1
|
Gholami S, Saffarfar H, Mehraban MR, Ardabili NS, Elhami A, Ebrahimi S, Ali-Khiavi P, Kheradmand R, Fattahpour SF, Mobed A. Targeting breast cancer: the promise of phage-based nanomedicines. Breast Cancer Res Treat 2025; 211:561-580. [PMID: 40244536 DOI: 10.1007/s10549-025-07696-5] [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: 01/12/2025] [Accepted: 03/23/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Breast cancer is a leading cause of cancer-related mortality among women worldwide, characterized by its aggressive nature, propensity for metastasis, and resistance to standard treatment modalities. Traditional therapies, including surgery, chemotherapy, and radiation, often encounter significant limitations such as systemic toxicity and lack of specificity. OBJECTIVE This review aims to evaluate the recent advancements in phage-based nanomedicines as a novel approach for targeted breast cancer therapy, focusing on their mechanisms of action, therapeutic benefits, and the challenges faced in clinical implementation. METHODS A comprehensive literature review was conducted, analyzing studies that investigate the application of bacteriophages in cancer therapy, particularly in breast cancer. The review highlights the integration of nanotechnology with phage therapy, examining the potential for enhanced targeting and reduced side effects. RESULTS Phage-based nanomedicines have shown promise in selectively targeting breast cancer cells while sparing healthy tissues, thereby improving therapeutic efficacy and safety profiles. The unique properties of bacteriophages, including their ability to be engineered for specific targeting and their natural ability to induce immune responses, present significant advantages over conventional treatments. CONCLUSION The integration of phage therapy with nanotechnology represents a promising frontier in the fight against breast cancer. This review underscores the need for continued research to address existing challenges and to explore the full potential of phage-based nanomedicines in improving patient outcomes in breast cancer treatment.
Collapse
Affiliation(s)
- Sarah Gholami
- Young Researcher and Elite Club, Islamic Azad University, Babol Branch, Babol, Iran
| | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Anis Elhami
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sara Ebrahimi
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Payam Ali-Khiavi
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Kheradmand
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahmad Mobed
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Talukdar A, Giri S, Doley R. Kraits of Indian subcontinent: Natural History, Risks, Venom Variation, Lethality and Treatment Strategies - A comprehensive review. Toxicon 2025:108406. [PMID: 40374096 DOI: 10.1016/j.toxicon.2025.108406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/30/2025] [Accepted: 05/11/2025] [Indexed: 05/17/2025]
Abstract
The World Health Organization (WHO) has re-classified "Snakebite" as a Neglected Tropical Disease in 2017, and estimated that as many as 5.4 million people suffer from snakebites every year. Out of this large number of snakebites, envenoming occurs in about 50% of the cases, and the number of resulting deaths could be as high as 138,000. The genus Bungarus commonly known as kraits are medically important elapid snakes widely distributed in the Indian subcontinent, southern China and the Southeast Asian countries (except Philippines). The Indian subcontinent (India, Bangladesh, Bhutan, Nepal, Pakistan, Sri Lanka and Maldives) is home to 8-9 krait species, among which B. caeruleus and B. niger are highly venomous. This review presents the current state of knowledge on krait bites in the Indian subcontinent. The risk of envenomation by kraits, the venom lethality and management in the Indian subcontinent have been critically analyzed. Moreover, the issue of dry bites from kraits and their management has also been reviewed. Furthermore, critical aspects, such as knowledge of snakebite management among healthcare workers, clinical symptoms of snakebite patients, and treatment in healthcare facilities including antivenom administration and their clinical efficacy, have helped us in identifying the critical knowledge gaps. Proposed preventive measures will help to reduce krait bite associated mortality and morbidity. Moreover, development and accessibility to affordable treatment options may help in the effective management of krait bites.
Collapse
Affiliation(s)
- Amit Talukdar
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam, 784028, India
| | - Surajit Giri
- Demow Rural Government Community Health Centre, Raichai, Konwar Dihingia Gaon, Sivasagar, Assam, India
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam, 784028, India.
| |
Collapse
|
3
|
Suksomboon P, Rattanasroi K, Osotprasit S, Chansap S, Prachasuphap A, Dhepakson P, Kueakhai P, Changklungmoa N. Construction, expression, and characterization of scFv fragment against Fasciola gigantica cathepsin L1H. Parasitol Res 2025; 124:51. [PMID: 40343526 PMCID: PMC12064615 DOI: 10.1007/s00436-025-08499-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Fasciola spp. infection is a significant zoonotic disease. Fasciola gigantica cathepsin L1H (FgCathL1H) is expressed across the life stages of Fasciola gigantica: newly excysted juvenile (NEJ), juvenile, and adult. An emerging tool for diagnosing fasciolosis in humans and cattle involves single-chain variable fragments (scFv) antibodies. These antibodies, consisting of linked variable regions of heavy chains (VHs) and light chains (VLs), retain binding specificity and affinity. This study aims to construct, express, and characterize an scFv antibody for use in a diagnostic kit for fasciolosis. The study successfully constructed and expressed recombinant scFv antibody genes derived from mouse spleen cells in Escherichia coli HB2151. Specific VH and VL fragments targeting recombinant FgCathL1H were amplified, inserted into a phagemid vector (pCANTAB5E), and transformed into E. coli TG1. Infection with the M13KO7 helper phage produced recombinant phages, and scFv clones with a high binding capacity were selected through three rounds of bio-panning. The expression of scFv proteins was induced with 1 mM IPTG, yielding antibodies detectable in the culture supernatant and periplasmic space. The indirect ELISA revealed strong binding in 10 scFv phage clones, which were sequenced and analyzed via computer-guided homology modeling and showed a similar classification to CDR1-3, consisting of VHs and VLs. The scFv DNA construct was approximately 747 bp in length. The SDS-PAGE, ELISA, and western blot confirmed the specificity of the scFv clone 1B, particularly at ~ 29 kDa. Docking studies showed epitopes on the scFv interacting with FgCathL1H. This scFv reacted specifically with F. gigantica antigens at 36 kDa (whole body (WB) of metacercaria and NEJ) and ~ 28 kDa (WB of 4-week-old juveniles and adults, and adult excretory-secretory protein (ES)). Immunolocalization showed positive staining in the cecal epithelium. Thus, scFv anti-rFgCathL1H shows promise for diagnosing fasciolosis.
Collapse
Affiliation(s)
- Phawiya Suksomboon
- Faculty of Allied Health Sciences, Burapha University, Long-Hard Bangsaen Road, Mueang District, Chonburi, 20131, Thailand
| | - Komsil Rattanasroi
- Faculty of Allied Health Sciences, Burapha University, Long-Hard Bangsaen Road, Mueang District, Chonburi, 20131, Thailand
| | - Supawadee Osotprasit
- Faculty of Allied Health Sciences, Burapha University, Long-Hard Bangsaen Road, Mueang District, Chonburi, 20131, Thailand
| | - Supanan Chansap
- Faculty of Allied Health Sciences, Burapha University, Long-Hard Bangsaen Road, Mueang District, Chonburi, 20131, Thailand
| | - Apichai Prachasuphap
- Department of Medical Sciences, Medical Life Sciences Institute, 88/7 Tiwanon Road, Talad Kwan Subdistrict, Muang District, Nonthaburi, 11000, Thailand
| | - Panadda Dhepakson
- Department of Medical Sciences, Medical Life Sciences Institute, 88/7 Tiwanon Road, Talad Kwan Subdistrict, Muang District, Nonthaburi, 11000, Thailand
| | - Pornanan Kueakhai
- Faculty of Allied Health Sciences, Burapha University, Long-Hard Bangsaen Road, Mueang District, Chonburi, 20131, Thailand
- Research Unit for Vaccine and Diagnosis of Parasitic Diseases, Burapha University, Long-Hard Bangsaen Road, Mueang District, Chonburi, 20131, Thailand
| | - Narin Changklungmoa
- Faculty of Allied Health Sciences, Burapha University, Long-Hard Bangsaen Road, Mueang District, Chonburi, 20131, Thailand.
- Research Unit for Vaccine and Diagnosis of Parasitic Diseases, Burapha University, Long-Hard Bangsaen Road, Mueang District, Chonburi, 20131, Thailand.
| |
Collapse
|
4
|
Menzies SK, Patel RN, Ainsworth S. Practical progress towards the development of recombinant antivenoms for snakebite envenoming. Expert Opin Drug Discov 2025:1-21. [PMID: 40302313 DOI: 10.1080/17460441.2025.2495943] [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: 11/04/2024] [Accepted: 04/16/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Snakebite envenoming is a neglected tropical disease that affects millions globally each year. In recent years, research into the potential production of recombinant antivenoms, formulated using mixtures of highly defined anti-toxin monoclonal antibodies, has rapidly moved from a theoretical concept to demonstrations of practical feasibility. AREAS COVERED This article examines the significant practical advancements in transitioning recombinant antivenoms from concept to potential clinical translation. The authors have based their review on literature obtained from Google Scholar and PubMed between September and November 2024. Coverage includes the development and validation of recombinant antivenom antibody discovery strategies, the characterization of the first broadly neutralizing toxin class antibodies, and recent translational proof-of-concept experiments. EXPERT OPINION The transition of recombinant antivenoms from a 'concept' to the current situation where high-throughput anti-venom mAb discovery is becoming routine, accompanied by increasing evidence of their broad neutralizing capacity in vivo, has been extraordinary. It is now important to build on this momentum by expanding the discovery of broadly neutralizing mAbs to encompass as many toxin classes as possible. It is anticipated that key demonstrations of whether recombinant antivenoms can match or surpass existing conventional polyvalent antivenoms in terms of neutralizing scope and capacity will be achieved in the next few years.
Collapse
Affiliation(s)
- Stefanie K Menzies
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, UK
| | - Rohit N Patel
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Stuart Ainsworth
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
5
|
Qu L, Chi Z, Ye BC. Development and application of an interbacterial DNA delivery system based on M13 bacteriophage. Arch Microbiol 2025; 207:113. [PMID: 40186660 DOI: 10.1007/s00203-025-04309-z] [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: 01/14/2025] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
With the rapid development in synthetic biology, bacterial therapy has been widely applied in disease diagnosis and treatment. A key step in regulating the execution of specific instructions by engineered bacteria in vivo involves the transfer of information between bacteria. Currently, small molecule compounds and proteins are commonly used as carriers for inter-bacterial information transfer. However, DNA, as an information carrier, boasts higher information capacity and ease of editing. Based on this, we have developed an engineered bacterial DNA delivery system using M13 bacteriophage to study the cooperative response mechanisms between bacteria. Firstly, we leveraged the ability of engineered bacteria to secrete phagemid DNA into the extracellular environment via bacteriophages and to acquire phagemid DNA through infection by extracellular phagemids, thereby developing a dynamic phagemid replenishment system. This system can maintain the stability of phagemid DNA in environments lacking antibiotic selection pressure, constructing a stable DNA delivery platform for phagemid output. Subsequently, using this engineered bacterial DNA delivery system, we modularized gene circuits and placed them in two different engineered bacteria. Through DNA transfer, we achieved a cooperative response to signals between the two bacteria, laying the groundwork for multi-bacterial joint regulation. Our research not only provides an effective tool for information transfer in engineered bacteria but also offers a novel approach for multi-bacterial therapy.
Collapse
Affiliation(s)
- Li Qu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhou Chi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Bang-Ce Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
6
|
Yang SQ, Yang K, Li XR, Zheng Y, Cao SJ, Yan QG, Huang XB, Wen YP, Zhao Q, Du SY, Lang YF, Zhao S, Li C, Wu R. A novel linear B cell epitope of the porcine circovirus type 3 capsid protein identified by phage display technology. J Virol Methods 2025; 333:115080. [PMID: 39638262 DOI: 10.1016/j.jviromet.2024.115080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Porcine circovirus type 3 (PCV3) is endemic in swine worldwide and causes reproductive disorders, dermatitis and nephrotic syndrome, and multi-organ inflammation. PCV3 capsid protein (Cap) can self-assemble into virus-like particles (VLPs), and is an ideal candidate for vaccines and diagnostic reagents.In this study, the recombinant PCV3 Cap protein was successfully expressed in E. coli by deleting the nuclear localization sequence (NLS). The PCV3 VLPs were observed by transmission electron microscopy, and its immunogenicity was evaluated in six-week-old female BALB/c mice. A monoclonal antibody was named mAb 2D6, and demonstrated strong reactivity and specificity to PCV3 Cap. The purified mAb 2D6 was further used for bio-panning to select phage expressing specific epitopes from phage-displayed 7 mer-peptide library. A novel linear B-cell epitope, recognized by mAb 2D6, was identified at the amino acid region 47-53 of Cap. The phage peptide sequences were analyzed using multiple sequence alignment and evaluated by peptide ELISA. These results provide insights for developing diagnostic tools and potential vaccines for PCV3.
Collapse
Affiliation(s)
- Shu-Qing Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ke Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin-Ran Li
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi Zheng
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - San-Jie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qi-Gui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Bo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi-Ping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Sen-Yan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi-Fei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chun Li
- Sichuan Animal Disease Prevention and Control Center, Chengdu 610047, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; National Animal Experiments Teaching Demonstration Center, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
7
|
Riaño-Umbarila L, Romero-Moreno JA, Possani LD, Becerril B. State of the art on the development of a recombinant antivenom against Mexican scorpion stings. Toxicon 2025; 257:108306. [PMID: 40049537 DOI: 10.1016/j.toxicon.2025.108306] [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: 01/13/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Around 2,750 species of scorpions have been recorded worldwide and classified into 21 families and 208 genera. Of these, the family Buthidae stands out as one of the largest, comprising several genera including the genus Centruroides with 102 recorded species. This genus is home to the largest number of species dangerous to humans as described in Mexico, where there are 55 species of the genus Centruroides, of which more than 24 are of medical importance. Envenoming in humans is caused by the presence of peptides (toxins) in the venom that modify the gating mechanism of Na+ voltage dependent ionic channels. Therefore, a rational approach to generate a new antivenom is to obtain neutralizing antibodies against these toxins, whose average abundance in venom is 10%. In this review paper, we document that from the characterization of the lethal venoms of Mexican scorpions, 30 lethal components have been identified, so their neutralization represents an enormous challenge. Thanks to phage display and directed evolution technologies, it has been possible to generate specific antibody fragments against several of these toxins, some of which exhibit broad cross-neutralization. Currently, progress has been made in neutralizing the venoms of 9 species with the use of recombinant antibody fragments, mainly of human origin. One of them has the potential to neutralize approximately 20 toxins.
Collapse
Affiliation(s)
- Lidia Riaño-Umbarila
- CONAHCYT, Mexico; Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - José Alberto Romero-Moreno
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Baltazar Becerril
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| |
Collapse
|
8
|
Chen Y, Liu X, Li J, Liu X. Development of a Sensitive Enzyme Immunoassay Using Phage-Displayed Antigen-Binding Fragments for Zearalenone Detection in Cereal Samples. Foods 2025; 14:746. [PMID: 40077448 PMCID: PMC11898766 DOI: 10.3390/foods14050746] [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: 01/24/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Zearalenone (ZEN), a non-steroidal estrogenic mycotoxin, contaminates animal feed and grain crops, thereby entering the food chain and posing a significant threat to human health. Consequently, there is an urgent need for a sensitive and rapid method for detecting trace levels of ZEN. In this study, we developed a phage-displayed antigen-binding fragment (Fab-phage) and established a Fab-phage-based enzyme-linked immunosorbent assay (Fab-pELISA) for ZEN detection. Under optimal conditions, this method exhibits a half-maximal inhibitory concentration of 0.36 ng/mL, with a linear range from 0.07 to 3.89 ng/mL and a detection limit of 0.03 ng/mL. The method demonstrates high selectivity towards ZEN and good recovery rates of 97.35-122.66% with relative standard deviations not exceeding 3.5%. Furthermore, the detection results obtained using Fab-pELISA on real cereal samples are consistent with those from high-performance liquid chromatography, meeting practical application requirements. Therefore, the Fab-phage serves as a valuable biochemical reagent, and the established Fab-pELISA represents a promising analytical strategy for detecting ZEN and other trace toxic contaminants in cereals.
Collapse
Affiliation(s)
| | | | | | - Xing Liu
- School of Food Science and Engineering, Hainan University, Haikou 570228, China (X.L.); (J.L.)
| |
Collapse
|
9
|
Atanasiu D, Saw WT, Friedman HM, Cohen GH. Targeting Herpes Simplex Virus Glycoprotein D with Bispecific Antibodies: Expanding Therapeutic Horizons by Searching for Synergy. Viruses 2025; 17:249. [PMID: 40007004 PMCID: PMC11860751 DOI: 10.3390/v17020249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Herpes simplex viruses (HSV-1 and HSV-2), which can be transmitted both orally and sexually, cause lifelong morbidity and in some cases, meningitis and encephalitis. While both the passive transfer of neutralizing antibodies and placental transfer of anti-HSV monoclonal antibodies (Mabs) have shown therapeutic promise in animal models, clinical trials have yet to identify approved immunotherapeutics for herpes infection. Here, we present strategies for the generation of recombinant bispecific antibodies (BsAbs) that target different domains of glycoprotein D (gD), crucial for HSV entry, that have the potential to outperform the effect of individual Mabs to curb herpes infection. Specifically, we selected three pairs of Mabs from our extensive panel for BsAb design and production based on their binding site and ability to block virus entry. Actual binding of BsAbs to gD and epitope availability on gD after BsAb binding were characterized using surface plasmon resonance (SPR) and inhibition by IgG Fab fragments generated from selected Mabs. While one BsAb exhibited an additive effect similar to that observed using a combination of the Mabs utilized for its generation, two showed antagonistic effects, suggesting that the simultaneous engagement of two epitopes or selective binding to one affected their activity against HSV. One BsAb (DL11/1D3) targeting the binding site for both nectin-1 and HVEM receptors demonstrated synergistic inhibitory activity against HSV, outperforming the effect of the individual antibodies. Recombinant DL11/1D3 antibody variants, in which the size of one or both paratopes was decreased to single chains (scFv-Fc), highlighted differences in potency depending on antibody size and format. We propose that BsAbs to individual glycoproteins offer a potential avenue for herpes therapeutics, but their design, mechanism of action, antibody format, and epitope engagement require careful consideration of structure for optimal efficacy.
Collapse
MESH Headings
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/therapeutic use
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/antagonists & inhibitors
- Humans
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/drug effects
- Antibodies, Viral/immunology
- Animals
- Virus Internalization/drug effects
- Herpes Simplex/therapy
- Herpes Simplex/virology
- Antibodies, Neutralizing/immunology
- Antibodies, Monoclonal/immunology
- Herpesvirus 2, Human/immunology
- Herpesvirus 2, Human/drug effects
- Epitopes/immunology
- Nectins
- Cell Adhesion Molecules/metabolism
Collapse
Affiliation(s)
- Doina Atanasiu
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Wan Ting Saw
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Harvey M. Friedman
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Gary H. Cohen
- Department of Basic and Translational Sciences, School of Dental Medicine, Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
10
|
Wang J, Zhou X, Elazab ST, Huang J, Hsu WH. Current Review of Monoclonal Antibody Therapeutics in Small Animal Medicine. Animals (Basel) 2025; 15:472. [PMID: 40002954 PMCID: PMC11852019 DOI: 10.3390/ani15040472] [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: 01/10/2025] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Monoclonal antibody therapy has been a cornerstone of human healthcare for nearly four decades, effectively treating a wide range of diseases including cancers, autoimmune disorders, and inflammatory conditions. However, its application in veterinary medicine is a relatively recent development, offering a promising therapeutic approach for managing chronic diseases in small animals. Dogs and cats, like humans, suffer from chronic conditions such as cancer, arthritis, allergies, and chronic pain, which mAb therapy could potentially address. This review aims to explore the therapeutic potential of mAb therapy in small animal medicine, focusing on currently authorized products, including their mechanisms of action, clinical efficacy, and safety concerns. A comprehensive review of the literature was conducted to evaluate the use of mAbs in veterinary medicine, specifically in the treatment of chronic disorders. While mAb therapy has shown significant benefits in human healthcare, challenges remain in its application to veterinary practice, including safety concerns and the limited availability of approved products. Despite these challenges, mAb therapy holds great promise for improving the management of chronic diseases in animals, with future research and development potentially expanding its clinical use.
Collapse
Affiliation(s)
- Jianzhong Wang
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China;
| | - Xueying Zhou
- Department of Veterinary Clinical Science, College of Veterinary Medicine, China Agricultural University, Beijing 100107, China;
| | - Sara T. Elazab
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Jian Huang
- Institute of Qinhai-Tibetan Plateau, College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China;
| | - Walter H. Hsu
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
11
|
Liu J, Wu L, Xie A, Liu W, He Z, Wan Y, Mao W. Unveiling the new chapter in nanobody engineering: advances in traditional construction and AI-driven optimization. J Nanobiotechnology 2025; 23:87. [PMID: 39915791 PMCID: PMC11800653 DOI: 10.1186/s12951-025-03169-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
Nanobodies (Nbs), miniature antibodies consisting solely of the variable region of heavy chains, exhibit unique properties such as small size, high stability, and strong specificity, making them highly promising for disease diagnosis and treatment. The engineering production of Nbs has evolved into a mature process, involving library construction, screening, and expression purification. Different library types, including immune, naïve, and synthetic/semi-synthetic libraries, offer diverse options for various applications, while display platforms like phage display, cell surface display, and non-surface display provide efficient screening of target Nbs. Recent advancements in artificial intelligence (AI) have opened new avenues in Nb engineering. AI's exceptional performance in protein structure prediction and molecular interaction simulation has introduced novel perspectives and tools for Nb design and optimization. Integrating AI with traditional experimental methods is anticipated to enhance the efficiency and precision of Nb development, expediting the transition from basic research to clinical applications. This review comprehensively examines the latest progress in Nb engineering, emphasizing library construction strategies, display platform technologies, and AI applications. It evaluates the strengths and weaknesses of various libraries and display platforms and explores the potential and challenges of AI in predicting Nb structure, antigen-antibody interactions, and optimizing physicochemical properties.
Collapse
Affiliation(s)
- Jiwei Liu
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China
| | - Lei Wu
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China
| | - Anqi Xie
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
| | - Weici Liu
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China
| | - Zhao He
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, 13850, USA.
- Department of Biomedical Engineering, The Pq Laboratory of BiomeDx/Rx, Binghamton University, Binghamton, NY, 13902, USA.
| | - Wenjun Mao
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China.
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China.
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, No. 299 Qingyang Rd., Wuxi, 214023, China.
| |
Collapse
|
12
|
Gencsoy Eker S, Inetas Yengin G, Tatar C, Oktem G. A Comprehensive Review of the Mechanisms and Clinical Development of Monoclonal Antibodies in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:181-203. [PMID: 39666264 DOI: 10.1007/5584_2024_838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Cancer is still the disease that ranks first in human mortality in the twenty-first century. In the last 20 years, the concept of molecular targeted therapy has come to the fore with the use of small molecule agents or signal transduction inhibitors that show anticancer effects for certain types of cancer. Monoclonal antibodies, which have a therapeutic effect, especially by providing signal transduction inhibition, are used clinically as first-line treatment in various types of cancer. Molecular targeted therapies are critical for eliminating the adverse effects and drug resistance problems that occur in traditional cancer treatments. This review summarizes current information on various targeted therapeutic agents, including the structure and classification of monoclonal antibodies, their production methods and mechanisms of action, the monoclonal antibodies used in clinical trials, the complement system mechanism and cancer relationship, and the relationship between complement-dependent cytotoxicity and monoclonal antibodies.
Collapse
Affiliation(s)
- Selen Gencsoy Eker
- Department of Stem Cell, Graduate School of Health Sciences, Ege University, Izmir, Turkey
| | - Gizem Inetas Yengin
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Cansu Tatar
- Department of Molecular Biology and Genetics, Yildiz Technical University, Istanbul, Turkey
| | - Gulperi Oktem
- Department of Stem Cell, Graduate School of Health Sciences, Ege University, Izmir, Turkey.
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey.
| |
Collapse
|
13
|
Gutiérrez JM, R Casewell N, Laustsen AH. Progress and Challenges in the Field of Snakebite Envenoming Therapeutics. Annu Rev Pharmacol Toxicol 2025; 65:465-485. [PMID: 39088847 DOI: 10.1146/annurev-pharmtox-022024-033544] [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: 08/03/2024]
Abstract
Snakebite envenoming kills and maims hundreds of thousands of people every year, especially in the rural settings of tropical regions. Envenomings are still treated with animal-derived antivenoms, which have prevented many lives from being lost but which are also medicines in need of innovation. Strides are being made to improve envenoming therapies, with promising efforts made toward optimizing manufacturing and quality aspects of existing antivenoms, accelerating research and development of recombinant antivenoms based on monoclonal antibodies, and repurposing of small-molecule inhibitors that block key toxins. Here, we review the most recent advances in these fields and discuss therapeutic opportunities and limitations for different snakebite treatment modalities. Finally, we discuss challenges related to preclinical and clinical evaluation, regulatory pathways, large-scale manufacture, and distribution and access that need to be addressed to fulfill the goals of the World Health Organization's global strategy to prevent and control snakebite envenoming.
Collapse
Affiliation(s)
- José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica;
| | - Nicholas R Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom;
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark;
| |
Collapse
|
14
|
McGonigle P. How Biologics Have Changed the Drug Discovery Landscape. Annu Rev Pharmacol Toxicol 2025; 65:29-46. [PMID: 39389098 DOI: 10.1146/annurev-pharmtox-061724-080811] [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: 10/12/2024]
Abstract
Advances in molecular biology and molecular genetics as well as major scientific breakthroughs in immunology and oncology have led to the rapid growth of biologic therapeutics. Their success has resulted in significant changes to virtually every step in the drug discovery and development process. Biologics are produced by living organisms, and screening libraries are generated by immunization or phage display. Lead optimization utilizes sophisticated protein engineering to improve drug-like properties and targeting specificity. The manufacturing process for biologics is complex and requires highly specialized facilities. Determination of pharmacology and safety must overcome the complications associated with species specificity. Initial clinical testing must proceed more slowly and carefully due to the limited predictive utility of preclinical data. In summary, the drug discovery and development process has been dramatically altered by biologic therapeutics and will continue to evolve with the introduction of messenger RNA-based therapeutics and the application of artificial intelligence.
Collapse
Affiliation(s)
- Paul McGonigle
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
15
|
El-Kalamawy HA, Awwad MH, Diab TM, Okasha H, Abdel-Kareim AM, Marawan MA, Shoulah SA, El-Dabaa E. Construction of Camelus dromedaries Immune Single Domain Antibodies Library for Development of Schistosoma mansoni Specific Nanobodies Using Phage Display Strategy. Recent Pat Biotechnol 2025; 19:69-83. [PMID: 39840412 DOI: 10.2174/0118722083275669231227063413] [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: 08/05/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2025]
Abstract
BACKGROUND Schistosoma mansoni poses a considerable global public health challenge. In Egypt, approximately 60% of the inhabitants in the Northern and Eastern areas of the Nile Delta are affected by this parasite, whereas the Southern region experiences a significantly lower infection rate of 6%. AIM Construction of an immune phage display Nbs library based on the VHH framework for selecting S. mansoni-specific Nbs for seeking cost-effective, sensitive, and specific diagnostic tools for rapidly detecting Schistosoma mansoni. METHODS Camel was immunized using soluble adult worm antigens (SAWP) for the production of Variable domains of heavy chains of camelid heavy-chain only antibodies (VHHs), which are known as nanobodies (Nb). The PBMCs repertoires VHH sequences library have been constructed with a high percentage of insertion and right orientation using pADL-23c phagmid and M13 phage followed by three rounds of bio-panning against SAWP using phage display technique. Evaluations using polyclonal phage ELISA and other techniques have been carried out to reveal the successful enrichment of anti-SAWP Nbs (VHH) clones. Evaluation of the diagnostic potentiality of these Nbs was carried out using ELISA on human serum samples confirmed for S. mansoni infection. Receiver Operator of Characteristics (ROC) curve analysis was used for discrimination between S. mansoni infection and both negative controls and the Fasciola hepatica group. RESULTS Using monoclonal ELISA, Nbs of 22 clones out of 24 selected clones showed binding affinity to SAWP. The cutoff values of the produced anti-S. mansoni Nbs was > 0.19, leading to 80% sensitivity, 95% specificity, and 90% accuracy. Sequence analysis of three of these Nbs with high binding affinities showed diversity in their targets, considering their CDR3 aa sequences. CONCLUSION This study successfully generated a diverse phage library enriched with anti-S. mansoni VHHs. The nanobodies produced exhibit high diagnostic potential for detecting S. mansoni infection in human patients, offering a promising avenue for the development of efficient diagnostic tools. The innovative approach described herein may have potential applications for patent considerations in the field of the field of diagnostic technology.
Collapse
Affiliation(s)
- Hadeer Adel El-Kalamawy
- Center of Excellence in Recombinant Biopharmaceutical Proteins, Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mohammed H Awwad
- Department of Zoology, Faculty of Science, Benha University, Benha, Egypt
| | - Tarek M Diab
- Department of Parasitology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Hend Okasha
- Center of Excellence in Recombinant Biopharmaceutical Proteins, Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | | | - Marawan A Marawan
- Department of Animal Medicine (Infectious Diseases), Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Salma A Shoulah
- Department of Animal Medicine (Infectious Diseases), Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Ehab El-Dabaa
- Center of Excellence in Recombinant Biopharmaceutical Proteins, Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|
16
|
Song BPC, Lai JY, Choong YS, Khanbabaei N, Latz A, Lim TS. Isolation of anti-Ancylostoma-secreted protein 5 (ASP5) antibody from a naïve antibody phage library. J Immunol Methods 2024; 535:113776. [PMID: 39551437 DOI: 10.1016/j.jim.2024.113776] [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: 06/19/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
Ancylostoma species are parasitic nematodes that release a multitude of proteins to manipulate host immune responses to facilitate their survival. Among the released proteins, Ancylostoma-secreted protein 5 (ASP5) plays a pivotal role in mediating host-parasite interactions, making it a promising target for interventions against canine hookworm infections caused by Ancylostoma species. Antibody phage display, a widely used method for generating human monoclonal antibodies was employed in this study. A bacterial expression system was used to produce ASP5 for biopanning. A single-chain fragment variable (scFv) monoclonal antibody against ASP5 was generated from the naïve Human AntibodY LibrarY (HAYLY). The resulting scFv antibody was characterized to elucidate its antigen-binding properties. The identified monoclonal antibody showed good specificity and binding characteristics which highlights its potential for diagnostic applications for hookworm infections.
Collapse
Affiliation(s)
- Brenda Pei Chui Song
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Jing Yi Lai
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Yee Siew Choong
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | | | - Andreas Latz
- Gold Standard Diagnostics Frankfurt GmbH, Dietzenbach, Germany
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia; Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800 Penang, Malaysia.
| |
Collapse
|
17
|
Istomina PV, Gorchakov AA, Paoin C, Yamabhai M. Phage display for discovery of anticancer antibodies. N Biotechnol 2024; 83:205-218. [PMID: 39186973 DOI: 10.1016/j.nbt.2024.08.506] [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: 02/24/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Antibodies and antibody-based immunotherapeutics are the mainstays of cancer immunotherapy. Expanding the repertoire of cancer-specific and cancer-associated epitopes targetable with antibodies represents an important area of research. Phage display is a powerful approach allowing the use of diverse antibody libraries to be screened for binding to a wide range of targets. In this review, we summarize the basics of phage display technology and highlight the advances in anticancer antibody identification and modification via phage display platform. Finally, we describe phage display-derived anticancer monoclonal antibodies that have been approved to date or are in clinical development.
Collapse
Affiliation(s)
- Polina V Istomina
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Suranaree, Muang, 111 University Avenue, Nakhon Ratchasima 30000, Thailand
| | - Andrey A Gorchakov
- Institute of Molecular and Cellular Biology of the Siberian Branch of the Russian Academy of Sciences, Lavrentieva 8/2, Novosibirsk 630090, Russia
| | - Chatchanok Paoin
- Medical Oncology Division, Institute of Medicine, Suranaree University of Technology, Suranaree, Muang, 111 University Avenue, Nakhon Ratchasima 30000, Thailand
| | - Montarop Yamabhai
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Suranaree, Muang, 111 University Avenue, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
18
|
Cui H, Elkord E. Turning Cancer Immunotherapy to the Emerging Immune Checkpoint TIGIT: Will This Break Through the Limitations of the Legacy Approach? Vaccines (Basel) 2024; 12:1306. [PMID: 39771968 PMCID: PMC11679306 DOI: 10.3390/vaccines12121306] [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: 10/24/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 01/04/2025] Open
Abstract
The discovery of immune checkpoints (ICs) has pushed cancer treatment into the next era. As an emerging immune checkpoint, the TIGIT/CD155 axis inhibits the cytotoxicity of T and NK cells through multiple pathways. Immune checkpoint inhibitors (ICIs) targeting TIGIT are hopefully expected to address the issue of unresponsiveness to anti-PD-(L)1 monoclonal antibodies (mAbs) by combination therapy. This paper presents insights on the expression, structure and mechanism of action of TIGIT, as well as the principles and methods of designing mAbs targeting TIGIT and their clinical data. The advantages and disadvantages of targeting TIGIT using mAbs, bispecific and tri-specific antibodies (bsAbs and tsAbs), peptides, and compounds, in addition to potential combination therapies of anti-TIGIT with anti-PD-1 or cancer vaccines, are addressed. Finally, perspectives on current immunotherapies targeting TIGIT are discussed.
Collapse
Affiliation(s)
- Haozhe Cui
- Department of Biosciences and Bioinformatics, School of Science, Suzhou Municipal Key Lab in Biomedical Sciences and Translational Immunology, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China;
| | - Eyad Elkord
- Department of Biosciences and Bioinformatics, School of Science, Suzhou Municipal Key Lab in Biomedical Sciences and Translational Immunology, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China;
- College of Health Sciences, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester M5 4WT, UK
| |
Collapse
|
19
|
Du K, He H, Zhao L, Gao L, Li T. Application of Anti-Immune Complex Reagents in Small Molecule Analyte Immunoassays. ACS OMEGA 2024; 9:45688-45705. [PMID: 39583695 PMCID: PMC11579784 DOI: 10.1021/acsomega.4c07268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024]
Abstract
The detection of small molecule analytes (SMAs) is of great significance for food and drug testing, environmental monitoring, and disease diagnosis. However, the performance of commercially available SMA immunoassays is limited by their low sensitivity and specificity due to the competitive format, leaving significant room for improvement. In recent years, the application of noncompetitive immunoassays for the detection of SMAs has become a hot topic, especially with the rapid evolution of antibody development technology. The remarkable development and application of anti-immune complex (anti-IC) reagents targeting antigen-specific antibodies have garnered significant interest from researchers and diagnostic companies, particularly in the field of SMA detection. The discovery and development history of anti-IC antibodies, the advantages and limitations of different anti-IC reagent preparation methods, and the mechanisms of interaction between ICs and anti-IC antibodies are reviewed. A comprehensive overview of the application of anti-IC antibodies in SMAs assay, including pesticide residue detection, mycotoxin detection, and clinical testing, as well as current challenges and potential solutions in noncompetitive immunoassays, is also summarized to provide a reference for the rapid and accurate detection of SMAs.
Collapse
Affiliation(s)
- Kai Du
- Shenzhen
New Industries Biomedical Engineering Company, Limited, Reagent Key Raw Materials R&D and Production Center, No. 16, Jinhui Road, Pingshan District, Shenzhen, Guangdong 518122, P. R. China
| | - Haihua He
- Shenzhen
New Industries Biomedical Engineering Company, Limited, Reagent Key Raw Materials R&D and Production Center, No. 16, Jinhui Road, Pingshan District, Shenzhen, Guangdong 518122, P. R. China
| | - Lan Zhao
- Shenzhen
New Industries Biomedical Engineering Company, Limited, Reagent Key Raw Materials R&D and Production Center, No. 16, Jinhui Road, Pingshan District, Shenzhen, Guangdong 518122, P. R. China
| | - Li Gao
- Shenzhen
New Industries Biomedical Engineering Company, Limited, Reagent Key Raw Materials R&D and Production Center, No. 16, Jinhui Road, Pingshan District, Shenzhen, Guangdong 518122, P. R. China
| | - Tinghua Li
- Shenzhen
New Industries Biomedical Engineering Company, Limited, Reagent Key Raw Materials R&D and Production Center, No. 16, Jinhui Road, Pingshan District, Shenzhen, Guangdong 518122, P. R. China
| |
Collapse
|
20
|
Yin H, Li L, Feng X, Wang Z, Zheng M, Zhao J, Fan X, Wu W, Gao L, Zhan Y, Zhao M, Lu Q. 2D4, a humanized monoclonal antibody targeting CD132, is a promising treatment for systemic lupus erythematosus. Signal Transduct Target Ther 2024; 9:323. [PMID: 39551768 PMCID: PMC11570697 DOI: 10.1038/s41392-024-02017-6] [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: 11/24/2023] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Current therapies for systemic lupus erythematosus that target a particular factor or cell type exhibit limited effectiveness. To address this limitation, our focus was on CD132, a subunit common to six inflammatory factor receptors implicated in SLE. Our study revealed heightened CD132 expression in SLE patients' lymphocytes, contributing to the production of pro-inflammatory cytokines and immunoglobulins. We developed a novel humanized anti-CD132 monoclonal antibody, named as 2D4. 2D4 efficiently blocked IL-21 and IL-15, with limited effectiveness against IL-2, thereby suppressing T and B cells without disrupting immune tolerance. In the mouse immunization model, 2D4 virtually inhibited T cell-dependent, antigen-specific B-cell response. In lupus murine models, 2D4 mitigated inflammation by suppressing multiple pro-inflammatory cytokines and anti-dsDNA antibody titers, also diminishing proteinuria and glomerulonephritis. Compared to Belimumab, 2D4 exhibited superior efficacy in ameliorating the inflammatory state and preserving renal function. Moreover, 2D4 exhibited the ability to inhibit the production of pro-inflammatory factors and autoantibodies in PBMCs from individuals with SLE, highlighting its therapeutic potential for SLE individuals. Potent, 2D4 has the potential to significantly improve clinical outcomes in SLE and other complex autoimmune disorders.
Collapse
Affiliation(s)
- Huiqi Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Liming Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiwei Feng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zijun Wang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Meiling Zheng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Junpeng Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinyu Fan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Wu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lingyu Gao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yijing Zhan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
21
|
de Souza CC, Glória JC, da Silva ERD, de Lima Guerra Corado A, de Alcântara KÁG, Cordeiro IB, de Andrade EV, Mariúba LAM. Single-Stranded Variable Fragment Gene Libraries Built for Phage Display: An Updated Review of Design, Selection and Application. J Microbiol Biotechnol 2024; 35:e2407049. [PMID: 39631781 PMCID: PMC11813352 DOI: 10.4014/jmb.2407.07049] [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: 07/25/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 12/07/2024]
Abstract
The development of the phage display technique has brought practicality and speed when selecting high-affinity molecules. It is used to obtain single-chain variable fragments (scFvs) and has revolutionized several branches of research and industry. These are developed from gene libraries that differ in their construction strategies, which causes a diversity of sequences, specificity and binding strength of the projected molecule to its antigen. In this review, we present the recent studies that demonstrate methods and approaches using immune, naïve, synthetic and semi-synthetic libraries to construct and select scFvs. Subsequently, the characteristics of these libraries, the functionality of the scFvs and the cost-benefits of production will be discussed. In addition, we highlight the methodological trends and challenges to be overcome in order to optimize the production and application of these antibody fragments.
Collapse
Affiliation(s)
- Caio Coutinho de Souza
- Programa de Pós-graduação em Biotecnologia (PPGBIOTEC), Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Laboratório de Diagnóstico e Controle de Doenças Infecciosas na Amazônia (DCDIA), Instituto Leônidas e Maria Deane (ILMD/Fiocruz-Amazônia), Manaus, AM, Brazil
| | - Juliane Corrêa Glória
- Laboratório de Diagnóstico e Controle de Doenças Infecciosas na Amazônia (DCDIA), Instituto Leônidas e Maria Deane (ILMD/Fiocruz-Amazônia), Manaus, AM, Brazil
- Programa de Pós-Graduação em Biologia da Interação Patógeno-Hospedeiro (PPGBIO-Interação), Instituto Leônidas e Maria Deane (ILMD/Fiocruz-Amazônia), Manaus, AM, Brazil
| | - Eliza Raquel Duarte da Silva
- Laboratório de Diagnóstico e Controle de Doenças Infecciosas na Amazônia (DCDIA), Instituto Leônidas e Maria Deane (ILMD/Fiocruz-Amazônia), Manaus, AM, Brazil
| | - André de Lima Guerra Corado
- Laboratório de Diagnóstico e Controle de Doenças Infecciosas na Amazônia (DCDIA), Instituto Leônidas e Maria Deane (ILMD/Fiocruz-Amazônia), Manaus, AM, Brazil
- Universidade Nilton Lins, Manaus, AM, Brazil
| | - Kelson Ávila Graça de Alcântara
- Laboratório de Diagnóstico e Controle de Doenças Infecciosas na Amazônia (DCDIA), Instituto Leônidas e Maria Deane (ILMD/Fiocruz-Amazônia), Manaus, AM, Brazil
- Faculdade Estácio do Amazonas, Manaus, AM, Brazil
| | - Isabelle Bezerra Cordeiro
- Programa de Pós-graduação em Biotecnologia (PPGBIOTEC), Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | - Edmar Vaz de Andrade
- Programa de Pós-graduação em Biotecnologia (PPGBIOTEC), Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | - Luis André Morais Mariúba
- Programa de Pós-graduação em Biotecnologia (PPGBIOTEC), Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Laboratório de Diagnóstico e Controle de Doenças Infecciosas na Amazônia (DCDIA), Instituto Leônidas e Maria Deane (ILMD/Fiocruz-Amazônia), Manaus, AM, Brazil
- Programa de Pós-Graduação em Biologia da Interação Patógeno-Hospedeiro (PPGBIO-Interação), Instituto Leônidas e Maria Deane (ILMD/Fiocruz-Amazônia), Manaus, AM, Brazil
- Programa de Pós-graduação em Imunologia Básica e Aplicada (PPGIBA), Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| |
Collapse
|
22
|
Nur A, Lai JY, Ch'ng ACW, Choong YS, Wan Isa WYH, Lim TS. A review of in vitro stochastic and non-stochastic affinity maturation strategies for phage display derived monoclonal antibodies. Int J Biol Macromol 2024; 277:134217. [PMID: 39069045 DOI: 10.1016/j.ijbiomac.2024.134217] [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: 05/15/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Monoclonal antibodies identified using display technologies like phage display occasionally suffers from a lack of affinity making it unsuitable for application. This drawback is circumvented with the application of affinity maturation. Affinity maturation is an essential step in the natural evolution of antibodies in the immune system. The evolution of molecular based methods has seen the development of various mutagenesis approaches. This allows for the natural evolutionary process during somatic hypermutation to be replicated in the laboratories for affinity maturation to fine-tune the affinity and selectivity of antibodies. In this review, we will discuss affinity maturation strategies for mAbs generated through phage display systems. The review will highlight various in vitro stochastic and non-stochastic affinity maturation approaches that includes but are not limited to random mutagenesis, site-directed mutagenesis, and gene synthesis.
Collapse
Affiliation(s)
- Alia Nur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Jing Yi Lai
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Angela Chiew Wen Ch'ng
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Yee Siew Choong
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Wan Yus Haniff Wan Isa
- School of Medical Sciences, Department of Medicine, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia; Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800 Penang, Malaysia.
| |
Collapse
|
23
|
Slavny P, Hegde M, Doerner A, Parthiban K, McCafferty J, Zielonka S, Hoet R. Advancements in mammalian display technology for therapeutic antibody development and beyond: current landscape, challenges, and future prospects. Front Immunol 2024; 15:1469329. [PMID: 39381002 PMCID: PMC11459229 DOI: 10.3389/fimmu.2024.1469329] [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: 07/23/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
The evolving development landscape of biotherapeutics and their growing complexity from simple antibodies into bi- and multi-specific molecules necessitates sophisticated discovery and engineering platforms. This review focuses on mammalian display technology as a potential solution to the pressing challenges in biotherapeutic development. We provide a comparative analysis with established methodologies, highlighting key aspects of mammalian display technology, including genetic engineering, construction of display libraries, and its pivotal role in hit selection and/or developability engineering. The review delves into the mechanisms underpinning developability-driven selection via mammalian display and their broader implications. Applications beyond antibody discovery are also explored, alongside advancements towards function-first screening technologies, precision genome engineering and AI/ML-enhanced libraries, situating them in the context of mammalian display. Overall, the review provides a comprehensive overview of the current mammalian display technology landscape, underscores the expansive potential of the technology for biotherapeutic development, addresses the critical challenges for the full realisation of this potential, and examines advances in related disciplines that might impact the future application of mammalian display technologies.
Collapse
Affiliation(s)
- Peter Slavny
- Discovery & Engineering Division, Iontas Ltd./FairJourney Biologics, Cambridge, United Kingdom
| | - Manjunath Hegde
- Technology Division, Iontas/FairJourney Biologics, Cambridge, United Kingdom
| | - Achim Doerner
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Kothai Parthiban
- Discovery & Engineering Division, Iontas Ltd./FairJourney Biologics, Cambridge, United Kingdom
| | - John McCafferty
- Maxion Therapeutics, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stefan Zielonka
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Rene Hoet
- Technology Division, Iontas/FairJourney Biologics, Cambridge, United Kingdom
- Technology Division, FairJourney Biologics, Porto, Portugal
| |
Collapse
|
24
|
Tulika T, Ruso-Julve F, Ahmadi S, Ljungars A, Rivera-de-Torre E, Wade J, Fernández-Quintero ML, Jenkins TP, Belfakir SB, Ross GMS, Boyens-Thiele L, Buell AK, Sakya SA, Sørensen CV, Bohn MF, Ledsgaard L, Voldborg BG, Francavilla C, Schlothauer T, Lomonte B, Andersen JT, Laustsen AH. Engineering of pH-dependent antigen binding properties for toxin-targeting IgG1 antibodies using light-chain shuffling. Structure 2024; 32:1404-1418.e7. [PMID: 39146931 PMCID: PMC11385703 DOI: 10.1016/j.str.2024.07.014] [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: 01/26/2024] [Revised: 06/07/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Immunoglobulin G (IgG) antibodies that bind their cognate antigen in a pH-dependent manner (acid-switched antibodies) can release their bound antigen for degradation in the acidic environment of endosomes, while the IgGs are rescued by the neonatal Fc receptor (FcRn). Thus, such IgGs can neutralize multiple antigens over time and therefore be used at lower doses than their non-pH-responsive counterparts. Here, we show that light-chain shuffling combined with phage display technology can be used to discover IgG1 antibodies with increased pH-dependent antigen binding properties, using the snake venom toxins, myotoxin II and α-cobratoxin, as examples. We reveal differences in how the selected IgG1s engage their antigens and human FcRn and show how these differences translate into distinct cellular handling properties related to their pH-dependent antigen binding phenotypes and Fc-engineering for improved FcRn binding. Our study showcases the complexity of engineering pH-dependent antigen binding IgG1s and demonstrates the effects on cellular antibody-antigen recycling.
Collapse
Affiliation(s)
- Tulika Tulika
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Fulgencio Ruso-Julve
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Shirin Ahmadi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Jack Wade
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Timothy P Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Selma B Belfakir
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; VenomAid Diagnostics ApS, Lyngby, Denmark
| | | | - Lars Boyens-Thiele
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Alexander K Buell
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Siri A Sakya
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Christoffer V Sørensen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Markus-Frederik Bohn
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Line Ledsgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Bjørn G Voldborg
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Chiara Francavilla
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiologia, Universidad de Costa Rica, San Jose, Costa Rica
| | - Jan Terje Andersen
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway.
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
25
|
Roth S, Ferrante T, Walt DR. Efficient discovery of antibody binding pairs using a photobleaching strategy for bead encoding. LAB ON A CHIP 2024; 24:4060-4072. [PMID: 39081159 DOI: 10.1039/d4lc00382a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Dye-encoded bead-based assays are widely used for diagnostics. Multiple bead populations are required for multiplexing and can be produced using different dye colors, labeling levels, or combinations of dye ratios. Ready-to-use multiplex bead populations restrict users to specific targets, are costly, or require specialized instrumentation. In-house methods produce few bead plexes or require many fine-tuning steps. To expand bead encoding strategies, we present a simple, safe, and cost-effective bench-top system for generating bead populations using photobleaching. By photobleaching commercially available dye-encoded magnetic beads for different durations, we produce three times as many differentiable bead populations on flow cytometry from a single dye color. Our photobleaching system uses a high-power LED module connected to a light concentrator and a heat sink. The beads are photobleached in solution homogeneously by constant mixing. We demonstrate this photobleaching method can be utilized for cross-testing antibodies, which is the first step in developing immunoassays. The assay uses multiple photobleached encoded beads conjugated with capture antibodies to test many binding pairs simultaneously. To further expand the number of antibodies that can be tested at once, several antibodies were conjugated to the same bead, forming a pooled assay. Our assay predicts the performance of antibody pairs used in ultrasensitive Simoa assays, narrowing the number of cross-tested pairs that need to be tested by at least two-thirds and, therefore, providing a rapid alternative for an initial antibody pair screening. The photobleaching system can be utilized for other applications, such as multiplexing, and for photobleaching other particles in solution.
Collapse
Affiliation(s)
- Shira Roth
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
- Harvard Medical School, Boston, MA 02115, USA
| | - Tom Ferrante
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - David R Walt
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Abdo AIK, Nordin F, Tye GJ. Selection and evaluation of single domain antibody against p19 subunit of IL-23 by phage display for potential use as an autoinflammatory therapeutic. Int Immunopharmacol 2024; 137:112371. [PMID: 38852516 DOI: 10.1016/j.intimp.2024.112371] [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: 03/07/2024] [Revised: 05/18/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
IL-23 is a double-subunit cytokine that plays an important role in shaping the immune response. IL-23 was found to be associated with several autoinflammatory diseases by generating sustained inflammatory loops that lead to tissue damage. Antibody neutralization of IL-23 was proven to be effective in ameliorating associated diseases. However, antibodies as large proteins have limited tissue penetration and tend to elicit anti-drug antibodies. Additionally, anti-IL-23 antibodies target only one subunit of IL-23 leaving the other one unneutralized. Here, we attempted to isolate a recycling single domain antibody by phage display. One of IL-23 subunits, p19, was expressed in E. coli fused to Gamillus protein to stabilize the α-helix-only p19. To remove Gamillus binders, two biopanning methods were investigated, first, preselection with Gamillus and second, challenge with IL-23 then on the subsequent round challenge with p19-Gam. The isolation of calcium-dependent and pH-dependent recycling binders was performed with EDTA and citrate buffers respectively. Both methods of panning failed to isolate high-affinity and specific p19 recycling binders, while from the second panning method, a high affinity and specific p19 standard binder, namely H11, was successfully isolated. H11 significantly inhibited the gene expression of IL-17 and IL-22 in IL-23-challenged PBMCs indicating H11 specificity and neutralizing ability for IL-23. The new binder due to its small size can overcome antibodies limitations, also, it can be further engineered in the future for antigen clearance such as fusing it to cell penetrating peptides, granting H11 the ability to clear excess IL-23 and enhancing its potential therapeutic effect.
Collapse
Affiliation(s)
- Ahmad Ismail Khaled Abdo
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia.
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia.
| |
Collapse
|
27
|
Wang SJ, Gupta R, Benegal A, Avula R, Huang YY, Vahey MD, Chakrabarty RK, Pappu RV, Singamaneni S, Puthussery JV, King MR. A High-Avidity, Thermostable, and Low-Cost Synthetic Capture for Ultrasensitive Detection and Quantification of Viral Antigens and Aerosols. ACS Sens 2024; 9:3096-3104. [PMID: 38753414 DOI: 10.1021/acssensors.4c00282] [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: 06/29/2024]
Abstract
Lateral flow assays (LFAs) are currently the most popular point-of-care diagnostics, rapidly transforming disease diagnosis from expensive doctor checkups and laboratory-based tests to potential on-the-shelf commodities. Yet, their sensitive element, a monoclonal antibody, is expensive to formulate, and their long-term storage depends on refrigeration technology that cannot be met in resource-limited areas. In this work, LCB1 affibodies (antibody mimetic miniproteins) were conjugated to bovine serum albumin (BSA) to afford a high-avidity synthetic capture (LCB1-BSA) capable of detecting the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein and virus like particles (VLPs). Substituting the monoclonal antibody 2B04 for LCB1-BSA (stable up to 60 °C) significantly improved the thermal stability, shelf life, and affordability of plasmonic-fluor-based LFAs (p-LFAs). Furthermore, this substitution significantly improved the sensitivity of p-LFAs toward the spike protein and VLPs with precise quantitative ability over 2 and 3 orders of magnitude, respectively. LCB1-BSA sensors could detect VLPs at 100-fold lower concentrations, and this improvement, combined with their robust nature, enabled us to develop an aerosol sampling technology to detect aerosolized viral particles. Synthetic captures like LCB1-BSA can increase the ultrasensitivity, availability, sustainability, and long-term accuracy of LFAs while also decreasing their manufacturing costs.
Collapse
Affiliation(s)
- Sean J Wang
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Rohit Gupta
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ananya Benegal
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Rohan Avula
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Yin-Yuan Huang
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Michael D Vahey
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Rajan K Chakrabarty
- Center for Aerosol Science and Engineering, Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Rohit V Pappu
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Joseph V Puthussery
- Center for Aerosol Science and Engineering, Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Matthew R King
- Department of Biomedical Engineering and Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
28
|
Sadraeian M, Maleki R, Moraghebi M, Bahrami A. Phage Display Technology in Biomarker Identification with Emphasis on Non-Cancerous Diseases. Molecules 2024; 29:3002. [PMID: 38998954 PMCID: PMC11243120 DOI: 10.3390/molecules29133002] [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: 03/12/2024] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 07/14/2024] Open
Abstract
In recent years, phage display technology has become vital in clinical research. It helps create antibodies that can specifically bind to complex antigens, which is crucial for identifying biomarkers and improving diagnostics and treatments. However, existing reviews often overlook its importance in areas outside cancer research. This review aims to fill that gap by explaining the basics of phage display and its applications in detecting and treating various non-cancerous diseases. We focus especially on its role in degenerative diseases, inflammatory and autoimmune diseases, and chronic non-communicable diseases, showing how it is changing the way we diagnose and treat illnesses. By highlighting important discoveries and future possibilities, we hope to emphasize the significance of phage display in modern healthcare.
Collapse
Affiliation(s)
- Mohammad Sadraeian
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Reza Maleki
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Mahta Moraghebi
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Abasalt Bahrami
- Department of Chemistry and Biochemistry, Bioengineering, and Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
Ghosh R, Arnheim A, van Zee M, Shang L, Soemardy C, Tang RC, Mellody M, Baghdasarian S, Sanchez Ochoa E, Ye S, Chen S, Williamson C, Karunaratne A, Di Carlo D. Lab on a Particle Technologies. Anal Chem 2024; 96:7817-7839. [PMID: 38650433 PMCID: PMC11112544 DOI: 10.1021/acs.analchem.4c01510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Affiliation(s)
- Rajesh Ghosh
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Alyssa Arnheim
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Mark van Zee
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Lily Shang
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Citradewi Soemardy
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Rui-Chian Tang
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Michael Mellody
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Sevana Baghdasarian
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Edwin Sanchez Ochoa
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Shun Ye
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Siyu Chen
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Cayden Williamson
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Amrith Karunaratne
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Dino Di Carlo
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Jonsson
Comprehensive Cancer Center, University
of California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
NanoSystems Institute, Los Angeles, California 90095, United States
| |
Collapse
|
30
|
Kazemzadeh H, Bagheri M, Sepehri M, Ebrahimi E, Wang H, Haider S, Kheirabadi M, Tohidkia MR. Isolation and Characterization of the Vascular Endothelial Growth Factor Receptor Targeting ScFv Antibody Fragments Derived from Phage Display Technology. ACS OMEGA 2024; 9:21964-21973. [PMID: 38799304 PMCID: PMC11112697 DOI: 10.1021/acsomega.3c10158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024]
Abstract
Angiogenesis, as a tumor hallmark, plays an important role in the growth and development of the tumor vasculature system. There is a huge amount of evidence suggesting that the vascular endothelial growth factor receptor (VEGFR-2)/VEGF-A axis is one of the main contributors to tumor angiogenesis and metastasis. Thus, inhibition of the VEGFR-2 signaling pathway by anti-VEGFR-2 mAb can retard tumor growth. In this study, we employ phage display technology and solution-phase biopanning (SPB) to isolate specific single-chain variable fragments (scFvs) against VEGFR-2 and report on the receptor binding characteristics of the candidate scFvs A semisynthetic phage antibody library to isolate anti-VEGFR-2 scFvs through an SPB performed with decreasing concentrations of the VEGFR-2-His tag and VEGFR-2-biotin. After successful expression and purification, the specificity of the selected scFv clones was further analyzed by enzyme-linked immunosorbent assay (ELISA), flow cytometry, and immunoblotting. The competition assay was undertaken to identify the VEGFR-2 receptor-blocking properties of the scFvs. Furthermore, the molecular binding characteristics of candidate scFvs were extensively studied by peptide-protein docking. Polyclonal ELISA analysis subsequent to four rounds of biopanning showed a significant enrichment of VEGFR-2-specific phage clones by increasing positive signals from the first round toward the fourth round of selection. The individual VEGFR-2-reactive scFv phage clones were identified by monoclonal phage ELISA. The sequence analysis and complementarity-determining region alignment identified the four unique anti-VEGFR-2-scFv clones. The soluble and purified scFvs displayed binding activity against soluble and cell-associated forms of VEGFR-2 protein in the ELISA and flow cytometry assays. Based on the inference from the molecular docking results, scFvs D3, E1, H1, and E9 recognized domains 2 and 3 on the VEGFR-2 protein and displayed competition with VEGF-A for binding to VEGFR-2. The competition assay confirmed that scFvs H1 and D3 can block the VEGFR-2/VEGF-A interaction. In conclusion, we identified novel VEGFR-2-blocking scFvs that perhaps exhibit the potential for angiogenesis inhibition in VEGFR-2-overexpressed tumor cells.
Collapse
Affiliation(s)
- Hamid Kazemzadeh
- Research
Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Mahsima Bagheri
- Research
Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Maryam Sepehri
- Research
Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Elham Ebrahimi
- Basic
Science Department, Faculty of Biology, Hakim Sabzevari University, P.O. Box 96179-76487, Sabzevar 571, Iran
| | - Huan Wang
- School
of Pharmacy, University College London, London WC1N 1AX, U.K.
| | - Shozeb Haider
- School
of Pharmacy, University College London, London WC1N 1AX, U.K.
| | - Mitra Kheirabadi
- Basic
Science Department, Faculty of Biology, Hakim Sabzevari University, P.O. Box 96179-76487, Sabzevar 571, Iran
| | - Mohammad Reza Tohidkia
- Research
Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| |
Collapse
|
31
|
Hampton JT, Liu WR. Diversification of Phage-Displayed Peptide Libraries with Noncanonical Amino Acid Mutagenesis and Chemical Modification. Chem Rev 2024; 124:6051-6077. [PMID: 38686960 PMCID: PMC11082904 DOI: 10.1021/acs.chemrev.4c00004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024]
Abstract
Sitting on the interface between biologics and small molecules, peptides represent an emerging class of therapeutics. Numerous techniques have been developed in the past 30 years to take advantage of biological methods to generate and screen peptide libraries for the identification of therapeutic compounds, with phage display being one of the most accessible techniques. Although traditional phage display can generate billions of peptides simultaneously, it is limited to expression of canonical amino acids. Recently, several groups have successfully undergone efforts to apply genetic code expansion to introduce noncanonical amino acids (ncAAs) with novel reactivities and chemistries into phage-displayed peptide libraries. In addition to biological methods, several different chemical approaches have also been used to install noncanonical motifs into phage libraries. This review focuses on these recent advances that have taken advantage of both biological and chemical means for diversification of phage libraries with ncAAs.
Collapse
Affiliation(s)
- J. Trae Hampton
- Texas
A&M Drug Discovery Center and Department of Chemistry, College
of Arts and Sciences, Texas A&M University, College Station, Texas 77843, United States
| | - Wenshe Ray Liu
- Texas
A&M Drug Discovery Center and Department of Chemistry, College
of Arts and Sciences, Texas A&M University, College Station, Texas 77843, United States
- Institute
of Biosciences and Technology and Department of Translational Medical
Sciences, College of Medicine, Texas A&M
University, Houston, Texas 77030, United States
- Department
of Biochemistry and Biophysics, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas 77843, United States
- Department
of Cell Biology and Genetics, College of Medicine, Texas A&M University, College
Station, Texas 77843, United States
- Department
of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
32
|
Upton C, Healey J, Rothnie AJ, Goddard AD. Insights into membrane interactions and their therapeutic potential. Arch Biochem Biophys 2024; 755:109939. [PMID: 38387829 DOI: 10.1016/j.abb.2024.109939] [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: 11/01/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Recent research into membrane interactions has uncovered a diverse range of therapeutic opportunities through the bioengineering of human and non-human macromolecules. Although the majority of this research is focussed on fundamental developments, emerging studies are showcasing promising new technologies to combat conditions such as cancer, Alzheimer's and inflammatory and immune-based disease, utilising the alteration of bacteriophage, adenovirus, bacterial toxins, type 6 secretion systems, annexins, mitochondrial antiviral signalling proteins and bacterial nano-syringes. To advance the field further, each of these opportunities need to be better understood, and the therapeutic models need to be further optimised. Here, we summarise the knowledge and insights into several membrane interactions and detail their current and potential uses therapeutically.
Collapse
Affiliation(s)
- Calum Upton
- School of Biosciences, Health & Life Science, Aston University, Birmingham, B4 7ET, UK
| | - Joseph Healey
- Nanosyrinx, The Venture Centre, University of Warwick Science Park, Coventry, CV4 7EZ, UK
| | - Alice J Rothnie
- School of Biosciences, Health & Life Science, Aston University, Birmingham, B4 7ET, UK
| | - Alan D Goddard
- School of Biosciences, Health & Life Science, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
33
|
Zheng X, Liu Q, Liang Y, Feng W, Yu H, Tong C, Song B. Advancement in the development of single chain antibodies using phage display technology. PeerJ 2024; 12:e17143. [PMID: 38618563 PMCID: PMC11015834 DOI: 10.7717/peerj.17143] [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: 12/11/2023] [Accepted: 02/29/2024] [Indexed: 04/16/2024] Open
Abstract
Phage display technology has become an important research tool in biological research, fundamentally changing the traditional monoclonal antibody preparation process, and has been widely used in the establishment of antigen-antibody libraries, drug design, vaccine research, pathogen detection, gene therapy, antigenic epitope research, and cellular signal transduction research.The phage display is a powerful platform for technology development. Using phage display technology, single chain fragment variable (scFv) can be screened, replacing the disadvantage of the large size of traditional antibodies. Phage display single chain antibody libraries have significant biological implications. Here we describe the types of antibodies, including chimeric antibodies, bispecific antibodies, and scFvs. In addition, we describe the phage display system, phage display single chain antibody libraries, screening of specific antibodies by phage libraries and the application of phage libraries.
Collapse
Affiliation(s)
- Xiaohui Zheng
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Qi Liu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yimin Liang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Wenzhi Feng
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Honghao Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Chunyu Tong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Bocui Song
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| |
Collapse
|
34
|
Moghadam MT, Mojtahedi A, Salamy S, Shahbazi R, Satarzadeh N, Delavar M, Ashoobi MT. Phage therapy as a glimmer of hope in the fight against the recurrence or emergence of surgical site bacterial infections. Infection 2024; 52:385-402. [PMID: 38308075 DOI: 10.1007/s15010-024-02178-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/05/2024] [Indexed: 02/04/2024]
Abstract
PURPOSE Over the last decade, surgery rates have risen alarmingly, and surgical-site infections are expanding these concerns. In spite of advances in infection control practices, surgical infections continue to be a significant cause of death, prolonged hospitalization, and morbidity. As well as the presence of bacterial infections and their antibiotic resistance, biofilm formation is one of the challenges in the treatment of surgical wounds. METHODS This review article was based on published studies on inpatients and laboratory animals receiving phage therapy for surgical wounds, phage therapy for tissue and bone infections treated with surgery to prevent recurrence, antibiotic-resistant wound infections treated with phage therapy, and biofilm-involved surgical wounds treated with phage therapy which were searched without date restrictions. RESULTS It has been shown in this review article that phage therapy can be used to treat surgical-site infections in patients and animals, eliminate biofilms at the surgical site, prevent infection recurrence in wounds that have been operated on, and eradicate antibiotic-resistant infections in surgical wounds, including multi-drug resistance (MDR), extensively drug resistance (XDR), and pan-drug resistance (PDR). A cocktail of phages and antibiotics can also reduce surgical-site infections more effectively than phages alone. CONCLUSION In light of these encouraging results, clinical trials and research with phages will continue in the near future to treat surgical-site infections, biofilm removal, and antibiotic-resistant wounds, all of which could be used to prescribe phages as an alternative to antibiotics.
Collapse
Affiliation(s)
- Majid Taati Moghadam
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Mojtahedi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shakiba Salamy
- Department of Microbiology, Faculty of Pharmacy, Islamic Azad University, Tehran, Iran
| | - Razieh Shahbazi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Naghmeh Satarzadeh
- Student Research Committee, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Delavar
- Vice President of Health and Executive Vice President, Rey Health Center, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
35
|
Mustafa MI, Mohammed A. Developing recombinant antibodies by phage display technology to neutralize viral infectious diseases. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100140. [PMID: 38182043 DOI: 10.1016/j.slasd.2024.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
The use of recombinant antibodies developed through phage display technology offers a promising approach for combating viral infectious diseases. By specifically targeting antigens on viral surfaces, these antibodies have the potential to reduce the severity of infections or even prevent them altogether. With the emergence of new and more virulent strains of viruses, it is crucial to develop innovative methods to counteract them. Phage display technology has proven successful in generating recombinant antibodies capable of targeting specific viral antigens, thereby providing a powerful tool to fight viral infections. In this mini-review article, we examine the development of these antibodies using phage display technology, and discuss the associated challenges and opportunities in developing novel treatments for viral infectious diseases. Furthermore, we provide an overview of phage display technology. As these methods continue to evolve and improve, novel and sophisticated tools based on phage display and peptide display systems are constantly emerging, offering exciting prospects for solving scientific, medical, and technological problems related to viral infectious diseases in the near future.
Collapse
Affiliation(s)
- Mujahed I Mustafa
- Department of Biotechnology, College of Applied and Industrial Sciences, University of Bahri, Khartoum, Sudan.
| | - Ahmed Mohammed
- Department of Biotechnology, School of Life Sciences and Technology, Omdurman Islamic university, Omdurman, Sudan
| |
Collapse
|
36
|
Aripov VS, Volkova NV, Ilyichev AA, Shcherbakov DN. Problems of creating antibody phage libraries and their solutions. Vavilovskii Zhurnal Genet Selektsii 2024; 28:249-257. [PMID: 38680186 PMCID: PMC11043502 DOI: 10.18699/vjgb-24-29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/18/2024] [Accepted: 01/19/2023] [Indexed: 05/01/2024] Open
Abstract
Phage display has become an efficient, reliable and popular molecular technique for generating libraries encompassing millions or even billions of clones of divergent peptides or proteins. The method is based on the correspondence between phage genotype and phenotype, which ensures the presentation of recombinant proteins of known amino acid composition on the surface of phage particles. The use of affinity selection allows one to choose variants with affinity for different targets from phage libraries. The implementation of the antibody phage display technique has revolutionized the field of clinical immunology, both for developing tools to diagnose infectious diseases and for producing therapeutic agents. It has also become the basis for efficient and relatively inexpensive methods for studying protein-protein interactions, receptor binding sites, as well as epitope and mimotope identification. The antibody phage display technique involves a number of steps, and the final result depends on their successful implementation. The diversity, whether natural or obtained by combinatorial chemistry, is the basis of any library. The choice of molecular techniques is critical to ensure that this diversity is maintained during the phage library preparation step and during the transformation of E. coli cells. After a helper phage is added to the suspension of transformed E. coli cells, a bacteriophage library is formed, which is a working tool for performing the affinity selection procedure and searching for individual molecules. Despite the apparent simplicity of generating phage antibody libraries, a number of subtleties need to be taken into account. First, there are the features of phage vector preparation. Currently, a large number of phagemid vectors have been developed, and their selection is also of great importance. The key step is preparing competent E. coli cells and the technology of their transformation. The choice of a helper phage and the method used to generate it is also important. This article discusses the key challenges faced by researchers in constructing phage antibody libraries.
Collapse
Affiliation(s)
- V S Aripov
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region, Russia
| | - N V Volkova
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region, Russia
| | - A A Ilyichev
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region, Russia
| | - D N Shcherbakov
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region, Russia
| |
Collapse
|
37
|
Park KS, Park TI, Lee JE, Hwang SY, Choi A, Pack SP. Aptamers and Nanobodies as New Bioprobes for SARS-CoV-2 Diagnostic and Therapeutic System Applications. BIOSENSORS 2024; 14:146. [PMID: 38534253 PMCID: PMC10968798 DOI: 10.3390/bios14030146] [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: 01/31/2024] [Revised: 03/10/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024]
Abstract
The global challenges posed by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic have underscored the critical importance of innovative and efficient control systems for addressing future pandemics. The most effective way to control the pandemic is to rapidly suppress the spread of the virus through early detection using a rapid, accurate, and easy-to-use diagnostic platform. In biosensors that use bioprobes, the binding affinity of molecular recognition elements (MREs) is the primary factor determining the dynamic range of the sensing platform. Furthermore, the sensitivity relies mainly on bioprobe quality with sufficient functionality. This comprehensive review investigates aptamers and nanobodies recently developed as advanced MREs for SARS-CoV-2 diagnostic and therapeutic applications. These bioprobes might be integrated into organic bioelectronic materials and devices, with promising enhanced sensitivity and specificity. This review offers valuable insights into advancing biosensing technologies for infectious disease diagnosis and treatment using aptamers and nanobodies as new bioprobes.
Collapse
Affiliation(s)
| | | | | | | | | | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (K.S.P.); (T.-I.P.); (J.E.L.); (S.-Y.H.); (A.C.)
| |
Collapse
|
38
|
Rivera‐de‐Torre E, Lampadariou S, Møiniche M, Bohn MF, Kazemi SM, Laustsen AH. Discovery of broadly-neutralizing antibodies against brown recluse spider and Gadim scorpion sphingomyelinases using consensus toxins as antigens. Protein Sci 2024; 33:e4901. [PMID: 38358130 PMCID: PMC10868436 DOI: 10.1002/pro.4901] [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: 04/19/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 02/16/2024]
Abstract
Broadly-neutralizing monoclonal antibodies are becoming increasingly important tools for treating infectious diseases and animal envenomings. However, designing and developing broadly-neutralizing antibodies can be cumbersome using traditional low-throughput iterative protein engineering methods. Here, we present a new high-throughput approach for the standardized discovery of broadly-neutralizing monoclonal antibodies relying on phage display technology and consensus antigens representing average sequences of related proteins. We showcase the utility of this approach by applying it to toxic sphingomyelinases from the venoms of species from very distant orders of the animal kingdom, the recluse spider and Gadim scorpion. First, we designed a consensus sphingomyelinase and performed three rounds of phage display selection, followed by DELFIA-based screening and ranking, and benchmarked this to a similar campaign involving cross-panning against recombinant versions of the native toxins. Second, we identified two scFvs that not only bind the consensus toxins, but which can also neutralize sphingomyelinase activity of native whole venom in vitro. Finally, we conclude that the phage display campaign involving the use of the consensus toxin was more successful in yielding cross-neutralizing scFvs than the phage display campaign involving cross-panning.
Collapse
Affiliation(s)
| | - Stefanos Lampadariou
- Department of Biotechnology and BiomedicineTechnical University of DenmarkKongens LyngbyDenmark
| | - Mark Møiniche
- Department of Biotechnology and BiomedicineTechnical University of DenmarkKongens LyngbyDenmark
| | - Markus F. Bohn
- Department of Biotechnology and BiomedicineTechnical University of DenmarkKongens LyngbyDenmark
| | | | - Andreas H. Laustsen
- Department of Biotechnology and BiomedicineTechnical University of DenmarkKongens LyngbyDenmark
| |
Collapse
|
39
|
Slagboom J, Lewis AH, Schouten WM, van Haperen R, Veltman M, Bittenbinder MA, Vonk FJ, Casewell NR, Grosveld F, Drabek D, Kool J. High throughput identification of human monoclonal antibodies and heavy-chain-only antibodies to treat snakebite. Toxicon X 2024; 21:100185. [PMID: 38425752 PMCID: PMC10901844 DOI: 10.1016/j.toxcx.2024.100185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/12/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Snakebite envenoming is a priority Neglected Tropical Disease that causes an estimated 81,000-135,000 fatalities each year. The development of a new generation of safer, affordable, and accessible antivenom therapies is urgently needed. With this goal in mind, rigorous characterisation of the specific toxins in snake venom is key to generating novel therapies for snakebite. Monoclonal antibodies directed against venom toxins are emerging as potentially strong candidates in the development of new snakebite diagnostics and treatment. Venoms comprise many different toxins of which several are responsible for their pathological effects. Due to the large variability of venoms within and between species, formulations of combinations of human antibodies are proposed as the next generation antivenoms. Here a high-throughput screening method employing antibody-based ligand fishing of venom toxins in 384 filter-well plate format has been developed to determine the antibody target/s The approach uses Protein G beads for antibody capture followed by exposure to a full venom or purified toxins to bind their respective ligand toxin(s). This is followed by a washing/centrifugation step to remove non-binding toxins and an in-well tryptic digest. Finally, peptides from each well are analysed by nanoLC-MS/MS and subsequent Mascot database searching to identify the bound toxin/s for each antibody under investigation. The approach was successfully validated to rapidly screen antibodies sourced from hybridomas, derived from venom-immunised mice expressing either regular human antibodies or heavy-chain-only human antibodies (HCAbs).
Collapse
Affiliation(s)
- Julien Slagboom
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| | - Abigail H. Lewis
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| | - Wietse M. Schouten
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| | - Rien van Haperen
- Department of Cell Biology and Genetics, Faculty of Medicine, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
- Harbour BioMed, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
| | - Mieke Veltman
- Department of Cell Biology and Genetics, Faculty of Medicine, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
- Harbour BioMed, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
| | - Mátyás A. Bittenbinder
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
- Naturalis Biodiversity Center, 2333 CR, Leiden, the Netherlands
| | - Freek J. Vonk
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
- Naturalis Biodiversity Center, 2333 CR, Leiden, the Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Frank Grosveld
- Department of Cell Biology and Genetics, Faculty of Medicine, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
- Harbour BioMed, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
| | - Dubravka Drabek
- Department of Cell Biology and Genetics, Faculty of Medicine, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
- Harbour BioMed, Erasmus Medical Center Rotterdam, 3000 DR, Rotterdam, the Netherlands
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, the Netherlands
| |
Collapse
|
40
|
Uko SO, Malami I, Ibrahim KG, Lawal N, Bello MB, Abubakar MB, Imam MU. Revolutionizing snakebite care with novel antivenoms: Breakthroughs and barriers. Heliyon 2024; 10:e25531. [PMID: 38333815 PMCID: PMC10850593 DOI: 10.1016/j.heliyon.2024.e25531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024] Open
Abstract
Snakebite envenoming (SBE) is a global public health concern, primarily due to the lack of effective antivenom for treating snakebites inflicted by medically significant venomous snakes prevalent across various geographic locations. The rising demand for safe, cost-effective, and potent snakebite treatments highlights the urgent need to develop alternative therapeutics targeting relevant toxins. This development could provide promising discoveries to create novel recombinant solutions, leveraging human monoclonal antibodies, synthetic peptides and nanobodies. Such technologies as recombinant DNA, peptide and epitope mapping phage display etc) have the potential to exceed the traditional use of equine polyclonal antibodies, which have long been used in antivenom production. Recombinant antivenom can be engineered to target certain toxins that play a critical role in snakebite pathology. This approach has the potential to produce antivenom with improved efficacy and safety profiles. However, there are limitations and challenges associated with these emerging technologies. Therefore, identifying the limitations is critical for overcoming the associated challenges and optimizing the development of recombinant antivenoms. This review is aimed at presenting a thorough overview of diverse technologies used in the development of recombinant antivenom, emphasizing their limitations and offering insights into prospects for advancing recombinant antivenoms.
Collapse
Affiliation(s)
- Samuel Odo Uko
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Biochemistry and Molecular Biology, Faculty of Chemical and Life Sciecnes, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Ibrahim Malami
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Pharmacognosy and Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Kasimu Ghandi Ibrahim
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, P. O. Box 2000, Zarqa, 13110, Jordan
| | - Nafiu Lawal
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Muhammad Bashir Bello
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Usmanu Danfodiyo University Sokoto, Nigeria
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Murtala Bello Abubakar
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Physiology, College of Health Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Physiology, College of Medicine and Health Sciences, Baze University, Abuja, Nigeria
| | - Mustapha Umar Imam
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Medical Biochemistry, College of Health Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
| |
Collapse
|
41
|
Dabiri H, Sadeghizadeh M, Ziaei V, Moghadasi Z, Maham A, Hajizadeh-Saffar E, Habibi-Anbouhi M. Development of an ostrich-derived single-chain variable fragment (scFv) against PTPRN extracellular domain. Sci Rep 2024; 14:3689. [PMID: 38355744 PMCID: PMC10866909 DOI: 10.1038/s41598-024-53386-5] [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: 10/15/2023] [Accepted: 01/31/2024] [Indexed: 02/16/2024] Open
Abstract
In type 1 diabetes, the immune system destroys pancreatic beta cells in an autoimmune condition. To overcome this disease, a specific monoclonal antibody that binds to pancreatic beta cells could be used for targeted immunotherapy. Protein tyrosine phosphatase receptor N (PTPRN) is one of the important surface antigen candidates. Due to its high sequence homology among mammals, so far, no single-chain monoclonal antibody has been produced against this receptor. In this study, we developed a novel single-chain variable fragment (scFv) against the PTPRN extracellular domain. To this aim, ostrich species was used as a host is far phylogenetically birds from mammals to construct a phage display library for the first time. An ostrich-derived scfv phage display library was prepared and biopanning steps were done to enrich and screen for isolating the best anti-PTPRN binders. An scFv with appropriate affinity and specificity to the PTPRN extracellular domain was selected and characterized by ELISA, western blotting, and flow cytometry. The anti-PTPRN scFv developed in this study could be introduced as an effective tool that can pave the way for the creation of antibody-based targeting systems in cooperation with the detection and therapy of type I diabetes.
Collapse
Affiliation(s)
- Hamed Dabiri
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Vahab Ziaei
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Moghadasi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Maham
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | | |
Collapse
|
42
|
Sørensen CV, Hofmann N, Rawat P, Sørensen FV, Ljungars A, Greiff V, Laustsen AH, Jenkins TP. ExpoSeq: simplified analysis of high-throughput sequencing data from antibody discovery campaigns. BIOINFORMATICS ADVANCES 2024; 4:vbae020. [PMID: 38425781 PMCID: PMC10902677 DOI: 10.1093/bioadv/vbae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/08/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
Summary High-throughput sequencing (HTS) offers a modern, fast, and explorative solution to unveil the full potential of display techniques, like antibody phage display, in molecular biology. However, a significant challenge lies in the processing and analysis of such data. Furthermore, there is a notable absence of open-access user-friendly software tools that can be utilized by scientists lacking programming expertise. Here, we present ExpoSeq as an easy-to-use tool to explore, process, and visualize HTS data from antibody discovery campaigns like an expert while only requiring a beginner's knowledge. Availability and implementation The pipeline is distributed via GitHub and PyPI, and it can either be installed as a package with pip or the user can choose to clone the repository.
Collapse
Affiliation(s)
- Christoffer V Sørensen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Nils Hofmann
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Puneet Rawat
- Department of Immunology, University of Oslo and Oslo University Hospital, NO-0316 Oslo, Norway
| | | | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Victor Greiff
- Department of Immunology, University of Oslo and Oslo University Hospital, NO-0316 Oslo, Norway
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Timothy P Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| |
Collapse
|
43
|
Liu Z, Kim D, Kang S, Jung JU. A Detailed Protocol for Constructing a Human Single-Chain Variable Fragment (scFv) Library and Downstream Screening via Phage Display. Methods Protoc 2024; 7:13. [PMID: 38392687 PMCID: PMC10893473 DOI: 10.3390/mps7010013] [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: 12/04/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
The development of monoclonal antibodies (mAbs) represents a significant milestone in both basic research and clinical applications due to their target specificity and versatility in therapeutic and diagnostic applications. The innovative strategy of mAb screening, utilizing phage display, facilitates the in vitro screening of antibodies with high affinity to target antigens. The single-chain variable fragment (scFv) is a subset of mAb derivatives, known for its high binding affinity and smaller size-just one-third of that of human IgG. This report outlines a detailed and comprehensive procedure for constructing a scFv phagemid library derived from human patients, followed by screening via phage display affinity selection. The protocol utilizes 348 primer combinations spanning the entire human antibody repertoire to minimize sequence bias and maintain library diversity during polymerase chain reaction (PCR) for scFv generation, resulting in a library size greater than 1 × 108. Furthermore, we describe a high-throughput phage display screening protocol using enzyme-linked immunosorbent assay (ELISA) to evaluate more than 1200 scFv candidates. The generation of a highly diverse scFv library, coupled with the implementation of a phage display screening methodology, is expected to provide a valuable resource for researchers in pursuit of scFvs with high affinity for target antigens, thus advancing both research and clinical endeavors.
Collapse
Affiliation(s)
- Ziyi Liu
- Cancer Biology Department, Infection Biology Program, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Z.L.); (D.K.); (S.K.)
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dokyun Kim
- Cancer Biology Department, Infection Biology Program, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Z.L.); (D.K.); (S.K.)
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Seokmin Kang
- Cancer Biology Department, Infection Biology Program, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Z.L.); (D.K.); (S.K.)
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jae U. Jung
- Cancer Biology Department, Infection Biology Program, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Z.L.); (D.K.); (S.K.)
- Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
44
|
Minatel VM, Prudencio CR, Barraviera B, Ferreira RS. Nanobodies: a promising approach to treatment of viral diseases. Front Immunol 2024; 14:1303353. [PMID: 38322011 PMCID: PMC10844482 DOI: 10.3389/fimmu.2023.1303353] [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: 09/29/2023] [Accepted: 12/12/2023] [Indexed: 02/08/2024] Open
Abstract
Since their discovery in the 1990s, heavy chain antibodies have garnered significant interest in the scientific community. These antibodies, found in camelids such as llamas and alpacas, exhibit distinct characteristics from conventional antibodies due to the absence of a light chain in their structure. Furthermore, they possess a single antigen-binding domain known as VHH or Nanobody (Nb). With a small size of approximately 15 kDa, these Nbs demonstrate improved characteristics compared to conventional antibodies, including greater physicochemical stability and enhanced biodistribution, enabling them to bind inaccessible epitopes more effectively. As a result, Nbs have found numerous applications in various medical and veterinary fields, particularly in diagnostics and therapeutics. Advances in biotechnology have made the production of recombinant antibodies feasible and compatible with large-scale manufacturing. Through the construction of immune phage libraries that display VHHs and subsequent selection through biopanning, it has become possible to isolate specific Nbs targeting pharmaceutical targets of interest, such as viruses. This review describes the processes involved in nanobody production, from hyperimmunization to purification, with the aim of their application in the pharmaceutical industry.
Collapse
Affiliation(s)
- Vitória Meneghetti Minatel
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP—Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | | | - Benedito Barraviera
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP—Univ Estadual Paulista), Botucatu, São Paulo, Brazil
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP—Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Rui Seabra Ferreira
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP—Univ Estadual Paulista), Botucatu, São Paulo, Brazil
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP—Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| |
Collapse
|
45
|
Li Y, Yang KD, Kong DC, Li XM, Duan HY, Ye JF. Harnessing filamentous phages for enhanced stroke recovery. Front Immunol 2024; 14:1343788. [PMID: 38299142 PMCID: PMC10829096 DOI: 10.3389/fimmu.2023.1343788] [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: 11/24/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Stroke poses a critical global health challenge, leading to substantial morbidity and mortality. Existing treatments often miss vital timeframes and encounter limitations due to adverse effects, prompting the pursuit of innovative approaches to restore compromised brain function. This review explores the potential of filamentous phages in enhancing stroke recovery. Initially antimicrobial-centric, bacteriophage therapy has evolved into a regenerative solution. We explore the diverse role of filamentous phages in post-stroke neurological restoration, emphasizing their ability to integrate peptides into phage coat proteins, thereby facilitating recovery. Experimental evidence supports their efficacy in alleviating post-stroke complications, immune modulation, and tissue regeneration. However, rigorous clinical validation is essential to address challenges like dosing and administration routes. Additionally, genetic modification enhances their potential as injectable biomaterials for complex brain tissue issues. This review emphasizes innovative strategies and the capacity of filamentous phages to contribute to enhanced stroke recovery, as opposed to serving as standalone treatment, particularly in addressing stroke-induced brain tissue damage.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
- School of Nursing, Jilin University, Changchun, China
| | - Kai-di Yang
- School of Nursing, Jilin University, Changchun, China
| | - De-cai Kong
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiao-meng Li
- School of Nursing, Jilin University, Changchun, China
| | - Hao-yu Duan
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
46
|
Cong Y, Devoogdt N, Lambin P, Dubois LJ, Yaromina A. Promising Diagnostic and Therapeutic Approaches Based on VHHs for Cancer Management. Cancers (Basel) 2024; 16:371. [PMID: 38254860 PMCID: PMC10814765 DOI: 10.3390/cancers16020371] [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: 11/29/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
The discovery of the distinctive structure of heavy chain-only antibodies in species belonging to the Camelidae family has elicited significant interest in their variable antigen binding domain (VHH) and gained attention for various applications, such as cancer diagnosis and treatment. This article presents an overview of the characteristics, advantages, and disadvantages of VHHs as compared to conventional antibodies, and their usage in diverse applications. The singular properties of VHHs are explained, and several strategies that can augment their utility are outlined. The preclinical studies illustrating the diagnostic and therapeutic efficacy of distinct VHHs in diverse formats against solid cancers are summarized, and an overview of the clinical trials assessing VHH-based agents in oncology is provided. These investigations demonstrate the enormous potential of VHHs for medical research and healthcare.
Collapse
Affiliation(s)
- Ying Cong
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, 1090 Brussels, Belgium;
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| |
Collapse
|
47
|
Kumar M, Jalota A, Sahu SK, Haque S. Therapeutic antibodies for the prevention and treatment of cancer. J Biomed Sci 2024; 31:6. [PMID: 38216921 PMCID: PMC10787459 DOI: 10.1186/s12929-024-00996-w] [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: 03/27/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024] Open
Abstract
The developments of antibodies for cancer therapeutics have made remarkable success in recent years. There are multiple factors contributing to the success of the biological molecule including origin of the antibody, isotype, affinity, avidity and mechanism of action. With better understanding of mechanism of cancer progression and immune manipulation, recombinant formats of antibodies are used to develop therapeutic modalities for manipulating the immune cells of patients by targeting specific molecules to control the disease. These molecules have been successful in minimizing the side effects instead caused by small molecules or systemic chemotherapy but because of the developing therapeutic resistance against these antibodies, combination therapy is thought to be the best bet for patient care. Here, in this review, we have discussed different aspects of antibodies in cancer therapy affecting their efficacy and mechanism of resistance with some relevant examples of the most studied molecules approved by the US FDA.
Collapse
Affiliation(s)
- Mukesh Kumar
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Akansha Jalota
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH, USA
| | - Sushil Kumar Sahu
- Department of Zoology, Siksha-Bhavana, Visva-Bharati, Santiniketan, West Bengal, India
| | - Shabirul Haque
- Center of Autoimmune Musculoskeletal and Hematopoietic Disease, Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
48
|
Yang Y, Chen IA. Visualization of Engineered M13 Phages Bound to Bacterial Targets by Transmission Electron Microscopy. Methods Mol Biol 2024; 2793:175-183. [PMID: 38526731 PMCID: PMC11296667 DOI: 10.1007/978-1-0716-3798-2_12] [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] [Indexed: 03/27/2024]
Abstract
The filamentous phage M13 is one of the most well-studied and characterized phages, particularly since it was introduced as a scaffold for phage display, a technique to express and evolve fusion proteins on the M13 phage's coat to study protein or peptide binding interactions. Since phages can be engineered or evolved to specifically bind to a variety of targets, engineered M13 phages have been explored for applications such as drug delivery, biosensing, and cancer therapy, among others. Specifically, with the rising challenge of antimicrobial resistance among bacteria, chimeric M13 phages have been explored both as detection and therapeutic agents due to the flexibility in tuning target specificity. Transmission electron microscopy (TEM) is a powerful tool enabling researchers to directly visualize and characterize binding of phages to bacterial surfaces. However, the filamentous phage structure poses a challenge for this technique, as the phages have similar morphology to bacterial structures such as pili. In order to differentiate between bacterial structures and the filamentous phages, here we describe a protocol to prepare TEM samples of engineered M13 phages bound to bacterial cells, in which the phage virions have been specifically labeled by decoration of the major capsid proteins with gold nanoparticles. This protocol enables clear visualization and unambiguous identification of attached filamentous phages within the context of bacterial cells expressing numerous pili.
Collapse
Affiliation(s)
- Yanxi Yang
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, USA
| | - Irene A Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, USA.
| |
Collapse
|
49
|
Zeppieri M, Musa M. Beyond the Dusty Fog: Local Eye Drop Therapy and Potentially New Treatment Alternatives in Pseudoexfoliative Glaucoma. Curr Med Chem 2024; 31:1608-1619. [PMID: 37855339 DOI: 10.2174/0109298673255220231010073215] [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: 03/27/2023] [Revised: 06/19/2023] [Accepted: 08/27/2023] [Indexed: 10/20/2023]
Abstract
Pseudoexfoliative glaucoma (PEG) is a type of secondary open-angle glaucoma characterized by the accumulation of whitish-gray material on the trabecular meshwork and lens, leading to an increase in intraocular pressure (IOP) and optic nerve damage. Local eye drop therapy is one of the first-line treatments for PEG, which include prostaglandin analogues, beta-blockers, and alpha-adrenergic agonists to lower IOP. New treatments beyond conventional techniques, however, are constantly being developed. One potential treatment proposed for PEG is based on magnetic phage display, which involves using magnetic nanoparticles conjugated to specific peptides or proteins selected using phage display techniques to remove aggregates in the anterior chamber of the eye or inflammatory cells and cytokines that contribute to PEG pathogenesis. Other potential treatments include microRNAs (miRNAs) that are involved in the regulation of gene expression at the post-transcription stages. Gene therapies, nanotechnology, immunotherapy and methods based on stem cells can also be potentially used to target and treat specific tissues and cells responsible for regulating IOP. In addition, photobiomodulation therapy (PBMT), a non-invasive procedure that utilizes low-level laser therapy to improve cellular function and promote tissue repair, can prove an interesting alternative in treating PEG. The aim of our mini-review is to provide a brief overview of these innovative methods that appear to offer potentially promising treatment options for PEG.
Collapse
Affiliation(s)
- Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Mutali Musa
- Department of Optometry, University of Benin, Benin City 300238, Edo State, Nigeria
| |
Collapse
|
50
|
Kumar A, Madni ZK, Chaturvedi S, Salunke DM. Recombinant human scFv antibody fragments against phospholipase A2 from Naja naja and Echis carinatus snake venoms: In vivo neutralization and mechanistic insights. Mol Immunol 2024; 165:55-67. [PMID: 38154407 DOI: 10.1016/j.molimm.2023.12.006] [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: 08/03/2023] [Revised: 11/07/2023] [Accepted: 12/17/2023] [Indexed: 12/30/2023]
Abstract
Snake envenomation results in a range of clinical sequelae, and widely used animal-based conventional antivenoms exhibit several limitations including the adverse immunological effects in human snake bite victims. Therefore, human monoclonal anti-snake venom antibodies or fragments can be an alternate therapy for overcoming the existing limitations. We developed venom-neutralizing humanized scFv antibodies and analyzed biochemical mechanisms associated with the inhibition of toxicity. Tomlinson I and J human scFv antibody libraries were screened against Naja naja and Echis carinatus venoms, and seven unique scFv antibodies were obtained. Further, specific toxins of snake venom interacting with each of these scFvs were identified, and phospholipase A2 (PLA2) was found to be prominently captured by the phage-anchored scFv antibodies. Our study indicated PLA2 to be one of the abundant toxins in Naja naja and Echis carinatus venom samples. The scFvs binding to PLA2 were used to perform in vivo survival assay using the mouse model and in vitro toxin inhibition assays. scFv N194, which binds to acidic PLA2, protected 50% of mice treated with Naja naja venom. Significant prolongation of survival time and 16% survival were observed in Echis carinatus venom-challenged mice treated with scFv E113 and scFv E10, respectively. However, a combination comprised of an equal amount of two scFvs, E113 and E10, both interacting with basic PLA2, exhibited synergistically enhanced survival of 33% in Echis carinatus venom-challenged mice. No such synergistically enhanced survival was observed in the case of combinatorial treatment with anti-Naja naja scFvs, N194, and N248. These scFvs demonstrated partial inhibition of venom-induced myotoxicity, and E113 also inhibited hemolysis by 50%, which corroborates the enhanced survival during combinatorial treatment in Echis carinatus venom-challenged mice.
Collapse
Affiliation(s)
- Amit Kumar
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Zaid Kamal Madni
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Shivam Chaturvedi
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Dinakar M Salunke
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|