1
|
Zhang W, McCartney F, Xu Y, Michalowski CB, Domingues I, Kambale EK, Moreels TG, Guilbaud L, Chen C, Marotti V, Brayden DJ, Beloqui A. An in situ bioadhesive foam as a large intestinal delivery platform for antibody fragment to treat inflammatory bowel disease. J Control Release 2024; 374:254-266. [PMID: 39151828 DOI: 10.1016/j.jconrel.2024.08.023] [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: 05/16/2024] [Revised: 07/22/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Biologics have been widely used as injectables in the treatment of inflammatory bowel disease (IBD). Different local treatment attempts have been developed in recent years. However, maintaining systemic levels of biologics is still crucial for achieving colitis remission. An equilibrium between systemic and local concentrations of biologics is therefore essential for treatment of colitis. Current formulations struggle to create optimal balance between drug concentrations in plasma and the colonic wall. Addressing this challenge, we developed a rectally delivered in situ foam that generates CO2via a reaction between potassium bicarbonate (PB) and citric acid (CA) without the aid of an external device. An anti-TNF-α antibody fragment (Fab) was loaded into the foam formulation, which promoted prolonged colon retention and improved Fab distribution up to proximal colon following rectal administration to mice. In addition, we observed increased plasma Fab concentrations in mice receiving the rectal Fab foam compared to a Fab solution. In a non-everted rat gut ex vivo model, a single exposure to the CO2-containing foam improved macromolecule transepithelial flux across colonic tissue by over ten-fold. Foam efficacy for Fab was investigated in a range of colitis mouse models, from acute to chronic. This non-invasive formulation platform demonstrates potential to overcome existing limitations in delivering biologics to inflamed colonic tissue.
Collapse
Affiliation(s)
- Wunan Zhang
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Fiona McCartney
- University College Dublin School of Veterinary Medicine and Conway Institute, Belfield, Dublin D4, Ireland
| | - Yining Xu
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Cécilia Bohns Michalowski
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Inês Domingues
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Espoir K Kambale
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Tom G Moreels
- UCLouvain, Université Catholique de Louvain, Institute of Experimental and Clinical Research, Laboratory of Hepato-Gastroenterology, 1200 Brussels, Belgium
| | - Léo Guilbaud
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Cheng Chen
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Valentina Marotti
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - David J Brayden
- University College Dublin School of Veterinary Medicine and Conway Institute, Belfield, Dublin D4, Ireland
| | - Ana Beloqui
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium; WEL Research Institute, Avenue Pasteur, 6, 1300 Wavre, Belgium.
| |
Collapse
|
2
|
Kim H, Taslakjian B, Kim S, Tirrell MV, Guler MO. Therapeutic Peptides, Proteins and their Nanostructures for Drug Delivery and Precision Medicine. Chembiochem 2024; 25:e202300831. [PMID: 38408302 DOI: 10.1002/cbic.202300831] [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: 12/08/2023] [Revised: 02/05/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Peptide and protein nanostructures with tunable structural features, multifunctionality, biocompatibility and biomolecular recognition capacity enable development of efficient targeted drug delivery tools for precision medicine applications. In this review article, we present various techniques employed for the synthesis and self-assembly of peptides and proteins into nanostructures. We discuss design strategies utilized to enhance their stability, drug-loading capacity, and controlled release properties, in addition to the mechanisms by which peptide nanostructures interact with target cells, including receptor-mediated endocytosis and cell-penetrating capabilities. We also explore the potential of peptide and protein nanostructures for precision medicine, focusing on applications in personalized therapies and disease-specific targeting for diagnostics and therapeutics in diseases such as cancer.
Collapse
Affiliation(s)
- HaRam Kim
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Boghos Taslakjian
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Sarah Kim
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Matthew V Tirrell
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Mustafa O Guler
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| |
Collapse
|
3
|
de Roode KE, Joosten L, Behe M. Towards the Magic Radioactive Bullet: Improving Targeted Radionuclide Therapy by Reducing the Renal Retention of Radioligands. Pharmaceuticals (Basel) 2024; 17:256. [PMID: 38399470 PMCID: PMC10892921 DOI: 10.3390/ph17020256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Targeted radionuclide therapy (TRT) is an emerging field and has the potential to become a major pillar in effective cancer treatment. Several pharmaceuticals are already in routine use for treating cancer, and there is still a high potential for new compounds for this application. But, a major issue for many radiolabeled low-to-moderate-molecular-weight molecules is their clearance via the kidneys and their subsequent reuptake. High renal accumulation of radioactive compounds may lead to nephrotoxicity, and therefore, the kidneys are often the dose-limiting organs in TRT with these radioligands. Over the years, different strategies have been developed aiming for reduced kidney retention and enhanced therapeutic efficacy of radioligands. In this review, we will give an overview of the efforts and achievements of the used strategies, with focus on the therapeutic potential of low-to-moderate-molecular-weight molecules. Among the strategies discussed here is coadministration of compounds that compete for binding to the endocytic receptors in the proximal tubuli. In addition, the influence of altering the molecular design of radiolabeled ligands on pharmacokinetics is discussed, which includes changes in their physicochemical properties and implementation of cleavable linkers or albumin-binding moieties. Furthermore, we discuss the influence of chelator and radionuclide choice on reabsorption of radioligands by the kidneys.
Collapse
Affiliation(s)
- Kim E. de Roode
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
| | - Martin Behe
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen, Switzerland
| |
Collapse
|
4
|
Liu Y, Corrales-Guerrero S, Kuo JC, Robb R, Nagy G, Cui T, Lee RJ, Williams TM. Improved Targeting and Safety of Doxorubicin through a Novel Albumin Binding Prodrug Approach. ACS OMEGA 2024; 9:977-987. [PMID: 38222540 PMCID: PMC10785662 DOI: 10.1021/acsomega.3c07163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/16/2024]
Abstract
Human serum albumin (HSA) improves the pharmacokinetic profile of drugs attached to it, making it an attractive carrier with proven clinical success. In our previous studies, we have shown that Caveolin-1 (Cav-1) and caveolae-mediated endocytosis play important roles in the uptake of HSA and albumin-bound drugs. Doxorubicin is an FDA-approved chemotherapeutic agent that is effective against multiple cancers, but its clinical applicability has been hampered by its high toxicity levels. In this study, a doxorubicin-prodrug was developed that could independently and avidly bind HSA in circulation, called IPBA-Dox. We first developed and characterized IPBA-Dox and confirmed that it can bind albumin in vitro while retaining a potent cytotoxic effect. We then verified that it efficiently binds to HSA in circulation, leading to an improvement in the pharmacokinetic profile of the drug. In addition, we tested our prodrug for Cav-1 selectivity and found that it preferentially affects cells that express relatively higher levels of Cav-1 in vitro and in vivo. Moreover, we found that our compound was well tolerated in vivo at concentrations at which doxorubicin was lethal. Altogether, we have developed a doxorubicin-prodrug that can successfully bind HSA, retaining a strong cytotoxic effect that preferentially targets Cav-1 positive cells while improving the general tolerability of the drug.
Collapse
Affiliation(s)
- Yang Liu
- Division
of Pharmaceutics and Pharmacology, The Ohio
State University, Columbus, Ohio 43210-1132, United States
| | - Sergio Corrales-Guerrero
- Biomedical
Sciences Graduate Program, The Ohio State
University, Columbus, Ohio 43210-1132, United States
| | - Jimmy C. Kuo
- Division
of Pharmaceutics and Pharmacology, The Ohio
State University, Columbus, Ohio 43210-1132, United States
| | - Ryan Robb
- University
of North Carolina, Chapel
Hill, North Carolina 27514-3916, United States
| | - Gregory Nagy
- Biomedical
Sciences Graduate Program, The Ohio State
University, Columbus, Ohio 43210-1132, United States
| | - Tiantian Cui
- Department
of Radiation Oncology, City of Hope National
Medical Center, Duarte, California 91010, United States
| | - Robert J. Lee
- Division
of Pharmaceutics and Pharmacology, The Ohio
State University, Columbus, Ohio 43210-1132, United States
| | - Terence M. Williams
- Department
of Radiation Oncology, City of Hope National
Medical Center, Duarte, California 91010, United States
| |
Collapse
|
5
|
Qian L, Lin X, Gao X, Khan RU, Liao JY, Du S, Ge J, Zeng S, Yao SQ. The Dawn of a New Era: Targeting the "Undruggables" with Antibody-Based Therapeutics. Chem Rev 2023. [PMID: 37186942 DOI: 10.1021/acs.chemrev.2c00915] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.
Collapse
Affiliation(s)
- Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544
| |
Collapse
|
6
|
Hall DB, Vakkasoglu AS, Hales LM, Soliman TM. D-VITylation: Harnessing the biology of vitamin D to improve the pharmacokinetic properties of peptides and small proteins. Int J Pharm 2022; 624:122031. [PMID: 35863594 DOI: 10.1016/j.ijpharm.2022.122031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 11/30/2022]
Abstract
Peptides have great potential to be potent and specific therapeutics, yet their small size leads to rapid glomerular filtration, which severely limits therapeutic applications. Although conjugation of small proteins to large polymers typically results in longer residence times, these conjugates often have a significant loss of biological activity due to steric hindrance. Here, we improve the pharmacokinetics (PK) of peptide therapeutics by harnessing the biology of vitamin D. Attachment of a small vitamin D-based molecule (D-VITylation) protects the conjugated peptide or protein from renal clearance by virtue of reversible binding to the serum-circulating vitamin D binding protein (DBP), without compromising bioactivity. Varying the conjugation site on vitamin D affects the binding to DBP, with higher affinity corresponding to a longer plasma half-life. We also demonstrate the important contribution of the peptide to the overall PK, likely due to alternative clearance mechanisms such as protease degradation and receptor-mediated cellular uptake. With a Fab antibody fragment, for which these alternate clearance mechanisms are not significant, D-VITylation increases the half-life of elimination from 14 to 61 h in rats. The PK profile in minipigs and projected lifetime in humans suggest that D-VITylation is a viable strategy to achieve once-weekly dosing of peptide therapeutics in humans.
Collapse
|
7
|
Nakatani Y, Ye Z, Ishizue Y, Higashi T, Imai T, Fujii I, Michigami M. “Human and Mouse Cross-Reactive” Albumin-Binding Helix–Loop–Helix Peptide Tag for Prolonged Bioactivity of Therapeutic Proteins. Mol Pharm 2022; 19:2279-2286. [PMID: 35635006 PMCID: PMC9257745 DOI: 10.1021/acs.molpharmaceut.2c00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The effectiveness
of protein and peptide pharmaceuticals depends
essentially on their intrinsic pharmacokinetics. Small-sized pharmaceuticals
in particular often suffer from short serum half-lives due to rapid
renal clearance. To improve the pharmacokinetics by association with
serum albumin (SA) in vivo, we generated an SA-binding
tag of a helix–loop–helix (HLH) peptide to be linked
with protein pharmaceuticals. For use in future preclinical studies,
screening of yeast-displayed HLH peptide libraries against human SA
(HSA) and mouse SA (MSA) was alternately repeated to give the SA-binding
peptide AY-VE, which exhibited cross-binding activities to HSA and
MSA with KD of 65 and 20 nM, respectively.
As a proof of concept, we site-specifically conjugated peptide AY-VE
with insulin to examine its bioactivity in vivo.
In mouse bioassay monitoring the blood glucose level, the AY-VE conjugate
was found to have a prolonged hypoglycemic effect for 12 h. The HLH
peptide tag is a general platform for extending the bioactivity of
therapeutic peptides or proteins.
Collapse
Affiliation(s)
- Yuto Nakatani
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Zhengmao Ye
- Interprotein Corporation, 3-10-2 Toyosaki, Kita-ku, Osaka 531-0072, Japan
| | - Yuki Ishizue
- Graduate School of Pharmaceutical Science, Kumamoto University, 5-1 Oe-Honmachi, Kumamoto 862-0973, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical Science, Kumamoto University, 5-1 Oe-Honmachi, Kumamoto 862-0973, Japan
| | - Teruko Imai
- Graduate School of Pharmaceutical Science, Kumamoto University, 5-1 Oe-Honmachi, Kumamoto 862-0973, Japan
- Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan
| | - Ikuo Fujii
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Masataka Michigami
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
8
|
Benešová M, Guzik P, Deberle LM, Busslinger SD, Landolt T, Schibli R, Müller C. Design and Evaluation of Novel Albumin-Binding Folate Radioconjugates: Systematic Approach of Varying the Linker Entities. Mol Pharm 2022; 19:963-973. [PMID: 35192367 DOI: 10.1021/acs.molpharmaceut.1c00932] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor targeting using folate radioconjugates is a promising strategy for theragnostics of folate receptor-positive tumors. The aim of this study was to investigate the impact of structural modifications of folate radioconjugates on their pharmacokinetic properties. Four novel folate radioconjugates ([177Lu]Lu-OxFol-2, [177Lu]Lu-OxFol-3, [177Lu]Lu-OxFol-4, and [177Lu]Lu-OxFol-5), modified with a lipophilic or hydrophilic linker entity in close proximity to the albumin-binding 4-(p-iodophenyl)butanoate entity or the DOTA chelator, respectively, were designed and evaluated for comparison with the previously developed [177Lu]Lu-OxFol-1. A hydrophobic 4-(aminomethyl)benzoic acid linker, incorporated in close proximity to the 4-(p-iodophenyl)butanoate entity, enhanced the albumin-binding properties (relative affinity 7.3) of [177Lu]Lu-OxFol-3 as compared to those of [177Lu]Lu-OxFol-1 (relative affinity set as 1.0). On the other hand, a hydrophilic d-glutamic acid (d-Glu) linker entity used in [177Lu]Lu-OxFol-2 compromised the albumin-binding properties. [177Lu]Lu-OxFol-4 and [177Lu]Lu-OxFol-5, in which the respective linker entities were incorporated adjacent to the DOTA chelator, showed similar albumin-binding properties (0.6 and 1.0, respectively) as [177Lu]Lu-OxFol-1. Biodistribution studies in KB tumor-bearing nude mice revealed twofold higher tumor-to-kidney ratios at 4 h and 24 h after injection of [177Lu]Lu-OxFol-3 (∼1.2) than after injection of [177Lu]Lu-OxFol-1 (∼0.6). The tumor-to-kidney ratios of [177Lu]Lu-OxFol-2 were, however, much lower (∼0.2) due to the high kidney retention of this radioconjugate. The tumor-to-kidney ratios of [177Lu]Lu-OxFol-5 were only slightly increased (∼0.9), and the ratios for [177Lu]Lu-OxFol-4 (∼0.7) were in the same range as for [177Lu]Lu-OxFol-1. SPECT/CT imaging studies demonstrated similar tumor uptake of all radioconjugates but a clearly improved tumor-to-kidney ratio for [177Lu]Lu-OxFol-3 as compared to that for [177Lu]Lu-OxFol-1. Based on these data, it can be concluded that the linker entity in close proximity to the 4-(p-iodophenyl)butanoate entity affects the radioconjugate's pharmacokinetic profile considerably due to the altered affinity to albumin. Changes in the linker entity, which connects the DOTA chelator with the folate molecule, do not have a major impact on the radioconjugate's tissue distribution profile, however. As a result of these findings, [177Lu]Lu-OxFol-3 had a comparable therapeutic effect to that of [177Lu]Lu-OxFol-1 but appeared advantageous in preventing kidney damage. Provided that the kidneys will present the dose-limiting organs in patients, [177Lu]Lu-OxFol-3 would be the preferred candidate for a clinical translation.
Collapse
Affiliation(s)
- Martina Benešová
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Patrycja Guzik
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Luisa M Deberle
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Tanja Landolt
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| |
Collapse
|
9
|
Xu YD, Tian L, Lai RY, Li Z, Procházková E, Ho J, Stenzel MH. Development of an Albumin–Polymer Bioconjugate via Covalent Conjugation and Supramolecular Interactions. Bioconjug Chem 2022; 33:321-332. [DOI: 10.1021/acs.bioconjchem.1c00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- You Dan Xu
- School of Chemistry, The University of New South Wales, Sydney 2052, NSW, Australia
| | - Linqing Tian
- School of Chemistry, The University of New South Wales, Sydney 2052, NSW, Australia
| | - Rebecca Yong Lai
- School of Chemistry, The University of New South Wales, Sydney 2052, NSW, Australia
| | - Zihao Li
- School of Chemistry, The University of New South Wales, Sydney 2052, NSW, Australia
| | - Eliška Procházková
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Junming Ho
- School of Chemistry, The University of New South Wales, Sydney 2052, NSW, Australia
| | - Martina H. Stenzel
- School of Chemistry, The University of New South Wales, Sydney 2052, NSW, Australia
| |
Collapse
|
10
|
Höltke C, Grewer M, Stölting M, Geyer C, Wildgruber M, Helfen A. Exploring the Influence of Different Albumin Binders on Molecular Imaging Probe Distribution. Mol Pharm 2021; 18:2574-2585. [PMID: 34048242 DOI: 10.1021/acs.molpharmaceut.1c00064] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The biodistribution of molecular imaging probes or tracers mainly depends on the chemical nature of the probe and the preferred metabolization and excretion routes. Small molecules have rather short half-lives while antibodies reside inside the organism for a longer period of time. An excretion via kidneys and bladder is faster than a mainly hepatobiliary elimination. To manipulate the biodistribution behavior of probes, different strategies have been pursued, including utilizing serum albumin as an inherent transport mechanism for small molecules. Here, we modified an existing small molecular fluorescent probe targeted to the endothelin-A receptor (ETAR) with three different albumin-binding moieties to search for an optimal modification strategy. A diphenylcyclohexyl (DPCH) group, a p-iodophenyl butyric acid (IPBA), and a fatty acid (FA) group were attached via amino acid linkers. All three modifications result in transient albumin binding of the developed compounds, as concluded from gel electrophoresis investigations. Spectrophotometric measurements applying variable amounts of bovine, murine, and human serum albumin (BSA, MSA, and HSA) reveal distinct variations of absorption and emission intensities and shifts of their maximum wavelengths. Binding to MSA results in the weakest effects, while binding to HSA leads to the strongest. Cell-based in vitro investigations utilizing ETAR-positive HT-1080 fibrosarcoma and ETAR-negative BT-20 breast adenocarcinoma cells support a retained specific target-binding capacity of the modified compounds and different degrees of unspecific binding. In vivo analysis of a HT-1080 xenograft model in nude mice over the course of 1 week by fluorescence reflectance imaging illustrates noticeable differences between the four examined probes. While the IPBA-modified probe shows the highest absolute signal intensity values, the FA-modified probe exhibits the most favorable tumor-to-organ ratios. In summary, reversible binding to albumin enhances the biological half-life of the designed probes substantially and enables near infrared optical imaging of subcutaneous tumors for several days in vivo. Because the unmodified probe already exhibits reasonable results, the attachment of albumin-binding moieties does not lead to a substantially improved imaging outcome in terms of target-to-background ratios. On the other hand, because the implemented transient albumin binding results in an overall higher amount of probe inside tumor lesions, this strategy might be adaptable for theranostic or therapeutic approaches in a future clinical routine.
Collapse
Affiliation(s)
- Carsten Höltke
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Martin Grewer
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Miriam Stölting
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Christiane Geyer
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Moritz Wildgruber
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany.,Department of Radiology, University Hospital, LMU Munich, 80539 Munich, Germany
| | - Anne Helfen
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| |
Collapse
|
11
|
Kramer V, Fernández R, Lehnert W, Jiménez-Franco LD, Soza-Ried C, Eppard E, Ceballos M, Meckel M, Benešová M, Umbricht CA, Kluge A, Schibli R, Zhernosekov K, Amaral H, Müller C. Biodistribution and dosimetry of a single dose of albumin-binding ligand [ 177Lu]Lu-PSMA-ALB-56 in patients with mCRPC. Eur J Nucl Med Mol Imaging 2021; 48:893-903. [PMID: 32949253 PMCID: PMC8036212 DOI: 10.1007/s00259-020-05022-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION PSMA-targeted radionuclide therapy with lutetium-177 has emerged as an effective treatment option for metastatic, castration-resistant prostate cancer (mCRPC). Recently, the concept of modifying PSMA radioligands with an albumin-binding entity was demonstrated as a promising measure to increase the tumor uptake in preclinical experiments. The aim of this study was to translate the concept to a clinical setting and evaluate the safety and dosimetry of [177Lu]Lu-PSMA-ALB-56, a novel PSMA radioligand with albumin-binding properties. METHODS Ten patients (71.8 ± 8.2 years) with mCRPC received an activity of 3360 ± 393 MBq (120-160 μg) [177Lu]Lu-PSMA-ALB-56 followed by whole-body SPECT/CT imaging over 7 days. Volumes of interest were defined on the SPECT/CT images for dosimetric evaluation for healthy tissue and tumor lesions. General safety and therapeutic efficacy were assessed by measuring blood biomarkers. RESULTS [177Lu]Lu-PSMA-ALB-56 was well tolerated, and no severe adverse events were observed. SPECT images revealed longer circulation of [177Lu]Lu-PSMA-ALB-56 in the blood with the highest uptake in tumor lesions at 48 h post injection. Compared with published data for other therapeutic PSMA radioligands (e.g. PSMA-617 and PSMA I&T), normalized absorbed doses of [177Lu]Lu-PSMA-ALB-56 were up to 2.3-fold higher in tumor lesions (6.64 ± 6.92 Gy/GBq) and similar in salivary glands (0.87 ± 0.43 Gy/GBq). Doses to the kidneys and red marrow (2.54 ± 0.94 Gy/GBq and 0.29 ± 0.07 Gy/GBq, respectively) were increased. CONCLUSION Our data demonstrated that the concept of albumin-binding PSMA-radioligands is feasible and leads to increased tumor doses. After further optimization of the ligand design, the therapeutic outcomes may be improved for patients with prostate cancer.
Collapse
Affiliation(s)
- Vasko Kramer
- Center for Nuclear Medicine & PET/CT Positronmed, Julio Prado 714, 7501068, Providencia, Santiago, Chile.
- Positronpharma SA, 7500921, Providencia, Santiago, Chile.
| | - René Fernández
- Center for Nuclear Medicine & PET/CT Positronmed, Julio Prado 714, 7501068, Providencia, Santiago, Chile
| | - Wencke Lehnert
- ABX-CRO, 01307, Dresden, Germany
- Department of Nuclear Medicine, University Medical Center Hamburg, 20251, Hamburg, Germany
| | | | - Cristian Soza-Ried
- Center for Nuclear Medicine & PET/CT Positronmed, Julio Prado 714, 7501068, Providencia, Santiago, Chile
| | | | - Matias Ceballos
- Center for Nuclear Medicine & PET/CT Positronmed, Julio Prado 714, 7501068, Providencia, Santiago, Chile
| | | | - Martina Benešová
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Christoph A Umbricht
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | | | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | | | - Horacio Amaral
- Center for Nuclear Medicine & PET/CT Positronmed, Julio Prado 714, 7501068, Providencia, Santiago, Chile
- Positronpharma SA, 7500921, Providencia, Santiago, Chile
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| |
Collapse
|
12
|
Preclinical Evaluation of [ 18F]FACH in Healthy Mice and Piglets: An 18F-Labeled Ligand for Imaging of Monocarboxylate Transporters with PET. Int J Mol Sci 2021; 22:ijms22041645. [PMID: 33562048 PMCID: PMC7915902 DOI: 10.3390/ijms22041645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 11/16/2022] Open
Abstract
The expression of monocarboxylate transporters (MCTs) is linked to pathophysiological changes in diseases, including cancer, such that MCTs could potentially serve as diagnostic markers or therapeutic targets. We recently developed [18F]FACH as a radiotracer for non-invasive molecular imaging of MCTs by positron emission tomography (PET). The aim of this study was to evaluate further the specificity, metabolic stability, and pharmacokinetics of [18F]FACH in healthy mice and piglets. We measured the [18F]FACH plasma protein binding fractions in mice and piglets and the specific binding in cryosections of murine kidney and lung. The biodistribution of [18F]FACH was evaluated by tissue sampling ex vivo and by dynamic PET/MRI in vivo, with and without pre-treatment by the MCT inhibitor α-CCA-Na or the reference compound, FACH-Na. Additionally, we performed compartmental modelling of the PET signal in kidney cortex and liver. Saturation binding studies in kidney cortex cryosections indicated a KD of 118 ± 12 nM and Bmax of 6.0 pmol/mg wet weight. The specificity of [18F]FACH uptake in the kidney cortex was confirmed in vivo by reductions in AUC0–60min after pre-treatment with α-CCA-Na in mice (−47%) and in piglets (−66%). [18F]FACH was metabolically stable in mouse, but polar radio-metabolites were present in plasma and tissues of piglets. The [18F]FACH binding potential (BPND) in the kidney cortex was approximately 1.3 in mice. The MCT1 specificity of [18F]FACH uptake was confirmed by displacement studies in 4T1 cells. [18F]FACH has suitable properties for the detection of the MCTs in kidney, and thus has potential as a molecular imaging tool for MCT-related pathologies, which should next be assessed in relevant disease models.
Collapse
|
13
|
Ito S, Senoo A, Nagatoishi S, Ohue M, Yamamoto M, Tsumoto K, Wakui N. Structural Basis for the Binding Mechanism of Human Serum Albumin Complexed with Cyclic Peptide Dalbavancin. J Med Chem 2020; 63:14045-14053. [DOI: 10.1021/acs.jmedchem.0c01578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sho Ito
- ROD (Single Crystal Analysis) Group, Application Laboratories, Rigaku Corporation, 3-9-12 Matsubara-cho, Akishima, Tokyo 196-8666, Japan
- Graduate School of Life Science, University of Hyogo, 3-2-1 Kouto, Kamigori, Ako, Hyogo 678-1297, Japan
| | - Akinobu Senoo
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Satoru Nagatoishi
- Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Masahito Ohue
- Department of Computer Science, School of Computing, Tokyo Institute of Technology, 4259-G3-56 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Masaki Yamamoto
- RIKEN SPring-8 Center, 1-1-1 Kouto, Sayo-cho, Sayo-gun, Hyogo 679-5148, Japan
- Graduate School of Life Science, University of Hyogo, 3-2-1 Kouto, Kamigori, Ako, Hyogo 678-1297, Japan
| | - Kouhei Tsumoto
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Naoki Wakui
- Department of Electrical and Electronic Systems Engineering, National Institute of Technology, Nagaoka College, 888 Nishikatakai, Nagaoka, Niigata 940-8532, Japan
| |
Collapse
|
14
|
Serum albumin: clinical significance of drug binding and development as drug delivery vehicle. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 123:193-218. [PMID: 33485484 DOI: 10.1016/bs.apcsb.2020.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human serum albumin, the primary transport and reservoir protein in the human circulatory system, interacts with numerous endogenous and exogenous ligands of varying structural characteristics. The mode of binding of drugs to albumin is central to understanding their pharmacokinetic profiles and has a major influence on their in vivo efficacy. Altered drug binding to albumin due to drug-drug interactions or abnormal physiology may result in marked changes in the active drug concentration, thus affecting its pharmacokinetic and pharmacodynamic properties. The propensity of drug-drug interaction to be clinically significant as well as possible exploitation of such interactions for therapeutic purposes is reviewed. Being the major organs of albumin metabolism, any impairment in the liver and kidney functions frequently alter the level of serum albumin, which affects the pharmacokinetic profiles of drugs and may have serious clinical implications. The natural function of serum albumin as a drug carrier is facilitated by its interaction with various cellular receptors. These receptors not only promote the uptake of drugs into cells but are also responsible for the extraordinarily long circulatory half-life of albumin. This property in combination with the presence of multiple ligand binding pockets have led to the emergence of serum albumin as an attractive vehicle for novel drug delivery systems. Here, we provide an overview of various albumin-based drug delivery strategies, classified according to their methods of drug attachment, and highlight their experimental and clinical successes.
Collapse
|
15
|
Pellegrino C, Favalli N, Sandholzer M, Volta L, Bassi G, Millul J, Cazzamalli S, Matasci M, Villa A, Myburgh R, Manz MG, Neri D. Impact of Ligand Size and Conjugation Chemistry on the Performance of Universal Chimeric Antigen Receptor T-Cells for Tumor Killing. Bioconjug Chem 2020; 31:1775-1783. [DOI: 10.1021/acs.bioconjchem.0c00258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Christian Pellegrino
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8093 Zurich, Switzerland
| | - Nicholas Favalli
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8093 Zurich, Switzerland
| | - Michael Sandholzer
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8093 Zurich, Switzerland
| | - Laura Volta
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8093 Zurich, Switzerland
| | - Gabriele Bassi
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8093 Zurich, Switzerland
| | - Jacopo Millul
- Philochem AG, Libernstrasse 3, 8112 Otelfingen, Switzerland
| | | | - Mattia Matasci
- Philochem AG, Libernstrasse 3, 8112 Otelfingen, Switzerland
| | | | - Renier Myburgh
- Department of Medical Oncology and Hematology, Comprehensive Cancer Center Zurich (CCCZ), University Hospital Zurich and University of Zürich, 8091 Zürich, Switzerland
| | - Markus G. Manz
- Department of Medical Oncology and Hematology, Comprehensive Cancer Center Zurich (CCCZ), University Hospital Zurich and University of Zürich, 8091 Zürich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), 8093 Zurich, Switzerland
| |
Collapse
|
16
|
Mimoun A, Delignat S, Peyron I, Daventure V, Lecerf M, Dimitrov JD, Kaveri SV, Bayry J, Lacroix-Desmazes S. Relevance of the Materno-Fetal Interface for the Induction of Antigen-Specific Immune Tolerance. Front Immunol 2020; 11:810. [PMID: 32477339 PMCID: PMC7240014 DOI: 10.3389/fimmu.2020.00810] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/08/2020] [Indexed: 12/26/2022] Open
Abstract
In humans, maternal IgGs are transferred to the fetus from the second trimester of pregnancy onwards. The transplacental delivery of maternal IgG is mediated by its binding to the neonatal Fc receptor (FcRn) after endocytosis by the syncytiotrophoblast. IgGs present in the maternal milk are also transferred to the newborn through the digestive epithelium upon binding to the FcRn. Importantly, the binding of IgGs to the FcRn is also responsible for the recycling of circulating IgGs that confers them with a long half-life. Maternally delivered IgG provides passive immunity to the newborn, for instance by conferring protective anti-flu or anti-pertussis toxin IgGs. It may, however, lead to the development of autoimmune manifestations when pathological autoantibodies from the mother cross the placenta and reach the circulation of the fetus. In recent years, strategies that exploit the transplacental delivery of antigen/IgG complexes or of Fc-fused proteins have been validated in mouse models of human diseases to impose antigen-specific tolerance, particularly in the case of Fc-fused factor VIII (FVIII) domains in hemophilia A mice or pre-pro-insulin (PPI) in the case of preclinical models of type 1 diabetes (T1D). The present review summarizes the mechanisms underlying the FcRn-mediated transcytosis of IgGs, the physiopathological relevance of this phenomenon, and the repercussion for drug delivery and shaping of the immune system during its ontogeny.
Collapse
Affiliation(s)
- Angelina Mimoun
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Sandrine Delignat
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Ivan Peyron
- HITh, INSERM, UMR_S1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Victoria Daventure
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Maxime Lecerf
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Jordan D Dimitrov
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Srinivas V Kaveri
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Jagadeesh Bayry
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | | |
Collapse
|
17
|
Cho J, Park J, Kim S, Kim JC, Tae G, Jin MS, Kwon I. Intramolecular distance in the conjugate of urate oxidase and fatty acid governs FcRn binding and serum half-life in vivo. J Control Release 2020; 321:49-58. [DOI: 10.1016/j.jconrel.2020.01.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/18/2020] [Indexed: 12/17/2022]
|
18
|
|
19
|
Albumin nanoparticles as nanocarriers for drug delivery: Focusing on antibody and nanobody delivery and albumin-based drugs. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101471] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
20
|
Sellers DL, Tan JKY, Pineda JMB, Peeler DJ, Porubsky VL, Olden BR, Salipante SJ, Pun SH. Targeting Ligands Deliver Model Drug Cargo into the Central Nervous System along Autonomic Neurons. ACS NANO 2019; 13:10961-10971. [PMID: 31589023 PMCID: PMC7651855 DOI: 10.1021/acsnano.9b01515] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
While biologic drugs such as proteins, peptides, or nucleic acids have shown promise in the treatment of neurodegenerative diseases, the blood-brain barrier (BBB) severely limits drug delivery to the central nervous system (CNS) after systemic administration. Consequently, drug delivery challenges preclude biological drug candidates from the clinical armamentarium. In order to target drug delivery and uptake into to the CNS, we used an in vivo phage display screen to identify peptides able to target drug-uptake by the vast array of neurons of the autonomic nervous system (ANS). Using next-generation sequencing, we identified 21 candidate targeted ANS-to-CNS uptake ligands (TACL) that enriched bacteriophage accumulation and delivered protein-cargo into the CNS after intraperitoneal (IP) administration. The series of TACL peptides were synthesized and tested for their ability to deliver a model enzyme (NeutrAvidin-horseradish peroxidase fusion) to the brain and spinal cord. Three TACL-peptides facilitated significant active enzyme delivery into the CNS, with limited accumulation in off-target organs. Peptide structure and serum stability is increased when internal cysteine residues are cyclized by perfluoroarylation with decafluorobiphenyl, which increased delivery to the CNS further. TACL-peptide was demonstrated to localize in parasympathetic ganglia neurons in addition to neuronal structures in the hindbrain and spinal cord. By targeting uptake into ANS neurons, we demonstrate the potential for TACL-peptides to bypass the blood-brain barrier and deliver a model drug into the brain and spinal cord.
Collapse
Affiliation(s)
- Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - James-Kevin Y. Tan
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Julio Marco B. Pineda
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - David J. Peeler
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Veronica L. Porubsky
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Brynn R. Olden
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington, 98195, USA
| |
Collapse
|
21
|
Lamb J, Fischer E, Rosillo-Lopez M, Salzmann CG, Holland JP. Multi-functionalised graphene nanoflakes as tumour-targeting theranostic drug-delivery vehicles. Chem Sci 2019; 10:8880-8888. [PMID: 32874485 PMCID: PMC7449665 DOI: 10.1039/c9sc03736e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 12/04/2022] Open
Abstract
Graphene nanoflakes (GNFs) consist of a graphene sheet approximately 30 nm in diameter with a pristine aromatic system and an edge terminated with carboxylic acid groups. Their high water solubility and relative ease of functionalisation using carboxylate chemistry means that GNFs are potential scaffolds for the synthesis of theranostic agents. In this work, GNFs were multi-functionalised with derivatives of (i) a peptide-based Glu-NH-C(O)-NH-Lys ligand that binds prostate-specific membrane antigen (PSMA), (ii) a potent anti-mitotic drug (R)-ispinesib, (iii) the chelate desferrioxamine B (DFO), and (iv) an albumin-binding tag reported to extend pharmacokinetic half-life in vivo. Subsequent 68Ga radiochemistry and experiments in vitro and in vivo were used to evaluate the performance of GNFs in theranostic drug design. Efficient 68Ga-radiolabelling was achieved and the particle-loading of (R)-ispinesib and Glu-NH-C(O)-NH-Lys was confirmed using cellular assays. Using dose-response curves and FACS analysis it was shown that GNFs loaded with (R)-ispinesib inhibited the kinesin spindle protein (KSP) and induced G2/M-phase cell cycle arrest. Cellular uptake and blocking experiments demonstrated that GNFs functionalised with the Glu-NH-C(O)-NH-Lys ligand showed specificity toward PSMA expressing cells (LNCaP). The distribution profile and excretion rates of 68Ga-radiolabelled GNFs in athymic nude mice was evaluated using time-activity curves derived from dynamic positron-emission tomography (PET). Image analysis indicated that GNFs have low accumulation and retention in background tissue, with rapid renal clearance. In summary, our study shows that GNFs are suitable candidates for use in theranostic drug design.
Collapse
Affiliation(s)
- Jennifer Lamb
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zurich , Switzerland . ; http://www.hollandlab.org ; Tel: +41 44 63 53 990
| | - Eliane Fischer
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zurich , Switzerland . ; http://www.hollandlab.org ; Tel: +41 44 63 53 990
| | - Martin Rosillo-Lopez
- Department of Chemistry , University College London , 20 Gordon Street , London WC1H 0AJ , UK
| | - Christoph G Salzmann
- Department of Chemistry , University College London , 20 Gordon Street , London WC1H 0AJ , UK
| | - Jason P Holland
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zurich , Switzerland . ; http://www.hollandlab.org ; Tel: +41 44 63 53 990
| |
Collapse
|
22
|
Zorzi A, Linciano S, Angelini A. Non-covalent albumin-binding ligands for extending the circulating half-life of small biotherapeutics. MEDCHEMCOMM 2019; 10:1068-1081. [PMID: 31391879 PMCID: PMC6644573 DOI: 10.1039/c9md00018f] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 06/01/2019] [Indexed: 12/13/2022]
Abstract
Peptides and small protein scaffolds are gaining increasing interest as therapeutics. Similarly to full-length antibodies, they can bind a target with a high binding affinity and specificity while remaining small enough to diffuse into tissues. However, despite their numerous advantages, small biotherapeutics often suffer from a relatively short circulating half-life, thus requiring frequent applications that ultimately restrict their ease of use and user compliance. To overcome this limitation, a large variety of half-life extension strategies have been developed in the last decades. Linkage to ligands that non-covalently bind to albumin, the most abundant serum protein with a circulating half-life of ∼19 days in humans, represents one of the most successful approaches for the generation of long-lasting biotherapeutics with improved pharmacokinetic properties and superior efficacy in the clinic.
Collapse
Affiliation(s)
- Alessandro Zorzi
- Institute of Chemical Sciences and Engineering , School of Basic Sciences , Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne CH-1015 , Switzerland
| | - Sara Linciano
- Department of Molecular Sciences and Nanosystems , Ca' Foscari University of Venice , Via Torino 155 , Venezia Mestre 30172 , Italy
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems , Ca' Foscari University of Venice , Via Torino 155 , Venezia Mestre 30172 , Italy
- European Centre for Living Technologies (ECLT) , San Marco 2940 , Venice 30124 , Italy .
| |
Collapse
|
23
|
Hoogenboezem EN, Duvall CL. Harnessing albumin as a carrier for cancer therapies. Adv Drug Deliv Rev 2018; 130:73-89. [PMID: 30012492 PMCID: PMC6200408 DOI: 10.1016/j.addr.2018.07.011] [Citation(s) in RCA: 368] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Serum albumin, a natural ligand carrier that is highly concentrated and long-circulating in the blood, has shown remarkable promise as a carrier for anti-cancer agents. Albumin is able to prolong the circulation half-life of otherwise rapidly cleared drugs and, importantly, promote their accumulation within tumors. The applications for using albumin as a cancer drug carrier are broad and include both traditional cancer chemotherapeutics and new classes of biologics. Strategies for leveraging albumin for drug delivery can be classified broadly into exogenous and in situ binding formulations that utilize covalent attachment, non-covalent association, or encapsulation in albumin-based nanoparticles. These methods have shown remarkable preclinical and clinical successes that are examined in this review.
Collapse
Affiliation(s)
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN.
| |
Collapse
|
24
|
Zvonova EA, Tyurin AA, Soloviev AA, Goldenkova-Pavlova IV. Strategies for Modulation of Pharmacokinetics of Recombinant Therapeutic Proteins. ACTA ACUST UNITED AC 2018. [DOI: 10.1134/s2079086418020093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
Improving long-term subcutaneous drug delivery by regulating material-bioenvironment interaction. Adv Drug Deliv Rev 2018; 127:20-34. [PMID: 29391221 DOI: 10.1016/j.addr.2018.01.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 02/08/2023]
Abstract
Subcutaneous long-acting release (LAR) formulations have been extensively developed in the clinic to increase patient compliance and reduce treatment cost. Despite preliminary success for some LAR systems, a major obstacle limiting the therapeutic effect remains on their interaction with surrounding tissues. In this review, we summarize how living bodies respond to injected or implanted materials, and highlight some typical strategies based on smart material design, which may significantly improve long-term subcutaneous drug delivery. Moreover, possible strategies to achieve ultra-long (months, years) subcutaneous drug delivery systems are proposed. Based on these discussions, we believe the well-designed subcutaneous long-acting formulations will hold great promise to improve patient quality of life in the clinic.
Collapse
|
26
|
Che Nordin MA, Teow SY. Review of Current Cell-Penetrating Antibody Developments for HIV-1 Therapy. Molecules 2018; 23:molecules23020335. [PMID: 29415435 PMCID: PMC6017373 DOI: 10.3390/molecules23020335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/06/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022] Open
Abstract
The discovery of highly active antiretroviral therapy (HAART) in 1996 has significantly reduced the global mortality and morbidity caused by the acquired immunodeficiency syndrome (AIDS). However, the therapeutic strategy of HAART that targets multiple viral proteins may render off-target toxicity and more importantly results in drug-resistant escape mutants. These have been the main challenges for HAART and refinement of this therapeutic strategy is urgently needed. Antibody-mediated treatments are emerging therapeutic modalities for various diseases. Most therapeutic antibodies have been approved by Food and Drug Administration (FDA) mainly for targeting cancers. Previous studies have also demonstrated the promising effect of therapeutic antibodies against HIV-1, but there are several limitations in this therapy, particularly when the viral targets are intracellular proteins. The conventional antibodies do not cross the cell membrane, hence, the pathogenic intracellular proteins cannot be targeted with this classical therapeutic approach. Over the years, the advancement of antibody engineering has permitted the therapeutic antibodies to comprehensively target both extra- and intra-cellular proteins in various infections and diseases. This review aims to update on the current progress in the development of antibody-based treatment against intracellular targets in HIV-1 infection. We also attempt to highlight the challenges and limitations in the development of antibody-based therapeutic modalities against HIV-1.
Collapse
Affiliation(s)
- Muhamad Alif Che Nordin
- Kulliyyah of Medicine and Health Sciences (KMHS), Kolej Universiti INSANIAH, 09300 Kuala Ketil, Kedah, Malaysia.
| | - Sin-Yeang Teow
- Sunway Institute for Healthcare Development (SIHD), School of Healthcare and Medical Sciences (SHMS), Sunway University, Jalan Universiti, Bandar Sunway, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
27
|
Benešová M, Umbricht CA, Schibli R, Müller C. Albumin-Binding PSMA Ligands: Optimization of the Tissue Distribution Profile. Mol Pharm 2018; 15:934-946. [PMID: 29400475 DOI: 10.1021/acs.molpharmaceut.7b00877] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The prostate-specific membrane antigen (PSMA) has emerged as an attractive prostate cancer associated target for radiotheragnostic application using PSMA-specific radioligands. The aim of this study was to design new PSMA ligands modified with an albumin-binding moiety in order to optimize their tissue distribution profile. The compounds were prepared by conjugation of a urea-based PSMA-binding entity, a DOTA chelator, and 4-( p-iodophenyl)butyric acid using multistep solid phase synthesis. The three ligands (PSMA-ALB-02, PSMA-ALB-05, and PSMA-ALB-07) were designed with varying linker entities. Radiolabeling with 177Lu was performed at a specific activity of up to 50 MBq/nmol resulting in radioligands of >98% radiochemical purity and high stability. In vitro investigations revealed high binding of all three PSMA radioligands to mouse (>64%) and human plasma proteins (>94%). Uptake and internalization into PSMA-positive PC-3 PIP tumor cells was equally high for all radioligands. Negligible accumulation was found in PSMA-negative PC-3 flu cells, indicating PSMA-specific binding of all radioligands. Biodistribution and imaging studies performed in PC-3 PIP/flu tumor-bearing mice showed enhanced blood circulation of the new radioligands when compared to the clinically employed 177Lu-PSMA-617. The PC-3 PIP tumor uptake of all three radioligands was very high (76.4 ± 2.5% IA/g, 79.4 ± 11.1% IA/g, and 84.6 ± 14.2% IA/g, respectively) at 24 h post injection (p.i.) resulting in tumor-to-blood ratios of ∼176, ∼48, and ∼107, respectively, whereas uptake into PC-3 flu tumors was negligible. Kidney uptake at 24 h p.i. was lowest for 177Lu-PSMA-ALB-02 (10.7 ± 0.92% IA/g), while 177Lu-PSMA-ALB-05 and 177Lu-PSMA-ALB-07 showed higher renal retention (23.9 ± 4.02% IA/g and 51.9 ± 6.34% IA/g, respectively). Tumor-to-background ratios calculated from values of the area under the curve (AUC) of time-dependent biodistribution data were in favor of 177Lu-PSMA-ALB-02 (tumor-to-blood, 46; tumor-to-kidney, 5.9) when compared to 177Lu-PSMA-ALB-05 (17 and 3.7, respectively) and 177Lu-PSMA-ALB-07 (39 and 2.1, respectively). The high accumulation of the radioligands in PC-3 PIP tumors was visualized on SPECT/CT images demonstrating increasing tumor-to-kidney ratios over time. Taking all of the characteristics into account, 177Lu-PSMA-ALB-02 emerged as the most promising candidate. The applied concept may be attractive for future clinical translation potentially enabling more potent and convenient prostate cancer radionuclide therapy.
Collapse
Affiliation(s)
- Martina Benešová
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ , Paul Scherrer Institut , 5232 Villigen-PSI , Switzerland.,Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 Zurich , Switzerland
| | - Christoph A Umbricht
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ , Paul Scherrer Institut , 5232 Villigen-PSI , Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ , Paul Scherrer Institut , 5232 Villigen-PSI , Switzerland.,Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 Zurich , Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ , Paul Scherrer Institut , 5232 Villigen-PSI , Switzerland.,Department of Chemistry and Applied Biosciences , ETH Zurich , 8093 Zurich , Switzerland
| |
Collapse
|
28
|
|
29
|
Müller C, Farkas R, Borgna F, Schmid RM, Benešová M, Schibli R. Synthesis, Radiolabeling, and Characterization of Plasma Protein-Binding Ligands: Potential Tools for Modulation of the Pharmacokinetic Properties of (Radio)Pharmaceuticals. Bioconjug Chem 2017; 28:2372-2383. [DOI: 10.1021/acs.bioconjchem.7b00378] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Cristina Müller
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
- Department
of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg
1-5/10, 8093 Zurich, Switzerland
| | - Renáta Farkas
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
| | - Francesca Borgna
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Marzolo 5, 35131 Padova, Italy
| | - Raffaella M. Schmid
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
| | - Martina Benešová
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
- Department
of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg
1-5/10, 8093 Zurich, Switzerland
| | - Roger Schibli
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
- Department
of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg
1-5/10, 8093 Zurich, Switzerland
| |
Collapse
|
30
|
Choy CJ, Ling X, Geruntho JJ, Beyer SK, Latoche JD, Langton-Webster B, Anderson CJ, Berkman CE. 177Lu-Labeled Phosphoramidate-Based PSMA Inhibitors: The Effect of an Albumin Binder on Biodistribution and Therapeutic Efficacy in Prostate Tumor-Bearing Mice. Am J Cancer Res 2017; 7:1928-1939. [PMID: 28638478 PMCID: PMC5479279 DOI: 10.7150/thno.18719] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/06/2017] [Indexed: 02/07/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) continues to be an active biomarker for small-molecule PSMA-targeted imaging and therapeutic agents for prostate cancer and various non-prostatic tumors that are characterized by PSMA expression on their neovasculature. One of the challenges for small-molecule PSMA inhibitors with respect to delivering therapeutic payloads is their rapid renal clearance. In order to overcome this pharmacokinetic challenge, we outfitted a 177Lu-labeled phosphoramidate-based PSMA inhibitor (CTT1298) with an albumin-binding motif (CTT1403) and compared its in vivo performance with that of an analogous compound lacking the albumin-binding motif (CTT1401). The radiolabeling of CTT1401 and CTT1403 was achieved using click chemistry to connect 177Lu-DOTA-N3 to the dibenzocyclooctyne (DBCO)-bearing CTT1298 inhibitor cores. A direct comparison in vitro and in vivo performance was made for CTT1401 and CTT1403; the specificity and efficacy by means of cellular uptake and internalization, biodistribution, and therapeutic efficacy were determined for both compounds. While both compounds displayed excellent uptake and rapid internalization in PSMA+ PC3-PIP cells, the albumin binding moiety in CTT1403 conferred clear advantages to the PSMA-inhibitor scaffold including increased circulating half-life and prostate tumor uptake that continued to increase up to 168 h post-injection. This increased tumor uptake translated into superior therapeutic efficacy of CTT1403 in PSMA+ PC3-PIP human xenograft tumors.
Collapse
|
31
|
Siwowska K, Haller S, Bortoli F, Benešová M, Groehn V, Bernhardt P, Schibli R, Müller C. Preclinical Comparison of Albumin-Binding Radiofolates: Impact of Linker Entities on the in Vitro and in Vivo Properties. Mol Pharm 2017; 14:523-532. [DOI: 10.1021/acs.molpharmaceut.6b01010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Klaudia Siwowska
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
| | - Stephanie Haller
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
| | - Francesca Bortoli
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
| | - Martina Benešová
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
- Department
of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Viola Groehn
- Merck & Cie Schaffhausen, 8200 Schaffhausen, Switzerland
| | - Peter Bernhardt
- Department
of Radiation Physics, The Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
- Department
of Medical Physics and Medical Bioengineering, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| | - Roger Schibli
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
- Department
of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Cristina Müller
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen-PSI, Switzerland
- Department
of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
32
|
Chen H, Jacobson O, Niu G, Weiss ID, Kiesewetter DO, Liu Y, Ma Y, Wu H, Chen X. Novel "Add-On" Molecule Based on Evans Blue Confers Superior Pharmacokinetics and Transforms Drugs to Theranostic Agents. J Nucl Med 2016; 58:590-597. [PMID: 27879373 DOI: 10.2967/jnumed.116.182097] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/09/2016] [Indexed: 12/14/2022] Open
Abstract
One of the major design considerations for a drug is its pharmacokinetics in the blood. A drug with a short half-life in the blood is less available at a target organ. Such a limitation dictates treatment with either high doses or more frequent doses, both of which may increase the likelihood of undesirable side effects. To address the need for additional methods to improve the blood half-life of drugs and molecular imaging agents, we developed an "add-on" molecule that contains 3 groups: a truncated Evans blue dye molecule that binds to albumin with a low micromolar affinity and provides a prolonged half-life in the blood; a metal chelate that allows radiolabeling for imaging and radiotherapy; and maleimide for easy conjugation to drug molecules. Methods: The truncated Evans blue molecule was conjugated with the chelator NOTA or DOTA, and the resulting conjugate was denoted as NMEB or DMEB, respectively. As a proof of concept, we coupled NMEB and DMEB to c(RGDfK), which is a small cyclic arginine-glycine-aspartic acid (RGD) peptide, for targeting integrin αvβ3 NMEB and DMEB were radiolabeled with 64Cu and 90Y, respectively, and tested in xenograft models. Results: The resulting radiolabeled conjugates showed a prolonged circulation half-life and enhanced tumor accumulation in integrin αvβ3-expressing tumors. Tumor uptake was markedly improved over that with NOTA- or DOTA-conjugated c(RGDfK). Tumor radiotherapy experiments in mice with 90Y-DMEB-RGD showed promising results; existing tumors were eliminated. Conclusion: Conjugation of our novel add-on molecule, NMEB or DMEB, to potential tracers or therapeutic agents improved blood half-life and tumor uptake and could transform such agents into theranostic entities.
Collapse
Affiliation(s)
- Haojun Chen
- Department of Nuclear Medicine, Xiamen Cancer Hospital of the First Affiliated Hospital of Xiamen University, Xiamen, China.,Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Ido D Weiss
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Dale O Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Yi Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Ying Ma
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Hua Wu
- Department of Nuclear Medicine, Xiamen Cancer Hospital of the First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
33
|
Davé E, Adams R, Zaccheo O, Carrington B, Compson JE, Dugdale S, Airey M, Malcolm S, Hailu H, Wild G, Turner A, Heads J, Sarkar K, Ventom A, Marshall D, Jairaj M, Kopotsha T, Christodoulou L, Zamacona M, Lawson AD, Heywood S, Humphreys DP. Fab-dsFv: A bispecific antibody format with extended serum half-life through albumin binding. MAbs 2016; 8:1319-1335. [PMID: 27532598 PMCID: PMC5058625 DOI: 10.1080/19420862.2016.1210747] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/19/2016] [Accepted: 07/05/2016] [Indexed: 11/24/2022] Open
Abstract
An antibody format, termed Fab-dsFv, has been designed for clinical indications that require monovalent target binding in the absence of direct Fc receptor (FcR) binding while retaining substantial serum presence. The variable fragment (Fv) domain of a humanized albumin-binding antibody was fused to the C-termini of Fab constant domains, such that the VL and VH domains were individually connected to the Cκ and CH1 domains by peptide linkers, respectively. The anti-albumin Fv was selected for properties thought to be desirable to ensure a durable serum half-life mediated via FcRn. The Fv domain was further stabilized by an inter-domain disulfide bond. The bispecific format was shown to be thermodynamically and biophysically stable, and retained good affinity and efficacy to both antigens simultaneously. In in vivo studies, the serum half-life of Fab-dsFv, 2.6 d in mice and 7.9 d in cynomolgus monkeys, was equivalent to Fab'-PEG.
Collapse
|
34
|
Adams R, Griffin L, Compson JE, Jairaj M, Baker T, Ceska T, West S, Zaccheo O, Davé E, Lawson AD, Humphreys DP, Heywood S. Extending the half-life of a fab fragment through generation of a humanized anti-human serum albumin Fv domain: An investigation into the correlation between affinity and serum half-life. MAbs 2016; 8:1336-1346. [PMID: 27315033 PMCID: PMC5058626 DOI: 10.1080/19420862.2016.1185581] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We generated an anti-albumin antibody, CA645, to link its Fv domain to an antigen-binding fragment (Fab), thereby extending the serum half-life of the Fab. CA645 was demonstrated to bind human, cynomolgus, and mouse serum albumin with similar affinity (1–7 nM), and to bind human serum albumin (HSA) when it is in complex with common known ligands. Importantly for half-life extension, CA645 binds HSA with similar affinity within the physiologically relevant range of pH 5.0 – pH 7.4, and does not have a deleterious effect on the binding of HSA to neonatal Fc receptor (FcRn). A crystal structure of humanized CA645 Fab in complex with HSA was solved and showed that CA645 Fab binds to domain II of HSA. Superimposition with the crystal structure of FcRn bound to HSA confirmed that CA645 does not block HSA binding to FcRn. In mice, the serum half-life of humanized CA645 Fab is 84.2 h. This is a significant extension in comparison with < 1 h for a non-HSA binding CA645 Fab variant. The Fab-HSA structure was used to design a series of mutants with reduced affinity to investigate the correlation between the affinity for albumin and serum half-life. Reduction in the affinity for MSA by 144-fold from 2.2 nM to 316 nM had no effect on serum half-life. Strikingly, despite a reduction in affinity to 62 µM, an extension in serum half-life of 26.4 h was still obtained. CA645 Fab and the CA645 Fab-HSA complex have been deposited in the Protein Data Bank (PDB) with accession codes, 5FUZ and 5FUO, respectively.
Collapse
Affiliation(s)
| | - Laura Griffin
- b Ashfield Healthcare Communications , Macclesfield , Cheshire , UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gunnoo SB, Madder A. Chemical Protein Modification through Cysteine. Chembiochem 2016; 17:529-53. [DOI: 10.1002/cbic.201500667] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Smita B. Gunnoo
- Organic & Biomimetic Chemistry Research Group; Department of Organic and Macromolecular Chemistry; Ghent University; Krijgslaan 281 9000 Gent Belgium
| | - Annemieke Madder
- Organic & Biomimetic Chemistry Research Group; Department of Organic and Macromolecular Chemistry; Ghent University; Krijgslaan 281 9000 Gent Belgium
| |
Collapse
|
36
|
Gunnoo SB, Madder A. Bioconjugation – using selective chemistry to enhance the properties of proteins and peptides as therapeutics and carriers. Org Biomol Chem 2016; 14:8002-13. [DOI: 10.1039/c6ob00808a] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Both peptide and protein therapeutics are becoming increasingly important for treating a wide range of diseases. Functionalisation of theseviasite-selective chemical modification leads to enhancement of their therapeutic properties.
Collapse
Affiliation(s)
- Smita B. Gunnoo
- Organic and Biomimetic Chemistry Research Group
- Department of Organic and Macromolecular Chemistry
- Ghent University
- Ghent
- Belgium
| | - Annemieke Madder
- Organic and Biomimetic Chemistry Research Group
- Department of Organic and Macromolecular Chemistry
- Ghent University
- Ghent
- Belgium
| |
Collapse
|
37
|
Dozier JK, Distefano MD. Site-Specific PEGylation of Therapeutic Proteins. Int J Mol Sci 2015; 16:25831-64. [PMID: 26516849 PMCID: PMC4632829 DOI: 10.3390/ijms161025831] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/11/2022] Open
Abstract
The use of proteins as therapeutics has a long history and is becoming ever more common in modern medicine. While the number of protein-based drugs is growing every year, significant problems still remain with their use. Among these problems are rapid degradation and excretion from patients, thus requiring frequent dosing, which in turn increases the chances for an immunological response as well as increasing the cost of therapy. One of the main strategies to alleviate these problems is to link a polyethylene glycol (PEG) group to the protein of interest. This process, called PEGylation, has grown dramatically in recent years resulting in several approved drugs. Installing a single PEG chain at a defined site in a protein is challenging. Recently, there is has been considerable research into various methods for the site-specific PEGylation of proteins. This review seeks to summarize that work and provide background and context for how site-specific PEGylation is performed. After introducing the topic of site-specific PEGylation, recent developments using chemical methods are described. That is followed by a more extensive discussion of bioorthogonal reactions and enzymatic labeling.
Collapse
Affiliation(s)
- Jonathan K Dozier
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
38
|
A biomimetic approach for enhancing the in vivo half-life of peptides. Nat Chem Biol 2015; 11:793-8. [PMID: 26344696 PMCID: PMC4575266 DOI: 10.1038/nchembio.1907] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/07/2015] [Indexed: 02/03/2023]
Abstract
The tremendous therapeutic potential of peptides has not yet been realized, mainly due to their short in vivo half-life. While conjugation to macromolecules has been a mainstay approach for enhancing the half-life of proteins, the steric hindrance of macromolecules often harms the binding of peptides to target receptors, compromising the in vivo efficacy. Here we report a new strategy for enhancing the in vivo half-life of peptides without compromising their potency. Our approach involves endowing peptides with a small-molecule that binds reversibly to the serum protein, transthyretin. Although there are few reversible albumin-binding molecules, we are unaware of designed small molecules that bind reversibly to other serum proteins and are used for half-life extension in vivo. We show here that our strategy was indeed effective in enhancing the half-life of an agonist for GnRH receptor while maintaining its binding affinity, which was translated into superior in vivo efficacy.
Collapse
|
39
|
Abstract
PEGylation is the covalent conjugation of PEG to therapeutic molecules. Protein PEGylation is a clinically proven approach for extending the circulation half-life and reducing the immunogenicity of protein therapeutics. Most clinically used PEGylated proteins are heterogeneous mixtures of PEG positional isomers conjugated to different residues on the protein main chain. Current research is focused to reduce product heterogeneity and to preserve bioactivity. Recent advances and possible future directions in PEGylation are described in this review. So far protein PEGylation has yielded more than 10 marketed products and in view of the lack of equally successful alternatives to extend the circulation half-life of proteins, PEGylation will still play a major role in drug delivery for many years to come.
Collapse
|
40
|
The role of albumin receptors in regulation of albumin homeostasis: Implications for drug delivery. J Control Release 2015; 211:144-62. [PMID: 26055641 DOI: 10.1016/j.jconrel.2015.06.006] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/02/2015] [Accepted: 06/04/2015] [Indexed: 12/20/2022]
Abstract
Albumin is the most abundant protein in blood and acts as a molecular taxi for a plethora of small insoluble substances such as nutrients, hormones, metals and toxins. In addition, it binds a range of medical drugs. It has an unusually long serum half-life of almost 3weeks, and although the structure and function of albumin has been studied for decades, a biological explanation for the long half-life has been lacking. Now, recent research has unravelled that albumin-binding cellular receptors play key roles in the homeostatic regulation of albumin. Here, we review our current understanding of albumin homeostasis with a particular focus on the impact of the cellular receptors, namely the neonatal Fc receptor (FcRn) and the cubilin-megalin complex, and we discuss their importance on uses of albumin in drug delivery.
Collapse
|
41
|
Lim SI, Kwon I. Bioconjugation of therapeutic proteins and enzymes using the expanded set of genetically encoded amino acids. Crit Rev Biotechnol 2015; 36:803-15. [DOI: 10.3109/07388551.2015.1048504] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sung In Lim
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, USA and
| | - Inchan Kwon
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, USA and
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| |
Collapse
|
42
|
Scheuermann J, Neri D. Dual-pharmacophore DNA-encoded chemical libraries. Curr Opin Chem Biol 2015; 26:99-103. [DOI: 10.1016/j.cbpa.2015.02.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/12/2015] [Accepted: 02/19/2015] [Indexed: 12/20/2022]
|
43
|
Sand KMK, Bern M, Nilsen J, Noordzij HT, Sandlie I, Andersen JT. Unraveling the Interaction between FcRn and Albumin: Opportunities for Design of Albumin-Based Therapeutics. Front Immunol 2015; 5:682. [PMID: 25674083 PMCID: PMC4306297 DOI: 10.3389/fimmu.2014.00682] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/17/2014] [Indexed: 01/08/2023] Open
Abstract
The neonatal Fc receptor (FcRn) was first found to be responsible for transporting antibodies of the immunoglobulin G (IgG) class from the mother to the fetus or neonate as well as for protecting IgG from intracellular catabolism. However, it has now become apparent that the same receptor also binds albumin and plays a fundamental role in homeostatic regulation of both IgG and albumin, as FcRn is expressed in many different cell types and organs at diverse body sites. Thus, to gain a complete understanding of the biological function of each ligand, and also their distribution in the body, an in-depth characterization of how FcRn binds and regulates the transport of both ligands is necessary. Importantly, such knowledge is also relevant when developing new drugs, as IgG and albumin are increasingly utilized in therapy. This review discusses our current structural and biological understanding of the relationship between FcRn and its ligands, with a particular focus on albumin and design of albumin-based therapeutics.
Collapse
Affiliation(s)
- Kine Marita Knudsen Sand
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Malin Bern
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Jeannette Nilsen
- Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway ; Institute of Clinical Medicine, University of Oslo , Oslo , Norway
| | - Hanna Theodora Noordzij
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Jan Terje Andersen
- Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| |
Collapse
|
44
|
|
45
|
|
46
|
Pollaro L, Raghunathan S, Morales-Sanfrutos J, Angelini A, Kontos S, Heinis C. Bicyclic Peptides Conjugated to an Albumin-Binding Tag Diffuse Efficiently into Solid Tumors. Mol Cancer Ther 2014; 14:151-61. [DOI: 10.1158/1535-7163.mct-14-0534] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Farajnia S, Ahmadzadeh V, Tanomand A, Veisi K, Khosroshahi SA, Rahbarnia L. Development trends for generation of single-chain antibody fragments. Immunopharmacol Immunotoxicol 2014; 36:297-308. [DOI: 10.3109/08923973.2014.945126] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
48
|
Improved PET imaging of tumors in mice using a novel (18) F-folate conjugate with an albumin-binding entity. Mol Imaging Biol 2014; 15:649-54. [PMID: 23760583 DOI: 10.1007/s11307-013-0651-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE The folate receptor (FR) is a promising target for nuclear imaging due to its overexpression in many different cancer types. A drawback of using folate radioconjugates is the high accumulation of radioactivity in the kidneys. Therefore, the aim of this study was to develop a (18) F-labeled folate conjugate with an albumin-binding entity to enhance the blood circulation time and hence improve the tumor-to-kidney ratio. PROCEDURES The novel (18) F-folate was prepared by conjugation of a (18) F-labeled glucose azide to an alkyne-functionalized folate precursor containing an albumin-binding entity via Cu(I)-catalyzed 1,3-dipolar cycloaddition. The radioconjugate was tested in vitro on FR-positive KB tumor cells and by biodistribution and positron emission tomography (PET) imaging studies using KB tumor-bearing mice. RESULTS The radiosynthesis of the albumin-binding [(18) F]fluorodeoxyglucose-folate ([(18) F]3) resulted in a radiochemical yield of 1-2 % decay corrected (d.c.) and a radiochemical purity of ≥95 %. The specific activity of [(18) F]3 ranged from 20 to 50 GBq/μmol. In vitro experiments revealed FR-specific binding of [(18) F]3 to KB tumor cells. In vivo we found an increasing uptake of [(18) F]3 into tumor xenografts over time reaching a value of ∼ 15 % injected dose (ID)/g at 4 h post-injection (p.i.). Uptake in the kidneys (∼ 13 % ID/g; 1 h p.i.) was approximately fourfold reduced compared to previously published (18) F-labeled folic acid derivatives. An excellent visualization of tumor xenografts with an unprecedentedly high tumor-to-kidney ratio (∼ 1) was obtained by PET imaging. CONCLUSIONS [(18) F]3 showed a favorable accumulation in tumor xenografts compared to the same folate conjugate without albumin-binding properties. Moreover, the increased tumor-to-kidney ratios improved the PET imaging quality significantly, in spite of a somewhat higher background radioactivity which was a consequence of the slower blood clearance of [(18) F]3.
Collapse
|
49
|
Morais M, Cantante C, Gano L, Santos I, Lourenço S, Santos C, Fontes C, Aires da Silva F, Gonçalves J, Correia JD. Biodistribution of a 67Ga-labeled anti-TNF VHH single-domain antibody containing a bacterial albumin-binding domain (Zag). Nucl Med Biol 2014; 41 Suppl:e44-8. [DOI: 10.1016/j.nucmedbio.2014.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 01/03/2014] [Accepted: 01/10/2014] [Indexed: 11/30/2022]
|
50
|
Sadavarte R, Spearman M, Okun N, Butler M, Ghosh R. Purification of chimeric heavy chain monoclonal antibody EG2-hFc using hydrophobic interaction membrane chromatography: an alternative to protein-A affinity chromatography. Biotechnol Bioeng 2014; 111:1139-49. [PMID: 24449405 DOI: 10.1002/bit.25193] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 01/13/2014] [Accepted: 01/15/2014] [Indexed: 01/11/2023]
Abstract
Heavy chain monoclonal antibodies are being considered as alternative to whole-IgG monoclonal antibodies for certain niche applications. Protein-A chromatography which is widely used for purifying IgG monoclonal antibodies is also used for purifying heavy chain monoclonal antibodies as these molecules possess fully functional Fc regions. However, the acidic conditions used to elute bound antibody may sometimes also leach protein-A, which is immunotoxic. Low pH conditions also tend to make the mAb molecules unstable and prone to aggregation. Moreover, protein-A affinity chromatography does not remove aggregates already present in the feed. Hydrophobic interaction membrane chromatography (or HIMC) has already been studied as an alternative to protein-A chromatography for purifying whole-IgG monoclonal antibodies. This paper describes the use of HIMC for capturing a humanized chimeric heavy chain monoclonal antibody (EG2-hFC). Binding and eluting conditions were suitably optimized using pure EG2-hFC. Based on this, an HIMC method was developed for capture of EG2-hFC directly from cell culture supernatant. The EG2-hFc purity obtained in this single-step process was high. The glycan profiles of protein-A and HIMC purified monoclonal antibody samples were similar, clearly demonstrating that both techniques captured similarly glycosylated population of EG2-hFc. Moreover, this technique was able to resolve aggregates from monomeric form of the EG2-hFc.
Collapse
Affiliation(s)
- Rahul Sadavarte
- Department of Chemical Engineering, McMaster University, Hamilton, ON, L8S 4L7, Canada
| | | | | | | | | |
Collapse
|