1
|
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
|
2
|
Wang WC, Chang J, Lee CH, Chiang YW, Leu SJ, Mao YC, Chiang JR, Yang CK, Wu CJ, Yang YY. Phage display-derived alpaca nanobodies as potential therapeutics for Naja atra snake envenomation. Appl Environ Microbiol 2024; 90:e0012124. [PMID: 38980046 PMCID: PMC11337809 DOI: 10.1128/aem.00121-24] [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: 01/24/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024] Open
Abstract
Naja atra, the Chinese cobra, is a major cause of snake envenomation in Asia, causing hundreds of thousands of clinical incidents annually. The current treatment, horse serum-derived antivenom, has unpredictable side effects and presents manufacturing challenges. This study focused on developing new-generation snake venom antidotes by using microbial phage display technology to derive nanobodies from an alpaca immunized with attenuated N. atra venom. Following confirmation of the immune response in the alpaca, we amplified VHH genes from isolated peripheral blood mononuclear cells and constructed a phage display VHH library of 1.0 × 107 transformants. After four rounds of biopanning, the enriched phages exhibited increased binding activity to N. atra venom. Four nanobody clones with high binding affinities were selected: aNAH1, aNAH6, aNAH7, and aNAH9. Specificity testing against venom from various snake species, including two Southeast Asian cobra species, revealed nanobodies specific to the genus Naja. An in vivo mouse venom neutralization assay demonstrated that all nanobodies prolonged mouse survival and aNAH6 protected 66.6% of the mice from the lethal dosage. These findings highlight the potential of phage display-derived nanobodies as valuable antidotes for N. atra venom, laying the groundwork for future applications in snakebite treatment.IMPORTANCEChinese cobra venom bites present a formidable medical challenge, and current serum treatments face unresolved issues. Our research applied microbial phage display technology to obtain a new, effective, and cost-efficient treatment approach. Despite interest among scientists in utilizing this technology to screen alpaca antibodies against toxins, the available literature is limited. This study makes a significant contribution by introducing neutralizing antibodies that are specifically tailored to Chinese cobra venom. We provide a comprehensive and unbiased account of the antibody construction process, accompanied by thorough testing of various nanobodies and an assessment of cross-reactivity with diverse snake venoms. These nanobodies represent a promising avenue for targeted antivenom development that bridges microbiology and biotechnology to address critical health needs.
Collapse
Affiliation(s)
- Wei-Chu Wang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jungshan Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Hsin Lee
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wei Chiang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
- Foundation for Poison Control, Taipei, Taiwan
| | - Sy-Jye Leu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yan-Chiao Mao
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- School of Medicine, National Defense Medical Centre, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Division of Clinical Toxicology, Department of Emergency Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jen-Ron Chiang
- Bioproduction Plants, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chun-Kai Yang
- Interdisciplinary Research on Ecology and Sustainability, National Dong Hwa University, Hualien, Taiwan
| | - Chao-Jung Wu
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yi-Yuan Yang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
3
|
Pierzynowska K, Morcinek-Orłowska J, Gaffke L, Jaroszewicz W, Skowron PM, Węgrzyn G. Applications of the phage display technology in molecular biology, biotechnology and medicine. Crit Rev Microbiol 2024; 50:450-490. [PMID: 37270791 DOI: 10.1080/1040841x.2023.2219741] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 10/17/2022] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
The phage display technology is based on the presentation of peptide sequences on the surface of virions of bacteriophages. Its development led to creation of sophisticated systems based on the possibility of the presentation of a huge variability of peptides, attached to one of proteins of bacteriophage capsids. The use of such systems allowed for achieving enormous advantages in the processes of selection of bioactive molecules. In fact, the phage display technology has been employed in numerous fields of biotechnology, as diverse as immunological and biomedical applications (in both diagnostics and therapy), the formation of novel materials, and many others. In this paper, contrary to many other review articles which were focussed on either specific display systems or the use of phage display in selected fields, we present a comprehensive overview of various possibilities of applications of this technology. We discuss an usefulness of the phage display technology in various fields of science, medicine and the broad sense of biotechnology. This overview indicates the spread and importance of applications of microbial systems (exemplified by the phage display technology), pointing to the possibility of developing such sophisticated tools when advanced molecular methods are used in microbiological studies, accompanied with understanding of details of structures and functions of microbial entities (bacteriophages in this case).
Collapse
Affiliation(s)
- Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | | | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Weronika Jaroszewicz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdańsk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| |
Collapse
|
4
|
Lee CH, Wu CJ, Yang YY, Wang WC, Leu SJ, Wu CT, Kao PS, Liu KJ, Tsai BY, Chiang YW, Mao YC, Benedict Dlamini N, Chang J. Characterization of chicken-derived antibody against Alpha-Enolase of Streptococcus pneumoniae. Int Immunopharmacol 2024; 128:111476. [PMID: 38185035 DOI: 10.1016/j.intimp.2023.111476] [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] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/09/2024]
Abstract
Streptococcus pneumoniae is a clinically relevant pathogen notorious for causing pneumonia, meningitis, and otitis media in immunocompromised patients. Currently, antibiotic therapy is the most efficient treatment for fighting pneumococcal infections. However, an arise in antimicrobial resistance in S. pneumoniae has become a serious health issue globally. To resolve the problem, alternative and cost-effective strategies, such as monoclonal antibody-based targeted therapy, are needed for combating bacterial infection. S. pneumoniae alpha-enolase (spEno1), which is thought to be a great target, is a surface protein that binds and converts human plasminogen to plasmin, leading to accelerated bacterial infections. We first purified recombinant spEno1 protein for chicken immunization to generate specific IgY antibodies. We next constructed two single-chain variable fragments (scFv) antibody libraries by phage display technology, containing 7.2 × 107 and 4.8 × 107 transformants. After bio-panning, ten scFv antibodies were obtained, and their binding activities to spEno1 were evaluated on ELISA, Western blot and IFA. The epitopes of spEno1 were identified by these scFv antibodies, which binding affinities were determined by competitive ELISA. Moreover, inhibition assay displayed that the scFv antibodies effectively inhibit the binding between spEno1 and human plasminogen. Overall, the results suggested that these scFv antibodies have the potential to serve as an immunotherapeutic drug against S. pneumoniae infections.
Collapse
Affiliation(s)
- Chi-Hsin Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Chao-Jung Wu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Yi-Yuan Yang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan; Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei 110301, Taiwan
| | - Wei-Chu Wang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Sy-Jye Leu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Cheng-Tsang Wu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Pei-Shih Kao
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan
| | - Bor-Yu Tsai
- Navi Bio-Therapeutics Inc., Taipei 10351, Taiwan
| | - Yu-Wei Chiang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yan-Chiao Mao
- Division of Clinical Toxicology, Department of Emergency Medicine, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Nhlanhla Benedict Dlamini
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Jungshan Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan; International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
5
|
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
|
6
|
León E, Ortiz V, Pérez A, Téllez J, Díaz GJ, Ramírez H MH, Contreras R LE. Anti-SpCas9 IgY Polyclonal Antibodies Production for CRISPR Research Use. ACS OMEGA 2023; 8:33809-33818. [PMID: 37744827 PMCID: PMC10515394 DOI: 10.1021/acsomega.3c04273] [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: 06/15/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023]
Abstract
The CRISPR/Cas adaptative immune system has been harnessed as an RNA-guided, programmable genome editing tool, allowing for diverse biotechnological applications. The implementation of the system relies on the ability to detect the Cas9 protein in biological samples. This task is facilitated by employing antibodies, which exhibit several advantageous features and applications in the context of tropical neglected diseases. This study reports a one-month immunization scheme with the Cas9 protein fromStreptococcus pyogenes to produce IgY polyclonal antibodies (anti-SpCas9), which can be rapidly isolated by combining yolk de-lipidation with protein salting out using pectin and ammonium sulfate, respectively. Immunodetection assays indicate that the antibodies are highly sensitive, specific, and useful for detecting the SpCas9 protein in promastigotes ofLeishmania braziliensisexpressing exogenous SpCas9. Thus, the simple method for producing anti-SpCas9 IgY antibodies will accelerate CRISPR/Cas-based studies in Leishmania spp. This approach serves as a valuable research tool in this parasite model and holds the potential for wide application in various other biological samples, promoting the implementation of the system. In fact, a bioinformatics approach based on the identification of antigenic determinants in the SpCas9 protein suggests the possibility of using the anti-SpCas9 IgY antibodies in applications such as Prime and Base editing.
Collapse
Affiliation(s)
- Esteban León
- Facultad
de Ciencias, Universidad Nacional de Colombia, 111311 Bogotá, Colombia
| | - Valentina Ortiz
- Facultad
de Ciencias, Universidad Nacional de Colombia, 111311 Bogotá, Colombia
| | - Alexander Pérez
- Facultad
de Ciencias, Universidad Nacional de Colombia, 111311 Bogotá, Colombia
| | - Jair Téllez
- Escuela
de Pregrado, Dirección Académica, Universidad Nacional de Colombia, 202017 sede La Paz, Colombia
| | - Gonzalo J. Díaz
- Facultad
de Medicina Veterinaria y de Zootecnia, Laboratorio de Toxicología, Universidad Nacional de Colombia, 111311 Bogotá, Colombia
| | - María H. Ramírez H
- Facultad
de Ciencias, Universidad Nacional de Colombia, 111311 Bogotá, Colombia
| | - Luis E. Contreras R
- Facultad
de Ciencias, Universidad Nacional de Colombia, 111311 Bogotá, Colombia
| |
Collapse
|
7
|
Thueng-In K, Theerawatanasirikul S, Meechan P, Lekcharoensuk P, Chaicumpa W. Cell-penetrating porcine single-chain antibodies (transbodies) against nonstructural protein 1β (NSP1β) of porcine reproductive and respiratory syndrome virus inhibit virus replication. Arch Virol 2023; 168:133. [PMID: 37029230 DOI: 10.1007/s00705-023-05760-3] [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: 11/15/2022] [Accepted: 03/09/2023] [Indexed: 04/09/2023]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) causes porcine reproductive and respiratory syndrome (PRRS) worldwide, especially in domestic pigs, with an enormous economic impact, estimated at $664 million in losses every year to the pig industry. Current vaccines confer limited protection, and no direct-acting anti-PRRS treatment is available. Non-structural protein (NSP) 1β, a cysteine-like protease (CLPro) of PRRSV plays an essential role in viral polyprotein processing, subgenomic RNA synthesis, and evasion of host innate immunity. Therefore, agents that interfere with the bioactivity of NSP1β would be expected to inhibit virus replication. In this study, a porcine single-chain antibody (scFv)-phage display library was constructed and used as a tool for production of NSP1β-specific porcine scFvs (pscFvs). The pscFvs to NSP1β were linked to a cell-penetrating peptide to form cell-penetrating pscFvs (transbodies), which could be internalized and inhibit PRRSV replication in infected cells. A computer simulation indicated that the effective pscFvs used several residues in multiple complementarity determining regions (CDRs) to interact with multiple residues in the CLPro and C-terminal motifs, which might explain the mechanism of pscFv-mediated inhibition of virus replication. Although experiments are needed to determine the antiviral mechanism of the transbodies, the current data indicate that transbodies can potentially be applied for treatment and prevention of PRRSV infection.
Collapse
Affiliation(s)
- K Thueng-In
- School of Pathology, Translational Medicine Program, Institute of Medicine, Suranaree University of Technology, Nakhonratchasima, Thailand.
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand.
| | - S Theerawatanasirikul
- Department of Anatomy, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - P Meechan
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Newcastle NSW, Australia
| | - P Lekcharoensuk
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - W Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Lee LP, Tan CH, Khomvilai S, Sitprija V, Chaiyabutr N, Tan KY. Characterizing and applying immunoglobulins in snakebite diagnostics: A simple and rapid venom detection assay for four medically important snake species in Southeast Asia. Int J Biol Macromol 2023; 236:123727. [PMID: 36863668 DOI: 10.1016/j.ijbiomac.2023.123727] [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: 11/07/2022] [Revised: 01/28/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Snakebite envenoming is a medical emergency requiring urgent and specific treatment. Unfortunately, snakebite diagnostics are scarce, time-consuming and lacking specificity. Hence, this study aimed to develop a simple, quick and specific snakebite diagnostic assay using animal antibodies. Anti-venom horse immunoglobulin G (IgG) and chicken immunoglobulin Y (IgY) were produced against the venoms of four major medically important snake species in Southeast Asia, i.e., the Monocled Cobra (Naja kaouthia), Malayan Krait (Bungarus candidus), Malayan Pit Viper (Calloselasma rhodostoma), and White-lipped Green Pit Viper (Trimeresurus albolabris). Different capture:detection configurations of double-antibody sandwich enzyme-linked immunosorbent assay (ELISA) were constructed using both immunoglobulins, and the horse IgG:IgG-HRP configuration was found to be most selective and sensitive in detecting the corresponding venoms. The method was further streamlined to develop a rapid immunodetection assay, which is able to produce a visual color change within 30 min for discrimination between different snake species. The study shows it is feasible to develop a simple, quick and specific immunodiagnostic assay using horse IgG, which can be derived directly from antisera prepared for antivenom production. The proof-of-concept indicates it is a sustainable and affordable approach in keeping with on-going antivenom manufacturing activities for specific species in the region.
Collapse
Affiliation(s)
- Louisa Pernee Lee
- Venom Research and Toxicology Laboratory, Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Choo Hock Tan
- Venom Research and Toxicology Laboratory, Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Sumana Khomvilai
- Queen Saovabha Memorial Institute, Thai Red Cross Society, 10330 Bangkok, Thailand
| | - Visith Sitprija
- Queen Saovabha Memorial Institute, Thai Red Cross Society, 10330 Bangkok, Thailand
| | - Narongsak Chaiyabutr
- Queen Saovabha Memorial Institute, Thai Red Cross Society, 10330 Bangkok, Thailand
| | - Kae Yi Tan
- Protein and Interactomics Laboratory, Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
9
|
Lee CH, Huang PN, Mwale PF, Wang WC, Leu SJ, Tseng SN, Shih SR, Chiang LC, Mao YC, Tsai BY, Dlamini NB, Nguyen TC, Tsai CH, Yang YY. The Bottlenecks of Preparing Virus Particles by Size Exclusion for Antibody Generation. Int J Mol Sci 2022; 23:12967. [PMID: 36361757 PMCID: PMC9653933 DOI: 10.3390/ijms232112967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
Enterovirus 71 (EV71) is the major etiological agent contributing to the development of hand-foot-mouth disease (HFMD). There are not any global available vaccines or antibody drugs against EV71 released yet. In this study, we perform the virus immunization in a cost-effective and convenient approach by preparing virus particles from size exclusion and immunization of chicken. Polyclonal yolk-immunoglobulin (IgY) was simply purified from egg yolk and monoclonal single-chain variable fragments (scFv) were selected via phage display technology with two scFv libraries containing 6.0 × 106 and 1.3 × 107 transformants. Specific clones were enriched after 5 rounds of bio-panning and four identical genes were classified after the sequence analysis. Moreover, the higher mutation rates were revealed in the CDR regions, especially in the CDR3. IgY showed specific binding activities to both EV71-infected and Coxsackievirus 16-infected cell lysates and high infectivity inhibitory activity of EV71. However, while IgY detected a 37 kDa protein, the selected scFv seemingly detected higher size proteins which could be cell protein instead of EV71 proteins. Despite the highly effective chicken antibody generation, the purity of virus particles prepared by size exclusion is the limitation of this study, and further characterization should be carried out rigorously.
Collapse
Affiliation(s)
- Chi-Hsin Lee
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Peng-Nien Huang
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 333423, Taiwan
- Division of Infectious Diseases, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Pharaoh Fellow Mwale
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Wei-Chu Wang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Sy-Jye Leu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Sung-Nien Tseng
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 333423, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 333423, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Liao-Chun Chiang
- College of Life Sciences, National Tsing Hua University, Hsinchu 300040, Taiwan
| | - Yan-Chiao Mao
- Division of Clinical Toxicology, Department of Emergency Medicine, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Bor-Yu Tsai
- Navi Bio-Therapeutics Inc., Taipei 10351, Taiwan
| | - Nhlanhla Benedict Dlamini
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Tien-Cuong Nguyen
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Chen-Hsin Tsai
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei 110301, Taiwan
| | - Yi-Yuan Yang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei 110301, Taiwan
| |
Collapse
|
10
|
Puzari U, Fernandes PA, Mukherjee AK. Advances in the Therapeutic Application of Small-Molecule Inhibitors and Repurposed Drugs against Snakebite. J Med Chem 2021; 64:13938-13979. [PMID: 34565143 DOI: 10.1021/acs.jmedchem.1c00266] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The World Health Organization has declared snakebite as a neglected tropical disease. Antivenom administration is the sole therapy against venomous snakebite; however, several limitations of this therapy reinforce the dire need for an alternative and/or additional treatment against envenomation. Inhibitors against snake venoms have been explored from natural resources and are synthesized in the laboratory; however, repurposing of small-molecule therapeutics (SMTs) against the principal toxins of snake venoms to inhibit their lethality and/or obnoxious effect of envenomation has been garnering greater attention owing to their established pharmacokinetic properties, low-risk attributes, cost-effectiveness, ease of administration, and storage stability. Nevertheless, SMTs are yet to be approved and commercialized for snakebite treatment. Therefore, we have systematically reviewed and critically analyzed the scenario of small synthetic inhibitors and repurposed drugs against snake envenomation from 2005 to date and proposed novel approaches and commercialization strategies for the development of efficacious therapies against snake envenomation.
Collapse
Affiliation(s)
- Upasana Puzari
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur-784028, Assam, India
| | - Pedro Alexandrino Fernandes
- LAQV@REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua Do Campo Alegre S/N, 4169-007 Porto, Portugal
| | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur-784028, Assam, India.,Institute of Advanced Study in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati-781035, Assam, India
| |
Collapse
|
11
|
Choraria A, Somasundaram R, Janani S, Rajendran S, Oukkache N, Michael A. Chicken egg yolk antibodies (IgY)-based antivenom for neutralization of snake venoms: a review. TOXIN REV 2021. [DOI: 10.1080/15569543.2021.1942063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Ankit Choraria
- Department of Microbiology, PSG College of Arts and Science, Coimbatore, India
| | | | - S. Janani
- Nanobiotechnology Laboratory, PSG Institute of Advanced Studies, Coimbatore, India
| | - Selvakumar Rajendran
- Nanobiotechnology Laboratory, PSG Institute of Advanced Studies, Coimbatore, India
| | - Naoual Oukkache
- Venoms and Toxins Laboratory, Institute Pasteur of Morocco, Casablanca, Morocco
| | - A. Michael
- Department of Microbiology, PSG College of Arts and Science, Coimbatore, India
| |
Collapse
|
12
|
Brilhante-da-Silva N, de Oliveira Sousa RM, Arruda A, Dos Santos EL, Marinho ACM, Stabeli RG, Fernandes CFC, Pereira SDS. Camelid Single-Domain Antibodies for the Development of Potent Diagnosis Platforms. Mol Diagn Ther 2021; 25:439-456. [PMID: 34146333 DOI: 10.1007/s40291-021-00533-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 11/26/2022]
Abstract
The distinct biophysical and pharmaceutical properties of camelid single-domain antibodies, referred to as VHHs or nanobodies, are associated with their nanometric dimensions, elevated stability, and antigen recognition capacity. These biomolecules can circumvent a number of diagnostic system limitations, especially those related to the size and stability of conventional immunoglobulins currently used in enzyme-linked immunosorbent assays and point-of-care, electrochemical, and imaging assays. In these formats, VHHs are directionally conjugated to different molecules, such as metallic nanoparticles, small peptides, and radioisotopes, which demonstrates their comprehensive versatility. Thus, the application of VHHs in diagnostic systems range from the identification of cancer cells to the detection of degenerative disease biomarkers, viral antigens, bacterial toxins, and insecticides. The improvements of sensitivity and specificity are among the central benefits resulting from the use of VHHs, which are indispensable parameters for high-quality diagnostics. Therefore, this review highlights the main biotechnological advances related to camelid single-domain antibodies and their use in in vitro and in vivo diagnostic approaches for human health.
Collapse
Affiliation(s)
- Nairo Brilhante-da-Silva
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, Brazil
| | - Rosa Maria de Oliveira Sousa
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil
| | - Andrelisse Arruda
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil
| | - Eliza Lima Dos Santos
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil
| | - Anna Carolina Machado Marinho
- Plataforma de Desenvolvimento de Anticorpos e Nanocorpos, Fundação Oswaldo Cruz, Fiocruz Ceará, Eusebio, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Rodrigo Guerino Stabeli
- Plataforma Bi-institucional de Medicina Translacional.Fundação Oswaldo Cruz-USP, Ribeirão Preto, São Paulo, Brazil
| | - Carla Freire Celedonio Fernandes
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, Brazil
- Plataforma de Desenvolvimento de Anticorpos e Nanocorpos, Fundação Oswaldo Cruz, Fiocruz Ceará, Eusebio, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Soraya Dos Santos Pereira
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil.
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, Brazil.
- Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, Brazil.
| |
Collapse
|
13
|
Lee CH, Liu CI, Leu SJ, Lee YC, Chiang JR, Chiang LC, Mao YC, Tsai BY, Hung CS, Chen CC, Yang YY. Chicken antibodies against venom proteins of Trimeresurus stejnegeri in Taiwan. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20200056. [PMID: 33281887 PMCID: PMC7682652 DOI: 10.1590/1678-9199-jvatitd-2020-0056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/06/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The venom of bamboo vipers (Trimeresurus stejnegeri - TS), commonly found in Taiwan, contains deadly hemotoxins that cause severe envenomation. Equine-derived antivenom is a specific treatment against snakebites, but its production costs are high and there are some inevitable side effects. The aim of the present work is to help in the development of an affordable and more endurable therapeutic strategy for snakebites. METHODS T. stejnegeri venom proteins were inactivated by glutaraldehyde in order to immunize hens for polyclonal immunoglobulin (IgY) antibodies production. After IgY binding assays, two antibody libraries were constructed expressing single-chain variable fragment (scFv) antibodies joined by the short or long linker for use in phage display antibody technology. Four rounds of biopanning were carried out. The selected scFv antibodies were then further tested for their binding activities and neutralization assays to TS proteins. RESULTS Purified IgY from egg yolk showed the specific binding ability to TS proteins. The dimensions of these two libraries contain 2.4 × 107 and 6.8 × 107 antibody clones, respectively. An increase in the titers of eluted phage indicated anti-TS clones remarkably enriched after 2nd panning. The analysis based on the nucleotide sequences of selected scFv clones indicated that seven groups of short linkers and four groups of long linkers were identified. The recombinant scFvs showed significant reactivity to TS venom proteins and a cross-reaction to Trimeresurus mucrosquamatus venom proteins. In in vivo studies, the data demonstrated that anti-TS IgY provided 100% protective effects while combined scFvs augmented partial survival time of mice injected with a lethal amount of TS proteins. CONCLUSION Chickens were excellent hosts for the production of neutralization antibodies at low cost. Phage display technology is available for generation of monoclonal antibodies against snake venom proteins. These antibodies could be applied in the development of diagnostic kits or as an alternative for snakebite envenomation treatment in the near future.
Collapse
Affiliation(s)
- Chi-Hsin Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-I Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Sy-Jye Leu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ching Lee
- The Center of Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jen-Ron Chiang
- Center for Research, Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taiwan
| | - Liao-Chun Chiang
- College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Yan-Chiao Mao
- Division of Clinical Toxicology, Department of Emergency Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | | | - Ching-Sheng Hung
- Department of Laboratory Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chi-Ching Chen
- Department of Pathology and Laboratory Medicine, Landseed Hospital, Taoyuan, Taiwan
| | - Yi-Yuan Yang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
14
|
Ge S, Xu L, Li B, Zhong F, Liu X, Zhang X. Canine Parvovirus is diagnosed and neutralized by chicken IgY-scFv generated against the virus capsid protein. Vet Res 2020; 51:110. [PMID: 32883344 PMCID: PMC7468180 DOI: 10.1186/s13567-020-00832-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
Canine parvovirus (CPV) can cause acute and highly contagious bloody enteritis in dog. To obtain antibodies against CPV, hens were immunized with virus-like particles (VLP) of CPV-VP2. The IgY single chain fragment variables (scFv) were generated by T7 phage display system and expressed in E. coli system. The titer of the primary scFv library reached to 1.5 × 106 pfu/mL, and 95% of the phages contained the target fragments. The CPV-VLP and CPV-VP2 protein showed similar reaction values to the purified scFv in the ELISA test, and the results of ELISA analysis using IgY-scFv toward CPV clinical samples were consistent with commercial immunochromatographic assay (ICA) and PCR detection, the scFv did not show cross reactivity with canine distemper virus (CDV) and canine coronavirus (CCV). IgY-scFv was successfully expressed in CRFK cells, and in the virus suppression assay, 55% of CPV infections were eliminated within 24 h. Docking results demonstrated that the number of amino acids of the binding sides between scFv and VP2 were AA37 and AA40, respectively. This study revealed the feasibility of a novel functional antibody fragment development strategy by generating diversified avian IgY-scFv libraries towards the pathogenic target of interest for both detection and therapeutic purposes in veterinary medicine.
Collapse
Affiliation(s)
- Shikun Ge
- Chinese-German Joint Laboratory for Natural Product Research, Key Laboratory of Biological Resources and Ecological Environment of Qinba Areas, School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Long Xu
- Chinese-German Joint Laboratory for Natural Product Research, Key Laboratory of Biological Resources and Ecological Environment of Qinba Areas, School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Ben Li
- Chinese-German Joint Laboratory for Natural Product Research, Key Laboratory of Biological Resources and Ecological Environment of Qinba Areas, School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Fagang Zhong
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
| | - Xiang Liu
- Chinese-German Joint Laboratory for Natural Product Research, Key Laboratory of Biological Resources and Ecological Environment of Qinba Areas, School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Xiaoying Zhang
- Chinese-German Joint Laboratory for Natural Product Research, Key Laboratory of Biological Resources and Ecological Environment of Qinba Areas, School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON Canada
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
| |
Collapse
|
15
|
Leu SJ, Lee YC, Lee CH, Liao PY, Chiang CW, Yang CM, Su CH, Ou TY, Liu KJ, Lo HJ, Tsai BY, Yang YY. Generation and Characterization of Single Chain Variable Fragment against Alpha-Enolase of Candida albicans. Int J Mol Sci 2020; 21:ijms21082903. [PMID: 32326294 PMCID: PMC7215377 DOI: 10.3390/ijms21082903] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/17/2020] [Accepted: 04/17/2020] [Indexed: 11/16/2022] Open
Abstract
Candida albicans (C. albicans) is an opportunistic human pathogen responsible for approximately a half of clinical candidemia. The emerging Candida spp. with resistance to azoles is a major challenge in clinic, suggesting an urgent demand for new drugs and therapeutic strategies. Alpha–enolase (Eno1) is a multifunctional protein and represents an important marker for invasive candidiasis. Thus, C. albicans Eno1 (CaEno1) is believed to be an important target for the development of therapeutic agents and antibody drugs. Recombinant CaEno1 (rCaEno1) was first used to immunize chickens. Subsequently, we used phage display technology to construct two single chain variable fragment (scFv) antibody libraries. A novel biopanning procedure was carried out to screen anti-rCaEno1 scFv antibodies, whose specificities were further characterized. The polyclonal IgY antibodies showed binding to rCaEno1 and native CaEno1. A dominant scFv (CaS1) and its properties were further characterized. CaS1 attenuated the growth of C. albicans and inhibited the binding of CaEno1 to plasminogen. Animal studies showed that CaS1 prolonged the survival rate of mice and zebrafish with candidiasis. The fungal burden in kidney and spleen, as well as level of inflammatory cytokines were significantly reduced in CaS1-treated mice. These results suggest CaS1 has potential of being immunotherapeutic drug against C. albicans infections.
Collapse
Affiliation(s)
- Sy-Jye Leu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-J.-L.); (C.-H.S.)
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (P.-Y.L.); (C.-W.C.); (C.-M.Y.)
- Center for Reproductive Medicine and Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Ching Lee
- The Center of Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Chi-Hsin Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-H.L.); (K.-J.L.)
| | - Po-Yen Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (P.-Y.L.); (C.-W.C.); (C.-M.Y.)
| | - Chen-Wei Chiang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (P.-Y.L.); (C.-W.C.); (C.-M.Y.)
| | - Chieh-Ming Yang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (P.-Y.L.); (C.-W.C.); (C.-M.Y.)
| | - Ching-Hua Su
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-J.-L.); (C.-H.S.)
| | - Tsong-Yih Ou
- Division of Infectious Diseases, Department of Internal Medicine, School of Medicine, College of Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan;
| | - Ko-Jiunn Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-H.L.); (K.-J.L.)
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan
| | - Hsiu-Jung Lo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan 70456, Taiwan;
- School of Dentistry, China Medical University, Taichung 40402, Taiwan
| | - Bor-Yu Tsai
- Navi Bio-Therapeutics Inc., Taipei 10351, Taiwan;
| | - Yi-Yuan Yang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-H.L.); (K.-J.L.)
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei 11031, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: ; Tel.: +886-2-27361661 (ext. 3325); Fax: +886-2-27324510
| |
Collapse
|
16
|
Slagboom J, Mladić M, Xie C, Kazandjian TD, Vonk F, Somsen GW, Casewell NR, Kool J. High throughput screening and identification of coagulopathic snake venom proteins and peptides using nanofractionation and proteomics approaches. PLoS Negl Trop Dis 2020; 14:e0007802. [PMID: 32236099 PMCID: PMC7153897 DOI: 10.1371/journal.pntd.0007802] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/13/2020] [Accepted: 03/01/2020] [Indexed: 11/19/2022] Open
Abstract
Snakebite is a neglected tropical disease that results in a variety of systemic and local pathologies in envenomed victims and is responsible for around 138,000 deaths every year. Many snake venoms cause severe coagulopathy that makes victims vulnerable to suffering life-threating haemorrhage. The mechanisms of action of coagulopathic snake venom toxins are diverse and can result in both anticoagulant and procoagulant effects. However, because snake venoms consist of a mixture of numerous protein and peptide components, high throughput characterizations of specific target bioactives is challenging. In this study, we applied a combination of analytical and pharmacological methods to identify snake venom toxins from a wide diversity of snake species that perturb coagulation. To do so, we used a high-throughput screening approach consisting of a miniaturised plasma coagulation assay in combination with a venom nanofractionation approach. Twenty snake venoms were first separated using reversed-phase liquid chromatography, and a post-column split allowed a small fraction to be analyzed with mass spectrometry, while the larger fraction was collected and dispensed onto 384-well plates. After fraction collection, any solvent present in the wells was removed by means of freeze-drying, after which it was possible to perform a plasma coagulation assay in order to detect coagulopathic activity. Our results demonstrate that many snake venoms simultaneously contain both procoagulant and anticoagulant bioactives that contribute to coagulopathy. In-depth identification analysis from seven medically-important venoms, via mass spectrometry and nanoLC-MS/MS, revealed that phospholipase A2 toxins are frequently identified in anticoagulant venom fractions, while serine protease and metalloproteinase toxins are often associated with procoagulant bioactivities. The nanofractionation and proteomics approach applied herein seems likely to be a valuable tool for the rational development of next-generation snakebite treatments by facilitating the rapid identification and fractionation of coagulopathic toxins, thereby enabling specific targeting of these toxins by new therapeutics such as monoclonal antibodies and small molecule inhibitors.
Collapse
Affiliation(s)
- Julien Slagboom
- Division of BioAnalytical Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, Amsterdam, The Netherlands
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Marija Mladić
- Animal Sciences and Health, Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Chunfang Xie
- Division of BioAnalytical Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, Amsterdam, The Netherlands
| | - Taline D. Kazandjian
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Freek Vonk
- Naturalis Biodiversity Center, Leiden, The Netherlands
| | - Govert W. Somsen
- Division of BioAnalytical Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, Amsterdam, The Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jeroen Kool
- Division of BioAnalytical Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Risk factors associated with snake antivenom reaction and the role of skin test. Acta Trop 2020; 203:105293. [PMID: 31821788 DOI: 10.1016/j.actatropica.2019.105293] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/22/2019] [Accepted: 12/06/2019] [Indexed: 11/21/2022]
Abstract
Antivenom reactions are a common complication of snake antivenom. This study aimed to identify predicators of antivenom reaction and the involvement of antivenom skin test in antivenom reaction development. This retrospective cohort study was conducted in six medical institutions in Taiwan. Data were extracted from the Chang Gung Research Database (CGRD) from January 2006 to December 2016. The association between antivenom reaction and patient demographics, type and dose of antivenom, and skin test results was analyzed. The study enrolled 799 patients, including 219 who developed antivenom reactions. Compared to patients receiving both freeze-dried hemorrhagic (FH) and freeze-dried neurotoxic (FN) antivenom, those administered a single type had a lower antivenom reaction risk (adjusted odds ratios [aORs]: 0.5 and 0.4, 95% confidence interval [CI]: 0.35-0.74 and 0.24-0.69, FH and FN respectively). Patients administered a higher antivenom dose (≥ 5 vials) had higher antivenom reaction risk (aOR: 1.8, 95% CI: 1.23-2.76). A positive skin test result was also associated with antivenom reaction (aOR: 16.7, 95% CI: 5.42-51.22). The skin test showed high specificity (98.5%, 95% CI: 97.49%-99.83%) but low sensitivity (17.5%, 95% CI: 10.74%-24.18%). The antivenom skin test should be abolished because of the extremely low sensitivity and possible misinterpretation of results because of the limitation of this examination.
Collapse
|
18
|
Ching KH, Berg K, Morales J, Pedersen D, Harriman WD, Abdiche YN, Leighton PA. Expression of human lambda expands the repertoire of OmniChickens. PLoS One 2020; 15:e0228164. [PMID: 31995598 PMCID: PMC6988971 DOI: 10.1371/journal.pone.0228164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/08/2020] [Indexed: 12/20/2022] Open
Abstract
Most of the approved monoclonal antibodies used in the clinic were initially discovered in mice. However, many targets of therapeutic interest are highly conserved proteins that do not elicit a robust immune response in mice. There is a need for non-mammalian antibody discovery platforms which would allow researchers to access epitopes that are not recognized in mammalian hosts. Recently, we introduced the OmniChicken®, a transgenic animal carrying human VH3-23 and VK3-15 at its immunoglobulin loci. Here, we describe a new version of the OmniChicken which carries VH3-23 and either VL1-44 or VL3-19 at its heavy and light chain loci, respectively. The Vλ-expressing birds showed normal B and T populations in the periphery. A panel of monoclonal antibodies demonstrated comparable epitope coverage of a model antigen compared to both wild-type and Vκ-expressing OmniChickens. Kinetic analysis identified binders in the picomolar range. The Vλ-expressing bird increases the antibody diversity available in the OmniChicken platform, further enabling discovery of therapeutic leads.
Collapse
Affiliation(s)
- Kathryn H. Ching
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | - Kimberley Berg
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | - Jacqueline Morales
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | - Darlene Pedersen
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | - William D. Harriman
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | | | - Philip A. Leighton
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| |
Collapse
|
19
|
Liu CC, Yu JS, Wang PJ, Hsiao YC, Liu CH, Chen YC, Lai PF, Hsu CP, Fann WC, Lin CC. Development of sandwich ELISA and lateral flow strip assays for diagnosing clinically significant snakebite in Taiwan. PLoS Negl Trop Dis 2018; 12:e0007014. [PMID: 30507945 PMCID: PMC6292642 DOI: 10.1371/journal.pntd.0007014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/13/2018] [Accepted: 11/21/2018] [Indexed: 01/01/2023] Open
Abstract
Taiwan is an island located in the south Pacific, a subtropical region that is home to 61 species of snakes. Of these snakes, four species—Trimeresurus stejnegeri, Protobothrops mucrosquamatus, Bungarus multicinctus and Naja atra—account for more than 90% of clinical envenomation cases. Currently, there are two types of bivalent antivenom: hemorrhagic antivenom against the venom of T. stejnegeri and P. mucrosquamatus, and neurotoxic antivenom for treatment of envenomation by B. multicinctus and N. atra. However, no suitable detection kits are available to precisely guide physicians in the use of antivenoms. Here, we sought to develop diagnostic assays for improving the clinical management of snakebite in Taiwan. A two-step affinity purification procedure was used to generate neurotoxic species-specific antibodies (NSS-Abs) and hemorrhagic species-specific antibodies (HSS-Abs) from antivenoms. These two SSAbs were then used to develop a sandwich ELISA (enzyme-linked immunosorbent assay) and a lateral flow assay comprising two test lines. The resulting ELISAs and lateral flow strip assays could successfully discriminate between neurotoxic and hemorrhagic venoms. The limits of quantification (LOQ) of the ELISA for neurotoxic venoms and hemorrhagic venoms were determined to be 0.39 and 0.78 ng/ml, respectively, and the lateral flow strips were capable of detecting neurotoxic and hemorrhagic venoms at concentrations lower than 5 and 50 ng/ml, respectively, in 10–15 min. Tests of lateral flow strips in 21 clinical snakebite cases showed 100% specificity and 100% sensitivity for neurotoxic envenomation, whereas the sensitivity for detecting hemorrhagic envenomation samples was 36.4%. We herein presented a feasible strategy for developing a sensitive sandwich ELISA and lateral flow strip assay for detecting and differentiating venom proteins from hemorrhagic and neurotoxic snakes. A useful snakebite diagnostic guideline according to the lateral flow strip results and clinical symptoms was proposed to help physicians to use antivenoms appropriately. The two-test-line lateral flow strip assay could potentially be applied in an emergency room setting to help physicians diagnose and manage snakebite victims. Snakebite is a public health issue that causes life-threatening medical emergencies. Rapid diagnosis of snakebite in the clinic is a critical necessity in many tropical and subtropical countries, where various venomous snakes are common. Venoms from different snake species contain distinct protein components that require treatment with different antivenoms. However, given the similarity in clinical symptoms among some snake envenomations, it is often challenging for physicians to precisely define the snake species responsible for envenomation. Thus, a reliable method or assay for rapidly diagnosing envenoming species is urgently needed. Here, we present a two-step affinity purification procedure for generating species-specific antibodies (SSAbs) from antivenom, followed by the development of a sandwich ELISA (enzyme-linked immunosorbent assay) and lateral flow strip assay using these SSAbs. This feasible and cost-effective strategy allowed us to develop workable assays for distinguishing between venom proteins from hemorrhagic and neurotoxic snakes in Taiwan. The usefulness of this strategy was demonstrated in the clinic, where both diagnostic assays were shown capable of detecting venoms in blood samples from snakebite patients. Together with the observation of clinical symptoms, the two-test-line lateral flow strip assay is potentially applicable in an emergency room setting to improve snakebite diagnosis and management.
Collapse
Affiliation(s)
- Chien-Chun Liu
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan
| | - Jau-Song Yu
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| | - Po-Jung Wang
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Yung-Chin Hsiao
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
| | - Chien-Hsin Liu
- Center for Research, Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taipei, Taiwan
| | - Yen-Chia Chen
- Department of Emergency medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Emergency Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Fang Lai
- Department of Emergency, Buddihist Tzu Chi Hospital, Hualien, Taiwan
| | - Chih-Po Hsu
- Department of Trauma and Emergency Surgery, Chang Gung Memorial Hospital, Chang Gung University, Tao-Yuan, Taiwan
| | - Wen-Chih Fann
- Department of Emergency Medicine, Chia-Yi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chih-Chuan Lin
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
- * E-mail:
| |
Collapse
|
20
|
Lee CH, Leu SJ, Lee YC, Liu CI, Lin LT, Mwale PF, Chiang JR, Tsai BY, Chen CC, Hung CS, Yang YY. Characterization of Chicken-Derived Single Chain Antibody Fragments against Venom of Naja Naja Atra. Toxins (Basel) 2018; 10:E383. [PMID: 30248928 PMCID: PMC6215181 DOI: 10.3390/toxins10100383] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 12/20/2022] Open
Abstract
Traditional, horse-derived antivenin is currently the most efficient treatment against snake bites. However, it is costly and has unpredictable side effects. Thus, alternative, cost-effective strategies for producing antivenin are needed. In this study, we immunized hens with inactivated NNA venom proteins from the cobra Naja naja atra (NNA). Purified yolk IgY antibodies showed specific anti-NNA binding activity comparable to that of the equine-derived antivenin. We used phage display technology to generate two antibody libraries containing 9.0 × 10⁸ and 8.4 × 10⁸ clones with a short or long linker, respectively. The phage ELISA indicated that anti-NNA clones displaying single-chain variable fragments (scFv) were significantly enriched after biopanning. The nucleotide sequences of the light and heavy chain genes of 30 monoclonal scFv antibodies were determined and classified into six groups with the short linker and nine groups with the long linker. These scFv clones specifically bound to NNA proteins but not to venom proteins from other snakes. Their binding affinities were further determined by competitive ELISA. Animal model studies showed that anti-NNA IgY antibodies exhibited complete protective effects, while a combination of scFv antibodies raised the survival rates and times of mice challenged with lethal doses of NNA venom proteins.
Collapse
Affiliation(s)
- Chi-Hsin Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Sy-Jye Leu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu-Ching Lee
- The Center of Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Chia-I Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Liang-Tzung Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Pharaoh Fellow Mwale
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Jen-Ron Chiang
- Center for Research, Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taipei 11561, Taiwan.
| | - Bor-Yu Tsai
- Navi Bio-Therapeutics Inc., Taipei 10351, Taiwan.
| | - Chi-Ching Chen
- Department of Pathology and Laboratory Medicine, Landseed Hospital, Taoyuan 32449, Taiwan.
| | - Ching-Sheng Hung
- Department of Laboratory Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan.
| | - Yi-Yuan Yang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
21
|
Nanobodies as novel therapeutic agents in envenomation. Biochim Biophys Acta Gen Subj 2018; 1862:2955-2965. [PMID: 30309831 DOI: 10.1016/j.bbagen.2018.08.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND An effective therapy against envenoming should be a priority in view of the high number scorpion stings and snakebites. Serum therapy is still widely applied to treat the envenomation victims; however this approach suffers from several shortcomings. The employment of monoclonal antibodies might be an outcome as these molecules are at the core of a variety of applications from protein structure determination to cancer treatment. The progress of activities in the twilight zone between genetic and antibody engineering have led to the development of a unique class of antibody fragments. These molecules possess several benefits and lack many possible disadvantages over classical antibodies. Within recombinant antibody formats, nanobodies or single domain antigen binding fragments derived from heavy chain only antibodies in camelids occupy a privileged position. SCOPE OF REVIEW In this paper we will briefly review the common methods of envenomation treatment and focus on details of various in vivo research activities that investigate the performance of recombinant, monoclonal nanobodies in venom neutralization. MAJOR CONCLUSIONS Nanobodies bind to their cognate target with high specificity and affinity, they can be produced in large quantities from microbial expression systems and are very robust even when challenged with harsh environmental conditions. Upon administering, they rapidly distribute throughout the body and seem to be well tolerated in humans posing low immunogenicity. GENERAL SIGNIFICANCE Scorpion and snake envenomation is a major issue in developing countries and nanobodies as a venom-neutralizing agent can be considered as a valuable and promising candidate in envenomation therapy.
Collapse
|
22
|
Chen CH, Changou CA, Hsieh TH, Lee YC, Chu CY, Hsu KC, Wang HC, Lin YC, Lo YN, Liu YR, Liou JP, Yen Y. Dual Inhibition of PIK3C3 and FGFR as a New Therapeutic Approach to Treat Bladder Cancer. Clin Cancer Res 2017; 24:1176-1189. [PMID: 29222162 DOI: 10.1158/1078-0432.ccr-17-2066] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/05/2017] [Accepted: 12/04/2017] [Indexed: 11/16/2022]
Abstract
Purpose: MPT0L145 has been developed as a FGFR inhibitor exhibiting significant anti-bladder cancer activity in vitro and in vivo via promoting autophagy-dependent cell death. Here, we aim to elucidate the underlying mechanisms.Experimental Design: Autophagy flux, morphology, and intracellular organelles were evaluated by Western blotting, transmission electron microscope, and fluorescence microscope. Molecular docking and surface plasmon resonance assay were performed to identify drug-protein interaction. Lentiviral delivery of cDNA or shRNA and CRISPR/Cas9-mediated genome editing was used to modulate gene expression. Mitochondrial oxygen consumption rate was measured by a Seahorse XFe24 extracellular flux analyzer, and ROS level was measured by flow cytometry.Results: MPT0L145 persistently increased incomplete autophagy and phase-lucent vacuoles at the perinuclear region, which were identified as enlarged and alkalinized late-endosomes. Screening of a panel of lipid kinases revealed that MPT0L145 strongly inhibits PIK3C3 with a Kd value of 0.53 nmol/L. Ectopic expression of PIK3C3 reversed MPT0L145-increased cell death and incomplete autophagy. Four residues (Y670, F684, I760, D761) at the ATP-binding site of PIK3C3 are important for the binding of MPT0L145. In addition, MPT0L145 promotes mitochondrial dysfunction, ROS production, and DNA damage, which may in part, contribute to cell death. ATG5-knockout rescued MPT0L145-induced cell death, suggesting simultaneous induction of autophagy is crucial to its anticancer activity. Finally, our data demonstrated that MPT0L145 is able to overcome cisplatin resistance in bladder cancer cells.Conclusions: MPT0L145 is a first-in-class PIK3C3/FGFR inhibitor, providing an innovative strategy to design new compounds that increase autophagy, but simultaneously perturb its process to promote bladder cancer cell death. Clin Cancer Res; 24(5); 1176-89. ©2017 AACR.
Collapse
Affiliation(s)
- Chun-Han Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun A Changou
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Translational Medicine, Taipei Medical University, Taipei, Taiwan.,Integrated Laboratory, Center of Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ching Lee
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Ying Chu
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kai-Cheng Hsu
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hao-Ching Wang
- Graduate Institute of Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Lin
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yan-Ni Lo
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yun Yen
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|