1
|
Heidarnejad K, Nooreddin Faraji S, Mahfoozi S, Ghasemi Z, Sadat Dashti F, Asadi M, Ramezani A. Breast cancer immunotherapy using scFv antibody-based approaches, a systematic review. Hum Immunol 2024; 85:111090. [PMID: 39214066 DOI: 10.1016/j.humimm.2024.111090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer is considered as the most common malignancy in women and the second leading cause of death related to cancer. Recombinant DNA technologies accelerated the development of antibody-based cancer therapy, which is effective in a broad range of cancers. The objective of the present study was to perform a systematic review on breast cancer immunotherapy using single-chain fragment variable (scFv) antibody formats. Searches were performed up to March 2023 using PubMed, Scopus, and Web of Science (ISI) databases. Three reviewers independently assessed study eligibility, data extraction, and evaluated the methodological quality of included primary studies. Different immunotherapy approaches have been identified and the most common approaches were scFv-conjugates, followed by simple scFvs and chimeric antigen receptor (CAR) therapy, respectively. Among breast cancer antigens, HER superfamily, CD family, and EpCAM were applied as the most important breast cancer immunotherapy targets. The present study shed more lights on scFv-based breast cancer immunotherapy approaches.
Collapse
Affiliation(s)
- Kamran Heidarnejad
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Nooreddin Faraji
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Shirin Mahfoozi
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Ghasemi
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fateme Sadat Dashti
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maryam Asadi
- School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Michalaki E, Chin R, Jeong K, Qi Z, Liebman LN, González-Vargas Y, Echeverri ES, Paunovska K, Muramatsu H, Pardi N, Tamburini BJ, Jakus Z, Dahlman JE, Dixon JB. Lymphatic endothelial cell-targeting lipid nanoparticles delivering VEGFC mRNA improve lymphatic function after injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605343. [PMID: 39131391 PMCID: PMC11312618 DOI: 10.1101/2024.07.31.605343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Dysfunction of the lymphatic system following injury, disease, or cancer treatment can lead to lymphedema, a debilitating condition with no cure. Advances in targeted therapy have shown promise for treating diseases where conventional therapies have been ineffective and lymphatic vessels have recently emerged as a new therapeutic target. Lipid nanoparticles (LNPs) have emerged as a promising strategy for tissue specific delivery of nucleic acids. Currently, there are no approaches to target LNPs to lymphatic endothelial cells, although it is well established that intradermal (ID) injection of nanoparticles will drain to lymphatics with remarkable efficiency. To design an LNP that would effectively deliver mRNA to LEC after ID delivery, we screened a library of 150 LNPs loaded with a reporter mRNA, for both self-assembly and delivery in vivo to lymphatic endothelial cells (LECs). We identified and validated several LNP formulations optimized for high LEC uptake when administered ID and compared their efficacy for delivery of functional mRNA with that of free mRNA and mRNA delivered with a commercially available MC3-based LNP (Onpattro™). The lead LEC-specific LNP was then loaded with VEGFC mRNA to test the therapeutic advantage of the LEC-specific LNP (namely, LNP7) for treating a mouse tail lymphatic injury model. A single dose of VEGFC mRNA delivered via LNP7 resulted in enhanced LEC proliferation at the site of injury, and an increase in lymphatic function up to 14-days post-surgery. Our results suggest a therapeutic potential of VEGFC mRNA lymphatic-specific targeted delivery in alleviating lymphatic dysfunction observed during lymphatic injury and could provide a promising approach for targeted, transient lymphangiogenic therapy.
Collapse
Affiliation(s)
- Eleftheria Michalaki
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology; Atlanta, GA, USA
| | - Rachel Chin
- Department of Biology, Georgia Institute of Technology; Atlanta, GA, USA
| | - Kiyoung Jeong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University; Atlanta, GA, USA
| | - Zhiming Qi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University; Atlanta, GA, USA
| | - Lauren N. Liebman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University; Atlanta, GA, USA
| | - Yarelis González-Vargas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University; Atlanta, GA, USA
| | - Elisa Schrader Echeverri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University; Atlanta, GA, USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University; Atlanta, GA, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Beth Jiron Tamburini
- University of Colorado School of Medicine, Department of Medicine, Aurora, CO, USA
| | - Zoltan Jakus
- Semmelweis University School of Medicine, Department of Physiology, Budapest, Hungary
| | - James E. Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University; Atlanta, GA, USA
| | - J. Brandon Dixon
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology; Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University; Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology; Atlanta, GA, USA
| |
Collapse
|
3
|
Zhang B, Swanson WB, Durdan M, Livingston HN, Dodd M, Vidanapathirana SM, Desai A, Douglas L, Mishina Y, Weivoda M, Greineder CF. Affinity targeting of therapeutic proteins to the bone surface-local delivery of sclerostin-neutralizing antibody enhances efficacy. J Bone Miner Res 2024; 39:717-728. [PMID: 38526976 PMCID: PMC11472147 DOI: 10.1093/jbmr/zjae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/02/2024] [Accepted: 03/23/2024] [Indexed: 03/27/2024]
Abstract
Currently available biotherapeutics for the treatment of osteoporosis lack explicit mechanisms for bone localization, potentially limiting efficacy and inducing off-target toxicities. While various strategies have been explored for targeting the bone surface, critical aspects remain poorly understood, including the optimal affinity ligand, the role of binding avidity and circulation time, and, most importantly, whether or not this strategy can enhance the functional activity of clinically relevant protein therapeutics. To investigate, we generated fluorescent proteins (eg, mCherry) with site-specifically attached small molecule (bisphosphonate) or peptide (deca-aspartate, D10) affinity ligands. While both affinity ligands successfully anchored fluorescent protein to the bone surface, quantitative radiotracing revealed only modest femoral and vertebral accumulation and suggested a need for enhanced circulation time. To achieve this, we fused mCherry to the Fc fragment of human IgG1 and attached D10 peptides to each C-terminus. The mCherry-Fc-D10 demonstrated an ~80-fold increase in plasma exposure and marked increases in femoral and vertebral accumulation (13.6% ± 1.4% and 11.4% ± 1.3% of the injected dose/g [%ID/g] at 24 h, respectively). To determine if bone surface targeting could enhance the efficacy of a clinically relevant therapeutic, we generated a bone-targeted sclerostin-neutralizing antibody, anti-sclerostin-D10. The targeted antibody demonstrated marked increases in bone accumulation and retention (20.9 ± 2.5% and 19.5 ± 2.5% ID/g in femur and vertebrae at 7 days) and enhanced effects in a murine model of ovariectomy-induced bone loss (bone volume/total volume, connectivity density, and structure model index all increased [P < .001] vs untargeted anti-sclerostin). Collectively, our results indicate the importance of both bone affinity and circulation time in achieving robust targeting of therapeutic proteins to the bone surface and suggest that this approach may enable lower doses and/or longer dosing intervals without reduction in biotherapeutic efficacy. Future studies will be needed to determine the translational potential of this strategy and its potential impact on off-site toxicities.
Collapse
Affiliation(s)
- Boya Zhang
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - William Benton Swanson
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margaret Durdan
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| | - Heather N Livingston
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michaela Dodd
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sachith M Vidanapathirana
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alec Desai
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lindsey Douglas
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Megan Weivoda
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Hematology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Colin F Greineder
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
4
|
Obeng EM, Steer DL, Fulcher A, Wagstaff KM. Steric-Deficient Oligoglycine Surrogates Facilitate Multivalent and Bifunctional Nanobody Synthesis via Combined Sortase A Transpeptidation and Click Chemistry. Bioconjug Chem 2023; 34:1667-1678. [PMID: 37534819 DOI: 10.1021/acs.bioconjchem.3c00319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Conferring multifunctional properties to proteins via enzymatic approaches has greatly facilitated recent progress in protein nanotechnology. In this regard, sortase (Srt) A transpeptidation has facilitated many of these developments due to its exceptional specificity, mild reaction conditions, and complementation with other bioorthogonal techniques, such as click chemistry. In most of these developments, Srt A is used to seamlessly tether oligoglycine-containing molecules to a protein of interest that is equipped with the enzyme's recognition sequence, LPXTG. However, the dependence on oligoglycine attacking nucleophiles and the associated cost of certain derivatives (e.g., cyclooctyne) limit the utility of this approach to lab-scale applications only. Thus, the quest to identify appropriate alternatives and understand their effectiveness remains an important area of research. This study identifies that steric and nucleophilicity-associated effects influence Srt A transpeptidation when two oligoglycine surrogates were examined. The approach was further used in complementation with click chemistry to synthesize bivalent and bifunctional nanobody conjugates for application in epithelial growth factor receptor targeting. The overall technique and tools developed here may facilitate the advancement of future nanotechnologies.
Collapse
Affiliation(s)
- Eugene M Obeng
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - David L Steer
- Monash Proteomics and Metabolomics Facility, Monash University, Clayton 3800, Victoria, Australia
| | - Alex Fulcher
- Monash Micro Imaging, Monash University, Clayton 3800, Victoria, Australia
| | - Kylie M Wagstaff
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| |
Collapse
|
5
|
Obeng EM, Fulcher AJ, Wagstaff KM. Harnessing sortase A transpeptidation for advanced targeted therapeutics and vaccine engineering. Biotechnol Adv 2023; 64:108108. [PMID: 36740026 DOI: 10.1016/j.biotechadv.2023.108108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The engineering of potent prophylactic and therapeutic complexes has always required careful protein modification techniques with seamless capabilities. In this light, methods that favor unobstructed multivalent targeting and correct antigen presentations remain essential and very demanding. Sortase A (SrtA) transpeptidation has exhibited these attributes in various settings over the years. However, its applications for engineering avidity-inspired therapeutics and potent vaccines have yet to be significantly noticed, especially in this era where active targeting and multivalent nanomedications are in great demand. This review briefly presents the SrtA enzyme and its associated transpeptidation activity and describes interesting sortase-mediated protein engineering and chemistry approaches for achieving multivalent therapeutic and antigenic responses. The review further highlights advanced applications in targeted delivery systems, multivalent therapeutics, adoptive cellular therapy, and vaccine engineering. These innovations show the potential of sortase-mediated techniques in facilitating the development of simple plug-and-play nanomedicine technologies against recalcitrant diseases and pandemics such as cancer and viral infections.
Collapse
Affiliation(s)
- Eugene M Obeng
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University, Clayton, VIC 3800, Australia
| | - Kylie M Wagstaff
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
6
|
Ferguson LT, Ma X, Myerson JW, Wu J, Glassman PM, Zamora ME, Hood ED, Zaleski M, Shen M, Essien EO, Shuvaev VV, Brenner JS. Mechanisms by Which Liposomes Improve Inhaled Drug Delivery for Alveolar Diseases. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200106. [PMID: 37266328 PMCID: PMC10231510 DOI: 10.1002/anbr.202200106] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/23/2022] [Indexed: 01/29/2023] Open
Abstract
Diseases of the pulmonary alveolus, such as pulmonary fibrosis, are leading causes of morbidity and mortality, but exceedingly few drugs are developed for them. A major reason for this gap is that after inhalation, drugs are quickly whisked away from alveoli due to their high perfusion. To solve this problem, the mechanisms by which nano-scale drug carriers dramatically improve lung pharmacokinetics using an inhalable liposome formulation containing nintedanib, an antifibrotic for pulmonary fibrosis, are studied. Direct instillation of liposomes in murine lung increases nintedanib's total lung delivery over time by 8000-fold and lung half life by tenfold, compared to oral nintedanib. Counterintuitively, it is shown that pulmonary surfactant neither lyses nor aggregates the liposomes. Instead, each lung compartment (alveolar fluid, alveolar leukocytes, and parenchyma) elutes liposomes over 24 h, likely serving as "drug depots." After deposition in the surfactant layer, liposomes are transferred over 3-6 h to alveolar leukocytes (which take up a surprisingly minor 1-5% of total lung dose instilled) in a nonsaturable fashion. Further, all cell layers of the lung parenchyma take up liposomes. These and other mechanisms elucidated here should guide engineering of future inhaled nanomedicine for alveolar diseases.
Collapse
Affiliation(s)
- Laura T. Ferguson
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Xiaonan Ma
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jacob W. Myerson
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jichuan Wu
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Patrick M. Glassman
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Marco E. Zamora
- School of Biomedical Engineering, Science, and Health SystemsDrexel UniversityPhiladelphiaPA19104USA
| | - Elizabeth D. Hood
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Michael Zaleski
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Mengwen Shen
- Emergency Medicine DepartmentYueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese Medicine200437ShanghaiChina
- Department of MicrobiologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Eno-Obong Essien
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Vladimir V. Shuvaev
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jacob S. Brenner
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Penn-CHOP Lung Biology InstitutePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| |
Collapse
|
7
|
Yu L, Shang Z, Jin Q, Chan SY, Hong W, Li N, Li P. Antibody-Antimicrobial Conjugates for Combating Antibiotic Resistance. Adv Healthc Mater 2023; 12:e2202207. [PMID: 36300640 DOI: 10.1002/adhm.202202207] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Indexed: 02/03/2023]
Abstract
As the development of new antibiotics lags far behind the emergence of drug-resistant bacteria, alternative strategies to resolve this dilemma are urgently required. Antibody-drug conjugate is a promising therapeutic platform to delivering cytotoxic payloads precisely to target cells for efficient disease treatment. Antibody-antimicrobial conjugates (AACs) have recently attracted considerable interest from researchers as they can target bacteria in the target sites and improve the effectiveness of drugs (i.e., reduced drug dosage and adverse effects), abating the upsurge of antimicrobial resistance. In this review, the selection and progress of three essential blocks that compose the AACs: antibodies, antimicrobial payloads, and linkers are discussed. The commonly used conjugation strategies and the latest applications of AACs in recent years are also summarized. The challenges and opportunities of this booming technology are also discussed at the end of this review.
Collapse
Affiliation(s)
- Luofeng Yu
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Zifang Shang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China.,Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, 518026, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology Chinese Academy of Sciences, Beijing, 100101, China
| | - Qizhe Jin
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Siew Yin Chan
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China.,Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Weilin Hong
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Nan Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| |
Collapse
|
8
|
Sarrett SM, Rodriguez C, Rymarczyk G, Hosny MM, Keinänen O, Delaney S, Thau S, Krantz BA, Zeglis BM. Lysine-Directed Site-Selective Bioconjugation for the Creation of Radioimmunoconjugates. Bioconjug Chem 2022; 33:1750-1760. [PMID: 35946495 DOI: 10.1021/acs.bioconjchem.2c00354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The synthesis of radioimmunoconjugates via the stochastic attachment of bifunctional chelators to lysines can yield heterogeneous products with suboptimal in vitro and in vivo behavior. In response to this, several site-selective approaches to bioconjugation have been developed, yet each has intrinsic drawbacks, such as the need for expensive reagents or the complexity of incorporating unnatural amino acids into IgGs. Herein, we describe the use of a simple and facile approach to lysine-directed site-selective bioconjugation for the generation of radioimmunoconjugates. This strategy relies upon on the selective modification of single lysine residues within each light chain of the monoclonal antibody (mAb) with a branched azide-bearing perfluorophenyl ester (PFP-bisN3) followed by the ligation of dibenzocyclooctyne (DBCO)-bearing payloads to these bioorthogonal handles via the strain-promoted azide-alkyne cycloaddition. This methodology was used to create [89Zr]Zr-SSKDFO-pertuzumab, a radioimmunoconjugate of the HER2-targeting mAb pertuzumab labeled with desferrioxamine (DFO) and the positron-emitting radiometal zirconium-89 (89Zr). [89Zr]Zr-SSKDFO-pertuzumab was compared to a pair of analogous probes: one synthesized via random lysine modification ([89Zr]Zr-DFO-pertuzumab) and another via thiol-maleimide chemistry ([89Zr]Zr-malDFO-pertuzumab). The bioconjugation strategy was assessed using ESI mass spectrometry, SDS-PAGE, and autoradiography. All three immunoconjugates demonstrated comparable binding to HER2 via flow cytometry and surface plasmon resonance (SPR), and 89Zr-labeled variants of each were synthesized in >99% radiochemical yield and molar activities of up to ∼55.5 GBq/μmol (10 mCi/mg). Subsequently, the in vivo behavior of this trio of 89Zr-immunoPET probes was interrogated in athymic nude mice bearing subcutaneous HER2-expressing BT-474 human breast cancer xenografts. [89Zr]Zr-SSKDFO-pertuzumab, [89Zr]Zr-malDFO-pertuzumab, and [89Zr]Zr-DFO-pertuzumab produced positron emission tomography (PET) images with high tumoral uptake and high tumor-to-healthy organ activity concentration ratios. A terminal biodistribution study complemented the PET results, revealing tumoral activity concentrations of 126.9 ± 50.3%ID/g, 86.9 ± 53.2%ID/g, and 92.5 ± 27.2%ID/g at 144 h post-injection for [89Zr]Zr-SSKDFO-pertuzumab, [89Zr]Zr-malDFO-pertuzumab, and [89Zr]Zr-DFO-pertuzumab, respectively. Taken together, the data clearly illustrate that this highly modular and facile approach to site-selective bioconjugation produces radioimmunoconjugates that are better-defined and more homogeneous than stochastically modified constructs and also exhibit excellent in vitro and in vivo performance. Furthermore, we contend that this lysine-directed strategy holds several key advantages over extant approaches to site-selective bioconjugation, especially in the context of production for the clinic.
Collapse
Affiliation(s)
- Samantha M Sarrett
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States.,Ph.D. Program in Biochemistry, Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, United States
| | - Cindy Rodriguez
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, United States.,Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, New York 10016, United States
| | - Grzegorz Rymarczyk
- Advanced Proteome Therapeutics Inc, Boston, Massachusetts 02118, United States
| | - Meena M Hosny
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States
| | - Outi Keinänen
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, United States.,Department of Chemistry, University of Helsinki, Helsinki, 00100, Finland
| | - Samantha Delaney
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States.,Ph.D. Program in Biochemistry, Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, United States
| | - Sarah Thau
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States
| | - Benjamin A Krantz
- Advanced Proteome Therapeutics Inc, Boston, Massachusetts 02118, United States.,Department of Medicine, Division of Hematology and Medical Oncology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Brian M Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States.,Ph.D. Program in Biochemistry, Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, United States.,Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States
| |
Collapse
|
9
|
Lynch DR, Stringham EN, Zhang B, Balbin-Cuesta G, Curtis BR, Palumbo JS, Greineder CF, Tourdot BE. Anchoring IgG-degrading enzymes to the surface of platelets selectively neutralizes antiplatelet antibodies. Blood Adv 2022; 6:4645-4656. [PMID: 35737875 PMCID: PMC9636316 DOI: 10.1182/bloodadvances.2022007195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/19/2022] [Indexed: 11/20/2022] Open
Abstract
Immune thrombocytopenia (ITP) is an acquired bleeding disorder characterized by immunoglobulin G (IgG)-mediated platelet destruction. Current therapies primarily focus on reducing antiplatelet antibodies using immunosuppression or increasing platelet production with thrombopoietin mimetics. However, there are no universally safe and effective treatments for patients presenting with severe life-threatening bleeding. The IgG-degrading enzyme of Streptococcus pyogenes (IdeS), a protease with strict specificity for IgG, prevents IgG-driven immune disorders in murine models, including ITP. In clinical trials, IdeS prevented IgG-mediated kidney transplant rejection; however, the concentration of IdeS used to remove pathogenic antibodies causes profound hypogammaglobulinemia, and IdeS is immunogenic, which limits its use. Therefore, this study sought to determine whether targeting IdeS to FcγRIIA, a low-affinity IgG receptor on the surface of platelets, neutrophils, and monocytes, would be a viable strategy to decrease the pathogenesis of antiplatelet IgG and reduce treatment-related complications of nontargeted IdeS. We generated a recombinant protein conjugate by site-specifically linking the C-terminus of a single-chain variable fragment from an FcγRIIA antibody, clone IV.3, to the N-terminus of IdeS (scIV.3-IdeS). Platelets treated with scIV.3-IdeS had reduced binding of antiplatelet IgG from patients with ITP and decreased platelet phagocytosis in vitro, with no decrease in normal IgG. Treatment of mice expressing human FcγRIIA with scIV.3-IdeS reduced thrombocytopenia in a model of ITP and significantly improved the half-life of transfused platelets expressing human FcγRIIA. Together, these data suggest that scIV.3-IdeS can selectively remove pathogenic antiplatelet IgG and may be a potential treatment for patients with ITP and severe bleeding.
Collapse
Affiliation(s)
- Donald R. Lynch
- Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Emily N. Stringham
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Boya Zhang
- Department of Pharmacology
- Department of Emergency Medicine
- BioInterfaces Institute
| | - Ginette Balbin-Cuesta
- Cellular and Molecular Biology Program
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
| | - Brian R. Curtis
- Platelet & Neutrophil Immunology Laboratory
- Blood Research Institute, Versiti, Milwaukee, WI
| | - Joseph S. Palumbo
- Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Colin F. Greineder
- Department of Pharmacology
- Department of Emergency Medicine
- BioInterfaces Institute
| | - Benjamin E. Tourdot
- Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
10
|
Glassman PM, Villa CH, Marcos-Contreras OA, Hood ED, Walsh LR, Greineder CF, Myerson JW, Shuvaeva T, Puentes L, Brenner JS, Siegel DL, Muzykantov VR. Targeted In Vivo Loading of Red Blood Cells Markedly Prolongs Nanocarrier Circulation. Bioconjug Chem 2022; 33:1286-1294. [PMID: 35710322 DOI: 10.1021/acs.bioconjchem.2c00196] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Engineering drug delivery systems for prolonged pharmacokinetics (PK) has been an ongoing pursuit for nearly 50 years. The gold standard for PK enhancement is the coating of nanoparticles with polymers, namely polyethylene glycol (PEGylation), which has been applied in several clinically used products. In the present work, we utilize the longest circulating and most abundant component of blood─the erythrocyte─to improve the PK behavior of liposomes. Antibody-mediated coupling of liposomes to erythrocytes was tested in vitro to identify a loading dose that did not adversely impact the carrier cells. Injection of erythrocyte targeting liposomes into mice resulted in a ∼2-fold improvement in the area under the blood concentration versus time profile versus PEGylated liposomes and a redistribution from the plasma into the cellular fraction of blood. These results suggest that in vivo targeting of erythrocytes is a viable strategy to improve liposome PK relative to current, clinically viable strategies.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Landis R Walsh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Laura Puentes
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Don L Siegel
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
11
|
Khursheed R, Paudel KR, Gulati M, Vishwas S, Jha NK, Hansbro PM, Oliver BG, Dua K, Singh SK. Expanding the arsenal against pulmonary diseases using surface-functionalized polymeric micelles: breakthroughs and bottlenecks. Nanomedicine (Lond) 2022; 17:881-911. [PMID: 35332783 DOI: 10.2217/nnm-2021-0451] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pulmonary diseases such as lung cancer, asthma and tuberculosis have remained one of the common challenges globally. Polymeric micelles (PMs) have emerged as an effective technique for achieving targeted drug delivery for a local as well as a systemic effect. These PMs encapsulate and protect hydrophobic drugs, increase pulmonary targeting, decrease side effects and enhance drug efficacy through the inhalation route. In the current review, emphasis has been placed on the different barriers encountered by the drugs given via the pulmonary route and the mechanism of PMs in achieving drug targeting. The applications of PMs in different pulmonary diseases have also been discussed in detail.
Collapse
Affiliation(s)
- Rubiya Khursheed
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Keshav R Paudel
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, 2007, Australia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.,Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Plot No. 32-34 Knowledge Park III Greater Noida, Uttar Pradesh, 201310, India
| | - Philip M Hansbro
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, 2007, Australia
| | - Brian G Oliver
- Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, 2007, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.,Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| |
Collapse
|
12
|
Ferguson LT, Hood ED, Shuvaeva T, Shuvaev VV, Basil MC, Wang Z, Nong J, Ma X, Wu J, Myerson JW, Marcos-Contreras OA, Katzen J, Carl JM, Morrisey EE, Cantu E, Villa CH, Mitragotri S, Muzykantov VR, Brenner JS. Dual Affinity to RBCs and Target Cells (DART) Enhances Both Organ- and Cell Type-Targeting of Intravascular Nanocarriers. ACS NANO 2022; 16:4666-4683. [PMID: 35266686 PMCID: PMC9339245 DOI: 10.1021/acsnano.1c11374] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
A long-standing goal of nanomedicine is to improve a drug's benefit by loading it into a nanocarrier that homes solely to a specific target cell and organ. Unfortunately, nanocarriers usually end up with only a small percentage of the injected dose (% ID) in the target organ, due largely to clearance by the liver and spleen. Further, cell-type-specific targeting is rarely achieved without reducing target organ accumulation. To solve these problems, we introduce DART (dual affinity to RBCs and target cells), in which nanocarriers are conjugated to two affinity ligands, one binding red blood cells and one binding a target cell (here, pulmonary endothelial cells). DART nanocarriers first bind red blood cells and then transfer to the target organ's endothelial cells as the bound red blood cells squeeze through capillaries. We show that within minutes after intravascular injection in mice nearly 70% ID of DART nanocarriers accumulate in the target organ (lungs), more than doubling the % ID ceiling achieved by a multitude of prior technologies, finally achieving a majority % ID in a target organ. Humanized DART nanocarriers in ex vivo perfused human lungs recapitulate this phenomenon. Furthermore, DART enhances the selectivity of delivery to target endothelial cells over local phagocytes within the target organ by 6-fold. DART's marked improvement in both organ- and cell-type targeting may thus be helpful in localizing drugs for a multitude of medical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Samir Mitragotri
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts 02138, United States
| | | | | |
Collapse
|
13
|
Implantable Immunosuppressant Delivery to Prevent Rejection in Transplantation. Int J Mol Sci 2022; 23:ijms23031592. [PMID: 35163514 PMCID: PMC8835747 DOI: 10.3390/ijms23031592] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
An innovative immunosuppressant with a minimally invasive delivery system has emerged in the biomedical field. The application of biodegradable and biocompatible polymer forms, such as hydrogels, scaffolds, microspheres, and nanoparticles, in transplant recipients to control the release of immunosuppressants can minimize the risk of developing unfavorable conditions. In this review, we summarized several studies that have used implantable immunosuppressant delivery to release therapeutic agents to prolong allograft survival. We also compared their applications, efficacy, efficiency, and safety/side effects with conventional therapeutic-agent administration. Finally, challenges and the future prospective were discussed. Collectively, this review will help relevant readers understand the different approaches to prevent transplant rejection in a new era of therapeutic agent delivery.
Collapse
|
14
|
Pornnoppadol G, Zhang B, Desai AA, Berardi A, Remmer HA, Tessier PM, Greineder CF. A hybridoma-derived monoclonal antibody with high homology to the aberrant myeloma light chain. PLoS One 2021; 16:e0252558. [PMID: 34634047 PMCID: PMC8504763 DOI: 10.1371/journal.pone.0252558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/20/2021] [Indexed: 11/23/2022] Open
Abstract
The identification of antibody variable regions in the heavy (VH) and light (VL) chains from hybridomas is necessary for the production of recombinant, sequence-defined monoclonal antibodies (mAbs) and antibody derivatives. This process has received renewed attention in light of recent reports of hybridomas having unintended specificities due to the production of non-antigen specific heavy and/or light chains for the intended antigen. Here we report a surprising finding and potential pitfall in variable domain sequencing of an anti-human CD63 hybridoma. We amplified multiple VL genes from the hybridoma cDNA, including the well-known aberrant Sp2/0 myeloma VK and a unique, full-length VL. After finding that the unique VL failed to yield a functional antibody, we discovered an additional full-length sequence with surprising similarity (~95% sequence identify) to the non-translated myeloma kappa chain but with a correction of its key frameshift mutation. Expression of the recombinant mAb confirmed that this highly homologous sequence is the antigen-specific light chain. Our results highlight the complexity of PCR-based cloning of antibody genes and strategies useful for identification of correct sequences.
Collapse
Affiliation(s)
- Ghasidit Pornnoppadol
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- BioInterfaces Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Boya Zhang
- BioInterfaces Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Alec A. Desai
- BioInterfaces Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Anthony Berardi
- BioInterfaces Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Macromolecular Science & Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Henriette A. Remmer
- Proteomics & Peptide Synthesis Core, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Peter M. Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- BioInterfaces Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Colin F. Greineder
- BioInterfaces Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Emergency Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
15
|
van Dorsten RT, Wagh K, Moore PL, Morris L. Combinations of Single Chain Variable Fragments From HIV Broadly Neutralizing Antibodies Demonstrate High Potency and Breadth. Front Immunol 2021; 12:734110. [PMID: 34603312 PMCID: PMC8481832 DOI: 10.3389/fimmu.2021.734110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Broadly neutralizing antibodies (bNAbs) are currently being assessed in clinical trials for their ability to prevent HIV infection. Single chain variable fragments (scFv) of bNAbs have advantages over full antibodies as their smaller size permits improved diffusion into mucosal tissues and facilitates vector-driven gene expression. We have previously shown that scFv of bNAbs individually retain significant breadth and potency. Here we tested combinations of five scFv derived from bNAbs CAP256-VRC26.25 (V2-apex), PGT121 (N332-supersite), 3BNC117 (CD4bs), 8ANC195 (gp120-gp41 interface) and 10E8v4 (MPER). Either two or three scFv were combined in equimolar amounts and tested in the TZM-bl neutralization assay against a multiclade panel of 17 viruses. Experimental IC50 and IC80 data were compared to predicted neutralization titers based on single scFv titers using the Loewe additive and the Bliss-Hill model. Like full-sized antibodies, combinations of scFv showed significantly improved potency and breadth compared to single scFv. Combinations of two or three scFv generally followed an independent action model for breadth and potency with no significant synergy or antagonism observed overall although some exceptions were noted. The Loewe model underestimated potency for some dual and triple combinations while the Bliss-Hill model was better at predicting IC80 titers of triple combinations. Given this, we used the Bliss-Hill model to predict the coverage of scFv against a 45-virus panel at concentrations that correlated with protection in the AMP trials. Using IC80 titers and concentrations of 1μg/mL, there was 93% coverage for one dual scFv combination (3BNC117+10E8v4), and 96% coverage for two of the triple combinations (CAP256.25+3BNC117+10E8v4 and PGT121+3BNC117+10E8v4). Combinations of scFv, therefore, show significantly improved breadth and potency over individual scFv and given their size advantage, have potential for use in passive immunization.
Collapse
Affiliation(s)
- Rebecca T. van Dorsten
- Center for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- Medical Research Council (MRC) Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kshitij Wagh
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, NM, United States
| | - Penny L. Moore
- Center for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- Medical Research Council (MRC) Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Center for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Lynn Morris
- Center for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- Medical Research Council (MRC) Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Center for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
16
|
Roman BI. The Expanding Role of Chemistry in Optimizing Proteins for Human Health Applications. J Med Chem 2021; 64:7179-7188. [PMID: 34014084 DOI: 10.1021/acs.jmedchem.1c00294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Over the past decades, therapeutics based on biological macromolecules and cells have successfully entered the clinical arena and progressively occupied an increasing share of what once was almost exclusively small molecule territory. This perspective explores the opportunities for chemists at the interface between biologics and small molecule-based products. It provides concrete examples by zooming in on the area of post-translational protein modification. The conclusion is that, rather than diminishing the relevance of chemistry in the pharmaceutical enterprise, the advent of the biologics has provided an additional playing field for synthetic and medicinal chemists, where they can contribute to the efficacy and scope of applicability of biological entities in a collaborative effort to transformatively address unmet medical needs.
Collapse
Affiliation(s)
- Bart I Roman
- Research Group SynBioC, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Universiteit Gent, Coupure Links 653, 9000 Gent, Belgium.,Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, 9000 Gent, Belgium
| |
Collapse
|
17
|
Qian M, Zhang Q, Lu J, Zhang J, Wang Y, Shangguan W, Feng M, Feng J. Long-Acting Human Interleukin 2 Bioconjugate Modified with Fatty Acids by Sortase A. Bioconjug Chem 2021; 32:615-625. [PMID: 33656323 DOI: 10.1021/acs.bioconjchem.1c00062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human Interleukin 2 (IL-2) has already achieved impressive results as a therapeutic agent for cancer and autoimmune diseases. However, one of the limitations associated with the clinical application of IL-2 is its short half-life owing to rapid clearance by the kidneys. Modification with fatty acids, as an albumin noncovalent ligand with the advantage of deep penetration into tissues and high activity-to-mass ratio, is a commonly used approach to improve the half-life of native peptides and proteins. In this investigation, we attempted to extend the half-life of IL-2 through conjugation with a fatty acid using sortase A (srtA). We initially designed and optimized three IL-2 analogues with different peptide linkers between the C-terminus of IL-2 and srtA recognition sequence (LPETG). Among these, analogue A3 was validated as the optimal IL-2 analogue for further modification. Next, six fatty acid moieties with the same fatty acid and different hydrophilic spacers were conjugated to A3 through srtA. The six bioconjugates generated were screened for in vitro biological activity, among which bioconjugate B6 was identified as near-optimal to IL-2. Additionally, B6 could effectively bind albumin through the conjugated fatty acid, which contributed to a significant improvement in its pharmacokinetic properties in vivo. In summary, we have developed a novel IL-2 bioconjugate, B6, modified with fatty acids using srtA, which may effectively serve as a new-generation long-acting IL-2 immunotherapeutic agent.
Collapse
Affiliation(s)
- Mengxin Qian
- Department of Microbiological & Biochemical Pharmacy, School of Pharmacy, Fudan University, 201203 Shanghai, China.,State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Qingbin Zhang
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Jianguang Lu
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China.,Shanghai Duomirui Biotechnology Co., Ltd., 201203 Shanghai, China
| | - Jinhua Zhang
- Department of Microbiological & Biochemical Pharmacy, School of Pharmacy, Fudan University, 201203 Shanghai, China.,State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Yapeng Wang
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Wenwen Shangguan
- Department of Microbiological & Biochemical Pharmacy, School of Pharmacy, Fudan University, 201203 Shanghai, China.,State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China
| | - Meiqing Feng
- Department of Microbiological & Biochemical Pharmacy, School of Pharmacy, Fudan University, 201203 Shanghai, China
| | - Jun Feng
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 201203 Shanghai, China.,Shanghai Duomirui Biotechnology Co., Ltd., 201203 Shanghai, China
| |
Collapse
|
18
|
Zhang B, Vidanapathirana SM, Greineder CF. Site-Specific Modification of Single-Chain Affinity Ligands for Fluorescence Labeling, Radiolabeling, and Bioconjugation. Methods Mol Biol 2021; 2355:163-173. [PMID: 34386959 PMCID: PMC9289842 DOI: 10.1007/978-1-0716-1617-8_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Single-chain protein affinity ligands are recombinant polypeptides that recreate the antigen-binding site of parental, monoclonal antibodies (mAbs) or present unique binding surfaces derived from display technologies, computational design, or other approaches. These diverse ligands have several advantages over full-length mAbs as agents for delivery of small molecule, protein, and nanoparticle cargoes to desired sites in the body. However, they present unique challenges for modification and bioconjugation. Fusion of a LPXTGG motif, or "sortag," and a 5-amino acid, flexible linker to the C-terminus of these affinity ligands enables high-efficiency transpeptidation by the bacterial enzyme, Sortase A, and site-specific addition of fluorophores, radiolabels, or functional groups for oriented and stoichiometrically controlled bioconjugation. We describe in detail this method and address several challenges and pitfalls in the purification and characterization of modified single-chain affinity ligands.
Collapse
Affiliation(s)
- Boya Zhang
- Departments of Pharmacology and Emergency Medicine, University of Michigan, Ann Arbor, MI, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sachith M Vidanapathirana
- Departments of Pharmacology and Emergency Medicine, University of Michigan, Ann Arbor, MI, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Colin F Greineder
- Departments of Pharmacology and Emergency Medicine, University of Michigan, Ann Arbor, MI, USA.
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
19
|
Park J, Lee S, Kim Y, Yoo TH. Methods to generate site-specific conjugates of antibody and protein. Bioorg Med Chem 2021; 30:115946. [DOI: 10.1016/j.bmc.2020.115946] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
|
20
|
Lieser RM, Yur D, Sullivan MO, Chen W. Site-Specific Bioconjugation Approaches for Enhanced Delivery of Protein Therapeutics and Protein Drug Carriers. Bioconjug Chem 2020; 31:2272-2282. [DOI: 10.1021/acs.bioconjchem.0c00456] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Rachel M. Lieser
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Daniel Yur
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Millicent O. Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| |
Collapse
|
21
|
Kiseleva RY, Glassman PG, LeForte KM, Walsh LR, Villa CH, Shuvaev VV, Myerson JW, Aprelev PA, Marcos-Contreras OA, Muzykantov VR, Greineder CF. Bivalent engagement of endothelial surface antigens is critical to prolonged surface targeting and protein delivery in vivo. FASEB J 2020; 34:11577-11593. [PMID: 32738178 DOI: 10.1096/fj.201902515rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022]
Abstract
Targeted drug delivery to the endothelium has the potential to generate localized therapeutic effects at the blood-tissue interface. For some therapeutic cargoes, it is essential to maintain contact with the bloodstream to exert protective effects. The pharmacokinetics (PK) of endothelial surface-targeted affinity ligands and biotherapeutic cargo remain a largely unexplored area, despite obvious translational implications for this strategy. To bridge this gap, we site-specifically radiolabeled mono- (scFv) and bivalent (mAb) affinity ligands specific for the endothelial cell adhesion molecules, PECAM-1 (CD31) and ICAM-1 (CD54). Radiotracing revealed similar lung biodistribution at 30 minutes post-injection (79.3% ± 4.2% vs 80.4% ± 10.6% ID/g for αICAM and 58.9% ± 3.6% ID/g vs. 47.7% ± 5.8% ID/g for αPECAM mAb vs. scFv), but marked differences in organ residence time, with antibodies demonstrating an order of magnitude greater area under the lung concentration vs. time curve (AUCinf 1698 ± 352 vs. 53.3 ± 7.9 ID/g*hrs for αICAM and 1023 ± 507 vs. 114 ± 37 ID/g*hrs for αPECAM mAb vs scFv). A physiologically based pharmacokinetic model, fit to and validated using these data, indicated contributions from both superior binding characteristics and prolonged circulation time supporting multiple binding-detachment cycles. We tested the ability of each affinity ligand to deliver a prototypical surface cargo, thrombomodulin (TM), using one-to-one protein conjugates. Bivalent mAb-TM was superior to monovalent scFv-TM in both pulmonary targeting and lung residence time (AUCinf 141 ± 3.2 vs 12.4 ± 4.2 ID/g*hrs for ICAM and 188 ± 90 vs 34.7 ± 19.9 ID/g*hrs for PECAM), despite having similar blood PK, indicating that binding strength is more important parameter than the kinetics of binding. To maximize bivalent target engagement, we synthesized an oriented, end-to-end anti-ICAM mAb-TM conjugate and found that this therapeutic had the best lung residence time (AUCinf 253 ± 18 ID/g*hrs) of all TM modalities. These observations have implications not only for the delivery of TM, but also potentially all therapeutics targeted to the endothelial surface.
Collapse
Affiliation(s)
- R Yu Kiseleva
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - P G Glassman
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - K M LeForte
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - L R Walsh
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C H Villa
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - V V Shuvaev
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - J W Myerson
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - P A Aprelev
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - O A Marcos-Contreras
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - V R Muzykantov
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C F Greineder
- Department of Emergency Medicine and Pharmacology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
22
|
Glassman PM, Myerson JW, Ferguson LT, Kiseleva RY, Shuvaev VV, Brenner JS, Muzykantov VR. Targeting drug delivery in the vascular system: Focus on endothelium. Adv Drug Deliv Rev 2020; 157:96-117. [PMID: 32579890 PMCID: PMC7306214 DOI: 10.1016/j.addr.2020.06.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
The bloodstream is the main transporting pathway for drug delivery systems (DDS) from the site of administration to the intended site of action. In many cases, components of the vascular system represent therapeutic targets. Endothelial cells, which line the luminal surface of the vasculature, play a tripartite role of the key target, barrier, or victim of nanomedicines in the bloodstream. Circulating DDS may accumulate in the vascular areas of interest and in off-target areas via mechanisms bypassing specific molecular recognition, but using ligands of specific vascular determinant molecules enables a degree of precision, efficacy, and specificity of delivery unattainable by non-affinity DDS. Three decades of research efforts have focused on specific vascular targeting, which have yielded a multitude of DDS, many of which are currently undergoing a translational phase of development for biomedical applications, including interventions in the cardiovascular, pulmonary, and central nervous systems, regulation of endothelial functions, host defense, and permeation of vascular barriers. We discuss the design of endothelial-targeted nanocarriers, factors underlying their interactions with cells and tissues, and describe examples of their investigational use in models of acute vascular inflammation with an eye on translational challenges.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America.
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Laura T Ferguson
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Raisa Y Kiseleva
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Vladimir V Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America.
| |
Collapse
|
23
|
Glassman PM, Walsh LR, Villa CH, Marcos-Contreras OA, Hood ED, Muzykantov VR, Greineder CF. Molecularly Engineered Nanobodies for Tunable Pharmacokinetics and Drug Delivery. Bioconjug Chem 2020; 31:1144-1155. [PMID: 32167754 DOI: 10.1021/acs.bioconjchem.0c00003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The use of single-domain antibody fragments, or nanobodies, has gained popularity in recent years as an alternative to traditional monoclonal antibody-based approaches. Relatively little is known, however, about the utility of nanobodies as targeting agents for delivery of therapeutic cargoes, particularly to vascular epitopes or in the setting of acute inflammatory conditions. We used a nanobody (VCAMelid) directed against mouse vascular cell adhesion molecule 1 (VCAM-1) and techniques for site-specific radiolabeling and bioconjugation to measure targeting to sites of constitutive and inducible antigen expression and investigate the impact of various characteristics (affinity, valence, circulation time) on nanobody biodistribution and pharmacokinetics. Engineering of VCAMelid for bivalent binding (BiVCAMelid) increased affinity by an order of magnitude and provided 2.8- and 3.6-fold enhancements in splenic and brain targeting in naive mice, with a further 2.6-fold increase in brain uptake in the setting of focal CNS inflammation. In contrast, introduction of an albumin-binding arm (VCAM/ALB8) did not affect binding affinity, but its prolonged circulation time resulted in 3.5-fold and 17.4-fold increases in splenic and brain uptake at 20 min post-dose and remarkable 40-, 25-, and 15-fold enhancements in overall exposure of blood, spleen, and brain, respectively, relative to both VCAMelid and BiVCAMelid. Both therapeutic protein (superoxide dismutase, SOD-1) and nanocarrier (liposome) delivery were enhanced by conjugation to VCAM-1 targeted nanobodies. The bispecific VCAM/ALB8 maintained its superiority over VCAMelid in enhancing both circulation time and organ targeting of SOD-1, but its advantages were largely blunted by conjugation to liposomes.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Landis R Walsh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Department of Emergency Medicine and Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
24
|
One-step site-specific antibody fragment auto-conjugation using SNAP-tag technology. Nat Protoc 2019; 14:3101-3125. [DOI: 10.1038/s41596-019-0214-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/07/2019] [Indexed: 12/13/2022]
|
25
|
|
26
|
Zhang Y, Park KY, Suazo KF, Distefano MD. Recent progress in enzymatic protein labelling techniques and their applications. Chem Soc Rev 2018; 47:9106-9136. [PMID: 30259933 PMCID: PMC6289631 DOI: 10.1039/c8cs00537k] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein-based conjugates are valuable constructs for a variety of applications. Conjugation of proteins to fluorophores is commonly used to study their cellular localization and the protein-protein interactions. Modification of therapeutic proteins with either polymers or cytotoxic moieties greatly enhances their pharmacokinetics or potency. To label a protein of interest, conventional direct chemical reaction with the side-chains of native amino acids often yields heterogeneously modified products. This renders their characterization complicated, requires difficult separation steps and may impact protein function. Although modification can also be achieved via the insertion of unnatural amino acids bearing bioorthogonal functional groups, these methods can have lower protein expression yields, limiting large scale production. As a site-specific modification method, enzymatic protein labelling is highly efficient and robust under mild reaction conditions. Significant progress has been made over the last five years in modifying proteins using enzymatic methods for numerous applications, including the creation of clinically relevant conjugates with polymers, cytotoxins or imaging agents, fluorescent or affinity probes to study complex protein interaction networks, and protein-linked materials for biosensing. This review summarizes developments in enzymatic protein labelling over the last five years for a panel of ten enzymes, including sortase A, subtiligase, microbial transglutaminase, farnesyltransferase, N-myristoyltransferase, phosphopantetheinyl transferases, tubulin tyrosin ligase, lipoic acid ligase, biotin ligase and formylglycine generating enzyme.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
27
|
Hood ED, Greineder CF, Shuvaeva T, Walsh L, Villa CH, Muzykantov VR. Vascular Targeting of Radiolabeled Liposomes with Bio-Orthogonally Conjugated Ligands: Single Chain Fragments Provide Higher Specificity than Antibodies. Bioconjug Chem 2018; 29:3626-3637. [PMID: 30240185 DOI: 10.1021/acs.bioconjchem.8b00564] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Liposomes are a proven, versatile, and clinically viable technology platform for vascular delivery of drugs and imaging probes. Although targeted liposomes have the potential to advance these applications, complex formulations and the need for optimal affinity ligands and conjugation strategies challenge their translation. Herein, we employed copper-free click chemistry functionalized liposomes to target platelet-endothelial cell adhesion molecule (PECAM-1) and intracellular adhesion molecule (ICAM-1) by conjugating clickable monoclonal antibodies (Ab) or their single chain variable fragments (scFv). For direct, quantitative tracing, liposomes were surface chelated with 111In to a >90% radiochemical yield and purity. Particle size and distribution, stability, ligand surface density, and specific binding to target cells were characterized in vitro. Biodistribution of liposomes after IV injection was characterized in mice using isotope detection in organs and by noninvasive imaging (single-photon emission computed tomography/computed tomography, SPECT/CT). As much as 20-25% of injected dose of liposomes carrying PECAM and ICAM ligands, but not control IgG accumulated in the pulmonary vasculature. The immunospecificity of pulmonary targeting of scFv/liposomes to PECAM-1 and ICAM-1, respectively, was 10-fold and 2.5-fold higher than of Ab/liposomes. Therefore, the combination of optimal ligands, benign conjugation, and labeling yields liposomal formulations that may be used for highly effective and specific vascular targeting.
Collapse
Affiliation(s)
- Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Landis Walsh
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| |
Collapse
|
28
|
Sago CD, Lokugamage MP, Paunovska K, Vanover DA, Monaco CM, Shah NN, Gamboa Castro M, Anderson SE, Rudoltz TG, Lando GN, Munnilal Tiwari P, Kirschman JL, Willett N, Jang YC, Santangelo PJ, Bryksin AV, Dahlman JE. High-throughput in vivo screen of functional mRNA delivery identifies nanoparticles for endothelial cell gene editing. Proc Natl Acad Sci U S A 2018; 115:E9944-E9952. [PMID: 30275336 PMCID: PMC6196543 DOI: 10.1073/pnas.1811276115] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Dysfunctional endothelium causes more disease than any other cell type. Systemically administered RNA delivery to nonliver tissues remains challenging, in large part because there is no high-throughput method to identify nanoparticles that deliver functional mRNA to cells in vivo. Here we report a system capable of simultaneously quantifying how >100 lipid nanoparticles (LNPs) deliver mRNA that is translated into functional protein. Using this system (named FIND), we measured how >250 LNPs delivered mRNA to multiple cell types in vivo and identified 7C2 and 7C3, two LNPs that efficiently deliver siRNA, single-guide RNA (sgRNA), and mRNA to endothelial cells. The 7C3 delivered Cas9 mRNA and sgRNA to splenic endothelial cells as efficiently as hepatocytes, distinguishing it from LNPs that deliver Cas9 mRNA and sgRNA to hepatocytes more than other cell types. These data demonstrate that FIND can identify nanoparticles with novel tropisms in vivo.
Collapse
Affiliation(s)
- Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Daryll A Vanover
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Christopher M Monaco
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Nirav N Shah
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Marielena Gamboa Castro
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Shannon E Anderson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Tobi G Rudoltz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Gwyneth N Lando
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Pooja Munnilal Tiwari
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Jonathan L Kirschman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Nick Willett
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
- Department of Orthopaedics, Emory University, Atlanta, GA 30322
- Atlanta Veterans Affairs Medical Center, Decatur, GA 30033
| | - Young C Jang
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332
| | - Anton V Bryksin
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332;
| |
Collapse
|