251
|
Kaltbeitzel J, Wich PR. Protein-based Nanoparticles: From Drug Delivery to Imaging, Nanocatalysis and Protein Therapy. Angew Chem Int Ed Engl 2023; 62:e202216097. [PMID: 36917017 DOI: 10.1002/anie.202216097] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/16/2023]
Abstract
Proteins and enzymes are versatile biomaterials for a wide range of medical applications due to their high specificity for receptors and substrates, high degradability, low toxicity, and overall good biocompatibility. Protein nanoparticles are formed by the arrangement of several native or modified proteins into nanometer-sized assemblies. In this review, we will focus on artificial nanoparticle systems, where proteins are the main structural element and not just an encapsulated payload. While under natural conditions, only certain proteins form defined aggregates and nanoparticles, chemical modifications or a change in the physical environment can further extend the pool of available building blocks. This allows the assembly of many globular proteins and even enzymes. These advances in preparation methods led to the emergence of new generations of nanosystems that extend beyond transport vehicles to diverse applications, from multifunctional drug delivery to imaging, nanocatalysis and protein therapy.
Collapse
Affiliation(s)
- Jonas Kaltbeitzel
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
252
|
Wu KW, Chen TH, Yang TC, Wang SC, Shameem M, Graham KS. Continuous monitoring of a monoclonal antibody by size exclusion chromatography reveals a correlation between system suitability parameters and column aging. J Pharm Biomed Anal 2023; 235:115622. [PMID: 37540994 DOI: 10.1016/j.jpba.2023.115622] [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: 05/25/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Size exclusion chromatography (SEC) is a foundational analytical method to assess product purity of biological molecules. To ensure accurate and reproducible data that meet regulatory agency standards, it is critical to monitor the chromatographic column with efficient and continuous approaches. In this study, 19 SEC columns (Waters Acquity BEH200) were evaluated using an in-house monoclonal antibody made at Regeneron. System suitability parameters (SSPs) were used to monitor the performance of the SEC assay, including USP resolution, USP plate count, USP tailing factor, asymmetry factor, elution time, peak width, and peak height. A general linear model was built and revealed that elution time, peak width, asymmetry factor, and tailing factor increased with injection number, while peak height, resolution, and plate count decreased. After 1000 injections, tailing factor and peak width increased by more than 10%, while resolution and plate count decreased by more than 10% from their respective starting values.
Collapse
Affiliation(s)
- Kai-Wei Wu
- Department of Pharmaceutics and Drug Delivery, The University of Mississippi, University, MS 38677, USA
| | - Tse-Hong Chen
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA.
| | - Teng-Chieh Yang
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Shao-Chun Wang
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Mohammed Shameem
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Kenneth S Graham
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| |
Collapse
|
253
|
Ma CH, Chen CL, Hsu CC. Real-time bottom-up characterization of protein mixtures enabled by online microdroplet-assisted enzymatic digestion (MAED). Chem Commun (Camb) 2023; 59:12585-12588. [PMID: 37789821 DOI: 10.1039/d3cc03509c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Enzymatic digestion remains one of the "rate-determining steps" in the bottom-up analysis of proteins. However, by performing digestion in microdroplets generated from electrosonic spray, the reaction could be accelerated to a timescale lower than milliseconds. Here, we describe a simple and rapid online digestion platform named online microdroplet-assisted enzymatic digestion (MAED). It involves the integration of intact protein separation with enzymatic digestion in microdroplets. Via online MAED, various protein standards, including an antibody standard, were characterized in a bottom-up manner without prior digestion, and high sequence coverages were obtained. We further extended the application of online MAED to a more complex sample, mouse brain extract, where protein identifications were successfully yielded. Compared with the conventional bottom-up approach, a more comprehensive characterization could be obtained particularly for low molecular weight proteins. In short, we provide a rapid and alternative bottom-up analysis in a top-down fashion as well as a new possibility for microdroplet chemistry.
Collapse
Affiliation(s)
- Cheng-Hua Ma
- Department of Chemistry, National Taiwan University, Taipei, 106216, Taiwan.
| | - Chih-Lin Chen
- Department of Chemistry, National Taiwan University, Taipei, 106216, Taiwan.
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, 106216, Taiwan.
| |
Collapse
|
254
|
Ren J, Jepson CE, Nealy SL, Kuhlmann CJ, Osuka S, Azolibe SU, Blucas MT, Nagaoka-Kamata Y, Kharlampieva E, Kamata M. Site-oriented conjugation of poly(2-methacryloyloxyethyl phosphorylcholine) for enhanced brain delivery of antibody. Front Cell Dev Biol 2023; 11:1214118. [PMID: 37920826 PMCID: PMC10618420 DOI: 10.3389/fcell.2023.1214118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023] Open
Abstract
Antibody therapeutics are limited in treating brain diseases due to poor blood-brain barrier (BBB) penetration. We have discovered that poly 2-methacryloyloxyethyl phosphorylcholine (PMPC), a biocompatible polymer, effectively facilitates BBB penetration via receptor-mediated transcytosis and have developed a PMPC-shell-based platform for brain delivery of therapeutic antibodies, termed nanocapsule. Yet, the platform results in functional loss of antibodies due to epitope masking by the PMPC polymer network, which necessitates the incorporation of a targeting moiety and degradable crosslinker to enable on-site antibody release. In this study, we developed a novel platform based on site-oriented conjugation of PMPC to the antibody, allowing it to maintain key functionalities of the original antibody. With an optimized PMPC chain length, the PMPC-antibody conjugate exhibited enhanced brain delivery while retaining epitope recognition, cellular internalization, and antibody-dependent cellular phagocytic activity. This simple formula incorporates only the antibody and PMPC without requiring additional components, thereby addressing the issues of the nanocapsule platform and paving the way for PMPC-based brain delivery strategies for antibodies.
Collapse
Affiliation(s)
- Jie Ren
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Chloe E. Jepson
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sarah L. Nealy
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Charles J. Kuhlmann
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Satoru Osuka
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stella Uloma Azolibe
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Madison T. Blucas
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yoshiko Nagaoka-Kamata
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eugenia Kharlampieva
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Masakazu Kamata
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
255
|
Wang P, Jin SY. Meta-analysis of the efficacy and safety of daratumumab in the treatment of multiple myeloma. World J Clin Cases 2023; 11:7091-7100. [PMID: 37946760 PMCID: PMC10631397 DOI: 10.12998/wjcc.v11.i29.7091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/12/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND The treatment of multiple myeloma has significantly progressed over the past half-century. The purpose of this study was to perform a systematic review and meta-analysis in order to explore the efficacy and safety of daratumumab in treating multiple myeloma. AIM To explore the efficacy and safety of daratumumab in treating multiple myeloma. METHODS A systematic literature search was performed using Chinese and English databases, including the China National Knowledge Infrastructure, Wanfang, China Biology Medicine, VIP, the Cochrane Library, Embase, and PubMed. The search encompassed studies in treating multiple myeloma with daratumumab, spanning from the inception of the database to June 2023. Revman 5.1 software was used for analysis. RESULTS Our analysis included eight English articles and one Chinese article of high quality. The meta-analysis results indicated that compared to other therapies, daratumumab could improve the overall response rate (ORR) [odds ratio (OR) = 2.67, 95% confidence interval (CI) = 2.01, 3.53, Z = 6.85, P < 0.00001], complete remission (CR) (OR = 2.87, 95%CI = 2.16, 3.83, Z = 7.23, P < 0.00001) and progression-free survival (PFS) time (hazard ratio = 0.48, 95%CI = 0.38,0.60, Z = 6.54, P < 0.00001) in patients with multiple myeloma. These differences were statistically significant. Additionally, these results suggested that daratumumab increases the risk of neutropenia and thrombocytopenia with minimal effect on the incidences of anemia and upper respiratory tract infections. CONCLUSION Daratumumab can improve ORR, CR rate, and PFS in patients with multiple myeloma. It also increases the risk of neutropenia and thrombocytopenia, necessitating careful monitoring during its clinical application.
Collapse
Affiliation(s)
- Pei Wang
- Department of Hematology, Yanbian University Hospital, Yanji 133000, Jilin Province, China
| | - Sheng-Yu Jin
- Department of Hematology, Yanbian University Hospital, Yanji 133000, Jilin Province, China
| |
Collapse
|
256
|
Beaumont VA, Liu L, Shi H, Rouse JC, Kim HY. Application of NMR and Chemometric Analyses to Better Understand the Quality Attributes in pH and Thermally Degraded Monoclonal Antibodies. Pharm Res 2023; 40:2457-2467. [PMID: 37798537 PMCID: PMC10661726 DOI: 10.1007/s11095-023-03600-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 08/30/2023] [Indexed: 10/07/2023]
Abstract
PURPOSE Nuclear magnetic resonance (NMR) spectroscopy provides the sensitivity and specificity to probe the higher order structure (HOS) of monoclonal antibodies (mAbs) for potential changes. This study demonstrates an application of chemometric tools to measure differences in the NMR spectra of mAbs after forced degradation relative to the respective unstressed starting materials. METHODS Samples of adalimumab (Humira, ADL-REF) and trastuzumab (Herceptin, TRA-REF) were incubated in three buffer-pH conditions at 40°C for 4 weeks to compare to a control sample that was left unstressed. Replicate 1D 1H and 2D 1H-13C HMQC NMR spectra were collected on all samples. Chemometric analyses such as Easy Comparability of HOS (ECHOS), PROtein FIngerprinting by Lineshape Enhancement (PROFILE), and Principal Component Analysis (PCA) were applied to capture and quantitate differences between the spectra. RESULTS Visual and statistical inspection of the 2D 1H-13C HMQC spectra of adalimumab and trastuzumab after forced degradation conditions shows no changes in the spectra relative to the unstressed material. Chemometric analysis of the 1D 1H NMR spectra shows only minor changes in the spectra of adalimumab after forced degradation, but significant differences in trastuzumab. CONCLUSION The chemometric analyses support the lack of statistical differences in the structure of pH-thermal stressed adalimumab, however, it reveals conformational changes or chemical modifications in trastuzumab after forced degradation. Application of chemometrics in comparative NMR studies enables HOS characterization and showcases the sensitivity and specificity in detecting differences in the spectra of mAbs after pH-thermal forced degradation with respect to local and global protein structure.
Collapse
Affiliation(s)
- Victor A Beaumont
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, 1 Burtt Road, Andover, MA, 01810, USA.
- Pfizer, Inc. Pharmaceutical Sciences Small Molecules, Analytical Research and Development, Discovery Park, Ramsgate Road, Sandwich, CT13 9FF, UK.
| | - Lucy Liu
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, 1 Burtt Road, Andover, MA, 01810, USA
| | - Heliang Shi
- Pfizer, Inc. Global Product Development, Oncology & Rare Disease Statistics, New York City, NY, 10001, USA
| | - Jason C Rouse
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, 1 Burtt Road, Andover, MA, 01810, USA
| | - Hai-Young Kim
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, 1 Burtt Road, Andover, MA, 01810, USA.
| |
Collapse
|
257
|
Zhou Y, Huang Z, Li W, Wei J, Jiang Q, Yang W, Huang J. Deep learning in preclinical antibody drug discovery and development. Methods 2023; 218:57-71. [PMID: 37454742 DOI: 10.1016/j.ymeth.2023.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/20/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
Antibody drugs have become a key part of biotherapeutics. Patients suffering from various diseases have benefited from antibody therapies. However, its development process is rather long, expensive and risky. To speed up the process, reduce cost and improve success rate, artificial intelligence, especially deep learning methods, have been widely used in all aspects of preclinical antibody drug development, from library generation to hit identification, developability screening, lead selection and optimization. In this review, we systematically summarize antibody encodings, deep learning architectures and models used in preclinical antibody drug discovery and development. We also critically discuss challenges and opportunities, problems and possible solutions, current applications and future directions of deep learning in antibody drug development.
Collapse
Affiliation(s)
- Yuwei Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Ziru Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Wenzhen Li
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jinyi Wei
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Qianhu Jiang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Wei Yang
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jian Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China.
| |
Collapse
|
258
|
Pang KT, Yang YS, Zhang W, Ho YS, Sormanni P, Michaels TCT, Walsh I, Chia S. Understanding and controlling the molecular mechanisms of protein aggregation in mAb therapeutics. Biotechnol Adv 2023; 67:108192. [PMID: 37290583 DOI: 10.1016/j.biotechadv.2023.108192] [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: 02/16/2023] [Revised: 05/09/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
In antibody development and manufacturing, protein aggregation is a common challenge that can lead to serious efficacy and safety issues. To mitigate this problem, it is important to investigate its molecular origins. This review discusses (1) our current molecular understanding and theoretical models of antibody aggregation, (2) how various stress conditions related to antibody upstream and downstream bioprocesses can trigger aggregation, and (3) current mitigation strategies employed towards inhibiting aggregation. We discuss the relevance of the aggregation phenomenon in the context of novel antibody modalities and highlight how in silico approaches can be exploited to mitigate it.
Collapse
Affiliation(s)
- Kuin Tian Pang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore; School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technology University, Singapore
| | - Yuan Sheng Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Wei Zhang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Pietro Sormanni
- Chemistry of Health, Yusuf Hamied Department of Chemistry, University of Cambridge, United Kingdom
| | - Thomas C T Michaels
- Department of Biology, Institute of Biochemistry, ETH Zurich, Otto-Stern-Weg 3, 8093 Zurich, Switzerland; Bringing Materials to Life Initiative, ETH Zurich, Switzerland
| | - Ian Walsh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Sean Chia
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
259
|
Rocamora F, Peralta AG, Shin S, Sorrentino J, Wu MYM, Toth EA, Fuerst TR, Lewis NE. Glycosylation shapes the efficacy and safety of diverse protein, gene and cell therapies. Biotechnol Adv 2023; 67:108206. [PMID: 37354999 PMCID: PMC11168894 DOI: 10.1016/j.biotechadv.2023.108206] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Over recent decades, therapeutic proteins have had widespread success in treating a myriad of diseases. Glycosylation, a near universal feature of this class of drugs, is a critical quality attribute that significantly influences the physical properties, safety profile and biological activity of therapeutic proteins. Optimizing protein glycosylation, therefore, offers an important avenue to developing more efficacious therapies. In this review, we discuss specific examples of how variations in glycan structure and glycoengineering impacts the stability, safety, and clinical efficacy of protein-based drugs that are already in the market as well as those that are still in preclinical development. We also highlight the impact of glycosylation on next generation biologics such as T cell-based cancer therapy and gene therapy.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Angelo G Peralta
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seunghyeon Shin
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - James Sorrentino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mina Ying Min Wu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric A Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
260
|
Prado NDR, Brilhante-Da-Silva N, Sousa RMO, Morais MSDS, Roberto SA, Luiz MB, Assis LCD, Marinho ACM, Araujo LFLD, Pontes RDS, Stabeli RG, Fernandes CFC, Pereira SDS. Single-domain antibodies applied as antiviral immunotherapeutics. J Virol Methods 2023; 320:114787. [PMID: 37516366 DOI: 10.1016/j.jviromet.2023.114787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Viral infections have been the cause of high mortality rates throughout different periods in history. Over the last two decades, outbreaks caused by zoonotic diseases and transmitted by arboviruses have had a significant impact on human health. The emergence of viral infections in different parts of the world encourages the search for new inputs to fight pathologies of viral origin. Antibodies represent the predominant class of new drugs developed in recent years and approved for the treatment of various human diseases, including cancer, autoimmune and infectious diseases. A promising group of antibodies are single-domain antibodies derived from camelid heavy chain immunoglobulins, or VHHs, are biomolecules with nanometric dimensions and unique pharmaceutical and biophysical properties that can be used in the diagnosis and immunotherapy of viral infections. For viral neutralization to occur, VHHs can act in different stages of the viral cycle, including the actual inhibition of infection, to hindering viral replication or assembly. This review article addresses advances involving the use of VHHs in therapeutic propositions aimed to battle different viruses that affect human health.
Collapse
Affiliation(s)
- Nidiane Dantas Reis Prado
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil
| | - Nairo Brilhante-Da-Silva
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil
| | - Rosa Maria Oliveira Sousa
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil
| | | | - Sibele Andrade Roberto
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil
| | - Marcos Barros Luiz
- Instituto Federal de Rondônia Campus Guajará-Mirim, IFRO, Guajará-Mirim, RO, Brazil
| | - Livia Coelho de Assis
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil
| | - Anna Carolina M Marinho
- Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Luiz Felipe Lemes de Araujo
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade de São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Rafael de Souza Pontes
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade de São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Rodrigo Guerino Stabeli
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil
| | - Carla Freire Celedonio Fernandes
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Soraya Dos Santos Pereira
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
261
|
Porębska N, Ciura K, Chorążewska A, Zakrzewska M, Otlewski J, Opaliński Ł. Multivalent protein-drug conjugates - An emerging strategy for the upgraded precision and efficiency of drug delivery to cancer cells. Biotechnol Adv 2023; 67:108213. [PMID: 37453463 DOI: 10.1016/j.biotechadv.2023.108213] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/20/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
With almost 20 million new cases per year, cancer constitutes one of the most important challenges for public health systems. Unlike traditional chemotherapy, targeted anti-cancer strategies employ sophisticated therapeutics to precisely identify and attack cancer cells, limiting the impact of drugs on healthy cells and thereby minimizing the unwanted side effects of therapy. Protein drug conjugates (PDCs) are a rapidly growing group of targeted therapeutics, composed of a cancer-recognition factor covalently coupled to a cytotoxic drug. Several PDCs, mainly in the form of antibody-drug conjugates (ADCs) that employ monoclonal antibodies as cancer-recognition molecules, are used in the clinic and many PDCs are currently in clinical trials. Highly selective, strong and stable interaction of the PDC with the tumor marker, combined with efficient, rapid endocytosis of the receptor/PDC complex and its subsequent effective delivery to lysosomes, is critical for the efficacy of targeted cancer therapy with PDCs. However, the bivalent architecture of contemporary clinical PDCs is not optimal for tumor receptor recognition or PDCs internalization. In this review, we focus on multivalent PDCs, which represent a rapidly evolving and highly promising therapeutics that overcome most of the limitations of current bivalent PDCs, enhancing the precision and efficiency of drug delivery to cancer cells. We present an expanding set of protein scaffolds used to generate multivalent PDCs that, in addition to folding into well-defined multivalent molecular structures, enable site-specific conjugation of the cytotoxic drug to ensure PDC homogeneity. We provide an overview of the architectures of multivalent PDCs developed to date, emphasizing their efficacy in the targeted treatment of various cancers.
Collapse
Affiliation(s)
- Natalia Porębska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Krzysztof Ciura
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Aleksandra Chorążewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| |
Collapse
|
262
|
Ariani A, Khotimah H, Nurdiana N, Rahayu M. Asiatic acid increased locomotor and head width by inducing brain-derived neurotrophic factor in intrauterine hypoxia-exposed zebrafish. Open Vet J 2023; 13:1326-1333. [PMID: 38027402 PMCID: PMC10658027 DOI: 10.5455/ovj.2023.v13.i10.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/21/2023] [Indexed: 12/01/2023] Open
Abstract
Background Hypoxia ischemia leads to abnormal behavior and growth. Prenatal hypoxia also decreases brain adaptive potential, which can cause fatal effects such as cell death. Asiatic acid (AA) in Centella asiatica is a neuroprotector through antioxidant and anti-inflammatory activities. Aim This study aimed to analyze the effect of AA as a neuroprotector against hypoxia during intrauterine development on locomotor activity, head width, and brain-derived neurotrophic factor (BDNF) expression. Methods The true experimental laboratory research used a posttest control-only design. Zebrafish embryos (Danio rerio) aged 0-2 dpf (days postfertilization) were exposed to hypoxia with oxygen levels reaching 1.5 mg/l. Then, AA was administered at successive concentrations, namely, 0.36, 0.72, and 1.45 μg/ml, at 2 hpf (hours postfertilization), 3, 6, and 9 dpf. Head width, velocity activity, and BDNF expression were observed. Results Intrauterine hypoxia significantly decreased head width, velocity rate, and BDNF expression (<0.001). Administration of AA at all concentrations and age 9 dpf to zebrafish larvae with intrauterine hypoxia exposure increased head width ( p < 0.0001), velocity (p < 0.05), and relative mRNA expression of BDNF (p < 0.05). Conclusion AA is potentially neuroprotective to the brain in zebrafish larvae exposed to hypoxia during intrauterine development.
Collapse
Affiliation(s)
- Ariani Ariani
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Husnul Khotimah
- Department of Pharmacology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Nurdiana Nurdiana
- Department of Pharmacology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Masruroh Rahayu
- Department of Neurology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
263
|
Shkunnikova S, Mijakovac A, Sironic L, Hanic M, Lauc G, Kavur MM. IgG glycans in health and disease: Prediction, intervention, prognosis, and therapy. Biotechnol Adv 2023; 67:108169. [PMID: 37207876 DOI: 10.1016/j.biotechadv.2023.108169] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023]
Abstract
Immunoglobulin (IgG) glycosylation is a complex enzymatically controlled process, essential for the structure and function of IgG. IgG glycome is relatively stable in the state of homeostasis, yet its alterations have been associated with aging, pollution and toxic exposure, as well as various diseases, including autoimmune and inflammatory diseases, cardiometabolic diseases, infectious diseases and cancer. IgG is also an effector molecule directly involved in the inflammation processes included in the pathogenesis of many diseases. Numerous recently published studies support the idea that IgG N-glycosylation fine-tunes the immune response and plays a significant role in chronic inflammation. This makes it a promising novel biomarker of biological age, and a prognostic, diagnostic and treatment evaluation tool. Here we provide an overview of the current state of knowledge regarding the IgG glycosylation in health and disease, and its potential applications in pro-active prevention and monitoring of various health interventions.
Collapse
Affiliation(s)
- Sofia Shkunnikova
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, Zagreb, Croatia
| | - Anika Mijakovac
- University of Zagreb, Faculty of Science, Department of Biology, Horvatovac 102a, Zagreb, Croatia
| | - Lucija Sironic
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, Zagreb, Croatia
| | - Maja Hanic
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, Zagreb, Croatia
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, Zagreb, Croatia; University of Zagreb, Faculty of Pharmacy and Biochemistry, Ulica Ante Kovačića 1, Zagreb, Croatia
| | | |
Collapse
|
264
|
Mena Lora AJ, Echeverria SL, Lindsey B, Li E, Sanchez L, Truesdell T, Takhsh E, Lavani R, Burgos R. Feasibility and impact of a monoclonal antibody infusion program in reaching vulnerable underserved communities. Infect Control Hosp Epidemiol 2023; 44:1690-1692. [PMID: 37855076 PMCID: PMC10587392 DOI: 10.1017/ice.2023.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 10/20/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has disproportionately impacted Black, indigenous, and people of color (BIPOC). Equitable access to therapeutics is key to addressing health disparities. We established a monoclonal infusion program in the emergency department of a safety-net hospital. Our program successfully reached underserved BIPOC communities and was sustained throughout the pandemic.
Collapse
Affiliation(s)
- Alfredo J. Mena Lora
- University of Illinois at Chicago, Chicago, Illinois
- Saint Anthony Hospital, Chicago, Illinois
| | | | | | - Ella Li
- Saint Anthony Hospital, Chicago, Illinois
| | | | | | | | | | | |
Collapse
|
265
|
Pan M, Liu Y, Sang T, Xie J, Lin H, Wei J, Shao S, Zheng Y, Zhang J. Enhanced antitumor and anti-metastasis by VEGFR2-targeted doxorubicin immunoliposome synergy with NK cell activation. Invest New Drugs 2023; 41:664-676. [PMID: 37542666 DOI: 10.1007/s10637-023-01372-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/08/2023] [Indexed: 08/07/2023]
Abstract
Liposomal doxorubicin exhibits stronger drug accumulation at the tumor site due to the Enhanced Permeability and Retention (EPR) effect. However, the prognosis for the patient is poor due to this drug's lack of targeting and tumor metastasis during treatment. Vascular epidermal growth factor receptor (VEGFR2) plays an important role in angiogenesis and cancer metastasis. To enhance antitumor efficacy of PEGylated liposomal doxorubicin, we constructed a VEGFR2-targeted and doxorubicin-loaded immunoliposome (Lipo-DOX-C00) by conjugating a VEGFR2-specific, single chain antibody fragment to DSPE-PEG2000-MAL, and then we inserted the antibody-conjugated polymer into liposomal doxorubicin (Lipo-DOX). The immunoliposome was formed uniformly with high affinity for VEGFR2. In vitro, Lipo-DOX-C00 enhanced doxorubicin internalization into LLC and 4T1 cells compared with non-conjugated, liposomal doxorubicin. In vivo, Lipo-DOX-C00 delivered DOX to tumor tissues effectively, which exhibited an improved antitumor and anti-metastasis efficacy in both LLC subcutaneous tumor models and 4T1 tumor models. In addition, the combined therapy of a VEGFR2-MICA bispecific antibody (JZC01) and Lipo-DOX-C00 achieved enhanced inhibition of cancer growth and metastasis due to activation of the immune system. Our study provides a promising approach to clinical application of liposomal doxorubicin.
Collapse
Affiliation(s)
- Mingzhu Pan
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yali Liu
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Tian Sang
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiajun Xie
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Huishu Lin
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Jianpeng Wei
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Shuai Shao
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yanying Zheng
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Juan Zhang
- Antibody Engineering Laboratory, School of Life Science & Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
266
|
Yoshikawa M, Nakamura H, Oda-Ueda N, Ohkuri T. Analysis of thermostability for seven Phe to Ala and six Pro to Gly mutants in the Fab constant region of adalimumab. J Biochem 2023; 174:345-353. [PMID: 37390406 DOI: 10.1093/jb/mvad047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023] Open
Abstract
To identify amino acids that play important roles in the structural stability of Fab, seven phenylalanine residues in the Fab constant region of the therapeutic antibody adalimumab were subjected to alanine mutagenesis. Six Fab mutants, H:F130A, H:F154A, H:F174A, L:F118A, L:F139A and L:F209A, showed decreased thermostability compared with wild-type Fab. In contrast, the Tm for the L:F116A mutant was 1.7°C higher than that of wild-type Fab, indicating that the F116 residue was unfavorable for Fab thermostability. Six proline mutants, H:P131G, H:P155G, H:P175G, L:P119G, L:P120G and L:P141G, were also prepared to investigate the effect of proline residues adjacent to mutated phenylalanine residues. The thermostability of the H:P155G and L:P141G mutants in particular was significantly reduced, with decreases in Tm of 5.0 and 3.0°C, respectively, compared with wild-type Fab. The H:P155 and L:P141 residues have a cis conformation, whereas the other mutated proline residues have a trans conformation. H:P155 and L:P141 had stacking interactions with the H:F154 and L:Y140, respectively, at the interface between the variable and constant regions. It is suggested that the interactions of the aromatic ring with a cis-form proline at the interface between the variable and constant regions is important for stability of Fab.
Collapse
Affiliation(s)
- Moeka Yoshikawa
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Hitomi Nakamura
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Naoko Oda-Ueda
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Takatoshi Ohkuri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| |
Collapse
|
267
|
Schoenfeld K, Harwardt J, Habermann J, Elter A, Kolmar H. Conditional activation of an anti-IgM antibody-drug conjugate for precise B cell lymphoma targeting. Front Immunol 2023; 14:1258700. [PMID: 37841262 PMCID: PMC10569071 DOI: 10.3389/fimmu.2023.1258700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/07/2023] [Indexed: 10/17/2023] Open
Abstract
Cancerous B cells are almost indistinguishable from their non-malignant counterparts regarding their surface antigen expression. Accordingly, the challenge to be faced consists in elimination of the malignant B cell population while maintaining a functional adaptive immune system. Here, we present an IgM-specific antibody-drug conjugate masked by fusion of the epitope-bearing IgM constant domain. Antibody masking impaired interaction with soluble pentameric as well as cell surface-expressed IgM molecules rendering the antibody cytotoxically inactive. Binding capacity of the anti-IgM antibody drug conjugate was restored upon conditional protease-mediated demasking which consequently enabled target-dependent antibody internalization and subsequent induction of apoptosis in malignant B cells. This easily adaptable approach potentially provides a novel mechanism of clonal B cell lymphoma eradication to the arsenal available for non-Hodgkin's lymphoma treatment.
Collapse
Affiliation(s)
- Katrin Schoenfeld
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jan Habermann
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Adrian Elter
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
268
|
Abdeldaim DT, Schindowski K. Fc-Engineered Therapeutic Antibodies: Recent Advances and Future Directions. Pharmaceutics 2023; 15:2402. [PMID: 37896162 PMCID: PMC10610324 DOI: 10.3390/pharmaceutics15102402] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Monoclonal therapeutic antibodies have revolutionized the treatment of cancer and other diseases. Fc engineering aims to enhance the effector functions or half-life of therapeutic antibodies by modifying their Fc regions. Recent advances in the Fc engineering of modern therapeutic antibodies can be considered the next generation of antibody therapy. Various strategies are employed, including altering glycosylation patterns via glycoengineering and introducing mutations to the Fc region, thereby enhancing Fc receptor or complement interactions. Further, Fc engineering strategies enable the generation of bispecific IgG-based heterodimeric antibodies. As Fc engineering techniques continue to evolve, an expanding portfolio of Fc-engineered antibodies is advancing through clinical development, with several already approved for medical use. Despite the plethora of Fc-based mutations that have been analyzed in in vitro and in vivo models, we focus here in this review on the relevant Fc engineering strategies of approved therapeutic antibodies to finetune effector functions, to modify half-life and to stabilize asymmetric bispecific IgGs.
Collapse
Affiliation(s)
- Dalia T. Abdeldaim
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
| |
Collapse
|
269
|
Kim JW, Kim HJ, Heo K, Lee Y, Jang HJ, Lee HY, Park JW, Cho YB, Lee JH, Shin HG, Yang HR, Choi HL, Shim HB, Lee S. A novel bispecific antibody dual-targeting approach for enhanced neutralization against fast-evolving SARS-CoV-2 variants. Front Immunol 2023; 14:1271508. [PMID: 37822941 PMCID: PMC10562541 DOI: 10.3389/fimmu.2023.1271508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction The emergence of new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has caused unprecedented health and socioeconomic crises, necessitating the immediate development of highly effective neutralizing antibodies. Despite recent advancements in anti-SARS-CoV-2 receptor-binding domain (RBD)-specific monoclonal antibodies (mAbs) derived from convalescent patient samples, their efficacy against emerging variants has been limited. In this study, we present a novel dual-targeting strategy using bispecific antibodies (bsAbs) that specifically recognize both the SARS-CoV-2 RBD and fusion peptide (FP), crucial domains for viral attachment to the host cell membrane and fusion in SARS-CoV-2 infection. Methods Using phage display technology, we rapidly isolated FP-specific mAbs from an established human recombinant antibody library, identifying K107.1 with a nanomolar affinity for SARS-CoV-2 FP. Furthermore, we generated K203.A, a new bsAb built in immunoglobulin G4-(single-chain variable fragment)2 forms and demonstrating a high manufacturing yield and nanomolar affinity to both the RBD and FP, by fusing K102.1, our previously reported RBD-specific mAb, with K107.1. Results Our comprehensive in vitro functional analyses revealed that the K203.A bsAb significantly outperformed the parental RBD-specific mAb in terms of neutralization efficacy against SARS-CoV-2 variants. Furthermore, intravenous monotherapy with K203.A demonstrated potent in vivo neutralizing activity without significant in vivo toxicity in a mouse model infected with a SARS-CoV-2 variant. Conclusion These findings present a novel bsAb dual-targeting strategy, directed at SARS-CoV-2 RBD and FP, as an effective approach for rapid development and management against continuously evolving SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ji Woong Kim
- Department of Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Hyun Jung Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Kyun Heo
- Department of Chemistry, Kookmin University, Seoul, Republic of Korea
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul, Republic of Korea
| | - Yoonwoo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Hui Jeong Jang
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, Kangwon National University, Chuncheon, Republic of Korea
| | - Yea Bin Cho
- Department of Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Ji Hyun Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Hye Lim Choi
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Hyun Bo Shim
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Sukmook Lee
- Department of Chemistry, Kookmin University, Seoul, Republic of Korea
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul, Republic of Korea
| |
Collapse
|
270
|
Sun Y, Zhang Y, Yu H, Saint Fleur A, Yu D, Yang Z, Feng H. A fine-tuned yeast surface-display/secretion platform enables the rapid discovery of neutralizing antibodies against Clostridioides difficile toxins. Microb Cell Fact 2023; 22:194. [PMID: 37749574 PMCID: PMC10519002 DOI: 10.1186/s12934-023-02200-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 09/06/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Neutralizing antibody plays a key role in protecting hosts from invasive pathogens and their virulent components. Current high-throughput assays for antibody screening are based on binding activities. However, those antibodies with high affinity may not have neutralizing activities. Subsequent functionality assays are necessary to identify neutralizing antibodies from binders with high affinity to their target antigens, which is laborious and time-consuming. Therefore, a versatile platform that can rapidly identify antibodies with both high binding affinity and neutralizing activity is desired to curb future pandemics like COVID-19. RESULTS In this proof-of-concept study, we adapted Saccharomyces cerevisiae to either display human antibodies on the yeast surface or secrete soluble antibodies into the cultivation supernatant under a controllable 'switch' through different carbon source induced promoters. Initially, an engineered chimeric-bispecific Fab antibody, derived from humanized nanobodies against both Clostridioides difficile toxin A and B (TcdA and TcdB), was successfully expressed either on the yeast cell surface or in the culture medium with intact bioactivity, suggesting the applicability of our system in antibody display and secretion. Next, a combinatorial Fab library was constructed from B cells isolated from a convalescent patient with a high serological neutralizing titer against TcdB. Following three rounds of magnetic bead enrichment and one round of flow cytometry sorting, antibodies against TcdB were enriched efficiently. We then sorted out single binders with high binding affinity and induced them to express soluble antibodies in culture medium. The neutralizing activity of culture supernatant was analyzed using cell-based assay immediately. This way, we rapidly identified two unique neutralizers (out of seven binders) that can neutralize the cytotoxicity of TcdB. CONCLUSION The antibody screening platform described here simplifies the neutralizing antibody discovery procedure and will be an attractive alternative for screening functional antibodies against infectious diseases.
Collapse
Affiliation(s)
- Ying Sun
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA.
- Department of Pathogen Biology, School of Basic Medical Sciences, China Medical University, Shenyang, 110122, China.
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA
| | - Hua Yu
- Fzata, Inc, Halethorpe, MD, 21227, USA
| | - Ashley Saint Fleur
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA
| | - Di Yu
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA
| | | | - Hanping Feng
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA.
- Fzata, Inc, Halethorpe, MD, 21227, USA.
| |
Collapse
|
271
|
Borhani SG, Levine MZ, Krumpe LH, Wilson J, Henrich CJ, O'Keefe BR, Lo DC, Sittampalam GS, Godfrey AG, Lunsford RD, Mangalampalli V, Tao D, LeClair CA, Thole AP, Frey D, Swartz J, Rao G. An approach to rapid distributed manufacturing of broad spectrum anti-viral griffithsin using cell-free systems to mitigate pandemics. N Biotechnol 2023; 76:13-22. [PMID: 37054948 PMCID: PMC10330340 DOI: 10.1016/j.nbt.2023.04.003] [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: 12/21/2022] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
This study describes the cell-free biomanufacturing of a broad-spectrum antiviral protein, griffithsin (GRFT) such that it can be produced in microgram quantities with consistent purity and potency in less than 24 h. We demonstrate GRFT production using two independent cell-free systems, one plant and one microbial. Griffithsin purity and quality were verified using standard regulatory metrics. Efficacy was demonstrated in vitro against SARS-CoV-2 and HIV-1 and was nearly identical to that of GRFT expressed in vivo. The proposed production process is efficient and can be readily scaled up and deployed wherever a viral pathogen might emerge. The current emergence of viral variants of SARS-CoV-2 has resulted in frequent updating of existing vaccines and loss of efficacy for front-line monoclonal antibody therapies. Proteins such as GRFT with its efficacious and broad virus neutralizing capability provide a compelling pandemic mitigation strategy to promptly suppress viral emergence at the source of an outbreak.
Collapse
Affiliation(s)
- Shayan G Borhani
- Center for Advanced Sensor Technology, Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA; Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - Max Z Levine
- Department of Chemical Engineering and Department of Bioengineering, Stanford University, Stanford, CA 94305-5025, USA
| | - Lauren H Krumpe
- Molecular Targets Program, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jennifer Wilson
- Molecular Targets Program, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Curtis J Henrich
- Molecular Targets Program, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Barry R O'Keefe
- Molecular Targets Program, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA; Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD 21702, USA
| | - Donald C Lo
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - G Sitta Sittampalam
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Alexander G Godfrey
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - R Dwayne Lunsford
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Venkata Mangalampalli
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Christopher A LeClair
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Aaron P Thole
- Center for Advanced Sensor Technology, Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA; Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - Douglas Frey
- Center for Advanced Sensor Technology, Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA; Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - James Swartz
- Department of Chemical Engineering and Department of Bioengineering, Stanford University, Stanford, CA 94305-5025, USA
| | - Govind Rao
- Center for Advanced Sensor Technology, Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA; Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA.
| |
Collapse
|
272
|
Hartman K, Steiner G, Siegel M, Looney CM, Hickling TP, Bray-French K, Springer S, Marban-Doran C, Ducret A. Expanding the MAPPs Assay to Accommodate MHC-II Pan Receptors for Improved Predictability of Potential T Cell Epitopes. BIOLOGY 2023; 12:1265. [PMID: 37759665 PMCID: PMC10525474 DOI: 10.3390/biology12091265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
A critical step in the immunogenicity cascade is attributed to human leukocyte antigen (HLA) II presentation triggering T cell immune responses. The liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based major histocompatibility complex (MHC) II-associated peptide proteomics (MAPPs) assay is implemented during preclinical risk assessments to identify biotherapeutic-derived T cell epitopes. Although studies indicate that HLA-DP and HLA-DQ alleles are linked to immunogenicity, most MAPPs studies are restricted to using HLA-DR as the dominant HLA II genotype due to the lack of well-characterized immunoprecipitating antibodies. Here, we address this issue by testing various commercially available clones of MHC-II pan (CR3/43, WR18, and Tü39), HLA-DP (B7/21), and HLA-DQ (SPV-L3 and 1a3) antibodies in the MAPPs assay, and characterizing identified peptides according to binding specificity. Our results reveal that HLA II receptor-precipitating reagents with similar reported specificities differ based on clonality and that MHC-II pan antibodies do not entirely exhibit pan-specific tendencies. Since no individual antibody clone is able to recover the complete HLA II peptide repertoire, we recommend a mixed strategy of clones L243, WR18, and SPV-L3 in a single immunoprecipitation step for more robust compound-specific peptide detection. Ultimately, our optimized MAPPs strategy improves the predictability and additional identification of T cell epitopes in immunogenicity risk assessments.
Collapse
Affiliation(s)
- Katharina Hartman
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland (C.M.L.)
| | - Guido Steiner
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland (C.M.L.)
| | - Michel Siegel
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland (C.M.L.)
| | - Cary M. Looney
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland (C.M.L.)
| | - Timothy P. Hickling
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland (C.M.L.)
| | - Katharine Bray-French
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland (C.M.L.)
| | - Sebastian Springer
- School of Science, Department of Biochemistry and Cell Biology, Constructor University, Campus Ring 1, 28759 Bremen, Germany
| | - Céline Marban-Doran
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland (C.M.L.)
| | - Axel Ducret
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland (C.M.L.)
| |
Collapse
|
273
|
Ayoup MS, Wahby Y, Abdel-Hamid H, Abu-Serie MM, Ramadan S, Barakat A, Teleb M, Ismail MMF. Reinvestigation of Passerini and Ugi scaffolds as multistep apoptotic inducers via dual modulation of caspase 3/7 and P53-MDM2 signaling for halting breast cancer. RSC Adv 2023; 13:27722-27737. [PMID: 37736568 PMCID: PMC10509784 DOI: 10.1039/d3ra04029a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/28/2023] [Indexed: 09/23/2023] Open
Abstract
Selective induction of breast cancer apoptosis is viewed as the mainstay of various ongoing oncology drug discovery programs. Passerini scaffolds have been recently exploited as selective apoptosis inducers via a caspase 3/7 dependent pathway. Herein, the optimized Passerini caspase activators were manipulated to synergistically induce P53-dependent apoptosis via modulating the closely related P53-MDM2 signaling axis. The adopted design rationale and synthetic routes relied on mimicking the general thematic features of lead MDM2 inhibitors incorporating multiple aromatic rings. Accordingly, the cyclization of representative Passerini derivatives and related Ugi compounds into the corresponding diphenylimidazolidine and spiro derivative was performed, resembling the nutlin-based and spiro MDM-2 inhibitors, respectively. The study was also extended to explore the apoptotic induction capacity of the scaffold after simplification and modifications. MTT assay on MCF-7 and MDA-MB231 breast cancer cells compared to normal fibroblasts (WI-38) revealed their promising cytotoxic activities. The flexible Ugi derivatives 3 and 4, cyclic analog 8, Passerini adduct 12, and the thiosemicarbazide derivative 17 were identified as the study hits regarding cytotoxic potency and selectivity, being over 10-folds more potent (IC50 = 0.065-0.096 μM) and safer (SI = 4.4-18.7) than doxorubicin (IC50 = 0.478 μM, SI = 0.569) on MCF-7 cells. They promoted apoptosis induction via caspase 3/7 activation (3.1-4.1 folds) and P53 induction (up to 4 folds). Further apoptosis studies revealed that these compounds enhanced gene expression of BAX by 2 folds and suppressed Bcl-2 expression by 4.29-7.75 folds in the treated MCF-7 cells. Docking simulations displayed their plausible binding modes with the molecular targets and highlighted their structural determinants of activities for further optimization studies. Finally, in silico prediction of the entire library was computationally performed, showing that most of them could be envisioned as drug-like candidates.
Collapse
Affiliation(s)
- Mohammed Salah Ayoup
- Chemistry Department, Faculty of Science, Alexandria University P. O. Box 426 Alexandria 21321 Egypt
| | - Yasmin Wahby
- Chemistry Department, Faculty of Science, Alexandria University P. O. Box 426 Alexandria 21321 Egypt
| | - Hamida Abdel-Hamid
- Chemistry Department, Faculty of Science, Alexandria University P. O. Box 426 Alexandria 21321 Egypt
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City) Egypt
| | - Sherif Ramadan
- Chemistry Department, Michigan State University East Lansing MI 48824 USA
- Department of Chemistry, Benha University Benha Egypt
| | - Assem Barakat
- Department of Chemistry, College of Science, King Saud University P. O. Box 2455 Riyadh 11451 Saudi Arabia
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University Alexandria 21521 Egypt
| | - Magda M F Ismail
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University Cairo 11754 Egypt
| |
Collapse
|
274
|
Wang Y, Lv H, Lei R, Yeung YH, Shen IR, Choi D, Teo QW, Tan TJ, Gopal AB, Chen X, Graham CS, Wu NC. An explainable language model for antibody specificity prediction using curated influenza hemagglutinin antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.11.557288. [PMID: 37745338 PMCID: PMC10515799 DOI: 10.1101/2023.09.11.557288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Despite decades of antibody research, it remains challenging to predict the specificity of an antibody solely based on its sequence. Two major obstacles are the lack of appropriate models and inaccessibility of datasets for model training. In this study, we curated a dataset of >5,000 influenza hemagglutinin (HA) antibodies by mining research publications and patents, which revealed many distinct sequence features between antibodies to HA head and stem domains. We then leveraged this dataset to develop a lightweight memory B cell language model (mBLM) for sequence-based antibody specificity prediction. Model explainability analysis showed that mBLM captured key sequence motifs of HA stem antibodies. Additionally, by applying mBLM to HA antibodies with unknown epitopes, we discovered and experimentally validated many HA stem antibodies. Overall, this study not only advances our molecular understanding of antibody response to influenza virus, but also provides an invaluable resource for applying deep learning to antibody research.
Collapse
Affiliation(s)
- Yiquan Wang
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Huibin Lv
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Ruipeng Lei
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Yuen-Hei Yeung
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Computer Science, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Ivana R. Shen
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Danbi Choi
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Qi Wen Teo
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Timothy J.C. Tan
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Akshita B. Gopal
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Xin Chen
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Claire S. Graham
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Nicholas C. Wu
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
275
|
Asher D, Dai D, Klimchak AC, Sedita LE, Gooch KL, Rodino-Klapac L. Paving the way for future gene therapies: A case study of scientific spillover from delandistrogene moxeparvovec. Mol Ther Methods Clin Dev 2023; 30:474-483. [PMID: 37674905 PMCID: PMC10477757 DOI: 10.1016/j.omtm.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Gene therapies have potential to improve outcomes of severe diseases after only a single administration. Novel therapies are continually being developed using knowledge gained from prior successes, a concept known as scientific spillover. Gene therapy advancement requires extensive development at each stage: preclinical work to create and evaluate vehicles for delivery of the therapy, design of clinical development programs, and establishment of a large-scale manufacturing process. Pioneering gene therapies are generating spillover as investigators confront myriad issues specific to this treatment modality. These include frameworks for construct engineering, dose evaluation, patient selection, outcome assessment, and safety monitoring. Consequently, the benefits of these therapies extend beyond offering knowledge for treating any one disease to establishing new platforms and paradigms that will accelerate advancement of future gene therapies. This impact is even more profound in rare diseases, where developing therapies in isolation may not be possible. This review describes some instances of scientific spillover in healthcare, and specifically gene therapy, using delandistrogene moxeparvovec (SRP-9001), a gene therapy recently approved by the US Food and Drug Administration for the treatment of ambulatory pediatric patients aged 4-5 years with Duchenne muscular dystrophy with a confirmed mutation in the DMD gene, as a case study.
Collapse
Affiliation(s)
- Damon Asher
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Daisy Dai
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Alexa C. Klimchak
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Lauren E. Sedita
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | | | | |
Collapse
|
276
|
Fekete S, Guillarme D. Ultra-short columns for the chromatographic analysis of large molecules. J Chromatogr A 2023; 1706:464285. [PMID: 37562104 DOI: 10.1016/j.chroma.2023.464285] [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: 06/23/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Today, reverse phase liquid chromatography (RPLC) analysis of proteins is almost exclusively performed on conventional columns (100-150 mm) in gradient elution mode. However, it was shown many years ago that large molecules present an on/off retention mechanism, and that only a very short inlet segment of the chromatographic column retains effectively the large molecules. Much shorter columns - like only a few centimetres or even a few millimetres - can therefore be used to efficiently analyse such macromolecules. The aim of this review is to summarise the historical and more recent works related to the use of very short columns for the analysis of model and therapeutic proteins. To this end, we have outlined the theoretical concepts behind the use of short columns, as well as the instrumental limitations and potential applications. Finally, we have shown that these very short columns were also possibly interesting for other chromatographic modes, such as ion exchange chromatography (IEX), hydrophilic interaction chromatography (HILIC) or hydrophobic interaction chromatography (HIC), as analyses in these chromatographic modes are performed in gradient elution mode.
Collapse
Affiliation(s)
| | - Davy Guillarme
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel Servet 1, 1211 Geneva 4, Switzerland; School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel Servet 1, 1211 Geneva 4, Switzerland.
| |
Collapse
|
277
|
França RKA, Studart IC, Bezerra MRL, Pontes LQ, Barbosa AMA, Brigido MM, Furtado GP, Maranhão AQ. Progress on Phage Display Technology: Tailoring Antibodies for Cancer Immunotherapy. Viruses 2023; 15:1903. [PMID: 37766309 PMCID: PMC10536222 DOI: 10.3390/v15091903] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The search for innovative anti-cancer drugs remains a challenge. Over the past three decades, antibodies have emerged as an essential asset in successful cancer therapy. The major obstacle in developing anti-cancer antibodies is the need for non-immunogenic antibodies against human antigens. This unique requirement highlights a disadvantage to using traditional hybridoma technology and thus demands alternative approaches, such as humanizing murine monoclonal antibodies. To overcome these hurdles, human monoclonal antibodies can be obtained directly from Phage Display libraries, a groundbreaking tool for antibody selection. These libraries consist of genetically engineered viruses, or phages, which can exhibit antibody fragments, such as scFv or Fab on their capsid. This innovation allows the in vitro selection of novel molecules directed towards cancer antigens. As foreseen when Phage Display was first described, nowadays, several Phage Display-derived antibodies have entered clinical settings or are undergoing clinical evaluation. This comprehensive review unveils the remarkable progress in this field and the possibilities of using clever strategies for phage selection and tailoring the refinement of antibodies aimed at increasingly specific targets. Moreover, the use of selected antibodies in cutting-edge formats is discussed, such as CAR (chimeric antigen receptor) in CAR T-cell therapy or ADC (antibody drug conjugate), amplifying the spectrum of potential therapeutic avenues.
Collapse
Affiliation(s)
- Renato Kaylan Alves França
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.F.); (M.M.B.)
- Graduate Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Igor Cabral Studart
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Biotechnology of Natural Resources, Federal University of Ceará, Fortaleza 60440-970, Brazil
| | - Marcus Rafael Lobo Bezerra
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Biotechnology of Natural Resources, Federal University of Ceará, Fortaleza 60440-970, Brazil
| | - Larissa Queiroz Pontes
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Biotechnology of Natural Resources, Federal University of Ceará, Fortaleza 60440-970, Brazil
| | - Antonio Marcos Aires Barbosa
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Applied Informatics, University of Fortaleza, Fortaleza 60811-905, Brazil
| | - Marcelo Macedo Brigido
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.F.); (M.M.B.)
| | - Gilvan Pessoa Furtado
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Biotechnology of Natural Resources, Federal University of Ceará, Fortaleza 60440-970, Brazil
| | - Andréa Queiroz Maranhão
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.F.); (M.M.B.)
| |
Collapse
|
278
|
Sharma P, Joshi RV, Pritchard R, Xu K, Eicher MA. Therapeutic Antibodies in Medicine. Molecules 2023; 28:6438. [PMID: 37764213 PMCID: PMC10535987 DOI: 10.3390/molecules28186438] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/05/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Antibody engineering has developed into a wide-reaching field, impacting a multitude of industries, most notably healthcare and diagnostics. The seminal work on developing the first monoclonal antibody four decades ago has witnessed exponential growth in the last 10-15 years, where regulators have approved monoclonal antibodies as therapeutics and for several diagnostic applications, including the remarkable attention it garnered during the pandemic. In recent years, antibodies have become the fastest-growing class of biological drugs approved for the treatment of a wide range of diseases, from cancer to autoimmune conditions. This review discusses the field of therapeutic antibodies as it stands today. It summarizes and outlines the clinical relevance and application of therapeutic antibodies in treating a landscape of diseases in different disciplines of medicine. It discusses the nomenclature, various approaches to antibody therapies, and the evolution of antibody therapeutics. It also discusses the risk profile and adverse immune reactions associated with the antibodies and sheds light on future applications and perspectives in antibody drug discovery.
Collapse
Affiliation(s)
- Prerna Sharma
- Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA
| | | | | | | | | |
Collapse
|
279
|
Kumari M, Acharya A, Krishnamurthy PT. Antibody-conjugated nanoparticles for target-specific drug delivery of chemotherapeutics. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:912-926. [PMID: 37701520 PMCID: PMC10494237 DOI: 10.3762/bjnano.14.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/22/2023] [Indexed: 09/14/2023]
Abstract
Nanotechnology provides effective methods for precisely delivering chemotherapeutics to cancer cells, thereby improving efficacy and reducing off-target side effects. The targeted delivery of nanoscale chemotherapeutics is accomplished by two different approaches, namely the exploitation of leaky tumor vasculature (EPR effect) and the surface modification of nanoparticles (NPs) with various tumor-homing peptides, aptamers, oligonucleotides, and monoclonal antibodies (mAbs). Because of higher binding affinity and specificity, mAbs have received a lot of attention for the detection of selective cancer biomarkers and also for the treatment of various types of cancer. Antibody-conjugated nanoparticles (ACNPs) are an effective targeted therapy for the efficient delivery of chemotherapeutics specifically to the targeted cancer cells. ACNPs combine the benefits of NPs and mAbs to provide high drug loads at the tumor site with better selectivity and delivery efficiency. The mAbs on the NP surfaces recognize their specific receptors expressed on the target cells and release the chemotherapeutic agent in a controlled manner. Appropriately designed and synthesized ACNPs are essential to fully realize their therapeutic benefits. In blood stream, ACNPs instantly interact with biological molecules, and a protein corona is formed. Protein corona formation triggers an immune response and affects the targeting ability of the nanoformulation. In this review, we provide recent findings to highlight several antibody conjugation methods such as adsorption, covalent conjugation, and biotin-avidin interaction. This review also provides an overview of the many effects of the protein corona and the theranostic applications of ACNPs for the treatment of cancer.
Collapse
Affiliation(s)
- Mamta Kumari
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| |
Collapse
|
280
|
Smith MD, Case MA, Makowski EK, Tessier PM. Position-Specific Enrichment Ratio Matrix scores predict antibody variant properties from deep sequencing data. Bioinformatics 2023; 39:btad446. [PMID: 37478351 PMCID: PMC10477941 DOI: 10.1093/bioinformatics/btad446] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/21/2023] [Accepted: 07/20/2023] [Indexed: 07/23/2023] Open
Abstract
MOTIVATION Deep sequencing of antibody and related protein libraries after phage or yeast-surface display sorting is widely used to identify variants with increased affinity, specificity, and/or improvements in key biophysical properties. Conventional approaches for identifying optimal variants typically use the frequencies of observation in enriched libraries or the corresponding enrichment ratios. However, these approaches disregard the vast majority of deep sequencing data and often fail to identify the best variants in the libraries. RESULTS Here, we present a method, Position-Specific Enrichment Ratio Matrix (PSERM) scoring, that uses entire deep sequencing datasets from pre- and post-selections to score each observed protein variant. The PSERM scores are the sum of the site-specific enrichment ratios observed at each mutated position. We find that PSERM scores are much more reproducible and correlate more strongly with experimentally measured properties than frequencies or enrichment ratios, including for multiple antibody properties (affinity and non-specific binding) for a clinical-stage antibody (emibetuzumab). We expect that this method will be broadly applicable to diverse protein engineering campaigns. AVAILABILITY AND IMPLEMENTATION All deep sequencing datasets and code to perform the analyses presented within are available via https://github.com/Tessier-Lab-UMich/PSERM_paper.
Collapse
Affiliation(s)
- Matthew D Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109-2200, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109-2200, United States
| | - Marshall A Case
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109-2200, United States
| | - Emily K Makowski
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109-2200, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109-2200, United States
| | - Peter M Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109-2200, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109-2200, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109-2200, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109-2200, United States
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, MI 48109-2200, United States
- Michigan Alzheimer’s Disease Center, University of Michigan, Ann Arbor, MI 48109-2200, United States
| |
Collapse
|
281
|
Ladiwala P, Dhara VG, Jenkins J, Kuang B, Hoang D, Yoon S, Betenbaugh MJ. Addressing amino acid-derived inhibitory metabolites and enhancing CHO cell culture performance through DOE-guided media modifications. Biotechnol Bioeng 2023; 120:2542-2558. [PMID: 37096798 DOI: 10.1002/bit.28403] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/22/2023] [Accepted: 04/10/2023] [Indexed: 04/26/2023]
Abstract
Previously, we identified six inhibitory metabolites (IMs) accumulating in Chinese hamster ovary (CHO) cultures using AMBIC 1.0 community reference medium that negatively impacted culture performance. The goal of the current study was to modify the medium to control IM accumulation through design of experiments (DOE). Initial over-supplementation of precursor amino acids (AAs) by 100% to 200% in the culture medium revealed positive correlations between initial AA concentrations and IM levels. A screening design identified 5 AA targets, Lys, Ile, Trp, Leu, Arg, as key contributors to IMs. Response surface design analysis was used to reduce initial AA levels between 13% and 33%, and these were then evaluated in batch and fed-batch cultures. Lowering AAs in basal and feed medium and reducing feed rate from 10% to 5% reduced inhibitory metabolites HICA and NAP by up to 50%, MSA by 30%, and CMP by 15%. These reductions were accompanied by a 13% to 40% improvement in peak viable cell densities and 7% to 50% enhancement in IgG production in batch and fed-batch processes, respectively. This study demonstrates the value of tuning specific AA levels in reference basal and feed media using statistical design methodologies to lower problematic IMs.
Collapse
Affiliation(s)
- Pranay Ladiwala
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Venkata Gayatri Dhara
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jackson Jenkins
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bingyu Kuang
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Duc Hoang
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
282
|
Vaskó D, Domján J, Szűcs B, Bakk L, Hajdinák P, Marosi G, Nagy ZK, Hirsch E, Fehér C. Development and Comparison of Alternative Methods for the Purification of Adalimumab Directly from Harvested Cell Culture Fluid. Food Technol Biotechnol 2023; 61:339-349. [PMID: 38022883 PMCID: PMC10666952 DOI: 10.17113/ftb.61.03.23.8094] [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: 01/23/2023] [Accepted: 07/11/2023] [Indexed: 12/01/2023] Open
Abstract
Research background Protein A affinity chromatography is a well-established method currently used in the pharmaceutical industry. However, the high costs usually associated with chromatographic separation of protein A and the difficulties in continuous operation make the investigation of alternative purification methods very important. Experimental approach In this study, extraction/back-extraction and precipitation/dissolution methods were developed and optimised. They were compared with protein A and cation exchange chromatography separations in terms of yield of monoclonal antibody (mAb) and amount of residual impurities, such as DNA and host cell proteins, and amount of mAb aggregates. For a comprehensive comparison of the different methods, experiments were carried out with the same cell-free fermentation broth containing adalimumab. Results and conclusions Protein A and cation exchange chromatographic separations resulted in high yield and purity of adalimumab. The precipitation-based process resulted in high yield but with lower purity. The extraction-based purification resulted in low yield and purity. Thus, the precipitation-based method proved to be more promising than the extraction-based method for direct purification of adalimumab from harvested cell culture fluid. Novelty and scientific contribution Although alternative purification methods may offer the advantages of simplicity and low-cost operation, further significant improvements are required to compete with the performance of chromatographic separations of adalimumab from true fermentation broth.
Collapse
Affiliation(s)
- Dorottya Vaskó
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - Júlia Domján
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - Bence Szűcs
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - László Bakk
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - Péter Hajdinák
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - György Marosi
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - Zsombor K. Nagy
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - Edit Hirsch
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - Csaba Fehér
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| |
Collapse
|
283
|
Hu Z, Cohen S, Swanson SJ. The immunogenicity of human-origin therapeutic antibodies are associated with V gene usage. Front Immunol 2023; 14:1237754. [PMID: 37720227 PMCID: PMC10502710 DOI: 10.3389/fimmu.2023.1237754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/18/2023] [Indexed: 09/19/2023] Open
Abstract
Therapeutic antibodies can elicit unwanted immune responses in a subset of patients, which leads to the production of anti-drug antibodies (ADA). Some of these ADAs have been reported to effect the pharmacokinetics, efficacy and/or safety of the therapeutic antibodies. The sequence diversity of antibodies are generated by VDJ recombination and mutagenesis. While the antibody generation process can create a large candidate pool for identifying high-affinity antibodies, it also could produce sequences that are foreign to the human immune system. However, it is not clear how VDJ recombination and mutagenesis impact the clinical ADA rate of therapeutic antibodies. In this study, we identified a positive correlation between the clinical ADA rate and the number of introduced mutations in the antibody sequences. We also found that the use of rare V alleles in human-origin antibody therapeutics is associated with higher risk of immunogenicity. The results suggest that antibody engineering projects should start with frameworks that contain commonly used V alleles and prioritize antibody candidates with low number of mutations to reduce the risk of immunogenicity.
Collapse
|
284
|
Quiroga LC, Sabourin AA. Review of Dual Biologics in Specialty Pharmacy Practice. Ann Pharmacother 2023; 57:1094-1110. [PMID: 36600576 DOI: 10.1177/10600280221135177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE To describe and review the published evidence on use of multiple biologics within specialty pharmacy practice. DATA SOURCES A search of PubMed and Embase was conducted from October 2021 through September 2022. Keywords included biologics for immune-mediated conditions along with the terms "dual," "add-on," and "combination." STUDY SELECTION AND DATA EXTRACTION All human studies in the English language were considered. Published abstracts, case reports, case series, randomized controlled trials, systematic reviews, and meta-analyses were included. DATA SYNTHESIS Although evidence is limited, there are published meta-analyses of combined biologic use within gastroenterology and rheumatology. There are also numerous case reports within dermatology. Clinical trials of dual biologics for severe rheumatologic conditions and inflammatory bowel disease are in progress. Existing evidence for use in pulmonology and allergy suggest dual biologic therapy can be safe and effective, but data are limited. Literature describing use of monoclonal antibodies for other overlapping conditions is lacking. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE This article reviews the evidence describing combination biologic use and outlines remaining knowledge gaps. It also describes the essential role that specialty pharmacists play in managing therapeutic mAbs. CONCLUSIONS High-quality evidence describing combination biologic use is limited and long-term safety data are lacking. Pharmacists should utilize their specialized training to assess appropriateness of therapy, provide patient counseling and monitor for safety and efficacy.
Collapse
Affiliation(s)
- Lauren C Quiroga
- Department of Pharmacy Services, University of Michigan Health, Ann Arbor, MI, USA
| | - Ashley A Sabourin
- Department of Pharmacy Services, University of Michigan Health, Ann Arbor, MI, USA
| |
Collapse
|
285
|
Bladt T, Vorup-Jensen T, Ebbesen M. Principles for Just Prioritization of Expensive Biological Therapies in the Danish Healthcare System. JOURNAL OF BIOETHICAL INQUIRY 2023; 20:523-542. [PMID: 37733175 PMCID: PMC10624754 DOI: 10.1007/s11673-023-10283-2] [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: 05/07/2022] [Accepted: 02/18/2023] [Indexed: 09/22/2023]
Abstract
The Danish healthcare system must meet the need for easy and equal access to healthcare for every citizen. However, investigations have shown unfair prioritization of cancer patients and unfair prioritization of resources for expensive medicines over care. What is needed are principles for proper prioritization. This article investigates whether American ethicists Tom Beauchamp and James Childress's principle of justice may be helpful as a conceptual framework for reflections on prioritization of expensive biological therapies in the Danish healthcare system. We present an empirical study exploring the principles for prioritizing new expensive biological therapies. This study includes qualitative interviews with key Danish stakeholders experienced in antibody therapy and prioritizing resources for expensive medicines. Beauchamp and Childress's model only covers government-funded primary and acute healthcare. Based on the interviews, this study indicates that to be helpful in a Danish context this model should include equal access for citizens to government-funded primary and acute healthcare, costly medicine, and other scarce treatments. We conclude that slightly modified, Beauchamp and Childress's principle of justice might be useful as a conceptual framework for reflections on the prioritization of expensive biological therapies in the Danish healthcare system.
Collapse
Affiliation(s)
- Tara Bladt
- Danish College of Pharmacy Technicians, Hillerød, Denmark
| | | | - Mette Ebbesen
- Techno-Anthropology & Participation, Department of Planning, Aalborg University, Rendsburggade 14, DK- 9000, Aalborg, Denmark.
| |
Collapse
|
286
|
Nema S, Nitika N. Monoclonal antibody: future of malaria control and prevention. Trans R Soc Trop Med Hyg 2023; 117:673-674. [PMID: 37093779 DOI: 10.1093/trstmh/trad027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/24/2023] [Accepted: 04/12/2023] [Indexed: 04/25/2023] Open
Abstract
Monoclonal antibodies (mAbs) are extremely specialized proteins that are cloned from B cells and bind to pathogen epitopes. There are currently no known prophylactic immune-based strategies or efficient, widespread treatments to stop the spread of malaria. In order to lower the prevalence of malaria and its associated mortality, we need mAbs that are capable of offering immediate passive protection against the disease. mAbs have become more crucial in the treatment or prevention of several other infectious diseases. Recently, mAb development for malaria prevention and control has greatly evolved and widespread use in public health settings is now a possibility.
Collapse
Affiliation(s)
- Shrikant Nema
- Parasite-Host Biology Group, ICMR-National Institute of Malaria Research, Sector 8, Dwarka 110077, New Delhi, India
| | - Nitika Nitika
- Parasite-Host Biology Group, ICMR-National Institute of Malaria Research, Sector 8, Dwarka 110077, New Delhi, India
| |
Collapse
|
287
|
Khairkhah N, Namvar A, Bolhassani A. Application of Cell Penetrating Peptides as a Promising Drug Carrier to Combat Viral Infections. Mol Biotechnol 2023; 65:1387-1402. [PMID: 36719639 PMCID: PMC9888354 DOI: 10.1007/s12033-023-00679-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023]
Abstract
Novel effective drugs or therapeutic vaccines have been already developed to eradicate viral infections. Some non-viral carriers have been used for effective drug delivery to a target cell or tissue. Among them, cell penetrating peptides (CPPs) attracted a special interest to enhance drug delivery into the cells with low toxicity. They were also applied to transfer peptide/protein-based and nucleic acids-based therapeutic vaccines against viral infections. CPPs-conjugated drugs or vaccines were investigated in several viral infections including poliovirus, Ebola, coronavirus, herpes simplex virus, human immunodeficiency virus, hepatitis B virus, hepatitis C virus, Japanese encephalitis virus, and influenza A virus. Some studies showed that the uptake of CPPs or CPPs-conjugated drugs can be performed through both non-endocytic and endocytic pathways. Despite high potential of CPPs for cargo delivery, there are some serious drawbacks such as non-tissue-specificity, instability, and suboptimal pharmacokinetics features that limit their clinical applications. At present, some solutions are utilized to improve the CPPs properties such as conjugation of CPPs with targeting moieties, the use of fusogenic lipids, generation of the proton sponge effect, etc. Up to now, no CPP or composition containing CPPs has been approved by the Food and Drug Administration (FDA) due to the lack of sufficient in vivo studies on stability, immunological assays, toxicity, and endosomal escape of CPPs. In this review, we briefly describe the properties, uptake mechanisms, advantages and disadvantages, and improvement of intracellular delivery, and bioavailability of cell penetrating peptides. Moreover, we focus on their application as an effective drug carrier to combat viral infections.
Collapse
Affiliation(s)
- Niloofar Khairkhah
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Namvar
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
288
|
Li J, Kang G, Wang J, Yuan H, Wu Y, Meng S, Wang P, Zhang M, Wang Y, Feng Y, Huang H, de Marco A. Affinity maturation of antibody fragments: A review encompassing the development from random approaches to computational rational optimization. Int J Biol Macromol 2023; 247:125733. [PMID: 37423452 DOI: 10.1016/j.ijbiomac.2023.125733] [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: 04/03/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Routinely screened antibody fragments usually require further in vitro maturation to achieve the desired biophysical properties. Blind in vitro strategies can produce improved ligands by introducing random mutations into the original sequences and selecting the resulting clones under more and more stringent conditions. Rational approaches exploit an alternative perspective that aims first at identifying the specific residues potentially involved in the control of biophysical mechanisms, such as affinity or stability, and then to evaluate what mutations could improve those characteristics. The understanding of the antigen-antibody interactions is instrumental to develop this process the reliability of which, consequently, strongly depends on the quality and completeness of the structural information. Recently, methods based on deep learning approaches critically improved the speed and accuracy of model building and are promising tools for accelerating the docking step. Here, we review the features of the available bioinformatic instruments and analyze the reports illustrating the result obtained with their application to optimize antibody fragments, and nanobodies in particular. Finally, the emerging trends and open questions are summarized.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Guangbo Kang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Jiewen Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Haibin Yuan
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Yili Wu
- Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and the Affiliated Kangning Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Oujiang Laboratory, Wenzhou, Zhejiang 325035, China
| | - Shuxian Meng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Ping Wang
- New Technology R&D Department, Tianjin Modern Innovative TCM Technology Company Limited, Tianjin 300392, China
| | - Miao Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; China Resources Biopharmaceutical Company Limited, Beijing 100029, China
| | - Yuli Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Tianjin Pharmaceutical Da Ren Tang Group Corporation Limited, Traditional Chinese Pharmacy Research Institute, Tianjin Key Laboratory of Quality Control in Chinese Medicine, Tianjin 300457, China; State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin 300193, China
| | - Yuanhang Feng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - He Huang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China.
| | - Ario de Marco
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Nova Gorica, Slovenia.
| |
Collapse
|
289
|
Bostanghadiri N, Ziaeefar P, Mofrad MG, Yousefzadeh P, Hashemi A, Darban-Sarokhalil D. COVID-19: An Overview of SARS-CoV-2 Variants-The Current Vaccines and Drug Development. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1879554. [PMID: 37674935 PMCID: PMC10480030 DOI: 10.1155/2023/1879554] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/07/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023]
Abstract
The world is presently in crisis facing an outbreak of a health-threatening microorganism known as COVID-19, responsible for causing uncommon viral pneumonia in humans. The virus was first reported in Wuhan, China, in early December 2019, and it quickly became a global concern due to the pandemic. Challenges in this regard have been compounded by the emergence of several variants such as B.1.1.7, B.1.351, P1, and B.1.617, which show an increase in transmission power and resistance to therapies and vaccines. Ongoing researches are focused on developing and manufacturing standard treatment strategies and effective vaccines to control the pandemic. Despite developing several vaccines such as Pfizer/BioNTech and Moderna approved by the U.S. Food and Drug Administration (FDA) and other vaccines in phase 4 clinical trials, preventive measures are mandatory to control the COVID-19 pandemic. In this review, based on the latest findings, we will discuss different types of drugs as therapeutic options and confirmed or developing vaccine candidates against SARS-CoV-2. We also discuss in detail the challenges posed by the variants and their effect on therapeutic and preventive interventions.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pardis Ziaeefar
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morvarid Golrokh Mofrad
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Parsa Yousefzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
290
|
Gaudreault F, Baardsnes J, Martynova Y, Dachon A, Hogues H, Corbeil CR, Purisima EO, Arbour M, Sulea T. Exploring rigid-backbone protein docking in biologics discovery: a test using the DARPin scaffold. Front Mol Biosci 2023; 10:1253689. [PMID: 37692063 PMCID: PMC10484509 DOI: 10.3389/fmolb.2023.1253689] [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: 07/05/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Accurate protein-protein docking remains challenging, especially for artificial biologics not coevolved naturally against their protein targets, like antibodies and other engineered scaffolds. We previously developed ProPOSE, an exhaustive docker with full atomistic details, which delivers cutting-edge performance by allowing side-chain rearrangements upon docking. However, extensive protein backbone flexibility limits its practical applicability as indicated by unbound docking tests. To explore the usefulness of ProPOSE on systems with limited backbone flexibility, here we tested the engineered scaffold DARPin, which is characterized by its relatively rigid protein backbone. A prospective screening campaign was undertaken, in which sequence-diversified DARPins were docked and ranked against a directed epitope on the target protein BCL-W. In this proof-of-concept study, only a relatively small set of 2,213 diverse DARPin interfaces were selected for docking from the huge theoretical library from mutating 18 amino-acid positions. A computational selection protocol was then applied for enrichment of binders based on normalized computed binding scores and frequency of binding modes against the predefined epitope. The top-ranked 18 designed DARPin interfaces were selected for experimental validation. Three designs exhibited binding affinities to BCL-W in the nanomolar range comparable to control interfaces adopted from known DARPin binders. This result is encouraging for future screening and engineering campaigns of DARPins and possibly other similarly rigid scaffolds against targeted protein epitopes. Method limitations are discussed and directions for future refinements are proposed.
Collapse
Affiliation(s)
- Francis Gaudreault
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Yuliya Martynova
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Aurore Dachon
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Hervé Hogues
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Christopher R. Corbeil
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Enrico O. Purisima
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Mélanie Arbour
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Traian Sulea
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
- Institute of Parasitology, McGill University, Montreal, QC, Canada
| |
Collapse
|
291
|
Herrera León C, Kalacas NA, Mier A, Sakhaii P, Merlier F, Prost E, Maffucci I, Montagna V, Mora-Radó H, Dhal PK, Tse Sum Bui B, Haupt K. Synthetic Peptide Antibodies as TNF-α Inhibitors: Molecularly Imprinted Polymer Nanogels Neutralize the Inflammatory Activity of TNF-α in THP-1 Derived Macrophages. Angew Chem Int Ed Engl 2023; 62:e202306274. [PMID: 37338464 DOI: 10.1002/anie.202306274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 06/21/2023]
Abstract
Tumor Necrosis Factor-α (TNF-α) is a cytokine that is normally produced by immune cells when fighting an infection. But, when too much TNF-α is produced as in autoimmune diseases, this leads to unwanted and persistent inflammation. Anti-TNF-α monoclonal antibodies have revolutionized the therapy of these disorders by blocking TNF-α and preventing its binding to TNF-α receptors, thus suppressing the inflammation. Herein, we propose an alternative in the form of molecularly imprinted polymer nanogels (MIP-NGs). MIP-NGs are synthetic antibodies obtained by nanomoulding the 3-dimensional shape and chemical functionalities of a desired target in a synthetic polymer. Using an in-house developed in silico rational approach, epitope peptides of TNF-α were generated and 'synthetic peptide antibodies' were prepared. The resultant MIP-NGs bind the template peptide and recombinant TNF-α with high affinity and selectivity, and can block the binding of TNF-α to its receptor. Consequently they were applied to neutralize pro-inflammatory TNF-α in the supernatant of human THP-1 macrophages, leading to a downregulation of the secretion of pro-inflammatory cytokines. Our results suggest that MIP-NGs, which are thermally and biochemically more stable and easier to manufacture than antibodies, and cost-effective, are very promising as next generation TNF-α inhibitors for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Claudia Herrera León
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| | - Noel Angelo Kalacas
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| | - Alejandra Mier
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| | - Peyman Sakhaii
- Global CMC Early Development, Synthetics Platform, Sanofi-Aventis Deutschland GmbH, Industrial Park Hoechst, Building G849, 65926, Frankfurt/Main, Germany
| | - Franck Merlier
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| | - Elise Prost
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| | - Irene Maffucci
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| | - Valentina Montagna
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| | - Helena Mora-Radó
- Global CMC Early Development, Synthetics Platform, Sanofi-Aventis Deutschland GmbH, Industrial Park Hoechst, Building G849, 65926, Frankfurt/Main, Germany
| | - Pradeep K Dhal
- Global CMC Early Development, Synthetics Platform, Sanofi Global R&D, 350 Water Street, Cambridge, MA 02141, USA
| | - Bernadette Tse Sum Bui
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| | - Karsten Haupt
- CNRS Enzyme and Cell Engineering Laboratory, Université de Technologie de Compiègne, Rue du Docteur Schweitzer, CS 60319, 60203, Compiègne Cedex, France
| |
Collapse
|
292
|
Rampuria P, Mosyak L, Root AR, Svenson K, Agostino MJ, LaVallie ER. Molecular insights into recognition of GUCY2C by T-cell engaging bispecific antibody anti-GUCY2CxCD3. Sci Rep 2023; 13:13408. [PMID: 37591971 PMCID: PMC10435522 DOI: 10.1038/s41598-023-40467-0] [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: 02/22/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023] Open
Abstract
The intestinal epithelial receptor Guanylyl Cyclase C (GUCY2C) is a tumor-associated cell surface antigen expressed across gastrointestinal malignancies that can serve as an efficacious target for colorectal cancer immunotherapy. Here, we describe a yeast surface-display approach combined with an orthogonal peptide-based mapping strategy to identify the GUCY2C binding epitope of a novel anti-GUCY2CxCD3 bispecific antibody (BsAb) that recently advanced into the clinic for the treatment of cancer. The target epitope was localized to the N-terminal helix H2 of human GUCY2C, which enabled the determination of the crystal structure of the minimal GUCY2C epitope in complex with the anti-GUCY2C antibody domain. To understand if this minimal epitope covers the entire antibody binding region and to investigate the impact of epitope position on the antibody's activity, we further determined the structure of this interaction in the context of the full-length extracellular domain (ECD) of GUCY2C. We found that this epitope is positioned on the protruding membrane-distal helical region of GUCY2C and that its specific location on the surface of GUCY2C dictates the close spatial proximity of the two antigen arms in a diabody arrangement essential to the tumor killing activity of GUCY2CxCD3 BsAb.
Collapse
Affiliation(s)
- Pragya Rampuria
- Biomedicine Design, Pfizer Inc., 610 Main St., Cambridge, MA, 02139, USA.
| | - Lidia Mosyak
- Biomedicine Design, Pfizer Inc., 610 Main St., Cambridge, MA, 02139, USA.
| | - Adam R Root
- Generate Biomedicines Inc, Cambridge, MA, USA
| | - Kristine Svenson
- Biomedicine Design, Pfizer Inc., 610 Main St., Cambridge, MA, 02139, USA
| | | | - Edward R LaVallie
- Biomedicine Design, Pfizer Inc., 610 Main St., Cambridge, MA, 02139, USA
| |
Collapse
|
293
|
Makowski EK, Chen HT, Tessier PM. Simplifying complex antibody engineering using machine learning. Cell Syst 2023; 14:667-675. [PMID: 37591204 PMCID: PMC10733906 DOI: 10.1016/j.cels.2023.04.009] [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: 10/28/2022] [Revised: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 08/19/2023]
Abstract
Machine learning is transforming antibody engineering by enabling the generation of drug-like monoclonal antibodies with unprecedented efficiency. Unsupervised algorithms trained on massive and diverse protein sequence datasets facilitate the prediction of panels of antibody variants with native-like intrinsic properties (e.g., high stability), greatly reducing the amount of subsequent experimentation needed to identify specific candidates that also possess desired extrinsic properties (e.g., high affinity). Additionally, supervised algorithms, which are trained on deep sequencing datasets obtained after enrichment of in vitro antibody libraries for one or more specific extrinsic properties, enable the prediction of antibody variants with desired combinations of extrinsic properties without the need for additional screening. Here we review recent advances using both machine learning approaches and how they are impacting the field of antibody engineering as well as key outstanding challenges and opportunities for these paradigm-changing methods.
Collapse
Affiliation(s)
- Emily K Makowski
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hsin-Ting Chen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter M Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
294
|
Lukšić F, Mijakovac A, Josipović G, Vičić Bočkor V, Krištić J, Cindrić A, Vinicki M, Rokić F, Vugrek O, Lauc G, Zoldoš V. Long-Term Culturing of FreeStyle 293-F Cells Affects Immunoglobulin G Glycome Composition. Biomolecules 2023; 13:1245. [PMID: 37627310 PMCID: PMC10452533 DOI: 10.3390/biom13081245] [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: 07/14/2023] [Revised: 08/09/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Glycosylation of IgG regulates the effector function of this antibody in the immune response. Glycosylated IgG is a potent therapeutic used for both research and clinical purposes. While there is ample research on how different cell culture conditions affect IgG glycosylation, the data are missing on the stability of IgG glycome during long cell passaging, i.e., cell "aging". To test this, we performed three independent time course experiments in FreeStyle 293-F cells, which secrete IgG with a human-like glycosylation pattern and are frequently used to generate defined IgG glycoforms. During long-term cell culturing, IgG glycome stayed fairly stable except for galactosylation, which appeared extremely variable. Cell transcriptome analysis revealed no correlation in galactosyltransferase B4GALT1 expression with galactosylation change, but with expression of EEF1A1 and SLC38A10, genes previously associated with IgG galactosylation through GWAS. The FreeStyle 293-F cell-based system for IgG production is a good model for studies of mechanisms underlying IgG glycosylation, but results from the present study point to the utmost importance of the need to control IgG galactosylation in both in vitro and in vivo systems. This is especially important for improving the production of precisely glycosylated IgG for therapeutic purposes, since IgG galactosylation affects the inflammatory potential of IgG.
Collapse
Affiliation(s)
- Fran Lukšić
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Anika Mijakovac
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| | - Goran Josipović
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| | - Vedrana Vičić Bočkor
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| | | | - Ana Cindrić
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| | - Martina Vinicki
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| | - Filip Rokić
- Laboratory for Advanced Genomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Oliver Vugrek
- Laboratory for Advanced Genomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia
| | - Vlatka Zoldoš
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| |
Collapse
|
295
|
Kalaitsidou I, Pasteli N, Venetis G, Poulopoulos A, Antoniades K. Immunohistochemical Expression of Epithelial Cell Adhesion Molecule (EpCAM) in Salivary Gland Cancer: Correlation with the Biological Behavior. Diagnostics (Basel) 2023; 13:2652. [PMID: 37627911 PMCID: PMC10453306 DOI: 10.3390/diagnostics13162652] [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: 07/19/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Salivary gland neoplasms comprise a diverse group of tumors with different biological behaviors and clinical outcomes. Understanding the underlying molecular alterations associated with these malignancies is critical for accurate diagnosis, prognosis, and treatment strategies. Among the many biomarkers under investigation, epithelial cell adhesion molecule (EpCAM) has emerged as a promising candidate in salivary gland cancer research. This article aims to provide a comprehensive overview of the differential expression of EpCAM in salivary gland cancer and its potential correlation with the biological behavior of these tumors. The clinical characteristics of 65 patients with salivary gland malignancy of different histopathological subtypes were included. We report the differential expression of EpCAM and the relationship between the clinical and histopathologic features of these tumors. Regarding the evaluation of the effect of EpCAM expression on survival, in our study, we showed that tumors with high EpCAM expression had reduced disease-free survival (DFS) and overall survival (OS) (p < 0.001) compared to patients with cancers with low EpCAM expression. In addition, the concurrent presence of perineural invasion and positive EpCAM expression appeared to be associated with shorter disease-free survival and overall survival. In conclusion, our study confirmed the prognostic value of detecting perineural invasion and EpCAM expression.
Collapse
Affiliation(s)
- Ioanna Kalaitsidou
- Department of Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.V.)
- Department of Cranio-Maxillofacial Surgery, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland
| | - Nikoleta Pasteli
- Pathology Department, G. Papanikolaou Hospital, 57010 Thessaloniki, Greece
| | - Gregory Venetis
- Department of Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.V.)
| | - Athanasios Poulopoulos
- Department of Oral Medicine and Maxillofacial Pathology, Aristotle University, 54124 Thessaloniki, Greece;
| | - Konstantinos Antoniades
- Department of Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.V.)
| |
Collapse
|
296
|
Bauer J, Rajagopal N, Gupta P, Gupta P, Nixon AE, Kumar S. How can we discover developable antibody-based biotherapeutics? Front Mol Biosci 2023; 10:1221626. [PMID: 37609373 PMCID: PMC10441133 DOI: 10.3389/fmolb.2023.1221626] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/10/2023] [Indexed: 08/24/2023] Open
Abstract
Antibody-based biotherapeutics have emerged as a successful class of pharmaceuticals despite significant challenges and risks to their discovery and development. This review discusses the most frequently encountered hurdles in the research and development (R&D) of antibody-based biotherapeutics and proposes a conceptual framework called biopharmaceutical informatics. Our vision advocates for the syncretic use of computation and experimentation at every stage of biologic drug discovery, considering developability (manufacturability, safety, efficacy, and pharmacology) of potential drug candidates from the earliest stages of the drug discovery phase. The computational advances in recent years allow for more precise formulation of disease concepts, rapid identification, and validation of targets suitable for therapeutic intervention and discovery of potential biotherapeutics that can agonize or antagonize them. Furthermore, computational methods for de novo and epitope-specific antibody design are increasingly being developed, opening novel computationally driven opportunities for biologic drug discovery. Here, we review the opportunities and limitations of emerging computational approaches for optimizing antigens to generate robust immune responses, in silico generation of antibody sequences, discovery of potential antibody binders through virtual screening, assessment of hits, identification of lead drug candidates and their affinity maturation, and optimization for developability. The adoption of biopharmaceutical informatics across all aspects of drug discovery and development cycles should help bring affordable and effective biotherapeutics to patients more quickly.
Collapse
Affiliation(s)
- Joschka Bauer
- Early Stage Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
| | - Nandhini Rajagopal
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Priyanka Gupta
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Pankaj Gupta
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Andrew E. Nixon
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| | - Sandeep Kumar
- In Silico Team, Boehringer Ingelheim, Hannover, Germany
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| |
Collapse
|
297
|
Li Y, Yang KD, Duan HY, Du YN, Ye JF. Phage-based peptides for pancreatic cancer diagnosis and treatment: alternative approach. Front Microbiol 2023; 14:1231503. [PMID: 37601380 PMCID: PMC10433397 DOI: 10.3389/fmicb.2023.1231503] [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: 05/30/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Pancreatic cancer is a devastating disease with a high mortality rate and a lack of effective therapies. The challenges associated with early detection and the highly aggressive nature of pancreatic cancer have limited treatment options, underscoring the urgent need for better disease-modifying therapies. Peptide-based biotherapeutics have become an attractive area of research due to their favorable properties such as high selectivity and affinity, chemical modifiability, good tissue permeability, and easy metabolism and excretion. Phage display, a powerful technique for identifying peptides with high affinity and specificity for their target molecules, has emerged as a key tool in the discovery of peptide-based drugs. Phage display technology involves the use of bacteriophages to express peptide libraries, which are then screened against a target of interest to identify peptides with desired properties. This approach has shown great promise in cancer diagnosis and treatment, with potential applications in targeting cancer cells and developing new therapies. In this comprehensive review, we provide an overview of the basic biology of phage vectors, the principles of phage library construction, and various methods for binding affinity assessment. We then describe the applications of phage display in pancreatic cancer therapy, targeted drug delivery, and early detection. Despite its promising potential, there are still challenges to be addressed, such as optimizing the selection process and improving the pharmacokinetic properties of phage-based drugs. Nevertheless, phage display represents a promising approach for the development of novel targeted therapies in pancreatic cancer and other tumors.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Kai-di Yang
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Hao-yu Duan
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Ya-nan Du
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
298
|
Qian Z, Song D, Ipsaro JJ, Bautista C, Joshua-Tor L, Yeh JTH, Tonks NK. Manipulating PTPRD function with ectodomain antibodies. Genes Dev 2023; 37:743-759. [PMID: 37669874 PMCID: PMC10546974 DOI: 10.1101/gad.350713.123] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/28/2023] [Indexed: 09/07/2023]
Abstract
Protein tyrosine phosphatases (PTPs) are critical regulators of signal transduction but have yet to be exploited fully for drug development. Receptor protein tyrosine phosphatase δ (RPTPδ/PTPRD) has been shown to elicit tumor-promoting functions, including elevating SRC activity and promoting metastasis in certain cell contexts. Dimerization has been implicated in the inhibition of receptor protein tyrosine phosphatases (RPTPs). We have generated antibodies targeting PTPRD ectodomains with the goal of manipulating their dimerization status ectopically, thereby regulating intracellular signaling. We have validated antibody binding to endogenous PTPRD in a metastatic breast cancer cell line, CAL51, and demonstrated that a monoclonal antibody, RD-43, inhibited phosphatase activity and induced the degradation of PTPRD. Similar effects were observed following chemically induced dimerization of its phosphatase domain. Mechanistically, RD-43 triggered the formation of PTPRD dimers in which the phosphatase activity was impaired. Subsequently, the mAb-PTPRD dimer complex was degraded through lysosomal and proteasomal pathways, independently of secretase cleavage. Consequently, treatment with RD-43 inhibited SRC signaling and suppressed PTPRD-dependent cell invasion. Together, these findings demonstrate that manipulating RPTP function via antibodies to the extracellular segments has therapeutic potential.
Collapse
Affiliation(s)
- Zhe Qian
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Graduate Program of Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York 11760, USA
| | - Dongyan Song
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Jonathan J Ipsaro
- Howard Hughes Medical Institute, W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | | | - Leemor Joshua-Tor
- Howard Hughes Medical Institute, W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Johannes T-H Yeh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Nicholas K Tonks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
| |
Collapse
|
299
|
Shi M, McHugh KJ. Strategies for overcoming protein and peptide instability in biodegradable drug delivery systems. Adv Drug Deliv Rev 2023; 199:114904. [PMID: 37263542 PMCID: PMC10526705 DOI: 10.1016/j.addr.2023.114904] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
The global pharmaceutical market has recently shifted its focus from small molecule drugs to peptide, protein, and nucleic acid drugs, which now comprise a majority of the top-selling pharmaceutical products on the market. Although these biologics often offer improved drug specificity, new mechanisms of action, and/or enhanced efficacy, they also present new challenges, including an increased potential for degradation and a need for frequent administration via more invasive administration routes, which can limit patient access, patient adherence, and ultimately the clinical impact of these drugs. Controlled-release systems have the potential to mitigate these challenges by offering superior control over in vivo drug levels, localizing these drugs to tissues of interest (e.g., tumors), and reducing administration frequency. Unfortunately, adapting controlled-release devices to release biologics has proven difficult due to the poor stability of biologics. In this review, we summarize the current state of controlled-release peptides and proteins, discuss existing techniques used to stabilize these drugs through encapsulation, storage, and in vivo release, and provide perspective on the most promising opportunities for the clinical translation of controlled-release peptides and proteins.
Collapse
Affiliation(s)
- Miusi Shi
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; Department of Chemistry, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
300
|
Hosseini SM, Mohammadnejad J, Yousefnia H, Alirezapour B, Rezayan AH. Development of 177Lu-Cetuximab-PAMAM dendrimeric nanosystem: a novel theranostic radioimmunoconjugate. J Cancer Res Clin Oncol 2023; 149:7779-7791. [PMID: 37029816 DOI: 10.1007/s00432-023-04724-z] [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: 01/28/2023] [Accepted: 03/22/2023] [Indexed: 04/09/2023]
Abstract
PURPOSE Epidermal growth factor receptors (EGFRs) are overexpressed in a wide range of tumors and are attractive candidates to target in targeted therapies. This study aimed to introduce a novel radiolabeled compound, 177Lu-cetuximab-PAMAM G4, for the treatment of EGFR-expressing tumors. METHODS In this study, the cetuximab mAb was bound to PAMAM G4 and labeled with 177Lu via DTPA-CHX chelator. The synthesized nanosystem was confirmed by different analyses such as DLS, FT-IR, TEM, and RT-LC. Cell viability of the radioimmunoconjugate was assessed over the EGFR-expressing cell line of SW480. The biodistribution of 177Lu-Cetuximab-PAMAMG4 was determined in different intervals after injection of the radiolabeled compound in normal and tumoral nude mice via scarification and SPECT images. RESULTS The average size of PAMAM G4 and PAMAM-Cetuximab-DTPA-CHX nanoparticles were 2 and 70 nm, respectively. 177Lu-Cetuximab-PAMAMG4 was prepared with radiochemical purity of more than 98%. The survival rates of SW480 cells at 24, 48, and 72 h post-treatment with177Lu-Cetuximab-PAMAMG4 (500 nM) were 18%, 15%, and 14%, respectively. The biodistribution studies showed a significant accumulation of 177Lu-Cetuximab-PAMAM in the EGFR-expressing tumor. CONCLUSION According to the results, this new agent can be considered as an efficient therapeutic complex for tumors expressing EGFR receptors.
Collapse
Affiliation(s)
- Seyed Mohammad Hosseini
- Department of Life Science Engineering, Faculty of Modern Science and Technology, Nano Biotechnology Group, University of Tehran, Tehran, 1439957131, Iran
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, 14155-1339, Iran
| | - Javad Mohammadnejad
- Department of Life Science Engineering, Faculty of Modern Science and Technology, Nano Biotechnology Group, University of Tehran, Tehran, 1439957131, Iran
| | - Hassan Yousefnia
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, 14155-1339, Iran.
| | - Behrouz Alirezapour
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, 14155-1339, Iran
| | - Ali Hossein Rezayan
- Department of Life Science Engineering, Faculty of Modern Science and Technology, Nano Biotechnology Group, University of Tehran, Tehran, 1439957131, Iran
| |
Collapse
|