
| | - Qinghui Sun
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Tropical Medicine, Hainan MedicalUniversity, Haikou, China
| | - Xiaodian Zhang
- Hainan Cancer Medical Center of The First Affiliated Hospital, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases, Hainan Medical University, Haikou, China
| | - Liping Zheng
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiaomei Yang
- Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Na He
- School of Tropical Medicine, Hainan MedicalUniversity, Haikou, China
| | - Yanyang Pang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, China
| | - Xi Wang
- Department of Anesthesiology, Haikou Third People's Hospital, Haikou, China
| | - Zhiheng Lai
- Department of Anorectal, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, China
| | - Wuping Zheng
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.
| | - Shaoping Zheng
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wu Wang
- Public Research Center, Hainan Medical University, Haikou, China.
| |
Collapse
|
4
|
Wang S, Wang H, Drabek A, Smith WS, Liang F, Huang ZR. Unleashing the Potential: Designing Antibody-Targeted Lipid Nanoparticles for Industrial Applications with CMC Considerations and Clinical Outlook. Mol Pharm 2024; 21:4-17. [PMID: 38117251 DOI: 10.1021/acs.molpharmaceut.3c00735] [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: 12/21/2023]
Abstract
Antibody-targeted lipid nanoparticles (Ab-LNPs) are rapidly gaining traction as multifaceted platforms in precision medicine, adept at delivering a diverse array of therapeutic agents, including nucleic acids and small molecules. This review provides an incisive overview of the latest developments in the field of Ab-LNP technology, with a special emphasis on pivotal design aspects such as antibody engineering, bioconjugation strategies, and advanced formulation techniques. Furthermore, it addresses critical chemistry, manufacturing, and controls (CMC) considerations and thoroughly examines the in vivo dynamics of Ab-LNPs, underscoring their promising potential for clinical application. By seamlessly blending scientific advancements with practical industrial perspectives, this review casts a spotlight on the burgeoning role of Ab-LNPs as an innovative and potent tool in the realm of targeted drug delivery.
Collapse
Affiliation(s)
- Sheryl Wang
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Hong Wang
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Andrew Drabek
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Wenwen Sha Smith
- FUSION BioVenture, 15 Presidential Way, Woburn, Massachusetts 01801, United States
| | - Feng Liang
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| | - Zhaohua Richard Huang
- Sanofi, Genomic Medicine Unit, 225 Second Avenue, Waltham, Massachusetts 02451, United States
| |
Collapse
|
5
|
Yang W, Pang Y, Wang X, Lai Z, Lu Y, Zheng S, Wang W. A novel CTLA-4 blocking strategy based on nanobody enhances the activity of dendritic cell vaccine-stimulated antitumor cytotoxic T lymphocytes. Cell Death Dis 2023; 14:406. [PMID: 37419930 PMCID: PMC10328924 DOI: 10.1038/s41419-023-05914-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023]
Abstract
Despite the great success of CTLA-4 blocking in cancer treatment, the use of anti-CTLA-4 monoclonal antibodies still faces many limitations. Now, immune checkpoint blocking coupled with adoptive cell therapy is gaining much attention. In this paper, we reported a strategy on the basis of anti-CTLA-4 nanobody (Nb)-modified liposomes to improve these obstacles. An Nb36/liposome complex was constructed and utilized as a blocker of the CTLA-4/B7 signal pathway in a combination with dendritic cell (DC)/tumor fusion vaccine to enhance the CD8+ T cell cytokine secretion, activation, proliferation, as well as specific cytotoxicity. Moreover, the CD8+ T cells induced by LPS-Nb36 and DC/tumor fusion vaccine led to higher CD8+ T cell effector function in vivo, which significantly retarded tumor growth and lengthened survival of tumor-bearing mice (HepG2, A549, and MGC-803). Our data demonstrate that the anti-CTLA-4 Nb-modified liposomes in connection with DC/tumor fusion vaccines enhance the CD8+ T cell antitumor activity in vitro and in vivo, and is expected to be an alternative therapy for patients with malignancies that have T cell dysfunction or have poor treatment against anti-CTLA-4 mAb.
Collapse
Affiliation(s)
- Wenli Yang
- Public Research Center of Hainan Medical University, Hainan Medical University, Haikou, 570100, China
- Tumor Institute, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 571199, China
- Department of Anatomy, Zunyi Medical University, Zunyi, 563006, China
| | - Yanyang Pang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, 570100, China
- Guangxi Key Laboratory of Nanobody Research, Guangxi Medical University, Nanning, 530021, China
| | - Xi Wang
- Department of Anesthesiology, Haikou Third People's Hospital, Haikou, 570100, China
| | - Zhiheng Lai
- Department of Anorectal, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, 570100, China
| | - Yanda Lu
- Tumor Institute, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
| | - Shaojiang Zheng
- Tumor Institute, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 571199, China.
| | - Wu Wang
- Public Research Center of Hainan Medical University, Hainan Medical University, Haikou, 570100, China.
| |
Collapse
|
6
|
Biabangard A, Asoodeh A, Jaafari MR, Moosavi F. AR13 peptide-conjugated liposomes improve the antitumor efficacy of doxorubicin in mice bearing C26 colon carcinoma; in silico, in vitro, and in vivo study. Toxicol Appl Pharmacol 2023; 466:116470. [PMID: 36933622 DOI: 10.1016/j.taap.2023.116470] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023]
Abstract
Currently, liposomes have emerged as efficient and safer nano-carriers for targeted therapy in different cancers. This work aimed to employ PEGylated liposomal doxorubicin (Doxil®/PLD), modified with AR13 peptide, to target Muc1 on the surface of colon cancerous cells. We performed molecular docking and simulation studies (using Gromacs package) of AR13 peptide against Muc1 to analyze and visualize the peptide-Muc1 binding combination. For in vitro analysis, the AR13 peptide was post-inserted into Doxil® and verified by TLC, 1H NMR, and HPLC techniques. The zeta potential, TEM, release, cell uptake, competition assay, and cytotoxicity studies were performed. In vivo antitumor activities and survival analysis on mice bearing C26 colon carcinoma were studied. Results showed that after 100 ns simulation, a stable complex between AR13 and Muc1 formed, and molecular dynamics analysis confirmed this interaction. In vitro analysis demonstrated significant enhancement of cellular binding and cell uptake. The results of in vivo study on BALB/c mice bearing C26 colon carcinoma, revealed an extended survival time to 44 days and higher tumor growth inhibition compared to Doxil®. Thus, the AR13 peptide could be explored as a potent ligand for Muc1, improving therapeutic antitumor efficiency in colon cancer cells.
Collapse
Affiliation(s)
- Atefeh Biabangard
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Fatemeh Moosavi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
7
|
de Freitas JVB, Reis AVF, Silva ADO, de Sousa ACC, Martins JRP, Nogueira KAB, da Silva Moreira T, Petrilli R, Eloy JO. Monoclonal Antibodies in Nanosystems as a Strategy for Cancer Treatment. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
8
|
Mishra AK, Pandey M, Dewangan HK, Sl N, Sahoo PK. A Comprehensive Review on Liver Targeting: Emphasis on Nanotechnology- based Molecular Targets and Receptors Mediated Approaches. Curr Drug Targets 2022; 23:1381-1405. [PMID: 36065923 DOI: 10.2174/1389450123666220906091432] [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: 11/10/2021] [Revised: 02/10/2022] [Accepted: 02/25/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND The pathogenesis of hepatic diseases involves several cells, which complicates the delivery of pharmaceutical agents. Many severe liver diseases affecting the worldwide population cannot be effectively treated. Major hindrances or challenges are natural physiological barriers and non-specific targeting of drugs administered, leading to inefficient treatment. Hence, there is an earnest need to look for novel therapeutic strategies to overcome these hindrances. A kind of literature has reported that drug safety and efficacy are incredibly raised when a drug is incorporated inside or attached to a polymeric material of either hydrophilic or lipophilic nature. This has driven the dynamic investigation for developing novel biodegradable materials, drug delivery carriers, target-specific drug delivery systems, and many other novel approaches. OBJECTIVE Present review is devoted to summarizing receptor-based liver cell targeting using different modified novel synthetic drug delivery carriers. It also highlights recent progress in drug targeting to diseased liver mediated by various receptors, including asialoglycoprotein, mannose and galactose receptor, Fc receptor, low-density lipoprotein, glycyrrhetinic, and bile acid receptor. The essential consideration is given to treating liver cancer targeting using nanoparticulate systems, proteins, viral and non-viral vectors, homing peptides and gene delivery. CONCLUSION Receptors based targeting approach is one such approach that was explored by researchers to develop novel formulations which can ensure site-specific drug delivery. Several receptors are on the surfaces of liver cells, which are highly overexpressed in various disease conditions. They all are helpful for the treatment of liver cancer.
Collapse
Affiliation(s)
- Ashwini Kumar Mishra
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Sector 3, MB Road Pushp Vihar, Delhi 110017, India
| | - Mukesh Pandey
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Sector 3, MB Road Pushp Vihar, Delhi 110017, India
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences (UIPS), Chandigarh University NH-05, Chandigarh Ludhiana Highway, Mohali Punjab, Pin: 160101, India
| | - Neha Sl
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Sector 3, MB Road Pushp Vihar, Delhi 110017, India
| | - Pravat Kumar Sahoo
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Sector 3, MB Road Pushp Vihar, Delhi 110017, India
| |
Collapse
|
9
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
10
|
Synthesis and evaluation of a novel adapter lipid derivative for preparation of cyclic peptide-modified PEGylated liposomes: Application of cyclic RGD peptide. Eur J Pharm Sci 2022; 176:106239. [PMID: 35714942 DOI: 10.1016/j.ejps.2022.106239] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/15/2022] [Accepted: 06/14/2022] [Indexed: 11/20/2022]
Abstract
Peptide ligand modified nanoparticles can simply prepared by post-insertion method to mix pre-formed nanoparticles with peptide-lipid conjugates in an aqueous solution at an optimal temperature. Therefore, water dispersibility of peptide-lipid conjugates is a very important factor for implementing the post-insertion method. We proposed that highly water dispersible peptide-lipid conjugates can be easily synthesized by separately designing novel adapter lipids with different water dispersibility and reacting them with ligands in a highly efficient manner. Adapter lipids have three critical roles; as spacers of ligand-conjugated lipids for efficient ligand presentation, as structures that form discrete molecular weight distributions, and as providing water dispersibility. In this study, we developed a novel adapter-lipid derivative that enables a variety of cyclic peptide modifications using the click reaction. The integrin αvβ3-targeted cyclic RGDfK (cRGD) peptide was selected as the cyclic peptide ligand. We designed a novel alkyne-tagged lipid with a discrete peptide spacer and bound the cRGD peptide using a click reaction to synthesize a cRGD-conjugated lipid with good water dispersibility for the preparation of cRGD-modified PEGylated liposomes using the post-insertion method. We also revealed that cRGD-modified PEGylated liposomes are efficiently associated with integrin αvβ3-expressing murine colon carcinoma (Colon-26) cells in a modification amount- and peptide sequence-dependent manner, showing high cytotoxicity upon loading with doxorubicin. This novel adapter lipid derivative can be used to synthesize various cyclic peptides by click reactions and will provide useful insights for the future development of cyclic peptide-modified PEGylated liposomes.
Collapse
|
11
|
Chen C, Zhou Y, Chen C, Zhu S, Yan X. Quantification of Available Ligand Density on the Surface of Targeted Liposomal Nanomedicines at the Single-Particle Level. ACS NANO 2022; 16:6886-6897. [PMID: 35394292 DOI: 10.1021/acsnano.2c02084] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Active targeting has been hailed as one of the most promising strategies to further enhance the therapeutic efficacy of liposomal nanomedicines. Owing to the critical role of ligand density in mediating cellular uptake and the intrinsic heterogeneity of liposomal formulations, precise quantification of the surface ligand density on a single-particle basis is of fundamental importance. In this work, we report a method to simultaneously measure the particle size and the number of ligands on the same liposomal nanoparticles by nanoflow cytometry. Then the ligand density for each individual liposome can be determined. With an analysis rate up to 10 000 particles per minute, a statistically representative distribution of ligand density could be determined in minutes. By utilizing fluorescently labeled recombinant receptors as the detection probe against the conjugated ligands, only those available for cell targeting can be exclusively detected. The influence of ligand input, conjugation strategy, and the polyethylene glycol spacer length on the available ligand density of folate-modified liposomes was investigated. The correlation between the available ligand density and cell targeting capability was assessed in a quantitative perspective for liposomes modified with three different targeting moieties. The optimal ligand density was determined to be 0.5-2.0, 0.7, and 0.2 ligand per 100 nm2 for folate-, transferrin-, and HER2-antibody-conjugated liposomes, respectively. These optimal values agreed well with the spike density of the natural counterparts, viruses. The as-developed approach is generally applicable to a wide range of active-targeting nanocarriers.
Collapse
Affiliation(s)
- Chaoxiang Chen
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, People's Republic of China
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Yingxing Zhou
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, People's Republic of China
| | - Chen Chen
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, People's Republic of China
| | - Shaobin Zhu
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, People's Republic of China
| | - Xiaomei Yan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, People's Republic of China
| |
Collapse
|
12
|
Swart LE, Koekman CA, Seinen CW, Issa H, Rasouli M, Schiffelers RM, Heidenreich O. A robust post-insertion method for the preparation of targeted siRNA LNPs. Int J Pharm 2022; 620:121741. [PMID: 35421533 DOI: 10.1016/j.ijpharm.2022.121741] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Targeted delivery of nucleic acids is gaining momentum due to improved efficacy, selectivity, increased circulation time and enhanced tissue retention in target cells. Using nucleic acid-based therapies previously undruggable targets have proven now to be amenable for treatment. Currently, several methods for preparing targeted or labelled delivery vehicles for nucleic acids are based on liposomal formulations. Lipid nanoparticles (LNPs) are structurally different from liposomes and these methods should therefore be evaluated before being translated to siRNA LNPs preparation protocols. Here, we describe a robust and facile method for the preparation of targeted or fluorescently labelled siRNA LNPs. Using a copper free strain-promoted azide-alkyne cycloaddition (SPAAC) we demonstrate that post-insertion of ligand-lipid conjugates into preformed LNPs is superior to direct-surface modification because it preserves the physicochemical parameters of the LNPs. We found that the time point of solvent removal by dialysis is critical and affects the hydrodynamic diameter of the LNPs; post-insertion after dialysis shows the smallest increase in hydrodynamic diameter and polydispersity index (PDI). The post-insertion of ligand-lipid conjugates also proceeded with rapid kinetics and high efficacy over a wide temperature range. Using this optimised protocol, we generated siRNA LNPs containing both targeting and fluorescent tracking ligands allowing us to monitor siRNA LNP uptake kinetics in dependence of the targeting ligand. In aggregate, we describe a robust approach for the generation of targeted and labelled siRNA LNPs that allows their controlled and facile decoration with ligand combinations.
Collapse
Affiliation(s)
- L E Swart
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - C A Koekman
- Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands
| | - C W Seinen
- Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands
| | - H Issa
- Department of Pediatrics, University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt (Main), Germany
| | - M Rasouli
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - R M Schiffelers
- Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands
| | - O Heidenreich
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
13
|
Poustforoosh A, Nematollahi MH, Hashemipour H, Pardakhty A. Recent advances in Bio-conjugated nanocarriers for crossing the Blood-Brain Barrier in (pre-)clinical studies with an emphasis on vesicles. J Control Release 2022; 343:777-797. [DOI: 10.1016/j.jconrel.2022.02.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022]
|
14
|
Carvalheiro M, Ferreira-Silva M, Holovanchuk D, Marinho HS, Moreira JN, Soares H, Corvo ML, Cruz MEM. Antagonist G-targeted liposomes for improved delivery of anticancer drugs in small cell lung carcinoma. Int J Pharm 2022; 612:121380. [PMID: 34915142 DOI: 10.1016/j.ijpharm.2021.121380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/19/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022]
Abstract
Ligand-mediated targeted liposomes have the potential to increase therapeutic efficacy of anticancer drugs. This work aimed to evaluate the ability of antagonist G, a peptide targeting agent capable of blocking the action of multiple neuropeptides, to selectivity improve targeting and internalization of liposomal formulations (long circulating liposomes, LCL, and stabilized antisense lipid particles containing ionizable amino lipid, SALP) to H69 and H82 small cell lung carcinoma (SCLC) cell lines. Antagonist G-targeted LCL and SALP were prepared by two different methods (either by direct covalent linkage at activated PEG grafted onto the liposomal surface or by post-insertion of DSPE-PEG-antagonist-G-conjugates into pre-formed liposomes). Association of the liposomal formulations with target SCLC cells was studied by fluorescence microscopy using fluorescence-labelled liposomes and confirmed quantitatively with [3H]-CHE-labelled liposomes. An antisense oligodeoxynucleotide against the overexpressed oncogene c-myc(as(c-myc)) was efficiently loaded into SALP formulations, the encapsulation efficiency decreased due to the inclusion of the targeting ligand. Also, liposome size was affected by as(c-myc) physical chemical properties. The amount of antagonist G linked to the surface of the liposomal formulations was dependent on the coupling method and lipid composition used. Covalent attachment of antagonist G increased liposomes cellular association and internalization via receptor-mediated and clathrin-dependent endocytosis, as assessed in SCLC cell lines. Biodistribution studies in healthy mice revealed a preferential lung accumulation of antagonist G-targeted SALP as compared to the non-targeted counterpart. Lung levels of the former were up to 3-fold higher 24 h after administration, highlighting their potential to be used as delivery vectors for SCLC treatment.
Collapse
Affiliation(s)
- Manuela Carvalheiro
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal; Departamento de Farmácia, Farmacologia e Tecnologias em Saúde, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Margarida Ferreira-Silva
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Denys Holovanchuk
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - H Susana Marinho
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal; Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - João Nuno Moreira
- CNC - Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; UC - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Helena Soares
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal
| | - M Luisa Corvo
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal; Departamento de Farmácia, Farmacologia e Tecnologias em Saúde, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Eugénia M Cruz
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
15
|
Geddie ML, Kirpotin DB, Kohli N, Kornaga T, Boll B, Razlog M, Drummond DC, Lugovskoy AA. Development of disulfide-stabilized Fabs for targeting of antibody-directed nanotherapeutics. MAbs 2022; 14:2083466. [PMID: 35708974 PMCID: PMC9225506 DOI: 10.1080/19420862.2022.2083466] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibody-directed nanotherapeutics (ADNs) represent a promising delivery platform for selective delivery of an encapsulated drug payload to the site of disease that improves the therapeutic index. Although both single-chain Fv (scFv) and Fab antibody fragments have been used for targeting, no platform approach applicable to any target has emerged. scFv can suffer from intrinsic instability, and the Fabs are challenging to use due to native disulfide over-reduction and resulting impurities at the end of the conjugation process. This occurs because of the close proximity of the disulfide bond connecting the heavy and light chain to the free cysteine at the C-terminus, which is commonly used as the conjugation site. Here we show that by engineering an alternative heavy chain-light chain disulfide within the Fab, we can maintain efficient conjugation while eliminating the process impurities and retaining stability. We have demonstrated the utility of this technology for efficient ADN delivery and internalization for a series of targets, including EphA2, EGFR, and ErbB2. We expect that this technology will be broadly applicable for targeting of nanoparticle encapsulated payloads, including DNA, mRNA, and small molecules.
Collapse
Affiliation(s)
- Melissa L Geddie
- Discovery, Merrimack Pharmaceuticals, Inc, Cambridge, Massachusetts, USA.,Research & Development, Diagonal Therapeutics, Cambridge, Massachusetts, USA
| | - Dmitri B Kirpotin
- Discovery, Merrimack Pharmaceuticals, Inc, Cambridge, Massachusetts, USA.,Research & Development, Akagera Medicines, San Francisco, CA, USA
| | - Neeraj Kohli
- Discovery, Merrimack Pharmaceuticals, Inc, Cambridge, Massachusetts, USA.,Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Tad Kornaga
- Discovery, Merrimack Pharmaceuticals, Inc, Cambridge, Massachusetts, USA
| | - Bjoern Boll
- Discovery, Merrimack Pharmaceuticals, Inc, Cambridge, Massachusetts, USA.,Drug Product Design, ten23 Health, Basel, Switzerland
| | - Maja Razlog
- Discovery, Merrimack Pharmaceuticals, Inc, Cambridge, Massachusetts, USA.,Research, Verseau Therapeutics, Bedford, Massachusetts, USA
| | - Daryl C Drummond
- Discovery, Merrimack Pharmaceuticals, Inc, Cambridge, Massachusetts, USA.,Research & Development, Akagera Medicines, San Francisco, CA, USA
| | - Alexey A Lugovskoy
- Discovery, Merrimack Pharmaceuticals, Inc, Cambridge, Massachusetts, USA.,Research & Development, Diagonal Therapeutics, Cambridge, Massachusetts, USA
| |
Collapse
|
16
|
Xie F, Zhang L, Shi S, Zheng A, Di J, Jin S, Miao X, Wu F, Chen X, Zhang Y, Wei X, Xu Y. Liposomal T cell engager and re-director for tumor cell eradication in cancer immunotherapy. MAbs 2022; 14:2115205. [PMID: 36041060 PMCID: PMC9450900 DOI: 10.1080/19420862.2022.2115205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
T cells are one of the most important effector cells in cancer immunotherapy. Various T cell-dependent bispecific antibody (TDB) drugs that engage T cells for targeted cancer cell lysis are being developed. Here, we describe supra-molecular T-cell redirecting antibody fragment-anchored liposomes (TRAFsomes) and report their immune modulation and anti-cancer effects. We found that TRAFsomes containing different copies of anti-CD3 fragments displayed different T cell modulation profiles, showing that optimization of surface density is needed to define the therapeutic window for potentiating cancer cell-specific immune reactions while minimizing nonspecific side effects. Moreover, small molecular immunomodulators may also be incorporated by liposomal encapsulation to drive CD8 + T cell biased immune responses. In vivo studies using human peripheral blood mononuclear cell reconstituted mouse models showed that TRAFsomes remained bounded to human T cells and persisted for more than 48 hours after injection. However, only TRAFsomes containing a few anti-CD3 (n = 9) demonstrated significant T cell-mediated anti-cancer activities to reverse tumor growth. Those with more anti-CD3s (n = 70) caused tumor growth and depletion of human T cells at the end of treatments. These data suggested that TRAFsomes can be as potent as traditional TDBs and the liposomal structure offers great potential for immunomodulation and improvement of the therapeutic index. Abbreviation: Chimeric antigen receptor T cells (CAR-T cells), Cytokine release syndrome (CRS) Cytotoxic T cell (CTL) Effector: target ratios (E:T ratios), Heavy chain (HC) Immune-related adverse events (irAE), Large unilamellar vesicle (LUV), Peripheral blood mononuclear cells (PBMCs, Single-chain variable fragment (scFv), T cell-dependent bispecific antibody (TDB), T cell redirecting antibody fragment-anchored liposomes (TRAFsomes), Methoxy poly-(ethylene glycol) (mPEG)
Collapse
Affiliation(s)
- Fang Xie
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, Jiangsu, China
| | - Luchen Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, Jiangsu, China
| | - Sanyuan Shi
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, Jiangsu, China
| | - Anjie Zheng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, Jiangsu, China
| | - Jiaxing Di
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, Jiangsu, China
| | - Shanshan Jin
- R&D department, Hangzhou Highfield Bipharmaceuticals Ltd, Hangzhou, Zhejiang, China
| | - Xuguang Miao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, Jiangsu, China
| | - Fenglan Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, Jiangsu, China
| | - Xiaolong Chen
- R&D department, Hangzhou Highfield Bipharmaceuticals Ltd, Hangzhou, Zhejiang, China
| | - Yanhong Zhang
- R&D department, Hangzhou Highfield Bipharmaceuticals Ltd, Hangzhou, Zhejiang, China
| | - Xiaohui Wei
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, Jiangsu, China
| | - Yuhong Xu
- R&D department, Hangzhou Highfield Bipharmaceuticals Ltd, Hangzhou, Zhejiang, China.,School of Pharmacy, Dali University, Dali, Yunnan, China
| |
Collapse
|
17
|
Fobian SF, Cheng Z, ten Hagen TLM. Smart Lipid-Based Nanosystems for Therapeutic Immune Induction against Cancers: Perspectives and Outlooks. Pharmaceutics 2021; 14:26. [PMID: 35056922 PMCID: PMC8779430 DOI: 10.3390/pharmaceutics14010026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy, a promising and widely applied mode of oncotherapy, makes use of immune stimulants and modulators to overcome the immune dysregulation present in cancer, and leverage the host's immune capacity to eliminate tumors. Although some success has been seen in this field, toxicity and weak immune induction remain challenges. Liposomal nanosystems, previously used as targeting agents, are increasingly functioning as immunotherapeutic vehicles, with potential for delivery of contents, immune induction, and synergistic drug packaging. These systems are tailorable, multifunctional, and smart. Liposomes may deliver various immune reagents including cytokines, specific T-cell receptors, antibody fragments, and immune checkpoint inhibitors, and also present a promising platform upon which personalized medicine approaches can be built, especially with preclinical and clinical potentials of liposomes often being frustrated by inter- and intrapatient variation. In this review, we show the potential of liposomes in cancer immunotherapy, as well as the methods for synthesis and in vivo progression thereof. Both preclinical and clinical studies are included to comprehensively illuminate prospects and challenges for future research and application.
Collapse
Affiliation(s)
| | | | - Timo L. M. ten Hagen
- Laboratory Experimental Oncology (LEO), Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (S.-F.F.); (Z.C.)
| |
Collapse
|
18
|
Liposomal Nanocarriers Designed for Sub-Endothelial Matrix Targeting under Vascular Flow Conditions. Pharmaceutics 2021; 13:pharmaceutics13111816. [PMID: 34834231 PMCID: PMC8618675 DOI: 10.3390/pharmaceutics13111816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
Vascular interventions result in the disruption of the tunica intima and the exposure of sub-endothelial matrix proteins. Nanoparticles designed to bind to these exposed matrices could provide targeted drug delivery systems aimed at inhibiting dysfunctional vascular remodeling and improving intervention outcomes. Here, we present the progress in the development of targeted liposomal nanocarriers designed for preferential collagen IV binding under simulated static vascular flow conditions. PEGylated liposomes (PLPs), previously established as effective delivery systems in vascular cells types, served as non-targeting controls. Collagen-targeting liposomes (CT-PLPs) were formed by conjugating established collagen-binding peptides to modified lipid heads via click chemistry (CTL), and inserting them at varying mol% either at the time of PLP assembly or via micellar transfer. All groups included fluorescently labeled lipid species for imaging and quantification. Liposomes were exposed to collagen IV matrices statically or via hemodynamic flow, and binding was measured via fluorometric analyses. CT-PLPs formed with 5 mol% CTL at the time of assembly demonstrated the highest binding affinity to collagen IV under static conditions, while maintaining a nanoparticle characterization profile of ~50 nm size and a homogeneity polydispersity index (PDI) of ~0.2 favorable for clinical translation. When liposomes were exposed to collagen matrices within a pressurized flow system, empirically defined CT-PLPs demonstrated significant binding at shear stresses mimetic of physiological through pathological conditions in both the venous and arterial architectures. Furthermore, when human saphenous vein explants were perfused with liposomes within a closed bioreactor system, CT-PLPs demonstrated significant ex vivo binding to diseased vascular tissue. Ongoing studies aim to further develop CT-PLPs for controlled targeting in a rodent model of vascular injury. The CT-PLP nanocarriers established here show promise as the framework for a spatially controlled delivery platform for future application in targeted vascular therapeutics.
Collapse
|
19
|
Banihashemi SR, Rahbarizadeh F, Zavaran Hosseini A, Ahmadvand D, Khoshtinat Nikkhoi S. Liposome-based nanocarriers loaded with anthrax lethal factor and armed with anti-CD19 VHH for effectively inhibiting MAPK pathway in B cells. Int Immunopharmacol 2021; 100:107927. [PMID: 34500284 DOI: 10.1016/j.intimp.2021.107927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 01/26/2023]
Abstract
OBJECTIVE One of the vital signaling pathways in cancer development and metastasis is mitogen-activated protein kinases (MAPKs). Bacillus anthracis Lethal Toxin (LT) is a potent MAPK signaling inhibitor. This toxin is comprised of two distinct domains, Lethal Factor (LF), MAPK inhibitor, and Protective Antigen (PA). To enter various cell lines, LF must be associated with the protective antigen (PA), which facilitates LF delivery. In the current study, to block MAPK signaling, LF was loaded into anti-CD19 immunoliposomes nanoparticle to deliver the cargo to Raji B cells. METHODS The liposome nanoparticle was prepared using classical lipid film formation, then conjugated to anti-CD19 VHH. The binding efficiency was measured through flow cytometry. The targeted cytotoxicity of LF immunoliposome was confirmed by BrdU lymphoproliferation assay. This was followed by Real-Time PCR to assess the effect of formulation on pro-apoptotic genes. The inhibitory effect of LF on MAPK signaling was confirmed by western blot. RESULTS Liposome nano-formulation was optimized to reach the maximum LF encapsulation and targeted delivery. Next, phosphorylation of MAPK pathway mediators like MEK1/2, P38 and JNK were inhibited following the treatment of Raji cells with LF-immunoliposome. The treatment also upregulated caspase genes, clearly illustrating cell death induced by LF through pyroptosis and caspase-dependent apoptosis. CONCLUSIONS In conclusion, anti-CD19 VHH immunoliposome was loaded with LF, a potent MAPK inhibitor targeting B cells, which curbs proliferation and ushers B cells toward apoptosis. Thus, immunoliposome presents as a versatile nanoparticle for delivery of LF to block aberrant MAPK activation. To use LF as a therapy, it would be necessary to materialize LF without PA. In the current study, PA was substituted with anti-CD19 immunoliposome to make it targeted to CD19+ while keeping the normal cells intact.
Collapse
Affiliation(s)
- S Reza Banihashemi
- Department of Medical Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran; Department of Immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran.
| | - Ahmad Zavaran Hosseini
- Department of Medical Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Davoud Ahmadvand
- School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
20
|
Surface Ligand Valency and Immunoliposome Binding: when More Is Not Always Better. Pharm Res 2021; 38:1593-1600. [PMID: 34463936 DOI: 10.1007/s11095-021-03092-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/03/2021] [Indexed: 01/19/2023]
Abstract
PURPOSE Nano-drug delivery systems are designed to contain surface ligands including antibodies for "active targeting". The number of ligands on each nanoparticle, known as the valency, is considered a critical determinant of the "targeting" property. We sought to understand the correlation between valency and binding properties using antibody conjugated liposomes, i.e. immunoliposomes (ILs), as the model. METHODS Anti-CD3 Fab containing a terminal cysteine residue were conjugated to DSPE-PEG-maleimide and incubated with preformed liposomes at 60°C. The un-incorporated antibodies were removed and the obtained ILs were characterized to contain in average 2-22 copies of anti-CD3 Fabs per liposome. The Biolayer Interferometry (BLI) probe surface was coated with various densities of CD3 epsilon&delta heterodimer (CD3D/E) to imitate different CD3 expression levels on target cells. The inference wavelength shifts upon anti-CD3 liposome binding were monitored and analyzed. RESULTS The data indicated ILs may bind either monovalently or multivalently, determined mainly by the surface ligand density rather than the ILs antibody valency. The ILs valency indeed correlated with the dissociation rate constant (Koff), but not with the association rate constant (Kon). Their binding capabilities also did not necessarily increase with the surface anti-CD3 valency. CONCLUSION We proposed a model for understanding the binding properties of ILs with different ligand valencies. The binding mode may change when the targeted surfaces had different antigen densities. The model should be important for the designing and optimization of active targeting drug delivery systems to fit different applications.
Collapse
|
21
|
Ahmed KS, Liu S, Mao J, Zhang J, Qiu L. Dual-Functional Peptide Driven Liposome Codelivery System for Efficient Treatment of Doxorubicin-Resistant Breast Cancer. Drug Des Devel Ther 2021; 15:3223-3239. [PMID: 34349500 PMCID: PMC8326382 DOI: 10.2147/dddt.s317454] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The active-targeted drug delivery systems had attracted more and more attention to efficiently overcome multidrug resistance (MDR) in cancer treatments. The aim of the work was to develop a multifunctional nano-structured liposomal system for co-delivery of doxorubicin hydrochloride (DOX) and celecoxib (CEL) to overcome doxorubicin resistance in breast cancer. METHODS A functional hybrid peptide (MTS-R8H3) with unique cellular penetrability, endo-lysosomal escape and mitochondrial targeting ability was successfully synthesized using solid phase synthesis technology. The peptide modified targeted liposomes (DOX/CEL-MTS-R8H3 lipo) for co-delivery of DOX and CEL were formulated to overcome the chemoresistance in MCF/ADR cells. RESULTS DOX/CEL-MTS-R8H3 lipo showed nanosized shape and displayed high stability for one month. The cytotoxicity effect of the co-delivery of DOX and CEL through peptide modified liposomes had remarkable treatment efficacy on killing MCF/ADR cells. Targeted liposome exhibited greater cellular entry ability about 5.72-fold stronger than DOX solution. Moreover, as compared with unmodified liposomes, the presence of MTS-R8H3 peptide entity on liposome surface enhanced the mitochondrial-targeting ability and achieved effective reactive oxygen species (ROS) production with significant inhibition of P-gp efflux activity. CONCLUSION The study suggested that the DOX/CEL-MTS-R8H3 lipo is a promising strategy for overcoming drug resistance in breast cancer treatments with high targeting inhibition efficiency.
Collapse
Affiliation(s)
- Kamel S Ahmed
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 19623, Egypt
| | - Shenhuan Liu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| | - Jing Mao
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| | - Jie Zhang
- The Jiaxing Key Laboratory of Oncological Photodynamic Therapy and the Targeted Drug Research, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, 314001, People’s Republic of China
| | - Lipeng Qiu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| |
Collapse
|
22
|
Moulahoum H, Ghorbanizamani F, Zihnioglu F, Timur S. Surface Biomodification of Liposomes and Polymersomes for Efficient Targeted Drug Delivery. Bioconjug Chem 2021; 32:1491-1502. [PMID: 34283580 DOI: 10.1021/acs.bioconjchem.1c00285] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chemotherapy has seen great progress in the development of performant treatment strategies. Nanovesicles such as liposomes and polymersomes demonstrated great potential in cancer therapy. However, these nanocarriers deliver their content passively, which faces a lot of constraints during blood circulation. The main challenge resides in degradation and random delivery to normal tissues. Hence, targeting drug delivery using specific molecules (such as antibodies) grafted over the surface of these nanocarriers came as the answer to overcome many problems faced before. The advantage of using antibodies is their antigen/antibody recognition, which provides a high level of specificity to reach treatment targets. This review discusses the many techniques of nanocarrier functionalization with antibodies. The aim is to recognize the various approaches by describing their advantages and deficiencies to create the most suitable drug delivery platform. Some methods are more suitable for other applications rather than drug delivery, which can explain the low success of some proposed targeted nanocarriers. In here, a critical analysis of how every method could impact the recognition and targeting capacity of some nanocarriers (liposomes and polymersomes) is discussed to make future research more impactful and advance the field of biomedicine further.
Collapse
Affiliation(s)
- Hichem Moulahoum
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Faezeh Ghorbanizamani
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Figen Zihnioglu
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Suna Timur
- Biochemistry Department, Faculty of Science, Ege University, 35100, Bornova, Izmir, Turkey.,Central Research Testing and Analysis Laboratory Research and Application Center, Ege University, 35100, Bornova, Izmir, Turkey
| |
Collapse
|
23
|
Quantitative paper-based dot blot assay for spike protein detection using fuchsine dye-loaded polymersomes. Biosens Bioelectron 2021; 192:113484. [PMID: 34246807 DOI: 10.1016/j.bios.2021.113484] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/12/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022]
Abstract
Real-time reverse transcriptase-polymerase chain reaction (RT-PCR)-based assays are the gold standard for virus diagnosis. Point-of-care (POC) technologies have shown great progress during this period. Herein, we propose a novel fuchsine dye-loaded polymersome for a colorimetric paper-based dot blot spike protein diagnostic assay for COVID-19 via smartphone-assisted sensing. The prepared platform aimed to create an adaptable tool that competes with traditional nanoparticle-based assays employing gold and silver. Analytical characterization and application of the testing platform showed high sensitivity (10 times better than gold nanoparticles), stability, fast turnaround, and reproducibility. The potential and possibilities demonstrated by the current platform could be observed in its adaptability for different markers and pathologies. In addition, smartphone-assisted sensing emphasizes the ability to use the tool at home by common peoples which can lower the burden on the healthcare facilities and reach more underdeveloped regions.
Collapse
|
24
|
Farahavar G, Abolmaali SS, Nejatollahi F, Safaie A, Javanmardi S, Khajeh Zadeh H, Yousefi R, Nadgaran H, Mohammadi-Samani S, Tamaddon AM, Ahadian S. Single-chain antibody-decorated Au nanocages@liposomal layer nanoprobes for targeted SERS imaging and remote-controlled photothermal therapy of melanoma cancer cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112086. [PMID: 33947576 DOI: 10.1016/j.msec.2021.112086] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 11/24/2022]
Abstract
The development of theranostic platforms combining surface-enhanced Raman spectroscopy (SERS) imaging with NIR-stimulated photothermal therapy (PTT) is of utmost importance for the precise diagnosis and selective treatment of cancers, especially in superficial solid tumors. For this purpose, a versatile theranostic nanoprobe of liposomal layer-coated Au nanocages (AuNCs) was decorated with an anti-MUC18 single-chain antibody (scFv). 4-mercapto benzoic acid (p-MBA)-labeled AuNCs (p-AuNCs) were coated by a liposomal layer (p-AuNCs@lip), followed by conjugating anti-MUC18 scFv via post-insertion method to form immuno-liposomal layer-coated AuNCs (p-AuNCs@scFv-lip). Physicochemical characterizations of the p-AuNCs@scFv-lip were investigated by transmission electron microscopy (TEM) and UV-vis and Raman spectroscopy. Furthermore, the targeting ability and theranostic efficiency of the nanoprobe were evaluated for specific diagnosis and treatment of cancerous melanoma cells by flow cytometry, SERS mapping, and live/dead assay. The formation of lipid layer on p-AuNCs surface was confirmed by TEM imaging. After decorating the liposomal layer with scFv, a relevant red shift was observed in the UV-vis spectrum. Moreover, p-AuNCs@lip presented characteristic peaks in the Raman spectrum, which exhibited only a minor change after scFv conjugation (p-AuNCs@scFv-lip). Interestingly, the cellular uptake of AuNCs@scFv-lip by A375 cell line (MUC18+) showed a 24-fold enhancement compared with SKBR3 cells (MUC18-). AuNCs@scFv-lip specifically identified A375 cells from SKBR cells via SERS mapping and effectively killed A375 cells through the PTT mechanism. Taken together, this theranostic platform can provide a promising tool for both in situ diagnosis and remote-controlled thermal ablation of cancer cells.
Collapse
Affiliation(s)
- Ghazal Farahavar
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz 71345, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Foroogh Nejatollahi
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amin Safaie
- Faculty of Science, Department of Physics, Shiraz University, Shiraz 71454, Iran.
| | - Sanaz Javanmardi
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | | | - Reza Yousefi
- Protein Chemistry Laboratory (PCL), Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Hamid Nadgaran
- Faculty of Science, Department of Physics, Shiraz University, Shiraz 71454, Iran.
| | - Soliman Mohammadi-Samani
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran; Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Ali Mohammad Tamaddon
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA; Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
25
|
Fisher RK, West PC, Mattern-Schain SI, Best MD, Kirkpatrick SS, Dieter RA, Arnold JD, Buckley MR, McNally MM, Freeman MB, Grandas OH, Mountain DJH. Advances in the Formulation and Assembly of Non-Cationic Lipid Nanoparticles for the Medical Application of Gene Therapeutics. NANOMATERIALS 2021; 11:nano11030825. [PMID: 33807086 PMCID: PMC8004789 DOI: 10.3390/nano11030825] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022]
Abstract
Lipid nanoparticles have become increasingly popular delivery platforms in the field of gene therapy, but bench-to-bedside success has been limited. Many liposomal gene vectors are comprised of synthetic cationic lipids, which are associated with lipid-induced cytotoxicity and immunogenicity. Natural, non-cationic PEGylated liposomes (PLPs) demonstrate favorable biocompatibility profiles but are not considered viable gene delivery vehicles due to inefficient nucleic acid loading and reduced cellular uptake. PLPs can be modified with cell-penetrating peptides (CPPs) to enhance the intracellular delivery of liposomal cargo but encapsulate leakage upon CPP-PLP assembly is problematic. Here, we aimed to identify parameters that overcome these performance barriers by incorporating nucleic acid condensers during CPP-PLP assembly and screening variable ethanol injection parameters for optimization. CPP-PLPs were formed with R8-amphiphiles via pre-insertion, post-insertion and post-conjugation techniques and liposomes were characterized for size, surface charge, homogeneity, siRNA encapsulation efficiency and retention and cell associative properties. Herein we demonstrate that pre-insertion of stearylated R8 into PLPs is an efficient method to produce non-cationic CPP-PLPs and we provide additional assembly parameter specifications for a modified ethanol injection technique that is optimized for siRNA encapsulation/retention and enhanced cell association. This assembly technique could provide improved clinical translation of liposomal based gene therapy applications.
Collapse
Affiliation(s)
- Richard K. Fisher
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Phillip C. West
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Samuel I. Mattern-Schain
- Department of Chemistry, University of Tennessee Knoxville, 1420 Circle Drive, Knoxville, TN 37996, USA; (S.I.M.-S.); (M.D.B.)
| | - Michael D. Best
- Department of Chemistry, University of Tennessee Knoxville, 1420 Circle Drive, Knoxville, TN 37996, USA; (S.I.M.-S.); (M.D.B.)
| | - Stacy S. Kirkpatrick
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Raymond A. Dieter
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Joshua D. Arnold
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Michael R. Buckley
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Michael M. McNally
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Michael B. Freeman
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Oscar H. Grandas
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Deidra J. H. Mountain
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
- Correspondence: ; Tel.: +1-865-305-9160
| |
Collapse
|
26
|
Pardridge WM. Brain Delivery of Nanomedicines: Trojan Horse Liposomes for Plasmid DNA Gene Therapy of the Brain. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:602236. [PMID: 35047884 PMCID: PMC8757841 DOI: 10.3389/fmedt.2020.602236] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Non-viral gene therapy of the brain is enabled by the development of plasmid DNA brain delivery technology, which requires the engineering and manufacturing of nanomedicines that cross the blood-brain barrier (BBB). The development of such nanomedicines is a multi-faceted problem that requires progress at multiple levels. First, the type of nanocontainer, e.g., nanoparticle or liposome, which encapsulates the plasmid DNA, must be developed. Second, the type of molecular Trojan horse, e.g., peptide or receptor-specific monoclonal antibody (MAb), must be selected for incorporation on the surface of the nanomedicine, as this Trojan horse engages specific receptors expressed on the BBB, and the brain cell membrane, to trigger transport of the nanomedicine from blood into brain cells beyond the BBB. Third, the plasmid DNA must be engineered without bacterial elements, such as antibiotic resistance genes, to enable administration to humans; the plasmid DNA must also be engineered with tissue-specific gene promoters upstream of the therapeutic gene, to insure gene expression in the target organ with minimal off-target expression. Fourth, upstream manufacturing of the nanomedicine must be developed and scalable so as to meet market demand for the target disease, e.g., annual long-term treatment of 1,000 patients with an orphan disease, short term treatment of 10,000 patients with malignant glioma, or 100,000 patients with new onset Parkinson's disease. Fifth, downstream manufacturing problems, such as nanomedicine lyophilization, must be solved to ensure the nanomedicine has a commercially viable shelf-life for treatment of CNS disease in humans.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
27
|
EGFR targeting for cancer therapy: Pharmacology and immunoconjugates with drugs and nanoparticles. Int J Pharm 2020; 592:120082. [PMID: 33188892 DOI: 10.1016/j.ijpharm.2020.120082] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/21/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
The epidermal growth factor receptor (EGFR) belongs to the tyrosine kinase receptors family and is present in the epithelial cell membrane. Its endogenous activation occurs through the binding of different endogenous ligands, including the epidermal growth factor (EGF), leading to signaling cascades able to maintain normal cellular functions. Although involved in the development and maintenance of tissues in normal conditions, when EGFR is overexpressed, it stimulates the growth and progression of tumors, resulting in angiogenesis, invasion and metastasis, through some main cascades such as Ras/Raf/MAPK, PIK-3/AKT, PLC-PKC and STAT. Besides, considering the limitations of conventional chemotherapy that result in high toxicity and low tumor specificity, EGFR is currently considered an important target. As a result, several monoclonal antibodies are currently approved for use in cancer treatment, such as cetuximab (CTX), panitumumab, nimotuzumab, necitumumab and others are in clinical trials. Aiming to combine the chemotherapeutic agent toxicity and specific targeting to EGFR overexpressing tumor tissues, two main strategies will be discussed in this review: antibody-drug conjugates (ADCs) and antibody-nanoparticle conjugates (ANCs). Briefly, ADCs consist of antibodies covalently linked through a spacer to the cytotoxic drug. Upon administration, binding to EGFR and endocytosis, ADCs suffer chemical and enzymatic reactions leading to the release and accumulation of the drug. Instead, ANCs consist of nanotechnology-based formulations, such as lipid, polymeric and inorganic nanoparticles able to protect the drug against inactivation, allowing controlled release and also passive accumulation in tumor tissues by the enhanced permeability and retention effect (EPR). Furthermore, ANCs undergo active targeting through EGFR receptor-mediated endocytosis, leading to the formation of lysosomes and drug release into the cytosol. Herein, we will present and discuss some important aspects regarding EGFR structure, its role on internal signaling pathways and downregulation aspects. Then, considering that EGFR is a potential therapeutic target for cancer therapy, the monoclonal antibodies able to target this receptor will be presented and discussed. Finally, ADCs and ANCs state of the art will be reviewed and recent studies and clinical progresses will be highlighted. To the best of our knowledge, this is the first review paper to address specifically the EGFR target and its application on ADCs and ANCs.
Collapse
|
28
|
Dadashi Noshahr K, Shamsi F, Valtchev P, Kokhaei P, Hemati M, Reza Akbari Eidgahi M, Khaleghian A. Optimization of post-insertion method to conjugate Doxil with anti-CD133 monoclonal antibodies: Investigating the specific binding and cytotoxicity to colorectal cancer cells in vitro. Saudi Pharm J 2020; 28:1392-1401. [PMID: 33250646 PMCID: PMC7679470 DOI: 10.1016/j.jsps.2020.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/07/2020] [Indexed: 10/25/2022] Open
Abstract
In this paper, Doxil coupled with anti-CD133 monoclonal antibodies made by either routine or optimized post-insertion technique, were compared with respect to their size, drug leakage, release pattern and the number of antibodies conjugated per single liposome. The results demonstrated that the number of antibodies conjugated per liposome in the optimized post-insertion technique was almost two times more than those in the routine post-insertion method. However, the drug release and leakage pattern was almost similar between the two methods. Furthermore, anti-tumor activity and therapeutic efficacy of the preferred CD133-targeted Doxil with Doxil was compared in terms of their in vitro binding, uptake, internalization and cytotoxicity against HT-29 (CD133+) and CHO (CD133-) cells. Flow cytometry analyses and confocal laser scanning microscopy results exhibited a significantly higher cellular uptake, binding and internalization of CD133-targeted Doxil in CD+133 cells relative to Doxil. Cytotoxicity results revealed a lower in vitro inhibitory concentration for CD133-targeted Doxil compared to Doxil. However, CHO (CD133-) cells displayed a similar uptake and in vitro cytotoxicity for both CD133-Doxil and non-targeted Doxil. Therefore, the results of this study can exhibit that specific recognition and binding of antibodies with CD133 receptors on HT-29 cells can result in enhanced cellular uptake, internalization and cytotoxicity. The research suggests further investigation for in vivo studies and may offer proof-of-principle for an active targeting concept.
Collapse
Affiliation(s)
- Karim Dadashi Noshahr
- Department of Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fahimeh Shamsi
- Department of Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- School of Chemical and Biomolecular Engineering, University of Sydney, NSW 2006, Australia
| | - Peter Valtchev
- School of Chemical and Biomolecular Engineering, University of Sydney, NSW 2006, Australia
| | - Parviz Kokhaei
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maral Hemati
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mohammad Reza Akbari Eidgahi
- Department of Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Biotechnology Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Khaleghian
- Department of Biochemistry, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
29
|
Rapid Target Binding and Cargo Release of Activatable Liposomes Bearing HER2 and FAP Single-Chain Antibody Fragments Reveal Potentials for Image-Guided Delivery to Tumors. Pharmaceutics 2020; 12:pharmaceutics12100972. [PMID: 33076292 PMCID: PMC7650594 DOI: 10.3390/pharmaceutics12100972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 01/04/2023] Open
Abstract
Liposomes represent suitable tools for the diagnosis and treatment of a variety of diseases, including cancers. To study the role of the human epidermal growth factor receptor 2 (HER2) as target in cancer imaging and image-guided deliveries, liposomes were encapsulated with an intrinsically quenched concentration of a near-infrared fluorescent dye in their aqueous interior. This resulted in quenched liposomes (termed LipQ), that were fluorescent exclusively upon degradation, dye release, and activation. The liposomes carried an always-on green fluorescent phospholipid in the lipid layer to enable tracking of intact liposomes. Additionally, they were functionalized with single-chain antibody fragments directed to fibroblast activation protein (FAP), a marker of stromal fibroblasts of most epithelial cancers, and to HER2, whose overexpression in 20–30% of all breast cancers and many other cancer types is associated with a poor treatment outcome and relapse. We show that both monospecific (HER2-IL) and bispecific (Bi-FAP/HER2-IL) formulations are quenched and undergo HER2-dependent rapid uptake and cargo release in cultured target cells and tumor models in mice. Thereby, tumor fluorescence was retained in whole-body NIRF imaging for 32–48 h post-injection. Opposed to cell culture studies, Bi-FAP/HER2-IL-based live confocal microscopy of a high HER2-expressing tumor revealed nuclear delivery of the encapsulated dye. Thus, the liposomes have potentials for image-guided nuclear delivery of therapeutics, and also for intraoperative delineation of tumors, metastasis, and tumor margins.
Collapse
|
30
|
Di J, Xie F, Xu Y. When liposomes met antibodies: Drug delivery and beyond. Adv Drug Deliv Rev 2020; 154-155:151-162. [PMID: 32926944 DOI: 10.1016/j.addr.2020.09.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
Drug encapsulated liposomes and monoclonal antibodies (Mabs) are two distinctively different classes of therapeutics, but both aim to become the ultimate "magic bullet". While PEGylated liposomes rely on the enhanced permeability and retention (EPR) effect for accumulation in solid tumor tissues, Mabs are designed to bind tightly to specific surface antigens on target cells to exert effector functions. Immunoliposome (IL) refers to the structural combination of liposomes and antibodies, whereas the antibodies are usually decorated on the liposome surface. ILs can therefore take advantage of interactions between antibodies and cancer cells for more efficient endocytosis and intracellular drug delivery. The antibody structure, affinity, density, as well as the liposome surface properties and drug to lipid ratios all contribute to the IL pharmacokinetic(PK) and pharmacodynamic(PD) behaviors. The optimal formulation parameters may vary for different target cells and tissues. Furthermore, besides the delivery of cytotoxic drugs to cancer cells, new ILs are being developed to interact with multiple target receptors, multiple target cells and trigger multiple therapeutic effects. We envision that the IL format can be a great platform for the molecular engineering of multi-valent, multi-specific interactions to achieve complex biological functions for therapeutic benefits, especially in the area of cancer immunotherapy.
Collapse
Affiliation(s)
- Jiaxing Di
- School of Pharmacy, Shanghai Jiao Tong University, China
| | - Fang Xie
- Department of Biomedical Engineering, Johns Hopkins University, United States of America
| | - Yuhong Xu
- College of Pharmacy and Chemistry, Dali University, China.
| |
Collapse
|
31
|
Sortase-A mediated chemoenzymatic lipidation of single-domain antibodies for cell membrane engineering. Eur J Pharm Biopharm 2020; 153:121-129. [DOI: 10.1016/j.ejpb.2020.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/11/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022]
|
32
|
Attia MF, Swasy MI, Akasov R, Alexis F, Whitehead DC. Strategies for High Grafting Efficiency of Functional Ligands to Lipid Nanoemulsions for RGD-Mediated Targeting of Tumor Cells In Vitro. ACS APPLIED BIO MATERIALS 2020; 3:5067-5079. [DOI: 10.1021/acsabm.0c00567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Mohamed F. Attia
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Maria I. Swasy
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Roman Akasov
- National University of Science and Technology “MISIS”, Leninskiy Prospect 4, 119991 Moscow, Russia
- Federal Scientific Research Center “Crystallography and Photonics”, Russian Academy of Sciences, Leninskiy Prospekt 59, 119333 Moscow, Russia
| | - Frank Alexis
- School of Biological Sciences and Engineering, Yachay Tech, San Miguel de Urcuquí 100650, Ecuador
| | - Daniel C. Whitehead
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| |
Collapse
|
33
|
Dual-action CXCR4-targeting liposomes in leukemia: function blocking and drug delivery. Blood Adv 2020; 3:2069-2081. [PMID: 31292126 DOI: 10.1182/bloodadvances.2019000098] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
CXC chemokine receptor 4 (CXCR4) is overexpressed by a broad range of hematological disorders, and its interaction with CXC chemokine ligand 12 (CXCL12) is of central importance in the retention and chemoprotection of neoplastic cells in the bone marrow and lymphoid organs. In this article, we describe the biological evaluation of a new CXCR4-targeting and -antagonizing molecule (BAT1) that we designed and show that, when incorporated into a liposomal drug delivery system, it can be used to deliver cancer therapeutics at high levels to chronic lymphocytic leukemia (CLL) cells. CXCR4 targeting and antagonism by BAT1 were demonstrated alone and following its incorporation into liposomes (BAT1-liposomes). Antagonism of BAT1 against the CXCR4/CXCL12 interaction was demonstrated through signaling inhibition and function blocking: BAT1 reduced ERK phosphorylation and cell migration to levels equivalent to those seen in the absence of CXCL12 stimulation (P < .001). Specific uptake of BAT1-liposomes and delivery of a therapeutic cargo to the cell nucleus was seen within 3 hours of incubation and induced significantly more CLL cell death after 24 hours than control liposomes (P = .004). The BAT1 drug-delivery system is modular, versatile, and highly clinically relevant, incorporating elements of proven clinical efficacy. The combined capabilities to block CXCL12-induced migration and intracellular signaling while simultaneously delivering therapeutic cargo mean that the BAT1-liposome drug-delivery system could be a timely and relevant treatment of a range of hematological disorders, particularly because the therapeutic cargo can be tailored to the disease being treated.
Collapse
|
34
|
Tansi FL, Rüger R, Kollmeier AM, Rabenhold M, Steiniger F, Kontermann RE, Teichgräber UK, Fahr A, Hilger I. Targeting the Tumor Microenvironment with Fluorescence-Activatable Bispecific Endoglin/Fibroblast Activation Protein Targeting Liposomes. Pharmaceutics 2020; 12:pharmaceutics12040370. [PMID: 32316521 PMCID: PMC7238156 DOI: 10.3390/pharmaceutics12040370] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/06/2020] [Accepted: 04/14/2020] [Indexed: 11/25/2022] Open
Abstract
Liposomes are biocompatible nanocarriers with promising features for targeted delivery of contrast agents and drugs into the tumor microenvironment, for imaging and therapy purposes. Liposome-based simultaneous targeting of tumor associated fibroblast and the vasculature is promising, but the heterogeneity of tumors entails a thorough validation of suitable markers for targeted delivery. Thus, we elucidated the potential of bispecific liposomes targeting the fibroblast activation protein (FAP) on tumor stromal fibroblasts, together with endoglin which is overexpressed on tumor neovascular cells and some neoplastic cells. Fluorescence-quenched liposomes were prepared by hydrating a lipid film with a high concentration of the self-quenching near-infrared fluorescent dye, DY-676-COOH, to enable fluorescence detection exclusively upon liposomal degradation and subsequent activation. A non-quenched green fluorescent phospholipid was embedded in the liposomal surface to fluorescence-track intact liposomes. FAP- and murine endoglin-specific single chain antibody fragments were coupled to the liposomal surface, and the liposomal potentials validated in tumor cells and mice models. The bispecific liposomes revealed strong fluorescence quenching, activatability, and selectivity for target cells and delivered the encapsulated dye selectively into tumor vessels and tumor associated fibroblasts in xenografted mice models and enabled their fluorescence imaging. Furthermore, detection of swollen lymph nodes during intra-operative simulations was possible. Thus, the bispecific liposomes have potentials for targeted delivery into the tumor microenvironment and for image-guided surgery.
Collapse
Affiliation(s)
- Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (A.M.K.); (U.K.T.)
- Correspondence: (F.L.T.); (R.R.); (I.H.); Tel.: +49-3641-9324993 (F.L.T.); +49-3641-949905 (R.R.); +49-3641-9325921 (I.H.)
| | - Ronny Rüger
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany (A.F.)
- Correspondence: (F.L.T.); (R.R.); (I.H.); Tel.: +49-3641-9324993 (F.L.T.); +49-3641-949905 (R.R.); +49-3641-9325921 (I.H.)
| | - Ansgar M. Kollmeier
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (A.M.K.); (U.K.T.)
| | - Markus Rabenhold
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany (A.F.)
| | - Frank Steiniger
- Center for Electron Microscopy, Jena University Hospital-Friedrich Schiller University Jena, Ziegelmuehlenweg 1, 07743 Jena, Germany;
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University Stuttgart, Allmandring 31, 70569 Stuttgart, Germany;
| | - Ulf K. Teichgräber
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (A.M.K.); (U.K.T.)
| | - Alfred Fahr
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany (A.F.)
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (A.M.K.); (U.K.T.)
- Correspondence: (F.L.T.); (R.R.); (I.H.); Tel.: +49-3641-9324993 (F.L.T.); +49-3641-949905 (R.R.); +49-3641-9325921 (I.H.)
| |
Collapse
|
35
|
Jiao J, Luo X, Wang C, Jiao X, Liu M, Liu X, Wei L, Deng Y, Song Y. Effects of Uncleavable and Cleavable PEG-Lipids with Different Molecular Weights on Accelerated Blood Clearance of PEGylated Emulsions in Beagle Dogs. AAPS PharmSciTech 2020; 21:106. [PMID: 32185548 DOI: 10.1208/s12249-020-1640-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/12/2020] [Indexed: 11/30/2022] Open
Abstract
To investigate the effect of polyethylene glycol (PEG) molecular weights on circulation time of PEGylated emulsions and the second injection of injected PEGylated emulsions, we studied the effect of molecular weights on the pharmacokinetic behavior of PEG-DSPE (modified emulsions with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-n-[methoxy (polyethyleneglycol)]) and PEG-CHMC (modified emulsions with poly(ethyleneglycol)-cholesteryl carbonate) emulsions in beagle dogs. The "accelerated blood clearance" (ABC) phenomenon was induced. Through this study, the contribution of PEG molecular weights on the ABC phenomenon was further clarified, and the results provided guidance for lessening or eliminating the ABC phenomenon. We injected different PEG-modified emulsions with 10% PEG-modified density into beagle dogs at 2 μmol phospholipids kg-1 and the blood samples were drawn after 1 min, 3 min, 5 min, 10 min, 15 min, 30 min, 60 min, 120 min, 240 min, 360 min, 600 min, and 24 h. Then, concentrations of the drug were assayed using high-performance liquid chromatography (HPLC). The results showed that the circulation times of PEG-DSPE-modified emulsions were significantly different because of the difference in molecular weights, whereas those of the PEG-CHMC modified emulsions were not. The spatial conformation of PEG with small molecular weights (PEG400, PEG600, and PEG800) was more likely to induce a strong ABC phenomenon. The results of our work suggest the interaction of circulation time and PEG molecular weights on the ABC phenomenon, implying that the spatial conformation of PEG has advantages that alleviate the ABC phenomenon. Importantly, the results have implications for the choice of molecular weights of PEG for PEGylated formulations.
Collapse
|
36
|
Bleher S, Buck J, Muhl C, Sieber S, Barnert S, Witzigmann D, Huwyler J, Barz M, Süss R. Poly(Sarcosine) Surface Modification Imparts Stealth-Like Properties to Liposomes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1904716. [PMID: 31722126 DOI: 10.1002/smll.201904716] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/23/2019] [Indexed: 06/10/2023]
Abstract
Circulation lifetime is a crucial parameter for a successful therapy with nanoparticles. Reduction and alteration of opsonization profiles by surface modification of nanoparticles is the main strategy to achieve this objective. In clinical settings, PEGylation is the most relevant strategy to enhance blood circulation, yet it has drawbacks, including hypersensitivity reactions in some patients treated with PEGylated nanoparticles, which fuel the search for alternative strategies. In this work, lipopolysarcosine derivatives (BA-pSar, bisalkyl polysarcosine) with precise chain lengths and low polydispersity indices are synthesized, characterized, and incorporated into the bilayer of preformed liposomes via a post insertion technique. Successful incorporation of BA-pSar can be realized in a clinically relevant liposomal formulation. Furthermore, BA-pSar provides excellent surface charge shielding potential for charged liposomes and renders their surface neutral. Pharmacokinetic investigations in a zebrafish model show enhanced circulation properties and reduction in macrophage recognition, matching the behavior of PEGylated liposomes. Moreover, complement activation, which is a key factor in hypersensitivity reactions caused by PEGylated liposomes, can be reduced by modifying the surface of liposomes with an acetylated BA-pSar derivative. Hence, this study presents an alternative surface modification strategy with similar benefits as the established PEGylation of nanoparticles, but with the potential of reducing its drawbacks.
Collapse
Affiliation(s)
- Stefan Bleher
- Department of Pharmaceutical Technology and Biopharmacy and Freiburger Materialforschungszentrum (FMF), Institute of Pharmaceutical Sciences, Albert Ludwig University of Freiburg, 79104, Freiburg, Germany
| | - Jonas Buck
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, 4056, Basel, Switzerland
| | - Christian Muhl
- Institute of Organic Chemistry, Johannes Gutenberg University Mainz, 55128, Mainz, Germany
| | - Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, 4056, Basel, Switzerland
| | - Sabine Barnert
- Department of Pharmaceutical Technology and Biopharmacy and Freiburger Materialforschungszentrum (FMF), Institute of Pharmaceutical Sciences, Albert Ludwig University of Freiburg, 79104, Freiburg, Germany
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, 4056, Basel, Switzerland
- Department of Biochemistry and Molecular Biology, University of British Columbia, Health Sciences Mall, Vancouver, V6T 1Z3, British Columbia, Canada
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, 4056, Basel, Switzerland
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg University Mainz, 55128, Mainz, Germany
| | - Regine Süss
- Department of Pharmaceutical Technology and Biopharmacy and Freiburger Materialforschungszentrum (FMF), Institute of Pharmaceutical Sciences, Albert Ludwig University of Freiburg, 79104, Freiburg, Germany
| |
Collapse
|
37
|
Obaid G, Bano S, Mallidi S, Broekgaarden M, Kuriakose J, Silber Z, Bulin AL, Wang Y, Mai Z, Jin W, Simeone D, Hasan T. Impacting Pancreatic Cancer Therapy in Heterotypic in Vitro Organoids and in Vivo Tumors with Specificity-Tuned, NIR-Activable Photoimmunonanoconjugates: Towards Conquering Desmoplasia? NANO LETTERS 2019; 19:7573-7587. [PMID: 31518145 PMCID: PMC6934365 DOI: 10.1021/acs.nanolett.9b00859] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Despite untiring efforts to develop therapies for pancreatic ductal adenocarcinoma (PDAC), survival statistics remain dismal, necessitating distinct approaches. Photodynamic priming (PDP), which improves drug delivery and combination regimens, as well as tumor photodestruction are key attributes of photodynamic therapy (PDT), making it a distinctive clinical option for PDAC. Localized, high-payload nanomedicine-assisted delivery of photosensitizers (PSs), with molecular specificity and controlled photoactivation, thus becomes critical in order to reduce collateral toxicity during more expansive photodynamic activation procedures with curative intent. As such, targeted photoactivable lipid-based nanomedicines are an ideal candidate but have failed to provide greater than two-fold cancer cell selectivity, if at all, due to their extensive multivariant physical, optical, and chemical complexity. Here, we report (1) a systematic multivariant tuning approach to engineer (Cet, anti-EGFR mAb) photoimmunonanoconjugates (PINs), and (2) stroma-rich heterotypic PDAC in vitro and in vivo models incorporating patient-derived pancreatic cancer-associated fibroblasts (PCAFs) that recapitulate the desmoplasia observed in the clinic. These offer a comprehensive, disease-specific framework for the development of Cet-PINs. Specificity-tuning of the PINs, in terms of PS lipid anchoring, electrostatic modulation, Cet orientation, and Cet surface densities, achieved ∼16-fold binding specificities and rapid penetration of the heterotypic organoids within 1 h, thereby providing a ∼16-fold enhancement in molecular targeted NIR photodestruction. As a demonstration of their inherent amenability for multifunctionality, encapsulation of high payloads of gemcitabine hydrochloride, 5-fluorouracil, and oxaliplatin within the Cet-PINs further improved their antitumor efficacy in the heterotypic organoids. In heterotypic desmoplastic tumors, the Cet-PINs efficiently penetrated up to 470 μm away from blood vessels, and photodynamic activation resulted in substantial tumor necrosis, which was not elicited in T47D tumors (low EGFR) or when using untargeted constructs in both tumor types. Photodynamic activation of the Cet-PINs in the heterotypic desmoplastic tumors resulted in collagen photomodulation, with a 1.5-fold reduction in collagen density, suggesting that PDP may also hold potential for conquering desmoplasia. The in vivo safety profile of photodynamic activation of the Cet-PINs was also substantially improved, as compared to the untargeted constructs. While treatment using the Cet-PINs did not cause any detriment to the mice's health or to healthy proximal tissue, photodynamic activation of untargeted constructs induced severe acute cachexia and weight loss in all treated mice, with substantial peripheral skin necrosis, muscle necrosis, and bowel perforation. This study is the first report demonstrating the true value of molecular targeting for NIR-activable PINs. These constructs integrate high payload delivery, efficient photodestruction, molecular precision, and collagen photomodulation in desmoplastic PDAC tumors in a single treatment using a single construct. Such combined PIN platforms and heterocellular models open up an array of further multiplexed combination therapies to synergistically control desmoplastic tumor progression and extend PDAC patient survival.
Collapse
Affiliation(s)
- Girgis Obaid
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Mans Broekgaarden
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Jerrin Kuriakose
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Zachary Silber
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Anne-Laure Bulin
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Yucheng Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Zhiming Mai
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Wendong Jin
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Diane Simeone
- Department of Surgery and Department of Pathology, Perlmutter Cancer Center, New York University Langone Health, New York, New York 10016, United States
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
38
|
Zhang Y, Li S, Zhou X, Sun J, Fan X, Guan Z, Zhang L, Yang Z. Construction of a Targeting Nanoparticle of 3',3″-Bis-Peptide-siRNA Conjugate/Mixed Lipid with Postinserted DSPE-PEG2000-cRGD. Mol Pharm 2019; 16:4920-4928. [PMID: 31642677 DOI: 10.1021/acs.molpharmaceut.9b00800] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cyclic Arg-Gly-Asp (cRGD) peptides are widely used as tumor-targeting ligands due to their specific binding ability to integrin αvβ3, which is overexpressed on the surface of various cancer cells and the endothelial cells of new blood vessels within tumor tissues. In this paper, the postinsertion strategy of DSPE-PEG2000-cRGD has been applied to the nanoparticles of 3',3″-bis-peptide-siRNA (pp-siRNA) encapsulated by gemini-like cationic lipid (CLD) and neutral cytosin-1-yl lipid (DNCA) from our lab. It was confirmed that the nanoparticles of pp-siRNA/CLD/DNCA/DSPE-PEG2000-cRGD (PCNR) were able to specifically target tumor cells with highly expressed integrin αvβ3; moreover, it efficiently downregulated the levels of BRAF mRNA and the BRAF protein and inhibited cell proliferation in A375 cells, in comparison with the nontargeted nanocomplex of pp-siRNA/CLD/DNCA/cRAD (PCNA). The uptake pathways of PCNR are mostly dependent on CvME-mediated endocytosis and macropinocytosis in A375 cells, which could bypass lysosome or quickly lead to the lysosomal escape to reduce siRNA degradation. Finally, the biodistribution study showed that PCNR exhibited a high ability to accumulate in tumor tissues. These results suggest that the nanocomplex of PCNR is promising to be highly effective in the treatment of melanomas including their mutation.
Collapse
Affiliation(s)
- Yanfen Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China.,School of Pharmaceutical Sciences, HeZe University, Heze, Shandong 274015, P. R. China
| | - Sixiu Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Xinyang Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Jing Sun
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Xinmeng Fan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Zhu Guan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
39
|
Lakkadwala S, Dos Santos Rodrigues B, Sun C, Singh J. Biodistribution of TAT or QLPVM coupled to receptor targeted liposomes for delivery of anticancer therapeutics to brain in vitro and in vivo. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 23:102112. [PMID: 31669083 DOI: 10.1016/j.nano.2019.102112] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/26/2019] [Accepted: 10/03/2019] [Indexed: 01/31/2023]
Abstract
Combination therapy has emerged as an efficient way to deliver chemotherapeutics for treatment of glioblastoma. It provides collaborative approach of targeting cancer cells by acting via multiple mechanisms, thereby reducing drug resistance. However, the presence of impermeable blood brain barrier (BBB) restricts the delivery of chemotherapeutic drugs into the brain. To overcome this limitation, we designed a dual functionalized liposomes by modifying their surface with transferrin (Tf) and a cell penetrating peptide (CPP) for receptor and adsorptive mediated transcytosis, respectively. In this study, we used two different CPPs (based on physicochemical properties) and investigated the influence of insertion of CPP to Tf-liposomes on biocompatibility, cellular uptake, and transport across the BBB both in vitro and in vivo. The biodistribution profile of Tf-CPP liposomes showed more than 10 and 2.7 fold increase in doxorubicin and erlotinib accumulation in mice brain, respectively as compared to free drugs with no signs of toxicity.
Collapse
Affiliation(s)
- Sushant Lakkadwala
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Bruna Dos Santos Rodrigues
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Chengwen Sun
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
40
|
Sun S, Zou H, Li L, Liu Q, Ding N, Zeng L, Li H, Mao S. CD123/CD33 dual-antibody modified liposomes effectively target acute myeloid leukemia cells and reduce antigen-negative escape. Int J Pharm 2019; 568:118518. [DOI: 10.1016/j.ijpharm.2019.118518] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/30/2019] [Accepted: 07/11/2019] [Indexed: 12/29/2022]
|
41
|
Unnikrishnan S, Du Z, Diakova GB, Klibanov AL. Formation of Microbubbles for Targeted Ultrasound Contrast Imaging: Practical Translation Considerations. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10034-10041. [PMID: 30509068 DOI: 10.1021/acs.langmuir.8b03551] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
For preparation of ligand-decorated microbubbles for targeted ultrasound contrast imaging, it is important to maximize the amount of ligand associated with the bubble shell. We describe optimization of the use of a biocompatible cosurfactant in the microbubble formulation media to maximize the incorporation of targeting ligand-lipid conjugate into the microbubble shell, and thus reduce the fraction of ligand not associated with microbubbles, following amalgamation preparation. The influence of the concentration of a helper cosurfactant propylene glycol (PG) on the efficacy of microbubble preparation by amalgamation and on the degree of association of fluorescent PEG-lipid with the microbubble shell was tested. Three sets of targeted bubbles were then prepared: with VCAM-1-targeting peptide VHPKQHRGGSK(FITC)GC-PEG-DSPE, cyclic RGDfK-PEG-DSPE, selective for αVβ3, and control cRADfK-PEG-DSPE, without such affinity. Microbubbles were prepared by 45 s amalgamation, with DSPC and PEG stearate as the main components of the shell, with 15% PG in aqueous saline. In vitro microbubble targeting was assessed with a parallel plate flow chamber with a recombinant receptor coated surface. In vivo targeting was assessed in MC-38 tumor-bearing mice (subcutaneous tumor in hind leg), 10 min after intravenous bolus of microbubble contrast agent (20 million particles per injection). Ultrasound imaging of the tumor and control nontarget muscle tissue in a contralateral leg was performed with a clinical scanner. Amalgamation technique with PG cosurfactant produced microbubbles at concentrations exceeding 2 × 109 particles/mL, and ∼50-60% or more of the added fluorescein-PEG-DSPE or VCAM-1-targeted fluorescent peptide was associated with microbubbles, about 2 times higher than that in the absence of PG. After intravenous injection, peptide-targeted bubbles selectively accumulated in the tumor vasculature, with negligible accumulation in nontumor contralateral leg muscle, or with control nontargeted microbubbles (assessed by contrast ultrasound imaging). For comparison, administration of RGD-decorated microbubbles prepared by traditional sonication, and purified from free peptide-PEG-lipid by repeated centrifugation, resulted in the same accumulation pattern as for translatable amalgamated microbubbles. Following amalgamation in the presence of PG, efficient transfer of ligand-PEG-lipid to microbubble shell was achieved and quantified. Purification of microbubbles from free peptide-PEG-lipid was not necessary, as proven by in vitro and in vivo targeting studies, so PG cosurfactant amalgamation technique generated peptide-targeted microbubbles are amenable for bedside preparation and clinical translation. The pathway to clinical translation is simplified by the fact that most of the materials used in this study either are on the United States Food and Drug Administration GRAS list or can be procured as pharmaceutical grade substances.
Collapse
Affiliation(s)
- Sunil Unnikrishnan
- Department of Biomedical Engineering , University of Virginia , Charlottesville , Virginia 22908 , United States
| | - Zhongmin Du
- Cardiovascular Division, Department of Medicine, Robert M. Berne Cardiovascular Research Center , University of Virginia School of Medicine , Charlottesville , Virginia 22908 , United States
| | - Galina B Diakova
- Cardiovascular Division, Department of Medicine, Robert M. Berne Cardiovascular Research Center , University of Virginia School of Medicine , Charlottesville , Virginia 22908 , United States
| | - Alexander L Klibanov
- Cardiovascular Division, Department of Medicine, Robert M. Berne Cardiovascular Research Center , University of Virginia School of Medicine , Charlottesville , Virginia 22908 , United States
- Department of Biomedical Engineering , University of Virginia , Charlottesville , Virginia 22908 , United States
| |
Collapse
|
42
|
Lakkadwala S, Dos Santos Rodrigues B, Sun C, Singh J. Dual functionalized liposomes for efficient co-delivery of anti-cancer chemotherapeutics for the treatment of glioblastoma. J Control Release 2019; 307:247-260. [PMID: 31252036 DOI: 10.1016/j.jconrel.2019.06.033] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/04/2019] [Accepted: 06/24/2019] [Indexed: 11/18/2022]
Abstract
Glioblastoma is a hostile brain tumor associated with high infiltration leading to poor prognosis. Anti-cancer chemotherapeutic agents have limited access into the brain due to the presence of the blood brain barrier (BBB). In this study, we designed a dual functionalized liposomal delivery system, surface modified with transferrin (Tf) for receptor mediated transcytosis and a cell penetrating peptide-penetratin (Pen) for enhanced cell penetration. We loaded doxorubicin and erlotinib into liposomes to enhance their translocation across the BBB to glioblastoma tumor. In vitro cytotoxicity and hemocompatibility studies demonstrated excellent biocompatibility for in vivo administration. Co-delivery of doxorubicin and erlotinib loaded Tf-Pen liposomes revealed significantly (p < 0.05) higher translocation (~15%) across the co-culture endothelial barrier resulting in regression of tumor in the in vitro brain tumor model. The biodistribution of Tf-Pen liposomes demonstrated ~12 and 3.3 fold increase in doxorubicin and erlotinib accumulation in mice brain, respectively compared to free drugs. In addition, Tf-Pen liposomes showed excellent antitumor efficacy by regressing ~90% of tumor in mice brain with significant increase in the median survival time (36 days) along with no toxicity. Thus, we believe that this study would have high impact for treating patients with glioblastoma.
Collapse
Affiliation(s)
- Sushant Lakkadwala
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Bruna Dos Santos Rodrigues
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Chengwen Sun
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA.
| |
Collapse
|
43
|
Krämer W, Grapentin C, Bouvain P, Temme S, Flögel U, Schubert R. Rational manufacturing of functionalized, long-term stable perfluorocarbon-nanoemulsions for site-specific 19F magnetic resonance imaging. Eur J Pharm Biopharm 2019; 142:114-122. [PMID: 31220572 DOI: 10.1016/j.ejpb.2019.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/29/2019] [Accepted: 06/13/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Perfluorocarbon (PFC)-nanoemulsions (NE) are a convenient tool for 19F magnetic resonance imaging in cell and animal experiments. Typical preparation methods, like high-pressure homogenization or microfluidization, produce nanoemulsions in mL-scale. However, experiments usually require only miniscule amounts of PFC-NE, several 100 µL. For site-specific imaging tissue-specific ligands, e.g. peptides or antibodies, are covalently bound to the NE surface. This requires the use of expensive functionalized phospholipids containing reactive groups (e.g. maleimide), which often deteriorate quickly in liquid storage, rendering the manufacturing process highly cost-inefficient. A technique to manufacture storage stable NE that maintain their functionality for coupling of various ligands is desired. METHODS AND RESULTS Different PFC-NE formulations and preparation techniques were compared and the most suitable of these was tested in short-, as well as long-term stability tests. Droplet size stability was investigated by dynamic light scattering and cryogenic transmission electron microscopy over 1.5 a. Surface modifiability was assessed by a fluorescence assay. The utility of these NE was proven in an in vitro model. CONCLUSION The established PFC-NE platform offers a cost-efficient way to produce larger amounts of long-term storable imaging agents, which can be surface-modified on demand for application in targeted 19F MRI.
Collapse
Affiliation(s)
- W Krämer
- Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University of Freiburg, Freiburg, Germany.
| | - C Grapentin
- Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University of Freiburg, Freiburg, Germany
| | - P Bouvain
- Department of Molecular Cardiology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - S Temme
- Department of Molecular Cardiology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - U Flögel
- Department of Molecular Cardiology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - R Schubert
- Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University of Freiburg, Freiburg, Germany
| |
Collapse
|
44
|
Kholodenko RV, Kalinovsky DV, Doronin II, Ponomarev ED, Kholodenko IV. Antibody Fragments as Potential Biopharmaceuticals for Cancer Therapy: Success and Limitations. Curr Med Chem 2019; 26:396-426. [DOI: 10.2174/0929867324666170817152554] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/17/2017] [Accepted: 07/24/2017] [Indexed: 12/23/2022]
Abstract
Monoclonal antibodies (mAbs) are an important class of therapeutic agents approved for the therapy of many types of malignancies. However, in certain cases applications of conventional mAbs have several limitations in anticancer immunotherapy. These limitations include insufficient efficacy and adverse effects. The antigen-binding fragments of antibodies have a considerable potential to overcome the disadvantages of conventional mAbs, such as poor penetration into solid tumors and Fc-mediated bystander activation of the immune system. Fragments of antibodies retain antigen specificity and part of functional properties of conventional mAbs and at the same time have much better penetration into the tumors and a greatly reduced level of adverse effects. Recent advantages in antibody engineering allowed to produce different types of antibody fragments with improved structure and properties for efficient elimination of tumor cells. These molecules opened up new perspectives for anticancer therapy. Here, we will overview the structural features of the various types of antibody fragments and their applications for anticancer therapy as separate molecules and as part of complex conjugates or structures. Mechanisms of antitumor action of antibody fragments as well as their advantages and disadvantages for clinical application will be discussed in this review.
Collapse
Affiliation(s)
- Roman V. Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho- Maklaya St., 16/10, Moscow 117997, Russian Federation
| | - Daniel V. Kalinovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho- Maklaya St., 16/10, Moscow 117997, Russian Federation
| | - Igor I. Doronin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho- Maklaya St., 16/10, Moscow 117997, Russian Federation
| | - Eugene D. Ponomarev
- School of Biomedical Sciences, Faculty of Medicine and Brain, The Chinese University of Hong Kong, Shatin NT, Hong Kong
| | - Irina V. Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho- Maklaya St., 16/10, Moscow 117997, Russian Federation
| |
Collapse
|
45
|
Saeed M, Zalba S, Seynhaeve ALB, Debets R, Ten Hagen TLM. Liposomes targeted to MHC-restricted antigen improve drug delivery and antimelanoma response. Int J Nanomedicine 2019; 14:2069-2089. [PMID: 30988609 PMCID: PMC6440454 DOI: 10.2147/ijn.s190736] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Purpose Melanoma is the most aggressive form of skin cancer. Chemotherapy at a late stage fails due to low accumulation in tumors, indicating the need for targeted therapy. Materials and methods To increase drug uptake by tumor cells, we have targeted doxorubicin-containing liposomes using a T-cell receptor (TCR)-like antibody (scFv G8 and Hyb3) directed against melanoma antigen A1 (MAGE-A1) presented by human leukocyte antigen A1 (M1/A1). With the use of flow cytometry and confocal microscopy, we have tested our formulation in vitro. In vivo pharmacokinetics was done in tumor-free nu/nu mice, while biodistribution and efficacy study was done in nu/nu mice xenograft. Results We demonstrated two to five times higher binding and internalization of these immunoliposomes by M1+/A1+ melanoma cells in vitro in comparison with nontargeted liposomes. Cytotoxicity assay showed significant tumor cell kill at 10 µM doxorubicin (DXR) for targeted vs nontargeted liposomes. In vivo pharmacokinetics of nontargeted and targeted liposomes were similar, while accumulation of targeted liposomes was 2- to 2.5-fold and 6.6-fold enhanced when compared with nontargeted liposomes and free drug, respectively. Notably, we showed a superior antitumor activity of MAGE-A1-targeted DXR liposomes toward M1+/A1+ expressing tumors in mice compared with the treatment of M1−/A1+ tumors. Our results indicate that targeted liposomes showed better cytotoxicity in vitro and pharmacokinetics in vivo. Conclusion Liposomes decorated with TCR-mimicking scFv antibodies effectively and selectively target antigen-positive melanoma. We showed that DXR-loaded liposomes coupled to anti-M1/-A1 scFv inflict a significant antitumor response. Targeting tumor cells specifically promotes internalization of drug-containing nanoparticles and may improve drug delivery and ultimately antitumor efficacy. Our data argue that targeting MAGE in A1 context, by nanosized carriers decorated with TCR-like antibodies mimicking scFv, can be used as a theragnostic platform for drug delivery, immunotherapy, and potentially imaging, and diagnosis of melanoma.
Collapse
Affiliation(s)
- Mesha Saeed
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, The Netherlands,
| | - Sara Zalba
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, The Netherlands,
| | - Ann L B Seynhaeve
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, The Netherlands,
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Timo L M Ten Hagen
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, The Netherlands,
| |
Collapse
|
46
|
Wöll S, Dickgiesser S, Rasche N, Schiller S, Scherließ R. Sortagged anti-EGFR immunoliposomes exhibit increased cytotoxicity on target cells. Eur J Pharm Biopharm 2019; 136:203-212. [DOI: 10.1016/j.ejpb.2019.01.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/19/2019] [Accepted: 01/21/2019] [Indexed: 10/27/2022]
|
47
|
Wöll S, Bachran C, Schiller S, Schröder M, Conrad L, Scherließ R, Swee LK. Sortagging of liposomes with a murine CD11b-specific VHH increases in vitro and in vivo targeting specificity of myeloid cells. Eur J Pharm Biopharm 2019; 134:190-198. [DOI: 10.1016/j.ejpb.2018.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
|
48
|
Cheng YA, Chen IJ, Su YC, Cheng KW, Lu YC, Lin WW, Hsieh YC, Kao CH, Chen FM, Roffler SR, Cheng TL. Enhanced drug internalization and therapeutic efficacy of PEGylated nanoparticles by one-step formulation with anti-mPEG bispecific antibody in intrinsic drug-resistant breast cancer. Biomater Sci 2019; 7:3404-3417. [DOI: 10.1039/c9bm00323a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
One-step formulation of BsAb with PLD is a simple method to enhance tumor specificity, internalization and the anti-cancer activity.
Collapse
|
49
|
Wu M, Huang T, Wang J, Chen P, Mi W, Ying Y, Wang H, Zhao D, Huang S. Antilung cancer effect of ergosterol and cisplatin-loaded liposomes modified with cyclic arginine-glycine-aspartic acid and octa-arginine peptides. Medicine (Baltimore) 2018; 97:e11916. [PMID: 30113492 PMCID: PMC6113040 DOI: 10.1097/md.0000000000011916] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/20/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Lung cancer is one of the most important diseases threatening human health, and targeted therapy has become the main research direction. This work, therefore, aimed to develop cyclic arginine-glycine-aspartic (RGD) and octa-arginine (R8) peptide-modified ergosterol (ERG)-combined cisplatin (diamminedichloridoplatinum(II) [DDP]) liposomes (LIP) as a drug delivery system. METHODS Soybean phospholipids (SPC) and cholesterol (Chol) were selected to prepare different LIPs: ERG-loaded LIP (ERG-LIP), DDP and ERG-LIP (DDP/ERG-LIP), R8 peptide-modified DDP and ERG-LIP (R8-DDP/ERG-LIP), and cyclic RGD and R8-DDP/ERG-LIP (RGD/R8-DDP/ERG-LIP). The quality, tumor sphere penetrating ability, in vitro cellular uptake, mechanism of cellular uptake, and in vitro cytotoxicity of RGD/R8-DDP/ERG-LIP were evaluated. RESULTS The LIP quality evaluation revealed that RGD/R8-DDP/ERG-LIP is round with a double-layer structure. The average particle size, dispersion coefficient of the polydispersity index (PDI), and zeta potential of RGD/R8-DDP/ERG-LIP were 155.2 ± 8.7 nm, 0.102, and 4.74 ± 0.7 mV, respectively. Furthermore, the LIPs were stable in the serum, and obviously inhibited the growth of A549 lung cancer cells with RGD/R8-DDP/ERG-LIP exhibiting the strongest inhibitory effect. The highest cellular uptake rate, which was at 4 hours, was exhibited by RGD/R8-DDP/ERG-LIP in a concentration-dependent manner. CONCLUSION The results showed that LIP uptake by A549 cells was mainly by the clathrin-mediated endocytosis pathway (chlorpromazine). The results also suggest that RGD/R8-DDP/ERG-LIP might be a promising drug delivery system to improve antilung cancer drug effect and tumor-targeting in vitro.
Collapse
Affiliation(s)
- Meijia Wu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University
| | - Ting Huang
- General Surgical Department, Hangzhou Red Cross Hospital
| | - Juan Wang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University
| | - Ping Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University
| | - Wanwan Mi
- College of Pharmaceutical Science, Zhejiang Chinese Medical University
| | - Yuanyuan Ying
- College of Pharmaceutical Science, Zhejiang Chinese Medical University
| | - Hangli Wang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University
| | - Dandan Zhao
- Pharmacy Department, Hangzhou Zhongxing Hospital, Hangzhou, Zhejiang, China
| | - Shengwu Huang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University
| |
Collapse
|
50
|
Lakkadwala S, Singh J. Dual Functionalized 5-Fluorouracil Liposomes as Highly Efficient Nanomedicine for Glioblastoma Treatment as Assessed in an In Vitro Brain Tumor Model. J Pharm Sci 2018; 107:2902-2913. [PMID: 30055226 DOI: 10.1016/j.xphs.2018.07.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 10/28/2022]
Abstract
Drug delivery to the brain has been a major challenge due to the presence of the blood-brain barrier, which limits the uptake of most chemotherapeutics into brain. We developed a dual-functionalized liposomal delivery system, conjugating cell penetrating peptide penetratin to transferrin-liposomes (Tf-Pen-conjugated liposomes) to enhance the transport of an anticancer chemotherapeutic drug, 5-fluorouracil (5-FU), across the blood-brain barrier into the tumor cells. The in vitro cellular uptake study showed that the dual-functionalized liposomes are capable of higher cellular uptake in glioblastoma (U87) and brain endothelial (bEnd.3) cells monolayer. In addition, dual-functionalized liposomes demonstrated significantly higher apoptosis in U87 cells. The liposomal nanoparticles showed excellent blood compatibility and in vitro cell viability, as studied by hemolysis and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, respectively. The 5-FU-loaded dual-functionalized liposomes demonstrated higher transport across the brain endothelial barrier and delivered 5-FU to tumor cells inside poly(lactic-co-glycolic acid)-chitosan scaffold (an in vitro brain tumor model), resulting in significant tumor regression.
Collapse
Affiliation(s)
- Sushant Lakkadwala
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105.
| |
Collapse
|