1
|
Meng F, Zhou N, Hu G, Liu R, Zhang Y, Jing M, Hou Q. A comprehensive overview of recent advances in generative models for antibodies. Comput Struct Biotechnol J 2024; 23:2648-2660. [PMID: 39027650 PMCID: PMC11254834 DOI: 10.1016/j.csbj.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Therapeutic antibodies are an important class of biopharmaceuticals. With the rapid development of deep learning methods and the increasing amount of antibody data, antibody generative models have made great progress recently. They aim to solve the antibody space searching problems and are widely incorporated into the antibody development process. Therefore, a comprehensive introduction to the development methods in this field is imperative. Here, we collected 34 representative antibody generative models published recently and all generative models can be divided into three categories: sequence-generating models, structure-generating models, and hybrid models, based on their principles and algorithms. We further studied their performance and contributions to antibody sequence prediction, structure optimization, and affinity enhancement. Our manuscript will provide a comprehensive overview of the status of antibody generative models and also offer guidance for selecting different approaches.
Collapse
Affiliation(s)
- Fanxu Meng
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Na Zhou
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
- National Institute of Health Data Science of China, Shandong University, Jinan 250100, China
| | - Guangchun Hu
- School of Information Science and Engineering, University of Jinan, Jinan 250022, China
| | - Ruotong Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
- National Institute of Health Data Science of China, Shandong University, Jinan 250100, China
| | - Yuanyuan Zhang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Ming Jing
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Provincial Key Laboratory of Computer Networks, Shandong Fundamental Research Center for Computer Science, Jinan 250000, China
| | - Qingzhen Hou
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
- National Institute of Health Data Science of China, Shandong University, Jinan 250100, China
| |
Collapse
|
2
|
Khezri H, Mostafavi M, Dabirmanesh B, Khajeh K. Peptibodies: Bridging the gap between peptides and antibodies. Int J Biol Macromol 2024; 278:134718. [PMID: 39142490 DOI: 10.1016/j.ijbiomac.2024.134718] [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: 05/04/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Peptides are a very critical class of pharmaceutical compounds that can control several signaling pathways and thereby affect many physiological and biochemical processes. Previous research suggests that both peptides and antibodies may serve as potent tools for research, diagnostics, vaccination, and therapeutics across diverse domains. The distinct attributes of peptides, like their profound tissue penetration, efficient cellular internalization, reduced immunogenicity, and adaptability to chemical modification, underscore their significance in biomedical applications. However, they also possess drawbacks such as lower affinity, poor absorption, low stability to proteolytic digestion, and rapid clearance. The advent of peptibodies is a significant advance that improves the limitations of both peptides and antibodies. Peptibodies, or Peptide-Fc fusions, represent a promising therapeutic modality comprising biologically active peptides fused to an Fc domain. The stability and efficacy of the peptide are enhanced by this fusion strategy, which overcomes some of the inherent limitations. Many peptibodies have been developed to treat conditions like cancer, diabetes, and lupus. Romiplostim and Dulaglutide are the only ones approved by the EMA and FDA, respectively. Given the growing significance of peptibodies in the pharmaceutical landscape, this investigation aims to explain key aspects encompassing the intrinsic properties of peptides, the intricacies of peptibody production, and their potential therapeutic applications.
Collapse
Affiliation(s)
- Hamidhossein Khezri
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdiyeh Mostafavi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Raja A, Kasana A, Verma V. Next-Generation Therapeutic Antibodies for Cancer Treatment: Advancements, Applications, and Challenges. Mol Biotechnol 2024:10.1007/s12033-024-01270-y. [PMID: 39222285 DOI: 10.1007/s12033-024-01270-y] [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: 06/04/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
The field of cancer treatment has evolved significantly over the last decade with the emergence of next-generation therapeutic antibodies. Conventional treatments like chemotherapy pose significant challenges, including adverse side effects. Monoclonal antibodies have paved the way for more targeted and effective interventions. The evolution from chimeric to humanized and fully human antibodies has led to a reduction in immunogenicity and enhanced tolerance in vivo. The advent of next-generation antibodies, including bispecific antibodies, nanobodies, antibody-drug conjugates, glyco-engineered antibodies, and antibody fragments, represents a leap forward in cancer therapy. These innovations offer increased potency, adaptability, and reduced drug resistance. Challenges such as target validation, immunogenicity, and high production costs exist. However, technological advancements in antibody engineering techniques provide optimism for addressing these issues. The future promises a paradigm shift, where ongoing research will propel these powerful antibodies to the forefront, revolutionizing the fight against cancer and creating new preventive and curative treatments. This review provides an overview of three next-generation antibody-based molecules, namely bispecific antibodies, antibody-drug conjugates, and nanobodies that have shown promising results in cancer treatment. It discusses the evolution of antibodies from conventional forms to next-generation molecules, along with their applications in cancer treatment, production methods, and associated challenges. The review aims to offer researchers insights into the evolving landscape of next-generation antibody-based cancer therapeutics and their potential to revolutionize treatment strategies.
Collapse
Affiliation(s)
- Abhavya Raja
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India
| | - Abhishek Kasana
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India
| | - Vaishali Verma
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India.
| |
Collapse
|
4
|
Imai M, Colas K, Suga H. Protein Grafting Techniques: From Peptide Epitopes to Lasso-Grafted Neobiologics. Chempluschem 2024; 89:e202400152. [PMID: 38693599 DOI: 10.1002/cplu.202400152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
Protein engineering techniques have vastly expanded their domain of impact, notably following the success of antibodies. Likewise, smaller peptide therapeutics have carved an increasingly significant niche for themselves in the pharmaceutical landscape. The concept of grafting such peptides onto larger protein scaffolds, thus harvesting the advantages of both, has given rise to a variety of protein engineering strategies that are reviewed herein. We also describe our own "Lasso-Grafting" approach, which combines traditional grafting concepts with mRNA display to streamline the production of multiple grafted drug candidates for virtually any target.
Collapse
Affiliation(s)
- Mikio Imai
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kilian Colas
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
5
|
Nijhawan HP, Prabhakar B, Misra A, Yadav KS. Fragmented antibodies in non-small cell lung cancer: A novel nano-engineered delivery system for detection and treatment of cancer. Drug Discov Today 2023; 28:103701. [PMID: 37453459 DOI: 10.1016/j.drudis.2023.103701] [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: 09/03/2022] [Revised: 05/08/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Non-small cell lung cancer (NSCLC) has a long history of defying traditional cytotoxic treatment. Significant advancements in biotechnology, cancer biology, and immunotherapy have provided new insights that have altered the landscape for the management of NSCLC, clearing the way for a new era of pharmaceuticals in the form of monoclonal antibodies and their fragments. Antibody fragments are superior to monoclonal antibodies because of their small size, which allows them to penetrate cells and tissues effectively. When combined with functional nanocarriers, antibody fragments can target cancer cells while offering improved efficacy and fewer off-target effects. We discuss current topics of interest including anti-CTLA-4 mAbs, Talactoferrin alfa (TLF), and the CYFRA 21-1 biomarker, with brief insights into its novel detection system.
Collapse
Affiliation(s)
- Harsh P Nijhawan
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile-Parle (W), Mumbai 400056, India
| | - Bala Prabhakar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile-Parle (W), Mumbai 400056, India
| | - Ambikanandan Misra
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile-Parle (W), Mumbai 400056, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed to be University, Vile-Parle (W), Mumbai 400056, India.
| |
Collapse
|
6
|
Bergonzo C, Hoopes JT, Kelman Z, Gallagher DT. Effects of glycans and hinge on dynamics in the IgG1 Fc. J Biomol Struct Dyn 2023:1-9. [PMID: 37897185 PMCID: PMC11055941 DOI: 10.1080/07391102.2023.2270749] [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: 07/11/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023]
Abstract
The crystallizable fragment (Fc) domain of immunoglobulin subclass IgG1 antibodies is engineered for a wide variety of pharmaceutical applications. Two important structural variables in Fc constructs are the hinge region connecting the Fc to the antigen binding fragments (Fab) and the glycans present in various glycoforms. These components affect receptor binding interactions that mediate immune activation. To design new antibody drugs, a robust in silico method for linking stability to structural changes is necessary. In this work, all-atom simulations were used to compare the dynamic behavior of the four structural variants arising from presence or absence of the hinge and glycans. We expressed the simplest of these constructs, the 'minimal Fc' with no hinge and no glycans, in Escherichia coli and report its crystal structure. The 'maximal Fc' that includes full hinge and G0F/G1F glycans is based on a previously reported structure, Protein Data Bank (PDB) ID: 5VGP. These, along with two intermediate structures (with only the glycans or with only the hinge) were used to independently measure the stability effects of the two structural variables using umbrella sampling simulations. Principal component analysis (PCA) was used to determine free energy effects along the Fc's dominant mode of motion. This work provides a comprehensive picture of the effects of hinge and glycans on Fc dynamics and stability.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Christina Bergonzo
- National Institute of Standards and Technology, 9600 Gudelsky Dr. Rockville, MD, 20850
- The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Dr. Rockville, MD, 20850
| | - J. Todd Hoopes
- The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Dr. Rockville, MD, 20850
- The Biomolecular Labeling Laboratory, 9600 Gudelsky Dr. Rockville, MD, 20850
| | - Zvi Kelman
- National Institute of Standards and Technology, 9600 Gudelsky Dr. Rockville, MD, 20850
- The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Dr. Rockville, MD, 20850
- The Biomolecular Labeling Laboratory, 9600 Gudelsky Dr. Rockville, MD, 20850
| | - D. Travis Gallagher
- National Institute of Standards and Technology, 9600 Gudelsky Dr. Rockville, MD, 20850
- The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Dr. Rockville, MD, 20850
| |
Collapse
|
7
|
Chew KS, Wells RC, Moshkforoush A, Chan D, Lechtenberg KJ, Tran HL, Chow J, Kim DJ, Robles-Colmenares Y, Srivastava DB, Tong RK, Tong M, Xa K, Yang A, Zhou Y, Akkapeddi P, Annamalai L, Bajc K, Blanchette M, Cherf GM, Earr TK, Gill A, Huynh D, Joy D, Knight KN, Lac D, Leung AWS, Lexa KW, Liau NPD, Becerra I, Malfavon M, McInnes J, Nguyen HN, Lozano EI, Pizzo ME, Roche E, Sacayon P, Calvert MEK, Daneman R, Dennis MS, Duque J, Gadkar K, Lewcock JW, Mahon CS, Meisner R, Solanoy H, Thorne RG, Watts RJ, Zuchero YJY, Kariolis MS. CD98hc is a target for brain delivery of biotherapeutics. Nat Commun 2023; 14:5053. [PMID: 37598178 PMCID: PMC10439950 DOI: 10.1038/s41467-023-40681-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/02/2023] [Indexed: 08/21/2023] Open
Abstract
Brain exposure of systemically administered biotherapeutics is highly restricted by the blood-brain barrier (BBB). Here, we report the engineering and characterization of a BBB transport vehicle targeting the CD98 heavy chain (CD98hc or SLC3A2) of heterodimeric amino acid transporters (TVCD98hc). The pharmacokinetic and biodistribution properties of a CD98hc antibody transport vehicle (ATVCD98hc) are assessed in humanized CD98hc knock-in mice and cynomolgus monkeys. Compared to most existing BBB platforms targeting the transferrin receptor, peripherally administered ATVCD98hc demonstrates differentiated brain delivery with markedly slower and more prolonged kinetic properties. Specific biodistribution profiles within the brain parenchyma can be modulated by introducing Fc mutations on ATVCD98hc that impact FcγR engagement, changing the valency of CD98hc binding, and by altering the extent of target engagement with Fabs. Our study establishes TVCD98hc as a modular brain delivery platform with favorable kinetic, biodistribution, and safety properties distinct from previously reported BBB platforms.
Collapse
Affiliation(s)
- Kylie S Chew
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Robert C Wells
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Arash Moshkforoush
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Darren Chan
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kendra J Lechtenberg
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Hai L Tran
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Johann Chow
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Do Jin Kim
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | - Devendra B Srivastava
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Raymond K Tong
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Mabel Tong
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kaitlin Xa
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Alexander Yang
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Yinhan Zhou
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Padma Akkapeddi
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Lakshman Annamalai
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kaja Bajc
- Department of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
| | - Marie Blanchette
- Department of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
| | - Gerald Maxwell Cherf
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Timothy K Earr
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Audrey Gill
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - David Huynh
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - David Joy
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kristen N Knight
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Diana Lac
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Amy Wing-Sze Leung
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Katrina W Lexa
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Nicholas P D Liau
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Isabel Becerra
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Mario Malfavon
- Department of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
| | - Joseph McInnes
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Hoang N Nguyen
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Edwin I Lozano
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Michelle E Pizzo
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Elysia Roche
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Patricia Sacayon
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Meredith E K Calvert
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Richard Daneman
- Department of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., La Jolla, 92093, CA, USA
| | - Mark S Dennis
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Joseph Duque
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kapil Gadkar
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Joseph W Lewcock
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Cathal S Mahon
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - René Meisner
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Hilda Solanoy
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Robert G Thorne
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Ryan J Watts
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Y Joy Yu Zuchero
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| | - Mihalis S Kariolis
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| |
Collapse
|
8
|
Sakai K, Sugano-Nakamura N, Mihara E, Rojas-Chaverra NM, Watanabe S, Sato H, Imamura R, Voon DCC, Sakai I, Yamasaki C, Tateno C, Shibata M, Suga H, Takagi J, Matsumoto K. Designing receptor agonists with enhanced pharmacokinetics by grafting macrocyclic peptides into fragment crystallizable regions. Nat Biomed Eng 2023; 7:164-176. [PMID: 36344661 PMCID: PMC9991925 DOI: 10.1038/s41551-022-00955-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 09/26/2022] [Indexed: 11/09/2022]
Abstract
Short half-lives in circulation and poor transport across the blood-brain barrier limit the utility of cytokines and growth factors acting as receptor agonists. Here we show that surrogate receptor agonists with longer half-lives in circulation and enhanced transport rates across the blood-brain barrier can be generated by genetically inserting macrocyclic peptide pharmacophores into the structural loops of the fragment crystallizable (Fc) region of a human immunoglobulin. We used such 'lasso-grafting' approach, which preserves the expression levels of the Fc region and its affinity for the neonatal Fc receptor, to generate Fc-based protein scaffolds with macrocyclic peptides binding to the receptor tyrosine protein kinase Met. The Met agonists dimerized Met, inducing biological responses that were similar to those induced by its natural ligand. Moreover, lasso-grafting of the Fc region of the mouse anti-transferrin-receptor antibody with Met-binding macrocyclic peptides enhanced the accumulation of the resulting Met agonists in brain parenchyma in mice. Lasso-grafting may allow for designer protein therapeutics with enhanced stability and pharmacokinetics.
Collapse
Affiliation(s)
- Katsuya Sakai
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.
- WPI-Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
| | - Nozomi Sugano-Nakamura
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Japan
| | - Emiko Mihara
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Japan
| | | | - Sayako Watanabe
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Japan
| | - Hiroki Sato
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Tumor Microenvironment Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Ryu Imamura
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI-Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Dominic Chih-Cheng Voon
- Inflammation and Epithelial Plasticity Unit, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Cancer Model Research Innovative Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Itsuki Sakai
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Chihiro Yamasaki
- Research and Development Department, PhoenixBio Co. Ltd, Higashihiroshima, Japan
| | - Chise Tateno
- Research and Development Department, PhoenixBio Co. Ltd, Higashihiroshima, Japan
| | - Mikihiro Shibata
- WPI-Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
- High-speed AFM for Biological Application Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Junichi Takagi
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Japan.
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.
- WPI-Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
- Tumor Microenvironment Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
9
|
Surface Design Options in Polymer- and Lipid-Based siRNA Nanoparticles Using Antibodies. Int J Mol Sci 2022; 23:ijms232213929. [PMID: 36430411 PMCID: PMC9692731 DOI: 10.3390/ijms232213929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
The mechanism of RNA interference (RNAi) could represent a breakthrough in the therapy of all diseases that arise from a gene defect or require the inhibition of a specific gene expression. In particular, small interfering RNA (siRNA) offers an attractive opportunity to achieve a new milestone in the therapy of human diseases. The limitations of siRNA, such as poor stability, inefficient cell uptake, and undesired immune activation, as well as the inability to specifically reach the target tissue in the body, can be overcome by further developments in the field of nanoparticulate drug delivery. Therefore, types of surface modified siRNA nanoparticles are presented and illustrate how a more efficient and safer distribution of siRNA at the target site is possible by modifying the surface properties of nanoparticles with antibodies. However, the development of such efficient and safe delivery strategies is currently still a major challenge. In consideration of that, this review article aims to demonstrate the function and targeted delivery of siRNA nanoparticles, focusing on the surface modification via antibodies, various lipid- and polymer-components, and the therapeutic effects of these delivery systems.
Collapse
|
10
|
Wang C, Hong J, Yang Z, Zhou X, Yang Y, Kong Y, Chen B, Wu H, Qian BZ, Dimitrov DS, Zhou X, Wu Y, Ying T. Design of a Novel Fab-Like Antibody Fragment with Enhanced Stability and Affinity for Clinical use. SMALL METHODS 2022; 6:e2100966. [PMID: 35174992 DOI: 10.1002/smtd.202100966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/05/2021] [Indexed: 06/14/2023]
Abstract
With increasing interest in applying recombinant monoclonal antibodies (mAbs) in human medicine, engineered mAb fragments with reduced size and improved stability are in demand to overcome current limitations in clinical use. Herein, a novel Fab-like antibody fragment generated via an in silico-based engineering approach where the CH1 and CL domains of Fab are replaced by the IgG1 CH3 domains is described. This construct, designated as FabCH3, maintains the natural N-terminus and C-terminus of IgG antibody, can be expressed at a high level in bacterial cells and, importantly, exhibits much higher stability and affinity than the parental Fab when tested in a mesothelin-specific Fab m912, as well as a vascular endothelial growth factor A (VEGFA)-specific Fab Ranibizumab (in vivo). The high-resolution crystal structures of m912 FabCH3 and m912 Fab are determined, and the comparative analysis reveals more rigid structures in both constant domains and complementarity-determining regions of FabCH3, explaining its enhanced stability and affinity. Overall, the stabilized FabCH3 described in this report provides a versatile platform for engineering Fab-like antibody fragments with higher stability and antigen-binding affinity that can be used as a distinct class of antibody therapeutics.
Collapse
Affiliation(s)
- Chunyu Wang
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiaxu Hong
- Department of Ophthalmology and Vision Science, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
- Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Zhenlin Yang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China
| | - Xujiao Zhou
- Department of Ophthalmology and Vision Science, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Yuhan Yang
- Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yu Kong
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Binfan Chen
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Huifang Wu
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Bin-Zhi Qian
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Dimiter S Dimitrov
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Xingtao Zhou
- Department of Ophthalmology and Vision Science, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Yanling Wu
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Tianlei Ying
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| |
Collapse
|
11
|
A Novel Anti-B7-H3 × Anti-CD3 Bispecific Antibody with Potent Antitumor Activity. Life (Basel) 2022; 12:life12020157. [PMID: 35207448 PMCID: PMC8879513 DOI: 10.3390/life12020157] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 12/11/2022] Open
Abstract
B7-H3 plays an important role in tumor apoptosis, proliferation, adhesion, angiogenesis, invasion, migration, and evasion of immune surveillance. It is overexpressed in various human solid tumor tissues. In patients, B7-H3 overexpression correlates with advanced stages, poor clinical outcomes, and resistance to therapy. The roles of B7-H3 in tumor progression make it a potential candidate for targeted therapy. Here, we generated a mouse anti-human B7-H3 antibody and demonstrated its binding activity via Tongji University Suzhou Instituteprotein-based and cell-based assays. We then developed a novel format anti-B7-H3 × anti-CD3 bispecific antibody based on the antibody-binding fragment of the anti-B7-H3 antibody and single-chain variable fragment structure of anti-CD3 antibody (OKT3) and demonstrated that this bispecific antibody mediated potent cytotoxic activities against various B7-H3-positive tumor cell lines in vitro by improving T cell activation and proliferation. This bispecific antibody also demonstrated potent antitumor activity in humanized mice xenograft models. These results revealed that the novel anti-B7-H3 × anti-CD3 bispecific antibody has the potential to be employed in treatment of B7-H3-positive solid tumors.
Collapse
|
12
|
Jäger S, Dickgiesser S, Tonillo J, Hecht S, Kolmar H, Schröter C. EGFR binding Fc domain-drug conjugates: stable and highly potent cytotoxic molecules mediate selective cell killing. Biol Chem 2021; 403:525-534. [PMID: 34535048 DOI: 10.1515/hsz-2021-0321] [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: 07/13/2021] [Accepted: 09/07/2021] [Indexed: 11/15/2022]
Abstract
The exposition of cancer cells to cytotoxic doses of payload is fundamental for the therapeutic efficacy of antibody drug conjugates (ADCs) in solid cancers. To maximize payload exposure, tissue penetration can be increased by utilizing smaller-sized drug conjugates which distribute deeper into the tumor. Our group recently explored small human epidermal growth factor receptor 2 (HER2) targeting Fc antigen binding fragments (Fcabs) for ADC applications in a feasibility study. Here, we expand this concept using epidermal growth factor receptor (EGFR) targeting Fcabs for the generation of site-specific auristatin-based drug conjugates. In contrast to HER2-targeting Fcabs, we identified novel conjugation sites in the EGFR-targeting Fcab scaffold that allowed for higher DAR enzymatic conjugation. We demonstrate feasibility of resultant EGFR-targeting Fcab-drug conjugates that retain binding to half-life prolonging neonatal Fc receptor (FcRn) and EGFR and show high serum stability as well as target receptor mediated cell killing at sub-nanomolar concentrations. Our results emphasize the applicability of the Fcab format for the generation of drug conjugates designed for increased penetration of solid tumors and potential FcRn-driven antibody-like pharmacokinetics.
Collapse
Affiliation(s)
- Sebastian Jäger
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, D-64293Darmstadt, Germany.,Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Str. 4, D-64287Darmstadt, Germany
| | - Stephan Dickgiesser
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, D-64293Darmstadt, Germany
| | - Jason Tonillo
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, D-64293Darmstadt, Germany
| | - Stefan Hecht
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, D-64293Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Str. 4, D-64287Darmstadt, Germany
| | - Christian Schröter
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, D-64293Darmstadt, Germany
| |
Collapse
|
13
|
Jäger S, Wagner TR, Rasche N, Kolmar H, Hecht S, Schröter C. Generation and Biological Evaluation of Fc Antigen Binding Fragment-Drug Conjugates as a Novel Antibody-Based Format for Targeted Drug Delivery. Bioconjug Chem 2021; 32:1699-1710. [PMID: 34185508 DOI: 10.1021/acs.bioconjchem.1c00240] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Fragment crystallizable (Fc) antigen binding fragments (Fcabs) represent a novel antibody format comprising a homodimeric Fc region with an engineered antigen binding site. In contrast to their full-length antibody offspring, Fcabs combine Fc-domain-mediated and antigen binding functions at only one-third of the size. Their reduced size is accompanied by elevated tissue penetration capabilities, which is an attractive feature for the treatment of solid tumors. In the present study, we explored for the first time Fcabs as a novel scaffold for antibody-drug conjugates (ADCs). As model, various HER2-targeting Fcab variants coupled to a pH-sensitive dye were used in internalization experiments. A selective binding on HER2-expressing tumor cells and receptor-mediated endocytosis could be confirmed for selected variants, indicating that these Fcabs meet the basic prerequisite for an ADC approach. Subsequently, Fcabs were site-specifically coupled to cytotoxic monomethyl auristatin E yielding homogeneous conjugates. The conjugates retained HER2 and FcRn binding behavior of the parent Fcabs, showed a selective in vitro cell killing and conjugation site-dependent serum stability. Moreover, Fcab conjugates showed elevated penetration in a spheroid model, compared to their full-length antibody and Trastuzumab counterparts. Altogether, the presented results emphasize the potential of Fcabs as a novel scaffold for targeted drug delivery in solid cancers and pave the way for future in vivo translation.
Collapse
Affiliation(s)
- Sebastian Jäger
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany.,Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Str. 4, 64287 Darmstadt, Germany
| | - Tim R Wagner
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Str. 4, 64287 Darmstadt, Germany
| | - Nicolas Rasche
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Str. 4, 64287 Darmstadt, Germany
| | - Stefan Hecht
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Christian Schröter
- ADCs & Targeted NBE Therapeutics, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| |
Collapse
|
14
|
Kariolis MS, Wells RC, Getz JA, Kwan W, Mahon CS, Tong R, Kim DJ, Srivastava A, Bedard C, Henne KR, Giese T, Assimon VA, Chen X, Zhang Y, Solanoy H, Jenkins K, Sanchez PE, Kane L, Miyamoto T, Chew KS, Pizzo ME, Liang N, Calvert MEK, DeVos SL, Baskaran S, Hall S, Sweeney ZK, Thorne RG, Watts RJ, Dennis MS, Silverman AP, Zuchero YJY. Brain delivery of therapeutic proteins using an Fc fragment blood-brain barrier transport vehicle in mice and monkeys. Sci Transl Med 2021; 12:12/545/eaay1359. [PMID: 32461332 DOI: 10.1126/scitranslmed.aay1359] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 02/10/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
Abstract
Effective delivery of protein therapeutics to the central nervous system (CNS) has been greatly restricted by the blood-brain barrier (BBB). We describe the development of a BBB transport vehicle (TV) comprising an engineered Fc fragment that exploits receptor-mediated transcytosis for CNS delivery of biotherapeutics by binding a highly expressed brain endothelial cell target. TVs were engineered using directed evolution to bind the apical domain of the human transferrin receptor (hTfR) without the use of amino acid insertions, deletions, or unnatural appendages. A crystal structure of the TV-TfR complex revealed the TV binding site to be away from transferrin and FcRn binding sites, which was further confirmed experimentally in vitro and in vivo. Recombinant expression of TVs fused to anti-β-secretase (BACE1) Fabs yielded antibody transport vehicle (ATV) molecules with native immunoglobulin G (IgG) structure and stability. Peripheral administration of anti-BACE1 ATVs to hTfR-engineered mice and cynomolgus monkeys resulted in substantially improved CNS uptake and sustained pharmacodynamic responses. The TV platform readily accommodates numerous additional configurations, including bispecific antibodies and protein fusions, yielding a highly modular CNS delivery platform.
Collapse
Affiliation(s)
- Mihalis S Kariolis
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA.
| | - Robert C Wells
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Jennifer A Getz
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Wanda Kwan
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Cathal S Mahon
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Raymond Tong
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Do Jin Kim
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Ankita Srivastava
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Catherine Bedard
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Kirk R Henne
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Tina Giese
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Victoria A Assimon
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Xiaocheng Chen
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Yin Zhang
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Hilda Solanoy
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Katherine Jenkins
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Pascal E Sanchez
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Lesley Kane
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Takashi Miyamoto
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Kylie S Chew
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Michelle E Pizzo
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Nicholas Liang
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Meredith E K Calvert
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Sarah L DeVos
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | | | - Sejal Hall
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Zachary K Sweeney
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Robert G Thorne
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Ryan J Watts
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Mark S Dennis
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Adam P Silverman
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Y Joy Yu Zuchero
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA.
| |
Collapse
|
15
|
Therapeutic Antibodies Targeting Potassium Ion Channels. Handb Exp Pharmacol 2021; 267:507-545. [PMID: 33963460 DOI: 10.1007/164_2021_464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Monoclonal antibodies combine specificity and high affinity binding with excellent pharmacokinetic properties and are rapidly being developed for a wide range of drug targets including clinically important potassium ion channels. Nonetheless, while therapeutic antibodies come with great promise, K+ channels represent particularly difficult targets for biologics development for a variety of reasons that include their dynamic structures and relatively small extracellular loops, their high degree of sequence conservation (leading to immune tolerance), and their generally low-level expression in vivo. The process is made all the more difficult when large numbers of antibody candidates must be screened for a given target, or when lead candidates fail to cross-react with orthologous channels in animal disease models due to their highly selective binding properties. While the number of antibodies targeting potassium channels in preclinical or clinical development is still modest, significant advances in the areas of protein expression and antibody screening are converging to open the field to an avalanche of new drugs. Here, the opportunities and constraints associated with the discovery of antibodies against K+ channels are discussed, with an emphasis on novel technologies that are opening the field to exciting new possibilities for biologics development.
Collapse
|
16
|
Lešnik S, Hodošček M, Podobnik B, Konc J. Loop Grafting between Similar Local Environments for Fc-Silent Antibodies. J Chem Inf Model 2020; 60:5475-5486. [PMID: 32379970 PMCID: PMC7686954 DOI: 10.1021/acs.jcim.9b01198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Reduction
of the affinity of the fragment crystallizable (Fc) region with immune
receptors by substitution of one or a few amino acids, known as Fc-silencing,
is an established approach to reduce the immune effector functions
of monoclonal antibody therapeutics. This approach to Fc-silencing,
however, is problematic as it can lead to instability and immunogenicity
of the developed antibodies. We evaluated loop grafting as a novel
approach to Fc-silencing in which the Fc loops responsible for immune
receptor binding were replaced by loops of up to 20 amino acids from
similar local environments in other human and mouse antibodies. Molecular
dynamics simulations of the designed variants of an Fc region in a
complex with the immune receptor FcγIIIa confirmed that loop
grafting potentially leads to a significant reduction in the binding
of the antibody variants to the receptor, while retaining their stability.
In comparison, standard variants with less than eight substituted
amino acids showed possible instability and a lower degree of Fc-silencing
due to the occurrence of compensatory interactions. The presented
approach to Fc-silencing is general and could be used to modulate
undesirable side effects of other antibody therapeutics without affecting
their stability or increasing their immunogenicity.
Collapse
Affiliation(s)
- Samo Lešnik
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Milan Hodošček
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Barbara Podobnik
- Biologics Technical Development Mengeš, Technical Research & Development Novartis, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - Janez Konc
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
17
|
Kraman M, Faroudi M, Allen NL, Kmiecik K, Gliddon D, Seal C, Koers A, Wydro MM, Batey S, Winnewisser J, Young L, Tuna M, Doody J, Morrow M, Brewis N. FS118, a Bispecific Antibody Targeting LAG-3 and PD-L1, Enhances T-Cell Activation Resulting in Potent Antitumor Activity. Clin Cancer Res 2020; 26:3333-3344. [PMID: 32299814 DOI: 10.1158/1078-0432.ccr-19-3548] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/22/2020] [Accepted: 03/27/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Although programmed death-ligand 1 (PD-L1) antibody-based therapy has improved the outcome of patients with cancer, acquired resistance to these treatments limits their clinical efficacy. FS118 is a novel bispecific, tetravalent antibody (mAb2) against human lymphocyte activation gene-3 (LAG-3) and PD-L1 with the potential to reinvigorate exhausted immune cells and overcome resistance mechanisms to PD-L1 blockade. Here, using FS118 and a murine surrogate, we characterized the activity and report a novel mechanism of action of this bispecific antibody. EXPERIMENTAL DESIGN This study characterizes the binding activity and immune function of FS118 in cell lines and human peripheral blood mononuclear cells and further investigates its antitumor activity and mechanism of action using a surrogate murine bispecific antibody (mLAG-3/PD-L1 mAb2). RESULTS FS118 demonstrated simultaneous binding to LAG-3 and PD-L1 with high affinity and comparable or better activity than the combination of the single component parts of the mAb2 in blocking LAG-3- and PD-L1-mediated immune suppression and enhancing T-cell activity. In syngeneic tumor mouse models, mLAG-3/PD-L1 mAb2 significantly suppressed tumor growth. Mechanistic studies revealed decreased LAG-3 expression on T cells following treatment with the mouse surrogate mLAG-3/PD-L1 mAb2, whereas LAG-3 expression increased upon treatment with the combination of mAbs targeting LAG-3 and PD-L1. Moreover, following binding of mLAG-3/PD-L1 mAb2 to target-expressing cells, mouse LAG-3 is rapidly shed into the blood. CONCLUSIONS This study demonstrates a novel benefit of the bispecific approach over a combination of mAbs and supports the further development of FS118 for the treatment of patients with cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Claire Seal
- F-star Therapeutics Ltd, Cambridge, United Kingdom
| | | | | | - Sarah Batey
- F-star Therapeutics Ltd, Cambridge, United Kingdom
| | | | - Lesley Young
- F-star Therapeutics Ltd, Cambridge, United Kingdom
| | | | | | | | - Neil Brewis
- F-star Therapeutics Ltd, Cambridge, United Kingdom
| |
Collapse
|
18
|
Direct Blood Culturing of Candida spp. on Solid Medium by a Rapid Enrichment Method with Magnetic Beads Coated with Recombinant Human Mannan-Binding Lectin. J Clin Microbiol 2020; 58:JCM.00057-20. [PMID: 32051260 PMCID: PMC7098737 DOI: 10.1128/jcm.00057-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/02/2020] [Indexed: 12/23/2022] Open
Abstract
A rapid and accurate method to identify the species and antibiotic resistance of Candida spp. in blood is vital to increase the survival rates of patients with bloodstream infections. However, the extremely low levels of Candida spp. in blood make rapid diagnosis by standard blood culture difficult. In this study, we constructed a direct blood culturing method (i.e., the M1 method) by a rapid enrichment method with magnetic beads coated with a recombined human mannan-binding lectin (rhMBL; i. A rapid and accurate method to identify the species and antibiotic resistance of Candida spp. in blood is vital to increase the survival rates of patients with bloodstream infections. However, the extremely low levels of Candida spp. in blood make rapid diagnosis by standard blood culture difficult. In this study, we constructed a direct blood culturing method (i.e., the M1 method) by a rapid enrichment method with magnetic beads coated with a recombined human mannan-binding lectin (rhMBL; i.e., M1 protein), which demonstrated much higher Candida sp.-binding capacity than that of full-length MBL expressed in vitro (i.e., M2). With the M1 method, individual colonies were obtained before the standard blood culture method for each species of Candida spp. tested at <1 CFU/ml (an average of 29 h earlier). Additionally, the clinical sensitivity of the M1 method was 90.5% compared with that of the standard blood culture method when detecting frozen plasma from patients. More significantly, the turnaround time of the M1 method for blood culture could be reduced by approximately 37 to 43 h compared with that of the standard blood culture method in clinical sample identification.
Collapse
|
19
|
Marques AC, Costa PJ, Velho S, Amaral MH. Functionalizing nanoparticles with cancer-targeting antibodies: A comparison of strategies. J Control Release 2020; 320:180-200. [PMID: 31978444 DOI: 10.1016/j.jconrel.2020.01.035] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 01/07/2023]
Abstract
Standard cancer therapies sometimes fail to deliver chemotherapeutic drugs to tumor cells in a safe and effective manner. Nanotechnology takes the lead in providing new therapeutic options for cancer due to major potential for selective targeting and controlled drug release. Antibodies and antibody fragments are attracting much attention as a source of targeting ligands to bind specific receptors that are overexpressed on cancer cells. Therefore, researchers are devoting time and effort to develop targeting strategies based on nanoparticles functionalized with antibodies, which hold great promise to enhance therapeutic efficacy and circumvent severe side effects. Several methods have been described to immobilize antibodies on the surface of nanoparticles. However, selecting the most appropriate for each application is challenging but also imperative to preserve antigen binding ability and yield stable antibody-conjugated nanoparticles. From this perspective, we aim to provide considerable knowledge on the most widely used methods of functionalization that can be helpful for decision-making and design of conjugation protocols as well. This review summarizes adsorption, covalent conjugation (carbodiimide, maleimide and "click" chemistries) and biotin-avidin interaction, while discussing the advantages, limitations and relevant therapeutic approaches currently under investigation.
Collapse
Affiliation(s)
- A C Marques
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - P J Costa
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - S Velho
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, R. Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - M H Amaral
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
20
|
Lim YY, Lim TS, Choong YS. Human IgG1 Fc pH-dependent optimization from a constant pH molecular dynamics simulation analysis. RSC Adv 2020; 10:13066-13075. [PMID: 35492131 PMCID: PMC9051383 DOI: 10.1039/c9ra10712f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/11/2020] [Indexed: 11/21/2022] Open
Abstract
The binding of IgG Fc with FcRn enables the long circulating half-life of IgG, where the Fc–FcRn complex interacts in a pH-dependent manner. This complex shows stronger interaction at pH ≤ 6.5 and weaker interaction at pH ≥ 7.4. The Fc–FcRn binding mechanism that promotes the long circulating half-life of IgG has prompted several IgG Fc-related mutational studies to focus on the pH-dependent Fc–FcRn complex interactions in order to improve the pharmacokinetic properties of Fc. Hence, in this study, we applied the in silico constant pH molecular dynamics (CpHMD) simulation approach to evaluate the human Fc–FcRn complex binding (pH 6.0) and dissociating (pH 7.5) mechanism at the molecular level. The analysis showed that the protonated state of the titratable residues changes from pH 6.0 to pH 7.5, where the disrupting energy for Fc–FcRn complex formation was found to be due to the electrostatic repulsion between the complex. According to the analysis, an Fc variant was computationally designed with an improved binding affinity at pH 6.0, which is still able to dissociate at pH 7.5 with FcRn at the in silico level. The binding free energy calculation via the MMPB/GBSA approach showed that the designed Fc mutant (MutM4) has increased binding affinity only at pH 6.0 compared with the reported mutant (YTE) Fc. This work demonstrates an alternative Fc design with better binding properties for FcRn, which can be useful for future experimental evaluation and validation. An in silico IgG-Fc variant with better affinity at pH 6.0 but retained the dissociation at pH 7.5 was designed.![]()
Collapse
Affiliation(s)
- Yee Ying Lim
- Institute for Research in Molecular Medicine (INFORMM)
- Universiti Sains Malaysia
- 11800 Minden
- Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine (INFORMM)
- Universiti Sains Malaysia
- 11800 Minden
- Malaysia
| | - Yee Siew Choong
- Institute for Research in Molecular Medicine (INFORMM)
- Universiti Sains Malaysia
- 11800 Minden
- Malaysia
| |
Collapse
|
21
|
Vázquez NAR. Adsorption of terbium ion on Fc/dymethylacrylamide: application of Monte Carlo simulation. POLIMEROS 2020. [DOI: 10.1590/0104-1428.08419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
22
|
Fu M, He Q, Guo Z, Zhou X, Li H, Zhao L, Tang H, Zhou X, Zhu H, Shen G, He Y, Lei P. Therapeutic Bispecific T-Cell Engager Antibody Targeting the Transferrin Receptor. Front Immunol 2019; 10:1396. [PMID: 31293575 PMCID: PMC6598450 DOI: 10.3389/fimmu.2019.01396] [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: 02/26/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022] Open
Abstract
Bispecific T-cell engager antibodies (BiTE) have been explored as a means to recruit cytolytic T cells to kill tumor cells. The transferrin receptor (TfR) is highly expressed on the surface of rapidly proliferating tumor cells. Therefore, it holds great potential in T cell redirecting therapies. In this research, we developed a BiTE targeting TfR and CD3 (TfR-BiTE) and studied its therapeutic impact on TfR-positive cancer. TfR-BiTE had the ability to induce the selective lysis of various TfR-positive cancer cells through the activation of T cells, the release of cytokines, and then the coming proliferation of T cells, whereas TfR-negative cells were not affected. In a subcutaneous HepG2 xenograft model, low concentrations of TfR-BiTE inhibited tumor growth. Overall, these results reveal that TfR-BiTE can selectively deplete TfR-positive HepG2 cells; hence, it represents a novel immunotherapeutic approach for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mingpeng Fu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi He
- Department of Transfusion Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zilong Guo
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoran Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heli Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Zhao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongling Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huifen Zhu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong He
- Department of Nuclear Medicine and PET Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Wright TA, Page RC, Konkolewicz D. Polymer conjugation of proteins as a synthetic post-translational modification to impact their stability and activity. Polym Chem 2019; 10:434-454. [PMID: 31249635 PMCID: PMC6596429 DOI: 10.1039/c8py01399c] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
For more than 40 years, protein-polymer conjugates have been widely used for many applications, industrially and biomedically. These bioconjugates have been shown to modulate the activity and stability of various proteins while introducing reusability and new activities that can be used for drug delivery, improve pharmacokinetic ability, and stimuli-responsiveness. Techniques such as RDRP, ROMP and "click" have routinely been utilized for development of well-defined bioconjugate and polymeric materials. Synthesis of bioconjugate materials often take advantage of natural amino acids present within protein and peptide structures for a host of coupling chemistries. Polymer modification may elicit increased or decreased activity, activity retention under harsh conditions, prolonged activity in vivo and in vitro, and introduce stimuli responsiveness. Bioconjugation has resulted to modulated thermal stability, chemical stability, storage stability, half-life and reusability. In this review we aim to provide a brief state of the field, highlight a wide range of behaviors caused by polymer conjugation, and provide areas of future work.
Collapse
Affiliation(s)
- Thaiesha A Wright
- Department of Chemistry and Biochemistry, Miami University Oxford, Ohio 45056, United States
| | - Richard C Page
- Department of Chemistry and Biochemistry, Miami University Oxford, Ohio 45056, United States
| | - Dominik Konkolewicz
- Department of Chemistry and Biochemistry, Miami University Oxford, Ohio 45056, United States
| |
Collapse
|
24
|
Carter LM, Poty S, Sharma SK, Lewis JS. Preclinical optimization of antibody-based radiopharmaceuticals for cancer imaging and radionuclide therapy-Model, vector, and radionuclide selection. J Labelled Comp Radiopharm 2018; 61:611-635. [PMID: 29412489 PMCID: PMC6081268 DOI: 10.1002/jlcr.3612] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/19/2017] [Accepted: 01/16/2018] [Indexed: 12/25/2022]
Abstract
Intact antibodies and their truncated counterparts (eg, Fab, scFv fragments) are generally exquisitely specific and selective vectors, enabling recognition of individual cancer-associated molecular phenotypes against a complex and dynamic biomolecular background. Complementary alignment of these advantages with unique properties of radionuclides is a defining paradigm in both radioimmunoimaging and radioimmunotherapy, which remain some of the most adept and promising tools for cancer diagnosis and treatment. This review discusses how translational potency can be maximized through rational selection of antibody-nuclide couples for radioimmunoimaging/therapy in preclinical models.
Collapse
Affiliation(s)
- Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sophie Poty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Departments of Radiology and Pharmacology, Weill Cornell Medical College, New York, New York, USA
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
25
|
Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs 2017; 9:182-212. [PMID: 28071970 PMCID: PMC5297537 DOI: 10.1080/19420862.2016.1268307] [Citation(s) in RCA: 605] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022] Open
Abstract
During the past two decades we have seen a phenomenal evolution of bispecific antibodies for therapeutic applications. The 'zoo' of bispecific antibodies is populated by many different species, comprising around 100 different formats, including small molecules composed solely of the antigen-binding sites of two antibodies, molecules with an IgG structure, and large complex molecules composed of different antigen-binding moieties often combined with dimerization modules. The application of sophisticated molecular design and genetic engineering has solved many of the technical problems associated with the formation of bispecific antibodies such as stability, solubility and other parameters that confer drug properties. These parameters may be summarized under the term 'developability'. In addition, different 'target product profiles', i.e., desired features of the bispecific antibody to be generated, mandates the need for access to a diverse panel of formats. These may vary in size, arrangement, valencies, flexibility and geometry of their binding modules, as well as in their distribution and pharmacokinetic properties. There is not 'one best format' for generating bispecific antibodies, and no single format is suitable for all, or even most of, the desired applications. Instead, the bispecific formats collectively serve as a valuable source of diversity that can be applied to the development of therapeutics for various indications. Here, a comprehensive overview of the different bispecific antibody formats is provided.
Collapse
Affiliation(s)
- Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Im Nonnenwald, Penzberg, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Nobelstraße, Stuttgart, Germany
| |
Collapse
|
26
|
|
27
|
Lobner E, Humm AS, Mlynek G, Kubinger K, Kitzmüller M, Traxlmayr MW, Djinović-Carugo K, Obinger C. Two-faced Fcab prevents polymerization with VEGF and reveals thermodynamics and the 2.15 Å crystal structure of the complex. MAbs 2017; 9:1088-1104. [PMID: 28816592 PMCID: PMC5627596 DOI: 10.1080/19420862.2017.1364825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Fcabs (Fc domain with antigen-binding sites) are promising novel therapeutics. By engineering of the C-terminal loops of the CH3 domains, 2 antigen binding sites can be inserted in close proximity. To elucidate the binding mode(s) between homodimeric Fcabs and small homodimeric antigens, the interaction between the Fcabs 448 and CT6 (having the AB, CD and EF loops and the C-termini engineered) with homodimeric VEGF was investigated. The crystal structures of these Fcabs, which form polymers with the antigen VEGF in solution, were determined. However, construction of heterodimeric Fcabs (JanusFcabs: one chain Fc-wt, one chain VEGF-binding) results in formation of distinct JanusFcab–VEGF complexes (2:1), which allowed elucidation of the crystal structure of the JanusCT6–VEGF complex at 2.15 Å resolution. VEGF binding to Janus448 and JanusCT6 is shown to be entropically unfavorable, but enthalpically favorable. Structure-function relationships are discussed with respect to Fcab design and engineering strategies.
Collapse
Affiliation(s)
- Elisabeth Lobner
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| | - Anne-Sophie Humm
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,c Department for Structural and Computational Biology , Max F. Perutz Laboratories, University of Vienna , Dr. Bohr-Gasse 9, Vienna , Austria
| | - Georg Mlynek
- c Department for Structural and Computational Biology , Max F. Perutz Laboratories, University of Vienna , Dr. Bohr-Gasse 9, Vienna , Austria
| | - Konstantin Kubinger
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| | - Michael Kitzmüller
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| | - Michael W Traxlmayr
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| | - Kristina Djinović-Carugo
- c Department for Structural and Computational Biology , Max F. Perutz Laboratories, University of Vienna , Dr. Bohr-Gasse 9, Vienna , Austria.,d Department of Biochemistry, Faculty of Chemistry and Chemical Technology , University of Ljubljana , Večna pot 113, Ljubljana , Slovenia
| | - Christian Obinger
- a Christian Doppler Laboratory for Antibody Engineering , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria.,b Department of Chemistry, Division of Biochemistry , BOKU - University of Natural Resources and Life Sciences , Muthgasse 18, Vienna , Austria
| |
Collapse
|
28
|
Fcab-HER2 Interaction: a Ménage à Trois. Lessons from X-Ray and Solution Studies. Structure 2017; 25:878-889.e5. [PMID: 28528777 DOI: 10.1016/j.str.2017.04.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/10/2017] [Accepted: 04/28/2017] [Indexed: 01/07/2023]
Abstract
The crystallizable fragment (Fc) of the immunoglobulin class G (IgG) is an attractive scaffold for the design of novel therapeutics. Upon engineering the C-terminal loops in the CH3 domains, Fcabs (Fc domain with antigen-binding sites) can be designed. We present the first crystal structures of Fcabs, i.e., of the HER2-binding clone H10-03-6 having the AB and EF loop engineered and the stabilized version STAB19 derived by directed evolution. Comparison with the crystal structure of the Fc wild-type protein reveals conservation of the overall domain structures but significant differences in EF-loop conformations. Furthermore, we present the first Fcab-antigen complex structures demonstrating the interaction between the engineered Fcab loops with domain IV of HER2. The crystal structures of the STAB19-HER2 and H10-03-6-HER2 complexes together with analyses by isothermal titration calorimetry, SEC-MALS, and fluorescence correlation spectroscopy show that one homodimeric Fcab binds two HER2 molecules following a negative cooperative binding behavior.
Collapse
|
29
|
Cultivation of Pichia pastoris carrying the scFv anti LDL (-) antibody fragment. Effect of preculture carbon source. Braz J Microbiol 2017; 48:419-426. [PMID: 28237678 PMCID: PMC5498413 DOI: 10.1016/j.bjm.2016.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 11/11/2016] [Accepted: 11/28/2016] [Indexed: 12/27/2022] Open
Abstract
Antibodies and antibody fragments are nowadays among the most important biotechnological products, and Pichia pastoris is one of the most important vectors to produce them as well as other recombinant proteins. The conditions to effectively cultivate a P. pastoris strain previously genetically modified to produce the single-chain variable fragment anti low density lipoprotein (−) under the control of the alcohol oxidase promoter have been investigated in this study. In particular, it was evaluated if, and eventually how, the carbon source (glucose or glycerol) used in the preculture preceding cryopreservation in 20% glycerol influences both cell and antibody fragment productions either in flasks or in bioreactor. Although in flasks the volumetric productivity of the antibody fragment secreted by cells precultured, cryopreserved and reactivated in glycerol was 42.9% higher compared with cells precultured in glucose, the use of glycerol in bioreactor led to a remarkable shortening of the lag phase, thereby increasing it by no less than thrice compared to flasks. These results are quite promising in comparison with those reported in the literature for possible future industrial applications of this cultivation, taking into account that the overall process time was reduced by around 8 h.
Collapse
|
30
|
Hendriks D, Choi G, de Bruyn M, Wiersma VR, Bremer E. Antibody-Based Cancer Therapy: Successful Agents and Novel Approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:289-383. [PMID: 28325214 DOI: 10.1016/bs.ircmb.2016.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since their discovery, antibodies have been viewed as ideal candidates or "magic bullets" for use in targeted therapy in the fields of cancer, autoimmunity, and chronic inflammatory disorders. A wave of antibody-dedicated research followed, which resulted in the clinical approval of a first generation of monoclonal antibodies for cancer therapy such as rituximab (1997) and cetuximab (2004), and infliximab (2002) for the treatment of autoimmune diseases. More recently, the development of antibodies that prevent checkpoint-mediated inhibition of T cell responses invigorated the field of cancer immunotherapy. Such antibodies induced unprecedented long-term remissions in patients with advanced stage malignancies, most notably melanoma and lung cancer, that do not respond to conventional therapies. In this review, we will recapitulate the development of antibody-based therapy, and detail recent advances and new functions, particularly in the field of cancer immunotherapy. With the advent of recombinant DNA engineering, a number of rationally designed molecular formats of antibodies and antibody-derived agents have become available, and we will discuss various molecular formats including antibodies with improved effector functions, bispecific antibodies, antibody-drug conjugates, antibody-cytokine fusion proteins, and T cells genetically modified with chimeric antigen receptors. With these exciting advances, new antibody-based treatment options will likely enter clinical practice and pave the way toward more successful control of malignant diseases.
Collapse
Affiliation(s)
- D Hendriks
- Department of Surgery, Translational Surgical Oncology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - G Choi
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - M de Bruyn
- Department of Obstetrics & Gynecology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - V R Wiersma
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| | - E Bremer
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands; University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
31
|
Bispecific Antibodies as a Development Platform for New Concepts and Treatment Strategies. Int J Mol Sci 2016; 18:ijms18010048. [PMID: 28036020 PMCID: PMC5297683 DOI: 10.3390/ijms18010048] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022] Open
Abstract
With the development of molecular cloning technology and the deep understanding of antibody engineering, there are diverse bispecific antibody formats from which to choose to pursue the optimal biological activity and clinical purpose. The single-chain-based bispecific antibodies usually bridge tumor cells with immune cells and form an immunological synapse because of their relatively small size. Bispecific antibodies in the IgG format include asymmetric bispecific antibodies and homodimerized bispecific antibodies, all of which have an extended blood half-life and their own crystalline fragment (Fc)-mediated functions. Besides retargeting effector cells to the site of cancer, new applications were established for bispecific antibodies. Bispecific antibodies that can simultaneously bind to cell surface antigens and payloads are a very ideal delivery system for therapeutic use. Bispecific antibodies that can inhibit two correlated signaling molecules at the same time can be developed to overcome inherent or acquired resistance and to be more efficient angiogenesis inhibitors. Bispecific antibodies can also be used to treat hemophilia A by mimicking the function of factor VIII. Bispecific antibodies also have broad application prospects in bone disorders and infections and diseases of the central nervous system. The latest developments of the formats and application of bispecific antibodies will be reviewed. Furthermore, the challenges and perspectives are summarized in this review.
Collapse
|
32
|
Martin F. Antibodies as leading tools to unlock the therapeutic potential in human disease. Immunol Rev 2016; 270:5-7. [PMID: 26864100 DOI: 10.1111/imr.12410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
33
|
Vega L JCM, Lee MK, Qin EC, Rich M, Lee KY, Kim DH, Chung HJ, Leckband DE, Kong H. Three Dimensional Conjugation of Recombinant N-Cadherin to a Hydrogel for In Vitro Anisotropic Neural Growth. J Mater Chem B 2016; 4:6803-6811. [PMID: 28503305 DOI: 10.1039/c6tb01814a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Living cells are extensively being studied to build functional tissues that are useful for both fundamental and applied bioscience studies. Increasing evidence suggests that cell-cell adhesion controlled by intercellular cadherin junction plays important roles in the quality of the resulting engineered tissue. These findings prompted efforts to interrogate biological effects of cadherin at a molecular scale; however, few efforts were made to harness the effects of cadherin on cells cultured in an in vivo-like three dimensional matrix. To this end, this study reports a hydrogel matrix three dimensionally functionalized with a controlled number of Fc-tagged recombinant N-cadherins (N-Cad-Fc). To retain the desired conformation of N-Cad, these cadherins were immobilized and oriented to the gel by anti-Fc-antibodies chemically coupled to gels. The gels were processed to present N-Cad-Fc in uniaxially aligned microchannels or randomly oriented micropores. Culturing cortical cells in the functionalized gels generated a large fraction of neurons that are functional as indicated by increased intracellular calcium ion concentrations with the microchanneled gel. In contrast, direct N-Cad-Fc immobilization to microchannel or micropore walls of the gel limited the growth of neurons and increased the glial to neuron ratio. The results of this study will be highly useful to organize a wide array of cadherin molecules in a series of biomaterials used for three-dimensional cell culture and to regulate phenotypic activities of tissue-forming cells in an elaborate manner.
Collapse
Affiliation(s)
- Johana C M Vega L
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Min Kyung Lee
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ellen C Qin
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Max Rich
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Kwan Young Lee
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Dong Hyun Kim
- Department of Human and Culture Convergence Technology R&BD Group, Korea Institute of Industrial Technology, Ansan-si Gyeonggi-do, 426-910 South Korea
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Deborah E Leckband
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
34
|
Nilvebrant J, Tessier PM, Sidhu SS. Engineered Autonomous Human Variable Domains. Curr Pharm Des 2016; 22:6527-6537. [PMID: 27655414 PMCID: PMC5326600 DOI: 10.2174/1381612822666160921143011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND The complex multi-chain architecture of antibodies has spurred interest in smaller derivatives that retain specificity but can be more easily produced in bacteria. Domain antibodies consisting of single variable domains are the smallest antibody fragments and have been shown to possess enhanced ability to target epitopes that are difficult to access using multidomain antibodies. However, in contrast to natural camelid antibody domains, human variable domains typically suffer from low stability and high propensity to aggregate. METHODS This review summarizes strategies to improve the biophysical properties of heavy chain variable domains from human antibodies with an emphasis on aggregation resistance. Several protein engineering approaches have targeted antibody frameworks and complementarity determining regions to stabilize the native state and prevent aggregation of the denatured state. CONCLUSION Recent findings enable the construction of highly diverse libraries enriched in aggregation-resistant variants that are expected to provide binders to diverse antigens. Engineered domain antibodies possess unique advantages in expression, epitope preference and flexibility of formatting over conventional immunoreagents and are a promising class of antibody fragments for biomedical development.
Collapse
Affiliation(s)
- Johan Nilvebrant
- Division of Protein Technology, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
- Donnelly Centre for Cellular and Biomolecular Research, Banting and Best Department of Medical Research, University of Toronto, Toronto, Canada
| | - Peter M. Tessier
- Center for Biotechnology and Interdisciplinary Studies, Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Sachdev S. Sidhu
- Donnelly Centre for Cellular and Biomolecular Research, Banting and Best Department of Medical Research, University of Toronto, Toronto, Canada
| |
Collapse
|