1
|
Gambles MT, Li S, Kendell I, Li J, Sborov D, Shami P, Yang J, Kopeček J. Multiantigen T-Cell Hybridizers: A Two-Component T-Cell-Activating Therapy. ACS NANO 2024; 18:23341-23353. [PMID: 39149859 DOI: 10.1021/acsnano.4c06500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Multispecific T-cell-engaging scaffolds have emerged as effective anticancer therapies for the treatment of hematological malignancies. Approaches that modulate cancer cell targeting and provide personalized, multispecific immunotherapeutics are needed. Here, we report on a modular, split antibody-like approach consisting of Fab' fragments modified with complementary morpholino oligonucleotides (MORFs). We synthesized a library of B-cell-targeting Fab'-MORF1 conjugates that self-assemble, via a Watson-Crick base pairing hybridization, with a complementary T-cell-engaging Fab'-MORF2 conjugate. We aptly titled our technology multiantigen T-cell hybridizers (MATCH). Using MATCH, cancer-specific T-cell recruitment was achieved utilizing four B-cell antigen targets: CD20, CD38, BCMA, and SLAMF7. The antigen expression profiles of various malignant B-cell lines were produced, and using these distinct profiles, cell-specific T-cell activation was attained on lymphoma, leukemia, and multiple myeloma cell lines in vitro. T-cell rechallenge experiments demonstrated the modular approach of MATCH by sequentially activating the same T-cell cohort against three different cancers using cancer antigen-specific Fab'-MORF1 conjugates. Furthermore, MATCH's efficacy was demonstrated in vivo by treating xenograft mouse models of human non-Hodgkin's lymphoma with CD20-directed MATCH therapy. In the pilot study, a single dose of MATCH allowed for long-term survival of all treated mice compared to saline control. In a second in vivo model, insights regarding optimal T-cell-to-target cell ratio were gleaned when a ratio of 5:1 T-cell-to-target cell MATCH-treated mice significantly delayed the onset of disease compared to higher and lower ratios.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Shannuo Li
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Isaac Kendell
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jiahui Li
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Douglas Sborov
- Huntsman Cancer Institute, University of Utah, Salt Lake City ,Utah 84112, United States
| | - Paul Shami
- Huntsman Cancer Institute, University of Utah, Salt Lake City ,Utah 84112, United States
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
2
|
Gambles MT, Yang J, Kopeček J. Multi-targeted immunotherapeutics to treat B cell malignancies. J Control Release 2023; 358:232-258. [PMID: 37121515 PMCID: PMC10330463 DOI: 10.1016/j.jconrel.2023.04.048] [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: 01/05/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The concept of multi-targeted immunotherapeutic systems has propelled the field of cancer immunotherapy into an exciting new era. Multi-effector molecules can be designed to engage with, and alter, the patient's immune system in a plethora of ways. The outcomes can vary from effector cell recruitment and activation upon recognition of a cancer cell, to a multipronged immune checkpoint blockade strategy disallowing evasion of the cancer cells by immune cells, or to direct cancer cell death upon engaging multiple cell surface receptors simultaneously. Here, we review the field of multi-specific immunotherapeutics implemented to treat B cell malignancies. The mechanistically diverse strategies are outlined and discussed; common B cell receptor antigen targeting strategies are outlined and summarized; and the challenges of the field are presented along with optimistic insights for the future.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
3
|
DOPE/CHEMS-Based EGFR-Targeted Immunoliposomes for Docetaxel Delivery: Formulation Development, Physicochemical Characterization and Biological Evaluation on Prostate Cancer Cells. Pharmaceutics 2023; 15:pharmaceutics15030915. [PMID: 36986777 PMCID: PMC10052572 DOI: 10.3390/pharmaceutics15030915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Docetaxel (DTX) is a non-selective antineoplastic agent with low solubility and a series of side effects. The technology of pH-sensitive and anti-epidermal growth factor receptor (anti-EGFR) immunoliposomes aims to increase the selective delivery of the drug in the acidic tumor environment to cells with EFGR overexpression. Thus, the study aimed to develop pH-sensitive liposomes based on DOPE (dioleoylphosphatidylethanolamine) and CHEMS (cholesteryl hemisuccinate), using a Box–Behnken factorial design. Furthermore, we aimed to conjugate the monoclonal antibody cetuximab onto liposomal surface, as well as to thoroughly characterize the nanosystems and evaluate them on prostate cancer cells. The liposomes prepared by hydration of the lipid film and optimized by the Box–Behnken factorial design showed a particle size of 107.2 ± 2.9 nm, a PDI of 0.213 ± 0.005, zeta potential of −21.9 ± 1.8 mV and an encapsulation efficiency of 88.65 ± 20.3%. Together, FTIR, DSC and DRX characterization demonstrated that the drug was properly encapsulated, with reduced drug crystallinity. Drug release was higher in acidic pH. The liposome conjugation with the anti-EGFR antibody cetuximab preserved the physicochemical characteristics and was successful. The liposome containing DTX reached an IC50 at a concentration of 65.74 nM in the PC3 cell line and 28.28 nM in the DU145 cell line. Immunoliposome, in turn, for PC3 cells reached an IC50 of 152.1 nM, and for the DU145 cell line, 12.60 nM, a considerable enhancement of cytotoxicity for the EGFR-positive cell line. Finally, the immunoliposome internalization was faster and greater than that of liposome in the DU145 cell line, with a higher EGFR overexpression. Thus, based on these results, it was possible to obtain a formulation with adequate characteristics of nanometric size, a high encapsulation of DTX and liposomes and particularly immunoliposomes containing DTX, which caused, as expected, a reduction in the viability of prostate cells, with high cellular internalization in EGFR overexpressing cells.
Collapse
|
4
|
Singla A, Simbassa SB, Chirra B, Gairola A, Southerland MR, Shah KN, Rose RE, Chen Q, Basharat A, Baeza J, Raina R, Chapman MJ, Hassan AM, Ivanov I, Sen A, Wu HJ, Cannon CL. Hetero-Multivalent Targeted Liposomal Drug Delivery to Treat Pseudomonas aeruginosa Infections. ACS APPLIED MATERIALS & INTERFACES 2022; 14:40724-40737. [PMID: 36018830 PMCID: PMC9480101 DOI: 10.1021/acsami.2c12943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
Pseudomonas aeruginosa is the leading nosocomial and community-acquired pathogen causing a plethora of acute and chronic infections. The Centers for Disease Control and Prevention has designated multidrug-resistant isolates of P. aeruginosa as a serious threat. A novel delivery vehicle capable of specifically targeting P. aeruginosa, and encapsulating antimicrobials, may address the challenges associated with these infections. We have developed hetero-multivalent targeted liposomes functionalized with host cell glycans to increase the delivery of antibiotics to the site of infection. Previously, we have demonstrated that compared with monovalent liposomes, these hetero-multivalent liposomes bind with higher affinity to P. aeruginosa. Here, compared with nontargeted liposomes, we have shown that greater numbers of targeted liposomes are found in the circulation, as well as at the site of P. aeruginosa (PAO1) infection in the thighs of CD-1 mice. No significant difference was found in the uptake of targeted, nontargeted, and PEGylated liposomes by J774.A1 macrophages. Ciprofloxacin-loaded liposomes were formulated and characterized for size, encapsulation, loading, and drug release. In vitro antimicrobial efficacy was assessed using CLSI broth microdilution assays and time-kill kinetics. Lastly, PAO1-inoculated mice treated with ciprofloxacin-loaded, hetero-multivalent targeted liposomes survived longer than mice treated with ciprofloxacin-loaded, monovalent targeted, or nontargeted liposomes and free ciprofloxacin. Thus, liposomes functionalized with host cell glycans target P. aeruginosa resulting in increased retention of the liposomes in the circulation, accumulation at the site of infection, and increased survival time in a mouse surgical site infection model. Consequently, this formulation strategy may improve outcomes in patients infected with P. aeruginosa.
Collapse
Affiliation(s)
- Akshi Singla
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Sabona B. Simbassa
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Bhagath Chirra
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Anirudh Gairola
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Marie R. Southerland
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Kush N. Shah
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Robert E. Rose
- Comparative
Medicine Program, Texas A&M University, College Station, Texas 77843, United States
| | - Qingquan Chen
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Ahmed Basharat
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Jaime Baeza
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Rohit Raina
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Morgan J. Chapman
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Adel M. Hassan
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| | - Ivan Ivanov
- Department
of Veterinary Physiology and Pharmacology, Texas A&M University, College
Station, Texas 77843, United States
| | - Anindito Sen
- Microscopy
and Imaging Center, Texas A&M University, College Station, Texas 77843, United States
| | - Hung-Jen Wu
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Carolyn L. Cannon
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan Texas 77807, United States
| |
Collapse
|
5
|
Sun Y, Davis E. Nanoplatforms for Targeted Stimuli-Responsive Drug Delivery: A Review of Platform Materials and Stimuli-Responsive Release and Targeting Mechanisms. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:746. [PMID: 33809633 PMCID: PMC8000772 DOI: 10.3390/nano11030746] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
To achieve the promise of stimuli-responsive drug delivery systems for the treatment of cancer, they should (1) avoid premature clearance; (2) accumulate in tumors and undergo endocytosis by cancer cells; and (3) exhibit appropriate stimuli-responsive release of the payload. It is challenging to address all of these requirements simultaneously. However, the numerous proof-of-concept studies addressing one or more of these requirements reported every year have dramatically expanded the toolbox available for the design of drug delivery systems. This review highlights recent advances in the targeting and stimuli-responsiveness of drug delivery systems. It begins with a discussion of nanocarrier types and an overview of the factors influencing nanocarrier biodistribution. On-demand release strategies and their application to each type of nanocarrier are reviewed, including both endogenous and exogenous stimuli. Recent developments in stimuli-responsive targeting strategies are also discussed. The remaining challenges and prospective solutions in the field are discussed throughout the review, which is intended to assist researchers in overcoming interdisciplinary knowledge barriers and increase the speed of development. This review presents a nanocarrier-based drug delivery systems toolbox that enables the application of techniques across platforms and inspires researchers with interdisciplinary information to boost the development of multifunctional therapeutic nanoplatforms for cancer therapy.
Collapse
Affiliation(s)
| | - Edward Davis
- Materials Engineering Program, Mechanical Engineering Department, Auburn University, 101 Wilmore Drive, Auburn, AL 36830, USA;
| |
Collapse
|
6
|
Sonju JJ, Dahal A, Singh SS, Jois SD. Peptide-functionalized liposomes as therapeutic and diagnostic tools for cancer treatment. J Control Release 2021; 329:624-644. [PMID: 33010333 PMCID: PMC8082750 DOI: 10.1016/j.jconrel.2020.09.055] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/26/2022]
Abstract
Clinically efficacious medication in anticancer therapy has been successfully designed with liposome-based nanomedicine. The liposomal formulation in cancer drug delivery can be facilitated with a functionalized peptide that mediates the specific drug delivery opportunities with increased drug penetrability, specific accumulation in the targeted site, and enhanced therapeutic efficacy. This review aims to focus on recent advances in peptide-functionalized liposomal formulation techniques in cancer diagnosis and treatment regarding recently published literature. It also will highlight different aspects of novel liposomal formulation techniques that incorporate surface functionalization with peptides for better anticancer effect and current challenges in peptide-functionalized liposomal drug formulation.
Collapse
Affiliation(s)
- Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
7
|
Biomedical nanoparticle design: What we can learn from viruses. J Control Release 2021; 329:552-569. [PMID: 33007365 PMCID: PMC7525328 DOI: 10.1016/j.jconrel.2020.09.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 01/02/2023]
Abstract
Viruses are nanomaterials with a number of properties that surpass those of many synthetic nanoparticles (NPs) for biomedical applications. They possess a rigorously ordered structure, come in a variety of shapes, and present unique surface elements, such as spikes. These attributes facilitate propitious biodistribution, the crossing of complex biological barriers and a minutely coordinated interaction with cells. Due to the orchestrated sequence of interactions of their stringently arranged particle corona with cellular surface receptors they effectively identify and infect their host cells with utmost specificity, while evading the immune system at the same time. Furthermore, their efficacy is enhanced by their response to stimuli and the ability to spread from cell to cell. Over the years, great efforts have been made to mimic distinct viral traits to improve biomedical nanomaterial performance. However, a closer look at the literature reveals that no comprehensive evaluation of the benefit of virus-mimetic material design on the targeting efficiency of nanomaterials exists. In this review we, therefore, elucidate the impact that viral properties had on fundamental advances in outfitting nanomaterials with the ability to interact specifically with their target cells. We give a comprehensive overview of the diverse design strategies and identify critical steps on the way to reducing them to practice. More so, we discuss the advantages and future perspectives of a virus-mimetic nanomaterial design and try to elucidate if viral mimicry holds the key for better NP targeting.
Collapse
|
8
|
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: 55] [Impact Index Per Article: 11.0] [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
|
9
|
Zhang X, Lin C, Chan W, Liu K, Lu A, Lin G, Hu R, Shi H, Zhang H, Yang Z. Dual-Functional Liposomes with Carbonic Anhydrase IX Antibody and BR2 Peptide Modification Effectively Improve Intracellular Delivery of Cantharidin to Treat Orthotopic Hepatocellular Carcinoma Mice. Molecules 2019; 24:molecules24183332. [PMID: 31547459 PMCID: PMC6767275 DOI: 10.3390/molecules24183332] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 01/08/2023] Open
Abstract
Liposomal nanotechnology has a great potential to overcome the current major problems of chemotherapy. However, the lack of penetrability and targetability retards the successful delivery of liposomal carriers. Previously, we showed that BR2 peptide modification endowed cantharidin-loaded liposomes with intracellular penetration that enhanced the drug cytotoxic effects. Here, we aimed to improve the targeting delivery of drugs into cancer cells via highly expressed carbonic anhydrase IX (CA IX) receptors by modifying our previous catharidin-loaded BR2-liposomes with anti-CA IX antibody. A higher cellular uptake of dual-functional liposomes (DF-Lp) than other treatments was observed. Induction of CA IX over-expressing resulted in a higher cellular binding of DF-Lp; subsequently, blocking with excess antibodies resulted in a decreased cancer-cell association, indicating a specific targeting property of our liposomes towards CA IX expressed cells. After 3h tracking, most of the liposomes were located around the nucleus which confirmed the involvement of targeting intracellular delivery. Cantharidin loaded DF-Lp exhibited enhanced cytotoxicity in vitro and was most effective in controlling tumor growth in vivo in an orthotopic hepatocellular carcinoma model compared to other groups. Collectively, our results presented the advantage of the BR2 peptide and CA IX antibody combination to elevate the therapeutic potential of cantharidin loaded DF-liposomes.
Collapse
Affiliation(s)
- Xue Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China.
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China.
| | - Congcong Lin
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| | - Waikei Chan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Kanglun Liu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu 215505, China.
| | - Ge Lin
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China.
| | - Rong Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China.
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China.
| | - Hongcan Shi
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China.
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China.
| | - Hongqi Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu 215505, China.
| |
Collapse
|
10
|
Abstract
Early researchers focussed on developing stimuli-responsive liposomes in order to manipulate drug release at the site of action or under certain conditions. In recent times, a great deal of efforts has been made to modify the surface of liposomes with ligands for the purpose of achieving targeted drug delivery. Due to the morphology of liposomes, their surfaces can be engineered by attaching molecules such as oligosaccharides, peptides, antibodies, antigens and oligonucleotides to the bilayer structure. Over the years, a number of techniques including the use of covalent and non-covalent linkages have been utilised in designing ligand-liposome conjugates. In this review, various strategies for the functionalisation of liposomes as well as the different types of ligand-liposome conjugates have been discussed. Finally, the pros and cons of conjugation in liposomes are concisely summarised.
Collapse
Affiliation(s)
- İpek Eroğlu
- Faculty of Pharmacy, Department of Basic Pharmaceutical Sciences, Hacettepe University, Ankara, Turkey
| | - Mamudu İbrahim
- Faculty of Pharmacy, Department of Basic Pharmaceutical Sciences, Hacettepe University, Ankara, Turkey
| |
Collapse
|
11
|
Wang D, Sun Y, Liu Y, Meng F, Lee RJ. Clinical translation of immunoliposomes for cancer therapy: recent perspectives. Expert Opin Drug Deliv 2018; 15:893-903. [PMID: 30169978 DOI: 10.1080/17425247.2018.1517747] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Liposomes have been extensively investigated as drug delivery vehicles. Immunoliposomes (ILs) are antibody-conjugated liposomes designed to selectively target antigen-expressing cells. ILs can be used to deliver drugs to tumor cells for improving efficacy and reducing toxicity. In addition, ILs can be used in immunoassays, immunotherapy, and imaging. Although there has been extensive coverage on ILs in the literature, only a limited number of clinical trials have been reported and no IL drug has been approved by the FDA. AREAS COVERED Factors to consider in developing ILs are discussed, including the choice of antibody or antibody fragment, the formulation of liposomes, and the conjugation chemistry. In addition, challenges and opportunities in clinical development of ILs are discussed. The purpose of this review is to provide an overview on the state of the art of ILs and to discuss potential future developments. EXPERT OPINION IL research has had a lengthy history and numerous preclinical studies have yielded encouraging results. However, there are a number of obstacles to clinical translation of ILs. Given the unique capabilities of ILs, its potential for clinical application is underexplored. There is great potential for expanded role for ILs in the clinic and further efforts to this end are warranted. ABBREVIATIONS Ab: antibody; ADCs: antibody-drug conjugates; API: active pharmaceutical ingredient; ADCC: antibody-dependent cellular cytotoxicity; CR: complete remission; cGMP: current good manufacturing practice; DSPE: distearoyl phosphatidylethanolamine; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; EPR: enhanced permeability and retention; Fc: fragment crystalline; Tf: transferrin; HACA: human-anti-chimeric antibody; HAHA: human-anti-human antibody; HAMA: human-anti-mouse antibody; HER2: human epidermal growth factor 2; IL: immunoliposome; LNPs: lipid nanoparticles; MRI: magnetic resonance imaging; MTD: maximum tolerated dose; PEG: polyethylene glycol; PET: positron emission tomography; PR: partial response; PSMA: prostate-specific membrane antigen; scFv: single-chain variable fragment; SPECT: single photon emission computed tomography; TTR: transthyretin.
Collapse
Affiliation(s)
- Di Wang
- a School of Life Sciences , Jilin University , Changchun , China
| | - Yating Sun
- a School of Life Sciences , Jilin University , Changchun , China
| | - Yange Liu
- a School of Life Sciences , Jilin University , Changchun , China
| | - Fanchao Meng
- a School of Life Sciences , Jilin University , Changchun , China
| | - Robert J Lee
- a School of Life Sciences , Jilin University , Changchun , China.,b Division of Pharmaceutics and Pharmaceutical Chemistry , The Ohio State University , Columbus , OH , USA
| |
Collapse
|
12
|
Belfiore L, Spenkelink LM, Ranson M, van Oijen AM, Vine KL. Quantification of ligand density and stoichiometry on the surface of liposomes using single-molecule fluorescence imaging. J Control Release 2018; 278:80-86. [DOI: 10.1016/j.jconrel.2018.03.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 03/21/2018] [Indexed: 12/01/2022]
|
13
|
Belfiore L, Saunders DN, Ranson M, Thurecht KJ, Storm G, Vine KL. Towards clinical translation of ligand-functionalized liposomes in targeted cancer therapy: Challenges and opportunities. J Control Release 2018; 277:1-13. [PMID: 29501721 DOI: 10.1016/j.jconrel.2018.02.040] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 01/03/2023]
Abstract
The development of therapeutic resistance to targeted anticancer therapies remains a significant clinical problem, with intratumoral heterogeneity playing a key role. In this context, improving the therapeutic outcome through simultaneous targeting of multiple tumor cell subtypes within a heterogeneous tumor is a promising approach. Liposomes have emerged as useful drug carriers that can reduce systemic toxicity and increase drug delivery to the tumor site. While clinically used liposomal drug formulations show marked therapeutic advantages over free drug formulations, ligand-functionalized liposomes that can target multiple tumor cell subtypes may further improve the therapeutic efficacy by facilitating drug delivery to a broader population of tumor cells making up the heterogeneous tumor tissue. Ligand-directed liposomes enable the so-called active targeting of cell receptors via surface-attached ligands that direct drug uptake into tumor cells or tumor-associated stromal cells, and so can increase the selectivity of drug delivery. Despite promising preclinical results demonstrating improved targeting and anti-tumor effects of ligand-directed liposomes, there has been limited translation of this approach to the clinic. Key challenges for translation include the lack of established methods to scale up production and comprehensively characterize ligand-functionalized liposome formulations, as well as the inadequate recapitulation of in vivo tumors in the preclinical models currently used to evaluate their performance. Herein, we discuss the utility of recent ligand-directed liposome approaches, with a focus on dual-ligand liposomes, for the treatment of solid tumors and examine the drawbacks limiting their progression to clinical adoption.
Collapse
Affiliation(s)
- Lisa Belfiore
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia
| | - Darren N Saunders
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology (AIBN), Centre for Advanced Imaging (CAI), Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, Australia
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, CG, The Netherlands
| | - Kara L Vine
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia.
| |
Collapse
|
14
|
Greene MK, Richards DA, Nogueira JCF, Campbell K, Smyth P, Fernández M, Scott CJ, Chudasama V. Forming next-generation antibody-nanoparticle conjugates through the oriented installation of non-engineered antibody fragments. Chem Sci 2017; 9:79-87. [PMID: 29629076 PMCID: PMC5869316 DOI: 10.1039/c7sc02747h] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/08/2017] [Indexed: 12/15/2022] Open
Abstract
Enabling oriented installation of non-engineered antibody fragments on nanoparticle surfaces to create next-generation antibody–nanoparticle conjugates.
The successful development of targeted nanotherapeutics is contingent upon the conjugation of therapeutic nanoparticles to target-specific ligands, with particular emphasis being placed on antibody-based ligands. Thus, new methods that enable the covalent and precise installation of targeting antibodies to nanoparticle surfaces are greatly desired, especially those which do not rely on costly and time-consuming antibody engineering techniques. Herein we present a novel method for the highly controlled and oriented covalent conjugation of non-engineered antibody F(ab) fragments to PLGA–PEG nanoparticles using disulfide-selective pyridazinedione linkers and strain-promoted alkyne–azide click chemistry. Exemplification of this method with trastuzumab and cetuximab showed significant improvements in both conjugation efficiency and antigen binding capability, when compared to commonly employed strategies for antibody–nanoparticle construction. This new approach paves the way for the development of antibody-targeted nanomedicines with improved paratope availability, reproducibility and uniformity to enhance both biological activity and ease of manufacture.
Collapse
Affiliation(s)
- Michelle K Greene
- Centre for Cancer Research and Cell Biology , School of Medicine , Dentistry and Biomedical Sciences , Queen's University Belfast , Belfast , UK .
| | | | | | - Katrina Campbell
- Institute for Global Food Security , School of Biological Sciences , Queen's University Belfast , Belfast , UK
| | - Peter Smyth
- Centre for Cancer Research and Cell Biology , School of Medicine , Dentistry and Biomedical Sciences , Queen's University Belfast , Belfast , UK .
| | - Marcos Fernández
- Department of Chemistry , University College London , London , UK .
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology , School of Medicine , Dentistry and Biomedical Sciences , Queen's University Belfast , Belfast , UK .
| | - Vijay Chudasama
- Department of Chemistry , University College London , London , UK . .,Research Institute for Medicines (iMed.ULisboa) , Faculty of Pharmacy , Universidade de Lisboa , Lisbon , Portugal
| |
Collapse
|
15
|
Richards DA, Maruani A, Chudasama V. Antibody fragments as nanoparticle targeting ligands: a step in the right direction. Chem Sci 2017; 8:63-77. [PMID: 28451149 PMCID: PMC5304706 DOI: 10.1039/c6sc02403c] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/05/2016] [Indexed: 12/13/2022] Open
Abstract
Recent advances in nanomedicine have shown that dramatic improvements in nanoparticle therapeutics and diagnostics can be achieved through the use of disease specific targeting ligands. Although immunoglobulins have successfully been employed for the generation of actively targeted nanoparticles, their use is often hampered by the suboptimal characteristics of the resulting complexes. Emerging data suggest that a switch in focus from full antibodies to antibody derived fragments could help to alleviate these problems and expand the potential of antibody-nanoparticle conjugates as biomedical tools. This review aims to highlight how antibody derived fragments have been utilised to overcome both fundamental and practical issues encountered during the design and application of antibody-targeted nanoparticles.
Collapse
Affiliation(s)
- Daniel A Richards
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; ; Tel: +44 (0)207 679 2077
| | - Antoine Maruani
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; ; Tel: +44 (0)207 679 2077
| | - Vijay Chudasama
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; ; Tel: +44 (0)207 679 2077
| |
Collapse
|
16
|
Dataset on FAP-induced emergence of spontaneous metastases and on the preparation of activatable FAP-targeting immunoliposomes to detect the metastases. Data Brief 2016; 9:143-8. [PMID: 27642620 PMCID: PMC5018079 DOI: 10.1016/j.dib.2016.08.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/18/2016] [Accepted: 08/29/2016] [Indexed: 11/23/2022] Open
Abstract
The underlying data demonstrates that fibroblast activation protein (FAP) paves the way for fibrosarcoma cells, which require the proteolysis of the extracellular matrix (ECM) and basement membranes to intravasate from implanted subcutaneous primary tumors into blood vessels, be transported to distant organs where they extravasate from the blood vessels, reattach and proliferate to metastases. The data additionally shows that FAP, when overexpressed on fibrosarcoma cells induces their invasion and formation of spontaneous metastases in multiple organs, particularly after subcutaneous co-implantation of the FAP-expressing and wildtype fibrosarcoma. The raw and processed data presented herein is related to a research article entitled "Potential of activatable FAP-targeting immunoliposomes in intraoperative imaging of spontaneous metastases" (F.L. Tansi, R. Rüger, C. Böhm, R.E. Kontermann, U.K. Teichgraeber, A. Fahr, I. Hilger, 2016) [1]. Furthermore, evidence for the detection of FAP-expressing tumor cells and cells of the tumor stroma by activatable FAP-targeting liposomes is presented in this dataset.
Collapse
|
17
|
Schuster L, Seifert O, Vollmer S, Kontermann RE, Schlosshauer B, Hartmann H. Immunoliposomes for Targeted Delivery of an Antifibrotic Drug. Mol Pharm 2015; 12:3146-57. [DOI: 10.1021/acs.molpharmaceut.5b00012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Liane Schuster
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Stefanie Vollmer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Burkhard Schlosshauer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | - Hanna Hartmann
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| |
Collapse
|
18
|
Rabenhold M, Steiniger F, Fahr A, Kontermann RE, Rüger R. Bispecific single-chain diabody-immunoliposomes targeting endoglin (CD105) and fibroblast activation protein (FAP) simultaneously. J Control Release 2015; 201:56-67. [DOI: 10.1016/j.jconrel.2015.01.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 01/09/2023]
|
19
|
Qiu Y, Deng D, Deng Q, Wu P, Zhang H, Cai C. Synthesis of magnetic Fe3O4–Au hybrids for sensitive SERS detection of cancer cells at low abundance. J Mater Chem B 2015; 3:4487-4495. [DOI: 10.1039/c5tb00638d] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A facile and rapid SERS-based immunoassay for living cancer cells using magnetic Fe3O4–Au hybrid nanoparticles is reported.
Collapse
Affiliation(s)
- Yanchun Qiu
- Jiangsu Key Laboratory of New Power Batteries
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials
- National and Local Joint Engineering Research Center of Biomedical Functional Materials
- College of Chemistry and Materials Science
- Nanjing Normal University
| | - Dan Deng
- Jiangsu Key Laboratory of New Power Batteries
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials
- National and Local Joint Engineering Research Center of Biomedical Functional Materials
- College of Chemistry and Materials Science
- Nanjing Normal University
| | - Qianwen Deng
- Jiangsu Key Laboratory of New Power Batteries
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials
- National and Local Joint Engineering Research Center of Biomedical Functional Materials
- College of Chemistry and Materials Science
- Nanjing Normal University
| | - Ping Wu
- Jiangsu Key Laboratory of New Power Batteries
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials
- National and Local Joint Engineering Research Center of Biomedical Functional Materials
- College of Chemistry and Materials Science
- Nanjing Normal University
| | - Hui Zhang
- Jiangsu Key Laboratory of New Power Batteries
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials
- National and Local Joint Engineering Research Center of Biomedical Functional Materials
- College of Chemistry and Materials Science
- Nanjing Normal University
| | - Chenxin Cai
- Jiangsu Key Laboratory of New Power Batteries
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials
- National and Local Joint Engineering Research Center of Biomedical Functional Materials
- College of Chemistry and Materials Science
- Nanjing Normal University
| |
Collapse
|
20
|
Wang X, Li S, Shi Y, Chuan X, Li J, Zhong T, Zhang H, Dai W, He B, Zhang Q. The development of site-specific drug delivery nanocarriers based on receptor mediation. J Control Release 2014; 193:139-53. [DOI: 10.1016/j.jconrel.2014.05.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/13/2014] [Accepted: 05/17/2014] [Indexed: 01/28/2023]
|
21
|
Seifert O, Pollak N, Nusser A, Steiniger F, Rüger R, Pfizenmaier K, Kontermann RE. Immuno-LipoTRAIL: Targeted delivery of TRAIL-functionalized liposomal nanoparticles. Bioconjug Chem 2014; 25:879-87. [PMID: 24766622 DOI: 10.1021/bc400517j] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The TNF-related apoptosis-inducing ligand (TRAIL) is a powerful inducer of apoptosis in tumor cells; however, clinical studies with recombinant soluble TRAIL were rather disappointing. Here, we developed TRAIL-functionalized liposomes (LipoTRAIL, LT) to mimic membrane-displayed TRAIL for efficient activation of death receptors DR4 and DR5 and enhanced induction of apoptosis, which were combined with an anti-EGFR single-chain Fv fragment (scFv) for targeted delivery to EGFR-positive tumor cells. These immuno-LipoTRAILs (ILTs) bound specifically to EGFR-expressing cells (Colo205) and exhibited increased cytotoxicity compared with that of nontargeted LTs. Compared to that of the soluble TRAIL, the plasma half-life of the functionalized liposomes was strongly extended, and increased antitumor activity of LT and ILT was demonstrated in a xenograft tumor model. Thus, we established a multifunctional liposomal TRAIL formulation (ILT) with improved pharmacokinetic and pharmacodynamic behavior, characterized by targeted delivery and increased induction of apoptosis due to multivalent TRAIL presentation.
Collapse
Affiliation(s)
- Oliver Seifert
- Institut für Zellbiologie und Immunologie, Universität Stuttgart , Allmandring 31, 70569 Stuttgart, Germany
| | | | | | | | | | | | | |
Collapse
|
22
|
Rüger R, Tansi FL, Rabenhold M, Steiniger F, Kontermann RE, Fahr A, Hilger I. In vivo near-infrared fluorescence imaging of FAP-expressing tumors with activatable FAP-targeted, single-chain Fv-immunoliposomes. J Control Release 2014; 186:1-10. [PMID: 24810115 DOI: 10.1016/j.jconrel.2014.04.050] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/17/2014] [Accepted: 04/24/2014] [Indexed: 12/01/2022]
Abstract
Molecular and cellular changes that precede the invasive growth of solid tumors include the release of proteolytic enzymes and peptides in the tumor stroma, the recruitment of phagocytic and lymphoid infiltrates and alteration of the extracellular matrix. The reactive tumor stroma consists of a large number of myofibroblasts, characterized by high expression of fibroblast activation protein alpha (FAP). FAP, a type-II transmembrane sialoglycoprotein is an attractive target in diagnosis and therapy of several pathologic disorders especially cancer. In the underlying work, a fluorescence-activatable liposome (fluorescence-quenched during circulation and fluorescence activation upon cellular uptake), bearing specific single-chain Fv fragments directed against FAP (scFv'FAP) was developed, and its potential for use in fluorescence diagnostic imaging of FAP-expressing tumor cells was evaluated by whole body fluorescence imaging. The liposomes termed anti-FAP-IL were prepared via post-insertion of ligand-phospholipid-conjugates into preformed DY-676-COOH-containing liposomes. The anti-FAP-IL revealed a homogeneous size distribution and showed specific interaction and binding with FAP-expressing cells in vitro. The high level of fluorescence quenching of the near-infrared fluorescent dye sequestered in the aqueous interior of the liposomes enables fluorescence imaging exclusively upon uptake and degradation by cells, which results in fluorescence activation. Only FAP-expressing cells were able to take up and activate fluorescence of anti-FAP-IL in vitro. Furthermore, anti-FAP-IL accumulated selectively in FAP-expressing xenograft models in vivo, as demonstrated by blocking experiments using free scFv'FAP. The local tumor fluorescence intensities were in agreement with the intrinsic degree of FAP-expression in different xenograft models. Thus, anti-FAP-IL can serve as a suitable in vivo diagnostic tool for pathological disorders accompanied by high FAP-expression.
Collapse
Affiliation(s)
- Ronny Rüger
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany.
| | - Felista L Tansi
- Dept. of Experimental Radiology, Institute of Diagnostic and Interventional Radiology I, Jena University Hospital-Friedrich Schiller University Jena, Erlanger Allee 101, 07747 Jena, Germany.
| | - Markus Rabenhold
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany
| | - 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
| | - Alfred Fahr
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany.
| | - Ingrid Hilger
- Dept. of Experimental Radiology, Institute of Diagnostic and Interventional Radiology I, Jena University Hospital-Friedrich Schiller University Jena, Erlanger Allee 101, 07747 Jena, Germany.
| |
Collapse
|
23
|
Musyanovych A, Landfester K. Polymer Micro- and Nanocapsules as Biological Carriers with Multifunctional Properties. Macromol Biosci 2014; 14:458-77. [DOI: 10.1002/mabi.201300551] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 02/03/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Anna Musyanovych
- Fraunhofer ICT-IMM; Carl-Zeiss-Str. 18-20 55129 Mainz Germany
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| | | |
Collapse
|
24
|
Modery-Pawlowski CL, Gupta AS. Heteromultivalent ligand-decoration for actively targeted nanomedicine. Biomaterials 2014; 35:2568-79. [PMID: 24411677 DOI: 10.1016/j.biomaterials.2013.12.047] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/19/2013] [Indexed: 12/25/2022]
Abstract
Active targeting has become an important component of nanomedicine design where nanovehicles are surface-decorated with cell receptor-specific or disease matrix-specific ligands to enable site-selective binding, retention and delivery of theranostic cargo. In this context, there have been numerous reports regarding surface-modification of nanovehicles with antibodies, antibody fragments, carbohydrates, aptamers and peptides as targeting ligands. However, majority of these reports have focused on using a single type of targeting moiety on the vehicle surface. In any disease development and progression, multiple receptors and proteins are often spatio-temporally upregulated simultaneously and heterogeneously. Rationalizing from this, a significant advantage can be envisioned in targeting multiple entities simultaneously using vehicle co-decoration with multiple types of ligands, to enhance binding activity and targeting specificity. To this end, we present a comprehensive up-to-date review on research endeavors in heteromultivalent ligand-modification of nanovehicles and provide a mechanistic rationale as well as an insightful discussion of this promising area, including findings from our own research.
Collapse
Affiliation(s)
| | - Anirban Sen Gupta
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA.
| |
Collapse
|
25
|
Gew LT, Misran M. Albumin-fatty acid interactions at monolayer interface. NANOSCALE RESEARCH LETTERS 2014; 9:218. [PMID: 24910574 PMCID: PMC4029981 DOI: 10.1186/1556-276x-9-218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/20/2014] [Indexed: 05/22/2023]
Abstract
The fluid mosaic model of Singer and Nicolson in 1972 shows how proteins are embedded in membranes. To elucidate the interactions between proteins and the surrounding lipids, stearic acid (SA) and bovine serum albumin (BSA) were used as lipid-protein components to mimic the normal membrane bilayer environment using the Langmuir-Blodgett technique. Surface pressure (π)-molecular area (A) isotherms were recorded for the SA monolayer in the presence of BSA on water. The mixed monolayer was successfully transferred onto an oxidized silicon wafer and imaged by tapping mode atomic force microscopy (AFM). Miscibility, compressibility and thermodynamic stability of the mixed system were examined. A large negative deviation of A ex, together with the minimum value of ΔG ex, was observed when the mole fraction of BSA (X BSA) was 0.8, indicating this to be the most stable mixture. In a compressibility analysis, X BSA was observed at below 50 mN m(-1), denoting a liquid-expanded phase and showing the occurrence of a strong interaction of SA with BSA molecules in this phase. AFM observations supported the quantitative data indicating that BSA was strongly attracted onto the membrane surface as predicted.
Collapse
Affiliation(s)
- Lai Ti Gew
- Department of Biological Sciences, Faculty of Science and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, Petaling Jaya, Selangor 46150, Malaysia
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Misni Misran
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|