1
|
Chen P, Yang Q, Li S, Liu X, Li B, Zhang J, Wang J, Yue X, Wang Y. Nanobody-Induced Aggregation of Gold Nanoparticles: A Mix-and-Read Strategy for the Rapid Detection of Cronobacter sakazakii. Anal Chem 2024; 96:17602-17611. [PMID: 39445484 DOI: 10.1021/acs.analchem.4c03463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Protein-nanoparticle interactions play a crucial role in both biomedical applications and the biosafety assessment of nanomaterials. Here, we found that nanobodies can induce citrate-capped gold nanoparticles (AuNPs) to aggregate into large clusters. Subsequently, we explored the mechanism behind this aggregation and proposed the "gold nucleation mechanism" to explain this phenomenon. Building on this observation, we developed a one-step label-free colorimetric method based on nanobody-induced AuNP aggregation. When nanobodies bind to target bacteria, spatial hindrance occurs, preventing further AuNPs aggregation. This alteration in surface plasmon resonance properties results in visible color changes. As an example, we present a simple and sensitive "mix-and-read" chromogenic immunosensor for Cronobacter sakazakii (C. sakazakii). The experiment can be completed within 20 min, with a visual detection limit of 103 CFU/mL and a quantitative detection limit of 136 CFU/mL. Importantly, our method exhibits no cross-reactivity with other bacterial species. This strategy harnesses the excellent properties of nanobodies and the optical characteristics of AuNPs for direct and rapid detection of foodborne pathogen.
Collapse
Affiliation(s)
- Pengyu Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qinkai Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Shurong Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xuan Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Bingzhi Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jiahao Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jianlong Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiaoyue Yue
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou, Henan Province 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control (Zhengzhou University of Light Industry), Ministry of Education, Zhengzhou 450001, China
| | - Yanru Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
2
|
Yang F, Yang Y, Li X, Aliyari S, Zhu G, Zhu Z, Zheng H, Zhang S. A Nanobody-based TRIM-away targets the intracellular protein degradation of African swine fever virus. Virology 2024; 600:110283. [PMID: 39488886 DOI: 10.1016/j.virol.2024.110283] [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: 08/22/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
African swine fever virus (ASFV) is the causative agent of African swine fever (ASF), a hemorrhagic illness with high fatality rates in domestic pigs that has resulted in a substantial socio-economic loss and threatens the global pork industry. Very few safe and efficient vaccines or compounds against ASF are commercially available, thus developing new antiviral strategies is urgently required. Targeted protein degradation (TPD) has emerged as one of the most innovative strategies for drug discovery. In this study, we generate Nanobody-based TRIM-aways specifically binding with and targeting ASFV-encoded structural proteins p30, p54, and p72 for degradation. Furthermore, nanobody-based trim-aways exhibit robust viral structural protein degradation capabilities in ASFV-infected iPAM and MA104 cells through both proteasomal and lysosomal pathways, concurrently demonstrating potent anti-ASFV activity with less viral production. Our study highlights the Nanobody-based TRIM-away targeting viral protein degradation as a potential candidate for the development of a novel antiviral strategy against ASF.
Collapse
Affiliation(s)
- Fayu Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Yuxi Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Xiaoyun Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Saba Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Guoliang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Zixiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China; African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China.
| | - Shilei Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China.
| |
Collapse
|
3
|
Hutchings CJ, Sato AK. Phage display technology and its impact in the discovery of novel protein-based drugs. Expert Opin Drug Discov 2024; 19:887-915. [PMID: 39074492 DOI: 10.1080/17460441.2024.2367023] [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: 11/28/2023] [Accepted: 06/07/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Phage display technology is a well-established versatile in vitro display technology that has been used for over 35 years to identify peptides and antibodies for use as reagents and therapeutics, as well as exploring the diversity of alternative scaffolds as another option to conventional therapeutic antibody discovery. Such successes have been responsible for spawning a range of biotechnology companies, as well as many complementary technologies devised to expedite the drug discovery process and resolve bottlenecks in the discovery workflow. AREAS COVERED In this perspective, the authors summarize the application of phage display for drug discovery and provide examples of protein-based drugs that have either been approved or are being developed in the clinic. The amenability of phage display to generate functional protein molecules to challenging targets and recent developments of strategies and techniques designed to harness the power of sampling diverse repertoires are highlighted. EXPERT OPINION Phage display is now routinely combined with cutting-edge technologies to deep-mine antibody-based repertoires, peptide, or alternative scaffold libraries generating a wealth of data that can be leveraged, e.g. via artificial intelligence, to enable the potential for clinical success in the discovery and development of protein-based therapeutics.
Collapse
|
4
|
Ding L, Hu J, Liu X, Zeng J, Hu Z, Chen J, Zhu K, Duan H, Huang X. Ultrasensitive dynamic light scattering immunodetection of alpha-fetoprotein using heptamer-amplified nanoparticle crosslinking aggregation. Mikrochim Acta 2024; 191:387. [PMID: 38869719 DOI: 10.1007/s00604-024-06437-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
A novel construction strategy is introduced for an ultrasensitive dynamic light scattering (DLS) immunosensor targeting alpha fetoprotein (AFP). This approach relies on a self-assembled heptamer fusion protein (A1-C4bpα), incorporating the dual functions of multivalent recognition and crosslinking aggregation amplification due to the presence of seven AFP-specific A1 nanobodies on the A1-C4bpα heptamer. Leveraging antibody-functionalized magnetic nanoparticles for target AFP capture and DLS signal output, the proposed heptamer-assisted DLS immunosensor offers high sensitivity, strong specificity, and ease of operation. Under the optimized conditions, the designed DLS immunosensor demonstrates excellent linear detection of AFP in the concentration range 0.06 ng mL-1 to 512 ng mL-1, with a detection limit of 15 pg mL-1. The selectivity, accuracy, precision, practicability, and reliability of this newly developed method were further validated through an assay of AFP levels in spiked and actual human serum samples. This work introduces a novel approach for constructing ultrasensitive DLS immunosensors, easily extendable to the sensitive determination of other targets via simply replacing the nanobody sequence, holding great promise in various applications, particularly in disease diagnosis.
Collapse
Affiliation(s)
- Lu Ding
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Hypertension Research Institute, Nanchang University, Nanchang, 330006, P. R. China
| | - Jiaqi Hu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
- Songzi Center for Inspection and Test, Songzi, 434200, P. R. China
| | - Xing Liu
- Key Laboratory of Tropical and Vegetables Quality and Safety for State Market Regulation, School of Food Science and Engineering, Hainan University, Haikou, 570228, P. R. China
| | - Junyi Zeng
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Hypertension Research Institute, Nanchang University, Nanchang, 330006, P. R. China
| | - Zhiwen Hu
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Hypertension Research Institute, Nanchang University, Nanchang, 330006, P. R. China
| | - Jing Chen
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Kang Zhu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Hong Duan
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University, Beijing, 100048, P. R. China.
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China.
- Jiangxi Medicine Academy of Nutrition and Health Management, Nanchang, 330006, P. R. China.
| |
Collapse
|
5
|
Kuravsky M, Gibbons GF, Joyce C, Scott-Tucker A, Macpherson A, Lawson ADG. Modular design of bi- and multi-specific knob domain fusions. Front Immunol 2024; 15:1384467. [PMID: 38605965 PMCID: PMC11008599 DOI: 10.3389/fimmu.2024.1384467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Introduction The therapeutic potential of bispecific antibodies is becoming widely recognised, with over a hundred formats already described. For many applications, enhanced tissue penetration is sought, so bispecifics with low molecular weight may offer a route to enhanced potency. Here we report the design of bi- and tri-specific antibody-based constructs with molecular weights as low as 14.5 and 22 kDa respectively. Methods Autonomous bovine ultra-long CDR H3 (knob domain peptide) modules have been engineered with artificial coiled-coil stalks derived from Sin Nombre orthohantavirus nucleocapsid protein and human Beclin-1, and joined in series to produce bi- and tri-specific antibody-based constructs with exceptionally low molecular weights. Results Knob domain peptides with coiled-coil stalks retain high, independent antigen binding affinity, exhibit exceptional levels of thermal stability, and can be readily joined head-to-tail yielding the smallest described multi-specific antibody format. The resulting constructs are able to bind simultaneously to all their targets with no interference. Discussion Compared to existing bispecific formats, the reduced molecular weight of the knob domain fusions may enable enhanced tissue penetration and facilitate binding to cryptic epitopes that are inaccessible to conventional antibodies. Furthermore, they can be easily produced at high yield as recombinant products and are free from the heavy-light chain mispairing issue. Taken together, our approach offers an efficient route to modular construction of minimalistic bi- and multi-specifics, thereby further broadening the therapeutic scope for knob domain peptides.
Collapse
|
6
|
Wang J, Kang G, Lu H, de Marco A, Yuan H, Feng Z, Gao M, Wang X, Wang H, Zhang X, Wang Y, Zhang M, Wang P, Feng Y, Liu Z, Cao X, Huang H. Novel bispecific nanobody mitigates experimental intestinal inflammation in mice by targeting TNF-α and IL-23p19 bioactivities. Clin Transl Med 2024; 14:e1636. [PMID: 38533646 PMCID: PMC10966562 DOI: 10.1002/ctm2.1636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/09/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Inflammatory bowel diseases (IBDs) pose significant challenges in terms of treatment non-response, necessitating the development of novel therapeutic approaches. Although biological medicines that target TNF-α (tumour necrosis factor-α) have shown clinical success in some IBD patients, a substantial proportion still fails to respond. METHODS We designed bispecific nanobodies (BsNbs) with the ability to simultaneously target human macrophage-expressed membrane TNF-α (hmTNF-α) and IL-23. Additionally, we fused the constant region of human IgG1 Fc (hIgG1 Fc) to BsNb to create BsNb-Fc. Our study encompassed in vitro and in vivo characterization of BsNb and BsNb-Fc. RESULTS BsNb-Fc exhibited an improved serum half-life, targeting capability and effector function than BsNb. It's demonstrated that BsNb-Fc exhibited superior anti-inflammatory effects compared to the anti-TNF-α mAb (infliximab, IFX) combined with anti-IL-12/IL-23p40 mAb (ustekinumab, UST) by Transwell co-culture assays. Notably, in murine models of acute colitis brought on by 2,4,6-trinitrobenzene sulfonic acid(TNBS) and dextran sulphate sodium (DSS), BsNb-Fc effectively alleviated colitis severity. Additionally, BsNb-Fc outperformed the IFX&UST combination in TNBS-induced colitis, significantly reducing colon inflammation in mice with colitis produced by TNBS and DSS. CONCLUSION These findings highlight an enhanced efficacy and improved biostability of BsNb-Fc, suggesting its potential as a promising therapeutic option for IBD patients with insufficient response to TNF-α inhibition. KEY POINTS A bispecific nanobody (BsNb) was created to target TNF-α and IL-23p19, exhibiting high affinity and remarkable stability. BsNb-Fc inhibited the release of cytokines in CD4+T cells during co-culture experiments. BsNb-Fc effectively alleviated colitis severity in mouse model with acute colitis induced by DSS or TNBS, outperforming the IFX&UST combination.
Collapse
Affiliation(s)
- Jiewen Wang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
| | - Guangbo Kang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
| | - Huiying Lu
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, The Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Ario de Marco
- Laboratory for Environmental and Life SciencesUniversity of Nova GoricaNova GoricaSlovenia
| | - Haibin Yuan
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
| | - Zelin Feng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General HospitalTianjin Medical UniversityTianjinChina
| | - Mengxue Gao
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
| | - Xiaoli Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General HospitalTianjin Medical UniversityTianjinChina
| | - Huahong Wang
- Department of GastroenterologyPeking University First HospitalBeijingChina
| | - Xiaolan Zhang
- Department of GastroenterologyThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yuli Wang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
- Tianjin Pharmaceutical Da Ren Tang Group Corporation Limited, Traditional Chinese Pharmacy Research InstituteTianjin Key Laboratory of Quality Control in Chinese MedicineTianjinChina
- State Key Laboratory of Drug Delivery Technology and PharmacokineticsTianjin Institute of Pharmaceutical ResearchTianjinChina
| | - Miao Zhang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
- China Resources Biopharmaceutical Company LimitedBeijingChina
| | - Ping Wang
- New Technology R&D DepartmentTianjin Modern Innovative TCM Technology Company LimitedTianjinChina
| | - Yuanhang Feng
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
| | - Zhanju Liu
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, The Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General HospitalTianjin Medical UniversityTianjinChina
| | - He Huang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and TechnologyTianjin UniversityTianjinChina
| |
Collapse
|
7
|
Sun M, Wang C, Luo H, Chen Y, Qu G, Chen J, Li L, Zhang M, Xue Q. Development and characterization of a novel nanobody with SRMV neutralizing activity. Microb Cell Fact 2024; 23:45. [PMID: 38341572 PMCID: PMC10858559 DOI: 10.1186/s12934-024-02311-6] [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: 08/27/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Peste des petits ruminants (PPR) is an acute, contact infectious disease caused by the small ruminant morbillivirus (SRMV), and its morbidity in goats and sheep can be up to 100% with significant mortality. Nanobody generated from camelid animals such as alpaca has attracted wide attention because of its unique advantages compared with conventional antibodies. The main objective of this study was to produce specific nanobodies against SRMV and identify its characteristics. To obtain the coding gene of SRMV-specific nanobodies, we first constructed an immune phage-displayed library from the VHH repertoire of alpaca that was immunized with SRMV-F and -H proteins. By using phage display technology, the target antigen-specific VHHs can be obtained after four consecutive rounds of biopanning. Results showed that the size of this VHH library was 2.26 × 1010 CFU/mL and the SRMV-F and -H specific phage particles were greatly enriched after four rounds of biopanning. The positive phage clones were selected and sequenced, and total of five independent different sequences of SRMV-specific nanobodies were identified. Subsequently, the DNA fragments of the five nanobodies were cloned into E. coli BL21(DE3), respectively, and three of them were successfully expressed and purified. Specificity and affinity towards inactivated SRMV of these purified nanobodies were then evaluated using the ELISA method. Results demonstrated that NbSRMV-1-1, NbSRMV-2-10, and NbSRMV-1-21 showed no cross-reactivity with other antigens, such as inactivated BTV, inactivated FMDV, His-tag labeled protein, and BSA. The ELISA titer of these three nanobodies against inactivated SRMV was up to 1:1000. However, only NbSRMV-1-21 displayed SRMV neutralizing activity at a maximum dilution of 1:4. The results indicate that the nanobodies against SRMV generated in this study could be useful in future applications. This study provided a novel antibody tool and laid a foundation for the treatment and detection of SRMV.
Collapse
Affiliation(s)
- Miao Sun
- Department of Viral Biologics, China Institute of Veterinary Drug Control, Beijing, China
| | - Changjiang Wang
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, China
| | - Huaye Luo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Yanfei Chen
- Department of Viral Biologics, China Institute of Veterinary Drug Control, Beijing, China
| | - Guanggang Qu
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, China
| | - Jian Chen
- Department of Viral Biologics, China Institute of Veterinary Drug Control, Beijing, China
| | - Ling Li
- Department of Viral Biologics, China Institute of Veterinary Drug Control, Beijing, China
| | - Min Zhang
- Tech-Bank Food Corporation Limited, Nanjing, China
| | - Qinghong Xue
- Department of Viral Biologics, China Institute of Veterinary Drug Control, Beijing, China.
| |
Collapse
|
8
|
Madsen AV, Pedersen LE, Kristensen P, Goletz S. Design and engineering of bispecific antibodies: insights and practical considerations. Front Bioeng Biotechnol 2024; 12:1352014. [PMID: 38333084 PMCID: PMC10850309 DOI: 10.3389/fbioe.2024.1352014] [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: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Bispecific antibodies (bsAbs) have attracted significant attention due to their dual binding activity, which permits simultaneous targeting of antigens and synergistic binding effects beyond what can be obtained even with combinations of conventional monospecific antibodies. Despite the tremendous therapeutic potential, the design and construction of bsAbs are often hampered by practical issues arising from the increased structural complexity as compared to conventional monospecific antibodies. The issues are diverse in nature, spanning from decreased biophysical stability from fusion of exogenous antigen-binding domains to antibody chain mispairing leading to formation of antibody-related impurities that are very difficult to remove. The added complexity requires judicious design considerations as well as extensive molecular engineering to ensure formation of high quality bsAbs with the intended mode of action and favorable drug-like qualities. In this review, we highlight and summarize some of the key considerations in design of bsAbs as well as state-of-the-art engineering principles that can be applied in efficient construction of bsAbs with diverse molecular formats.
Collapse
Affiliation(s)
- Andreas V. Madsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lasse E. Pedersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Peter Kristensen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
9
|
Dorosky RJ, Schreier JE, Lola SL, Sava RL, Coryell MP, Akue A, KuKuruga M, Carlson PE, Dreher-Lesnick SM, Stibitz S. Nanobodies as potential tools for microbiological testing of live biotherapeutic products. AMB Express 2024; 14:9. [PMID: 38245586 PMCID: PMC10799837 DOI: 10.1186/s13568-023-01659-z] [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: 12/14/2023] [Accepted: 12/23/2023] [Indexed: 01/22/2024] Open
Abstract
Nanobodies are highly specific binding domains derived from naturally occurring single chain camelid antibodies. Live biotherapeutic products (LBPs) are biological products containing preparations of live organisms, such as Lactobacillus, that are intended for use as drugs, i.e. to address a specific disease or condition. Demonstrating potency of multi-strain LBPs can be challenging. The approach investigated here is to use strain-specific nanobody reagents in LBP potency assays. Llamas were immunized with radiation-killed Lactobacillus jensenii or L. crispatus whole cell preparations. A nanobody phage-display library was constructed and panned against bacterial preparations to identify nanobodies specific for each species. Nanobody-encoding DNA sequences were subcloned and the nanobodies were expressed, purified, and characterized. Colony immunoblots and flow cytometry showed that binding by Lj75 and Lj94 nanobodies were limited to a subset of L. jensenii strains while binding by Lc38 and Lc58 nanobodies were limited to L. crispatus strains. Mass spectrometry was used to demonstrate that Lj75 specifically bound a peptidase of L. jensenii, and that Lc58 bound an S-layer protein of L. crispatus. The utility of fluorescent nanobodies in evaluating multi-strain LBP potency assays was assessed by evaluating a L. crispatus and L. jensenii mixture by fluorescence microscopy, flow cytometry, and colony immunoblots. Our results showed that the fluorescent nanobody labelling enabled differentiation and quantitation of the strains in mixture by these methods. Development of these nanobody reagents represents a potential advance in LBP testing, informing the advancement of future LBP potency assays and, thereby, facilitation of clinical investigation of LBPs.
Collapse
Affiliation(s)
- Robert J Dorosky
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| | - Jeremy E Schreier
- Department of Marine Sciences, University of Georgia, Athens, GA, USA
| | - Stephanie L Lola
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Rosa L Sava
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Michael P Coryell
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Adovi Akue
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Mark KuKuruga
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Paul E Carlson
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Sheila M Dreher-Lesnick
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Scott Stibitz
- Office of Vaccines Research and Review, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
10
|
Yanakieva D, Vollmer L, Evers A, Siegmund V, Arras P, Pekar L, Doerner A, Valldorf B, Kolmar H, Zielonka S, Krah S. Cattle-derived knob paratopes grafted onto peripheral loops of the IgG1 Fc region enable the generation of a novel symmetric bispecific antibody format. Front Immunol 2023; 14:1238313. [PMID: 37942319 PMCID: PMC10628450 DOI: 10.3389/fimmu.2023.1238313] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
In this work we present a novel symmetric bispecific antibody format based on engraftments of cattle-derived knob paratopes onto peripheral loops of the IgG1 Fc region. For this, knob architectures obtained from bovine ultralong CDR-H3 antibodies were inserted into the AB loop or EF loop of the CH3 domain, enabling the introduction of an artificial binding specificity into an IgG molecule. We demonstrate that inserted knob domains largely retain their binding affinities, resulting into bispecific antibody derivatives versatile for effector cell redirection. Essentially, generated bispecifics demonstrated adequate biophysical properties and were not compromised in their Fc mediated functionalities such as FcRn or FcγRIIIa binding.
Collapse
Affiliation(s)
- Desislava Yanakieva
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Lena Vollmer
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Andreas Evers
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Vanessa Siegmund
- Early Protein Supply and Characterization, Merck Healthcare KGaA, Darmstadt, Germany
| | - Paul Arras
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Lukas Pekar
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Achim Doerner
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Stefan Zielonka
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Simon Krah
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
11
|
Li Z, Zhang W, Zhang Q, Li P, Tang X. Self-Assembly Multivalent Fluorescence-Nanobody Coupled Multifunctional Nanomaterial with Colorimetric Fluorescence and Photothermal to Enhance Immunochromatographic Assay. ACS NANO 2023; 17:19359-19371. [PMID: 37782130 DOI: 10.1021/acsnano.3c06930] [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: 10/03/2023]
Abstract
The multimodal lateral flow immunoassay (LFIA) has provided accurate and reliable results for fast and immediate detection. Nonetheless, multimodal LFIA remains challenging to develop biosensors with high sensitivity and tolerance to matrix interference in agro-food. In this study, we developed a self-assembled multivalent fluorescence-nanobody (Nb26-EGFP-H6) with 16.5-fold and 30-fold higher affinity and sensitivity than a monovalent nanobody (Nb26). Based on the Nb26-EGFP-H6, we synthesized enhanced immune-probes Zn-CN@Nb26-EGFP-H6 by pyrolyzing and oxidizing an imidazolating zeolite framework-8 (ZIF-8) to obtain photothermal metal-carbon nanomaterials (Zn-CN) for immobilizing Nb26-EGFP-H6. The rough and porous structure of Zn-CN with a large surface area facilitates the enrichment and immobilization of antibodies. A trimodal lateral flow immunoassay (tLFIA) with colorimetric, fluorescent, and photothermal triple signal outputs was constructed for the detection of aflatoxin B1 (AFB1) in maize. Attractively, the Zn-CN-based tLFIA's multiplex guarantees accurate and sensitive detection of AFB1, with triple signal detection limits of 0.0012 ng/mL (colorimetric signals), 0.0094 ng/mL (fluorescent signals), and 0.252 ng/mL (photothermal signals). The sensitivity of the trimode immunosensor was 628-fold and 42-fold higher than that of the original Nb26-based ELISA (IC50) and the unimodal LFIA (LOD). This work provides an idea for constructing a sensitive, tolerant matrix and efficient and accurate analytical platform for rapidly detecting AFB1 in food.
Collapse
Affiliation(s)
- Zhiqiang Li
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs; Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
| | - Wen Zhang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs; Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
| | - Qi Zhang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs; Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Food Safety Research Institute, HuBei University, Wuhan 430062, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Peiwu Li
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs; Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Food Safety Research Institute, HuBei University, Wuhan 430062, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Xianghu Laboratory, Hangzhou, 311231, China
| | - Xiaoqian Tang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs; Laboratory of Risk Assessment for Oilseed Products (Wuhan), Ministry of Agriculture and Rural Affairs; Quality Inspection and Test Center for Oilseed Products, Ministry of Agriculture and Rural Affairs; Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
- Food Safety Research Institute, HuBei University, Wuhan 430062, China
| |
Collapse
|
12
|
Buzas D, Bunzel AH, Staufer O, Milodowski EJ, Edmunds GL, Bufton JC, Vidana Mateo BV, Yadav SKN, Gupta K, Fletcher C, Williamson MK, Harrison A, Borucu U, Capin J, Francis O, Balchin G, Hall S, Vega MV, Durbesson F, Lingappa S, Vincentelli R, Roe J, Wooldridge L, Burt R, Anderson RJL, Mulholland AJ, Bristol UNCOVER Group, Hare J, Bailey M, Davidson AD, Finn A, Morgan D, Mann J, Spatz J, Garzoni F, Schaffitzel C, Berger I. In vitro generated antibodies guide thermostable ADDomer nanoparticle design for nasal vaccination and passive immunization against SARS-CoV-2. Antib Ther 2023; 6:277-297. [PMID: 38075238 PMCID: PMC10702856 DOI: 10.1093/abt/tbad024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 01/10/2024] Open
Abstract
Background Due to COVID-19, pandemic preparedness emerges as a key imperative, necessitating new approaches to accelerate development of reagents against infectious pathogens. Methods Here, we developed an integrated approach combining synthetic, computational and structural methods with in vitro antibody selection and in vivo immunization to design, produce and validate nature-inspired nanoparticle-based reagents against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Results Our approach resulted in two innovations: (i) a thermostable nasal vaccine called ADDoCoV, displaying multiple copies of a SARS-CoV-2 receptor binding motif derived epitope and (ii) a multivalent nanoparticle superbinder, called Gigabody, against SARS-CoV-2 including immune-evasive variants of concern (VOCs). In vitro generated neutralizing nanobodies and electron cryo-microscopy established authenticity and accessibility of epitopes displayed by ADDoCoV. Gigabody comprising multimerized nanobodies prevented SARS-CoV-2 virion attachment with picomolar EC50. Vaccinating mice resulted in antibodies cross-reacting with VOCs including Delta and Omicron. Conclusion Our study elucidates Adenovirus-derived dodecamer (ADDomer)-based nanoparticles for use in active and passive immunization and provides a blueprint for crafting reagents to combat respiratory viral infections.
Collapse
Affiliation(s)
- Dora Buzas
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Adrian H Bunzel
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Oskar Staufer
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- Leibniz Institute for New Materials, Helmholtz Institute for Pharmaceutical Research and Center for Biophysics, Saarland University, Saarbrücken 66123, Germany
| | | | - Grace L Edmunds
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Joshua C Bufton
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | | - Kapil Gupta
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | | | - Maia K Williamson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Ufuk Borucu
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Julien Capin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Ore Francis
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Georgia Balchin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Sophie Hall
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Mirella V Vega
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Fabien Durbesson
- Architecture et Fonction des Macromolécules Biologiques, UMR 7257, CNRS, Aix-Marseille Université, Marseille, France
| | | | - Renaud Vincentelli
- Architecture et Fonction des Macromolécules Biologiques, UMR 7257, CNRS, Aix-Marseille Université, Marseille, France
| | - Joe Roe
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Linda Wooldridge
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Rachel Burt
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | | | | | | | - Jonathan Hare
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Mick Bailey
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Andrew D Davidson
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | - Adam Finn
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
- Children's Vaccine Centre, Bristol Medical School, Bristol BS2 8EF UK
| | - David Morgan
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | - Jamie Mann
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Joachim Spatz
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Frederic Garzoni
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Christiane Schaffitzel
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
| | - Imre Berger
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
| |
Collapse
|
13
|
Harmsen MM, Cornelissen JC, van der Wal FJ, Bergervoet JHW, Koene M. Single-Domain Antibody Multimers for Detection of Botulinum Neurotoxin Serotypes C, D, and Their Mosaics in Endopep-MS. Toxins (Basel) 2023; 15:573. [PMID: 37755999 PMCID: PMC10535107 DOI: 10.3390/toxins15090573] [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: 08/21/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are highly toxic proteins that require high-affinity immunocapture reagents for use in endopeptidase-based assays. Here, 30 novel and 2 earlier published llama single-domain antibodies (VHHs) against the veterinary-relevant BoNT serotypes C and D were yeast-produced. These VHHs recognized 10 independent antigenic sites, and many cross-reacted with the BoNT/DC and CD mosaic variants. As VHHs are highly suitable for genetically linking to increase antigen-binding affinity, 52 VHH multimers were produced and their affinity for BoNT/C, D, DC, and CD was determined. A selection of 15 multimers with high affinity (KD < 0.1 nM) was further shown to be resilient to a high salt wash that is used for samples from complex matrices and bound native BoNTs from culture supernatants as shown by Endopep-MS. High-affinity multimers suitable for further development of a highly sensitive Endopep-MS assay include four multimers that bind both BoNT/D and CD with KD of 14-99 pM, one multimer for BoNT/DC (65 pM) that also binds BoNT/C (75 pM), and seven multimers for BoNT/C (<1-19 pM), six of which also bind BoNT/DC with lower affinity (93-508 pM). In addition to application in diagnostic tests, these VHHs could be used for the development of novel therapeutics for animals or humans.
Collapse
Affiliation(s)
- Michiel M. Harmsen
- Wageningen Bioveterinary Research, Wageningen University & Research, 8221 RA Lelystad, The Netherlands (F.J.v.d.W.)
| | - Jan C. Cornelissen
- Wageningen Bioveterinary Research, Wageningen University & Research, 8221 RA Lelystad, The Netherlands (F.J.v.d.W.)
| | - Fimme J. van der Wal
- Wageningen Bioveterinary Research, Wageningen University & Research, 8221 RA Lelystad, The Netherlands (F.J.v.d.W.)
| | - Jan H. W. Bergervoet
- Wageningen Plant Research, Wageningen University & Research, 6708 PB Wageningen, The Netherlands
| | - Miriam Koene
- Wageningen Bioveterinary Research, Wageningen University & Research, 8221 RA Lelystad, The Netherlands (F.J.v.d.W.)
| |
Collapse
|
14
|
Shim J, Chen J, Carrasco-Triguero M, Fischer SK. Overcoming Soluble Target Interference in Measurement of Total Bispecific Therapeutic Antibody Concentrations. AAPS J 2023; 25:82. [PMID: 37594571 DOI: 10.1208/s12248-023-00848-9] [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: 05/19/2023] [Accepted: 08/04/2023] [Indexed: 08/19/2023] Open
Abstract
The measurement of therapeutic drug concentrations is used to assess drug exposure and the relationship between therapeutic pharmacokinetics (PK) and pharmacodynamics (PD), which help determine the optimal dose for patients. Ligand binding assays (LBAs) are often the method of choice for evaluation of drug concentration and use either the therapeutic target protein or antibodies to the therapeutic as capture and/or detection reagents. Due to the bivalency of antibody therapeutics, heterogeneous states of the drug/target complex can exist in the presence of soluble targets which can complicate measurement of unbound drug. In the case of bispecific antibodies, measurement of drug can be even more complicated and depend upon the levels of both targets to each arm. Measuring the total drug allows for PKPD modeling prediction of human dose projections in addition to overcoming challenges associated with measuring free drug for bispecific antibodies. Here, we present a study in which a sandwich ELISA format was used to measure total anti-KLK5/KLK7 antibody concentrations. This assay utilized a non-blocking anti-idiotype (ID) antibody to one arm of the antibody for capture and an antibody to target bound to the other arm of the antibody for detection. Our qualified assay showed acceptable precision, accuracy, dilutional linearity, and reproducibility and enabled detection of a total bispecific antibody at high levels of two targets. To confirm that our assay was detecting total drug, a subset of samples was evaluated in a generic total LC-MS/MS assay.
Collapse
Affiliation(s)
- Jeongsup Shim
- BioAnalytical Sciences, Development Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Jessica Chen
- BioAnalytical Sciences, Development Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Montserrat Carrasco-Triguero
- BioAnalytical Sciences, Development Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Saloumeh K Fischer
- BioAnalytical Sciences, Development Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| |
Collapse
|
15
|
Hughes AC, Kirkland M, Du W, Rasooly R, Hernlem B, Tam C, Zhang Y, He X. Development of Thermally Stable Nanobodies for Detection and Neutralization of Staphylococcal Enterotoxin B. Toxins (Basel) 2023; 15:400. [PMID: 37368700 DOI: 10.3390/toxins15060400] [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: 05/04/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
In this study, sixteen unique staphylococcal enterotoxin B (SEB)-reactive nanobodies (nbs), including ten monovalent and six bivalent nbs, were developed. All characterized nbs were highly specific for SEB and did not cross-react with other staphylococcal enterotoxins (SE). Several formats of highly sensitive enzyme-linked immunosorbent assays (ELISAs) were established using SEB nbs and a polyclonal antibody (pAb). The lowest limit of detection (LOD) reached 50 pg/mL in PBS. When applied to an ELISA to detect SEB-spiked milk (a commonly contaminated foodstuff), a LOD as low as 190 pg/mL was obtained. The sensitivity of ELISA was found to increase concurrently with the valency of nbs used in the assay. In addition, a wide range of thermal tolerance was observed among the sixteen nbs, with a subset of nbs, SEB-5, SEB-9, and SEB-62, retaining activity even after exposure to 95 °C for 10 min, whereas the conventional monoclonal and polyclonal antibodies exhibited heat-labile properties. Several nbs demonstrated a long shelf-life, with one nb (SEB-9) retaining 93% of its activity after two weeks of storage at room temperature. In addition to their usage in toxin detection, eleven out of fifteen nbs were capable of neutralizing SEB's super-antigenic activity, demonstrated by their inhibition on IL-2 expression in an ex vivo human PBMC assay. Compared to monoclonal and polyclonal antibodies, the nbs are relatively small, thermally stable, and easy to produce, making them useful in applications for sensitive, specific, and cost-effective detection and management of SEB contamination in food products.
Collapse
Affiliation(s)
- Anna C Hughes
- Western Regional Research Center United States Department of Agriculture, Agricultural Research Service, 800 Buchanan St., Albany, CA 94710, USA
| | - Marina Kirkland
- Western Regional Research Center United States Department of Agriculture, Agricultural Research Service, 800 Buchanan St., Albany, CA 94710, USA
| | - Wenxian Du
- Western Regional Research Center United States Department of Agriculture, Agricultural Research Service, 800 Buchanan St., Albany, CA 94710, USA
| | - Reuven Rasooly
- Western Regional Research Center United States Department of Agriculture, Agricultural Research Service, 800 Buchanan St., Albany, CA 94710, USA
| | - Bradley Hernlem
- Western Regional Research Center United States Department of Agriculture, Agricultural Research Service, 800 Buchanan St., Albany, CA 94710, USA
| | - Christina Tam
- Western Regional Research Center United States Department of Agriculture, Agricultural Research Service, 800 Buchanan St., Albany, CA 94710, USA
| | - Yuzhu Zhang
- Western Regional Research Center United States Department of Agriculture, Agricultural Research Service, 800 Buchanan St., Albany, CA 94710, USA
| | - Xiaohua He
- Western Regional Research Center United States Department of Agriculture, Agricultural Research Service, 800 Buchanan St., Albany, CA 94710, USA
| |
Collapse
|
16
|
Yong J, Mellick AS, Whitelock J, Wang J, Liang K. A Biomolecular Toolbox for Precision Nanomotors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2205746. [PMID: 36055646 DOI: 10.1002/adma.202205746] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/31/2022] [Indexed: 06/15/2023]
Abstract
The application of nanomotors for cancer diagnosis and therapy is a new and exciting area of research, which when combined with precision nanomedicine, promises to solve many of the issues encountered by previous development of passive nanoparticles. The goal of this article is to introduce nanomotor and nanomedicine researchers to the deep pool of knowledge available regarding cancer cell biology and biochemistry, as well as provide a greater appreciation of the complexity of cell membrane compositions, extracellular surfaces, and their functional consequences. A short description of the nanomotor state-of-art for cancer therapy and diagnosis is first provided, as well as recommendations for future directions of the field. Then, a biomolecular targeting toolbox has been collated for researchers looking to apply their nanomaterial of choice to a biological setting, as well as providing a glimpse into currently available clinical therapies and technologies. This toolbox contains an overview of different classes of targeting molecules available for high affinity and specific targeting and cell surface targets to aid researchers in the selection of a clinical disease model and targeting methodology. It is hoped that this review will provide biological context, inspiration, and direction to future nanomotor and nanomedicine research.
Collapse
Affiliation(s)
- Joel Yong
- School of Chemical Engineering and Australian Centre for NanoMedicine, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| | - Albert S Mellick
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, 2170, Australia
| | - John Whitelock
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kang Liang
- School of Chemical Engineering and Australian Centre for NanoMedicine, The University of New South Wales, Kensington, New South Wales, 2052, Australia
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| |
Collapse
|
17
|
Kiyose N, Miyazaki N, Furuhata K, Ito Y. Sensitive immunoassay of Legionella using multivalent conjugates of engineered VHHs. J Biochem 2023; 173:185-195. [PMID: 36525357 DOI: 10.1093/jb/mvac102] [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: 03/18/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
VHH antibodies or nanobodies, which are antigen-binding domains of heavy chain antibodies from camelid species, have several advantageous characteristics, including compact molecular size, high productibility in bacteria and easy engineering for functional improvement. Focusing on these advantages of VHHs, we attempted to establish an immunoassay system for detection of Legionella, the causative pathogen of Legionnaires' disease. A VHH phage display library was constructed using cDNA from B cells of alpacas immunized with Legionella pneumophila serogroup1 (LpSG1). Through biopanning, two specific VHH clones were isolated and used to construct a Legionella detection system based on the latex agglutination assay. After engineering the VHHs and improving the assay system, the sensitive detection system was successfully established for the LpSG1 antigen. The immunoassay developed in this study should be useful in easy and sensitive detection of Legionella, the causative agent of Legionnaires' disease, which is a potentially fatal pneumonia.
Collapse
Key Words
- VHH.Abbreviations: Abs, antibodies; BSA, bovine serum albumin; CDR, complementarity determining region; CFU, colony forming unit; DBCO, dibenzylcyclooctyne; ELISA, enzyme-linked immunosorbent assay; FR, framework region; HcAbs, heavy chain antibodies; KLH, keyhole limpet hemocyanin; LpSG1, Legionella pneumophila serogroup1; MALDI-TOFMS, matrix assisted laser desorption/ionization time of flight mass spectrometry; NHS, N-hydroxysuccinimide; PBMC, peripheral blood mononuclear cells; PCR, polymerase chain reaction; RT-PCR, reverse transcription PCR; SDS-PAGE, sodium do-decyl sulphate-polyacrylamide gel electrophoresis; TMB, 3,3′,5,5′-tetramethylbenzidine solution
- alpaca
- antibody
- engineering
- immunoassay
Collapse
Affiliation(s)
- Norihiko Kiyose
- Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima 890-0065, Japan.,ARK Resource Co., Ltd., 383-2, Nakahara-machi, Nishi-ku, Kumamoto 861-5271, Japan
| | - Nobuo Miyazaki
- ARK Resource Co., Ltd., 383-2, Nakahara-machi, Nishi-ku, Kumamoto 861-5271, Japan
| | - Katsunori Furuhata
- School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa 252-5201, Japan
| | - Yuji Ito
- Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima 890-0065, Japan
| |
Collapse
|
18
|
Shukla AK, Misra S. Bispecific antibodies and its applications: a novel approach for targeting SARS-Cov-2. J Basic Clin Physiol Pharmacol 2023; 34:161-168. [PMID: 36607905 DOI: 10.1515/jbcpp-2022-0068] [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: 03/08/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023]
Abstract
The COVID-19 pandemic remains a severe global threat, with the world engulfed in the struggle against the disease's second or third waves, which are approaching frightening proportions in terms of cases and mortality in many nations. Despite the critical need for effective therapy, there is still uncertainty about the optimal practices for treating COVID-19 with various pharmaceutical approaches. This being third year, global immunity and eradication of SARS-CoV-2 is currently seems to be out of reach. Efforts to produce safe and effective vaccinations have shown promise, and progress is being made. Additional therapeutic modalities, as well as vaccine testing in children, are required for prophylaxis and treatment of high-risk individuals. As a result, neutralising antibodies and other comparable therapeutic options offer a lot of promise as immediate and direct antiviral medications. Bispecific antibodies offer a lot of potential in COVID-19 treatment because of their qualities including stability, small size and ease of manufacture. These can be used to control the virus's infection of the lungs because they are available in an inhalational form. To combat the COVID-19 pandemic, innovative approaches with effective nanobodies, high-expression yield and acceptable costs may be required.
Collapse
Affiliation(s)
- Ajay Kumar Shukla
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, India
| | - Saurav Misra
- Department of Pharmacology, Kalpana Chawla Government Medical College, Karnal, India
| |
Collapse
|
19
|
Hambach J, Mann AM, Bannas P, Koch-Nolte F. Targeting multiple myeloma with nanobody-based heavy chain antibodies, bispecific killer cell engagers, chimeric antigen receptors, and nanobody-displaying AAV vectors. Front Immunol 2022; 13:1005800. [PMID: 36405759 PMCID: PMC9668101 DOI: 10.3389/fimmu.2022.1005800] [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: 07/28/2022] [Accepted: 10/21/2022] [Indexed: 11/29/2022] Open
Abstract
Nanobodies are well suited for constructing biologics due to their high solubility. We generated nanobodies directed against CD38, a tumor marker that is overexpressed by multiple myeloma and other hematological malignancies. We then used these CD38-specific nanobodies to construct heavy chain antibodies, bispecific killer cell engagers (BiKEs), chimeric antigen receptor (CAR)-NK cells, and nanobody-displaying AAV vectors. Here we review the utility of these nanobody-based constructs to specifically and effectively target CD38-expressing myeloma cells. The promising results of our preclinical studies warrant further clinical studies to evaluate the potential of these CD38-specific nanobody-based constructs for treatment of multiple myeloma.
Collapse
Affiliation(s)
- Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Marei Mann
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Bannas
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Friedrich Koch-Nolte,
| |
Collapse
|
20
|
Mei Y, Chen Y, Sivaccumar JP, An Z, Xia N, Luo W. Research progress and applications of nanobody in human infectious diseases. Front Pharmacol 2022; 13:963978. [PMID: 36034845 PMCID: PMC9411660 DOI: 10.3389/fphar.2022.963978] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/04/2022] [Indexed: 01/18/2023] Open
Abstract
Infectious diseases, caused by pathogenic microorganisms, are capable of affecting crises. In addition to persistent infectious diseases such as malaria and dengue fever, the vicious outbreaks of infectious diseases such as Neocon, Ebola and SARS-CoV-2 in recent years have prompted the search for more efficient and convenient means for better diagnosis and treatment. Antibodies have attracted a lot of attention due to their good structural characteristics and applications. Nanobodies are the smallest functional single-domain antibodies known to be able to bind stably to antigens, with the advantages of high stability, high hydrophilicity, and easy expression and modification. They can directly target antigen epitopes or be constructed as multivalent nanobodies or nanobody fusion proteins to exert therapeutic effects. This paper focuses on the construction methods and potential functions of nanobodies, outlines the progress of their research, and highlights their various applications in human infectious diseases.
Collapse
Affiliation(s)
- Yaxian Mei
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, School of Life Science, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
| | - Yuanzhi Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, School of Life Science, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
| | - Jwala P. Sivaccumar
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, United States
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, United States
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, School of Life Science, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
| | - Wenxin Luo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, School of Life Science, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- *Correspondence: Wenxin Luo,
| |
Collapse
|
21
|
Chen J, Huang C, Zhao W, Ren J, Ji F, Jia L. SnoopLigase Enables Highly Efficient Generation of C-C-Linked Bispecific Nanobodies Targeting TNF-α and IL-17A. Bioconjug Chem 2022; 33:1446-1455. [PMID: 35938675 DOI: 10.1021/acs.bioconjchem.2c00143] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bispecific antibodies (bis-Nbs) have been extensively developed since the concept was devised over the decades. Taking advantage of the superior characteristics of nanobodies, bis-Nbs exhibit an emerging tendency to become the new generation of research and diagnostic tools. Traditional strategies to connect the homo- or heterogeneous monomers are commonly applied, but there are still technical issues to generate the bispecific molecules as efficiently as designed. Here, we utilize SnoopLigase to directly tether the C terminus (C-C) of the tagged nanobodies against tumor necrosis factor-α (TNF-α) and interleukin-17A (IL-17A). Under optimal conditions, the yield of C-C-linked bis-Nbs can reach as high as 70% due to the existence of SnoopLigase. The prepared bis-Nbs possessed similar or even higher affinity as the monomers and significantly inhibited the proliferation and migration of rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS) induced by TNF-α and IL-17A. This study provides an innovative route for using SnoopLigase to realize a highly efficient generation of C-C-linked bis-Nbs. The approach can be applied to different and multicomponent systems for their potential applications in disease diagnosis and treatment.
Collapse
Affiliation(s)
- Jiewen Chen
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Chundong Huang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Wei Zhao
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Jun Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Fangling Ji
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| |
Collapse
|
22
|
Li L, Liu X, Su B, Zhang H, Li R, Liu Z, Chen Q, Huang T, Cao H. An innovative electrochemical immunosensor based on nanobody heptamer and AuNPs@ZIF-8 nanocomposites as support for the detection of alpha fetoprotein in serum. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
23
|
Fan W, Ji P, Sun X, Kong M, Zhou N, Zhang Q, Wang Y, Liu Q, Li X, Zhou EM, Zhao Q, Sun Y. Screening and identification of nucleocapsid protein-nanobodies that inhibited Newcastle disease virus replication in DF-1 cells. Front Microbiol 2022; 13:956561. [PMID: 36051768 PMCID: PMC9426676 DOI: 10.3389/fmicb.2022.956561] [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: 05/30/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Newcastle disease (ND) is an acute and highly contagious infectious disease found in poultry. Although commercial ND virus (NDV) vaccines are universally used, some case reports persistently documented vaccination failure. Therefore, novel strategies are still required to control the occurrence of the disease in chickens. Recently, nanobodies (Nbs), which have the advantages of small molecular weight and low production costs, have been shown to be promising therapeutics against viral infection. In the present study, a total of 16 Nbs against NDV nucleocapsid protein (NP) were screened from two libraries against NDV using phage display technology. Of the 16 screened Nbs, eight were prevented from binding to NDV NP protein through administering positive chicken sera for anti-NDV antibodies, indicating that the epitopes recognized by these eight Nbs were able to induce the immune response after the chickens were infected with NDV stock. Subsequently, transfection assay, construction of recombinant DF-1 cells capable of expressing different nanobodies and viral inhibition assay were used to screen the nanobodies inhibiting NDV replication. The results demonstrated that Nb18, Nb30, and Nb88 significantly inhibited the replication of Class I and different genotypes of Class II NDV strains in DF-1 cells when they were expressed in the cytoplasm. Collectively, these nanobodies provided new tools for researching the functions of NDV NP protein and may be used as a novel strategy for designing drugs against NDV infection in chickens.
Collapse
Affiliation(s)
- Wenqi Fan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Pinpin Ji
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Xuwen Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Min Kong
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Ning Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Qiang Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Ying Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Qianqian Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Xiaoxuan Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - En-Min Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Qin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
| | - Yani Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Xianyang, China
- Scientific Observing and Experimental Station of Veterinary Pharmacology and Diagnostic Technology, Ministry of Agriculture, Yangling, China
- *Correspondence: Yani Sun,
| |
Collapse
|
24
|
Blocking phospholamban with VHH intrabodies enhances contractility and relaxation in heart failure. Nat Commun 2022; 13:3018. [PMID: 35641497 PMCID: PMC9156741 DOI: 10.1038/s41467-022-29703-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 03/28/2022] [Indexed: 12/19/2022] Open
Abstract
The dysregulated physical interaction between two intracellular membrane proteins, the sarco/endoplasmic reticulum Ca2+ ATPase and its reversible inhibitor phospholamban, induces heart failure by inhibiting calcium cycling. While phospholamban is a bona-fide therapeutic target, approaches to selectively inhibit this protein remain elusive. Here, we report the in vivo application of intracellular acting antibodies (intrabodies), derived from the variable domain of camelid heavy-chain antibodies, to modulate the function of phospholamban. Using a synthetic VHH phage-display library, we identify intrabodies with high affinity and specificity for different conformational states of phospholamban. Rapid phenotypic screening, via modified mRNA transfection of primary cells and tissue, efficiently identifies the intrabody with most desirable features. Adeno-associated virus mediated delivery of this intrabody results in improvement of cardiac performance in a murine heart failure model. Our strategy for generating intrabodies to investigate cardiac disease combined with modified mRNA and adeno-associated virus screening could reveal unique future therapeutic opportunities. Here the authors use modified RNA and VHH libraries to generate intrabodies that target dysregulated interactions between two calcium handling proteins in failing cardiomyocytes. Heart specific expression of the intrabodies in a murine heart failure model results in improved cardiac function.
Collapse
|
25
|
Li B, Qin X, Mi LZ. Nanobodies: from structure to applications in non-injectable and bispecific biotherapeutic development. NANOSCALE 2022; 14:7110-7122. [PMID: 35535618 DOI: 10.1039/d2nr00306f] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The increasing demand for convenient, miniaturized and multifunctional antibodies necessitates the development of novel antigen-recognition molecules for biological and medical studies. Nanobodies, the functional variable regions of camelid heavy-chain-only antibodies, as a new tool, complement the conventional antibodies and are in the stage of rapid development. The outstanding advantages of nanobodies include a stable structure, easy production, excellent water solubility, high affinity toward antigens and low immunogenicity. With promising application potential, nanobodies are now increasingly applied to various studies, including protein structure analysis, microscopic imaging, medical diagnosis, and drug development. The approval of the first nanobody drug Caplacizumab by the FDA disclosed the therapeutic potential of nanobodies. The outbreak of COVID-19 accelerated the development of nanobody drugs in non-injectable and bispecific biotherapeutic applications. Herein, we reviewed recent studies on the nanobody structure, screening and their applications in protein structure analysis and nanobody drugs, especially on non-injectable nanobody and bispecific nanobody development.
Collapse
Affiliation(s)
- Bingxuan Li
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | - Xiaohong Qin
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | - Li-Zhi Mi
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| |
Collapse
|
26
|
Van Fossen EM, Bednar RM, Jana S, Franklin R, Beckman J, Karplus PA, Mehl RA. Nanobody assemblies with fully flexible topology enabled by genetically encoded tetrazine amino acids. SCIENCE ADVANCES 2022; 8:eabm6909. [PMID: 35522749 PMCID: PMC9075797 DOI: 10.1126/sciadv.abm6909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/23/2022] [Indexed: 06/14/2023]
Abstract
Assembling nanobodies (Nbs) into polyvalent multimers is a powerful strategy for improving the effectiveness of Nb-based therapeutics and biotechnological tools. However, generally effective approaches to Nb assembly are currently restricted to the amino or carboxyl termini, greatly limiting the diversity of Nb multimer topologies that can be produced. Here, we show that reactive tetrazine groups-site-specifically inserted by genetic code expansion at Nb surface sites-are compatible with Nb folding and function, enabling Nb assembly at any desired point. Using two anti-SARS-CoV-2 Nbs with viral neutralization ability, we created Nb homo- and heterodimers with improved properties compared with conventionally linked Nb homodimers, which, in the case of our tetrazine-conjugated trimer, translated into enhanced viral neutralization. Thus, this tetrazine-based approach is a generally applicable strategy that greatly increases the accessible range of Nb assembly topologies, and thereby adds the optimization of topology as an effective avenue to generate Nb assemblies with improved efficacy.
Collapse
Affiliation(s)
- Elise M. Van Fossen
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Riley M. Bednar
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Subhashis Jana
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Rachel Franklin
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Joseph Beckman
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
- e-MSion, Inc., 2121 NE Jack London Drive, Corvallis, OR 97330, USA
| | - P. Andrew Karplus
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Ryan A. Mehl
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
27
|
Barakat S, Berksöz M, Zahedimaram P, Piepoli S, Erman B. Nanobodies as molecular imaging probes. Free Radic Biol Med 2022; 182:260-275. [PMID: 35240292 DOI: 10.1016/j.freeradbiomed.2022.02.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 12/12/2022]
Abstract
Camelidae derived single-domain antibodies (sdAbs), commonly known as nanobodies (Nbs), are the smallest antibody fragments with full antigen-binding capacity. Owing to their desirable properties such as small size, high specificity, strong affinity, excellent stability, and modularity, nanobodies are on their way to overtake conventional antibodies in terms of popularity. To date, a broad range of nanobodies have been generated against different molecular targets with applications spanning basic research, diagnostics, and therapeutics. In the field of molecular imaging, nanobody-based probes have emerged as a powerful tool. Radioactive or fluorescently labeled nanobodies are now used to detect and track many targets in different biological systems using imaging techniques. In this review, we provide an overview of the use of nanobodies as molecular probes. Additionally, we discuss current techniques for the generation, conjugation, and intracellular delivery of nanobodies.
Collapse
Affiliation(s)
- Sarah Barakat
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Melike Berksöz
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Pegah Zahedimaram
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Tuzla, Istanbul, Turkey.
| | - Sofia Piepoli
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bogazici University, 34342, Bebek, Istanbul, Turkey.
| | - Batu Erman
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bogazici University, 34342, Bebek, Istanbul, Turkey.
| |
Collapse
|
28
|
Ji F, Ren J, Vincke C, Jia L, Muyldermans S. Nanobodies: From Serendipitous Discovery of Heavy Chain-Only Antibodies in Camelids to a Wide Range of Useful Applications. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2446:3-17. [PMID: 35157266 DOI: 10.1007/978-1-0716-2075-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The presence of unique heavy chain-only antibodies (HCAbs) in camelids was discovered at Vrije Universiteit Brussel (VUB, Brussels, Belgium) at a time when many researchers were exploring the cloning and expression of smaller antigen-binding fragments (Fv and Fab) from hybridoma-derived antibodies. The potential importance of this discovery was anticipated, and efforts were immediately undertaken to understand the emergence and ontogeny of these HCAbs as well as to investigate the applications of the single-domain antigen-binding variable domains of HCAbs (nanobodies). Nanobodies were demonstrated to possess multiple biochemical and biophysical advantages over other antigen-binding antibody fragments and alternative scaffolds. Today, nanobodies have a significant and growing impact on research, biotechnology, and medicine.
Collapse
Affiliation(s)
- Fangling Ji
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Jun Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Cécile Vincke
- Cellular and Molecular Immunology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Serge Muyldermans
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China. .,Cellular and Molecular Immunology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
29
|
Staufer O, Gupta K, Hernandez Bücher JE, Kohler F, Sigl C, Singh G, Vasileiou K, Yagüe Relimpio A, Macher M, Fabritz S, Dietz H, Cavalcanti Adam EA, Schaffitzel C, Ruggieri A, Platzman I, Berger I, Spatz JP. Synthetic virions reveal fatty acid-coupled adaptive immunogenicity of SARS-CoV-2 spike glycoprotein. Nat Commun 2022; 13:868. [PMID: 35165285 PMCID: PMC8844029 DOI: 10.1038/s41467-022-28446-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 infection is a major global public health concern with incompletely understood pathogenesis. The SARS-CoV-2 spike (S) glycoprotein comprises a highly conserved free fatty acid binding pocket (FABP) with unknown function and evolutionary selection advantage1,2. Deciphering FABP impact on COVID-19 progression is challenged by the heterogenous nature and large molecular variability of live virus. Here we create synthetic minimal virions (MiniVs) of wild-type and mutant SARS-CoV-2 with precise molecular composition and programmable complexity by bottom-up assembly. MiniV-based systematic assessment of S free fatty acid (FFA) binding reveals that FABP functions as an allosteric regulatory site enabling adaptation of SARS-CoV-2 immunogenicity to inflammation states via binding of pro-inflammatory FFAs. This is achieved by regulation of the S open-to-close equilibrium and the exposure of both, the receptor binding domain (RBD) and the SARS-CoV-2 RGD motif that is responsible for integrin co-receptor engagement. We find that the FDA-approved drugs vitamin K and dexamethasone modulate S-based cell binding in an FABP-like manner. In inflammatory FFA environments, neutralizing immunoglobulins from human convalescent COVID-19 donors lose neutralization activity. Empowered by our MiniV technology, we suggest a conserved mechanism by which SARS-CoV-2 dynamically couples its immunogenicity to the host immune response.
Collapse
Affiliation(s)
- Oskar Staufer
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany.
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany.
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany.
| | - Kapil Gupta
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK
| | - Jochen Estebano Hernandez Bücher
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
| | - Fabian Kohler
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Christian Sigl
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Gunjita Singh
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Kate Vasileiou
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Ana Yagüe Relimpio
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
| | - Meline Macher
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Sebastian Fabritz
- Department for Chemical Biology, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Hendrik Dietz
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Elisabetta Ada Cavalcanti Adam
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Christiane Schaffitzel
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK
- Halo Therapeutics Ltd, Science Creates, Albert Road St. Philips Central, Bristol, BS2 0XJ, UK
| | - Alessia Ruggieri
- Department of Infectious Diseases, Molecular Virology, Center for Integrated Infectious Disease Research, University of Heidelberg, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany
| | - Ilia Platzman
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Imre Berger
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK.
- Halo Therapeutics Ltd, Science Creates, Albert Road St. Philips Central, Bristol, BS2 0XJ, UK.
| | - Joachim P Spatz
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany.
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany.
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany.
| |
Collapse
|
30
|
Wichgers Schreur PJ, van de Water S, Kortekaas J. Creation of Multimeric Single-Domain Antibodies Using Bacterial Superglues. Methods Mol Biol 2022; 2446:313-326. [PMID: 35157280 DOI: 10.1007/978-1-0716-2075-5_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Multimerization of single-domain antibodies (sdAbs) is instrumental for construction of antibody molecules with high avidity, extended in vivo half-life, and tailor-made biological activity. Two-component superglues, based on bacterium-derived peptides (Tags) and small protein domains (Catchers) that form isopeptide bonds when in close proximity, enable the creation of multimers by simply mixing of the individual components. Here, we provide detailed methods for the construction of sdAbs and scaffolds bearing genetically fused superglue components and their assembly into multimeric complexes.
Collapse
Affiliation(s)
- Paul J Wichgers Schreur
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, The Netherlands.
| | - Sandra van de Water
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Jeroen Kortekaas
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
- Laboratory of Virology, Wageningen University, Lelystad, The Netherlands
| |
Collapse
|
31
|
Soleimanizadeh A, Dinter H, Schindowski K. Central Nervous System Delivery of Antibodies and Their Single-Domain Antibodies and Variable Fragment Derivatives with Focus on Intranasal Nose to Brain Administration. Antibodies (Basel) 2021; 10:antib10040047. [PMID: 34939999 PMCID: PMC8699001 DOI: 10.3390/antib10040047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
IgG antibodies are some of the most important biopharmaceutical molecules with a high market volume. In spite of the fact that clinical therapies with antibodies are broadly utilized in oncology, immunology and hematology, their delivery strategies and biodistribution need improvement, their limitations being due to their size and poor ability to penetrate into tissues. In view of their small size, there is a rising interest in derivatives, such as single-domain antibodies and single-chain variable fragments, for clinical diagnostic but also therapeutic applications. Smaller antibody formats combine several benefits for clinical applications and can be manufactured at reduced production costs compared with full-length IgGs. Moreover, such formats have a relevant potential for targeted drug delivery that directs drug cargo to a specific tissue or across the blood–brain barrier. In this review, we give an overview of the challenges for antibody drug delivery in general and focus on intranasal delivery to the central nervous system with antibody formats of different sizes.
Collapse
Affiliation(s)
- Arghavan Soleimanizadeh
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Faculty of Medicine, University of Ulm, 89081 Ulm, Germany
| | - Heiko Dinter
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Department of Pharmacy and Biochemistry, University of Tübingen, 72076 Tübingen, Germany
| | - Katharina Schindowski
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Correspondence:
| |
Collapse
|
32
|
Wagner TR, Rothbauer U. Nanobodies - Little helpers unravelling intracellular signaling. Free Radic Biol Med 2021; 176:46-61. [PMID: 34536541 DOI: 10.1016/j.freeradbiomed.2021.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/26/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022]
Abstract
The identification of diagnostic and therapeutic targets requires a comprehensive understanding of cellular processes, for which advanced technologies in biomedical research are needed. The emergence of nanobodies (Nbs) derived from antibody fragments of camelid heavy chain-only antibodies as intracellular research tools offers new possibilities to study and modulate target antigens in living cells. Here we summarize this rapidly changing field, beginning with a brief introduction of Nbs, followed by an overview of how target-specific Nbs can be generated, and introduce the selection of intrabodies as research tools. Intrabodies, by definition, are intracellular functional Nbs that target ectopic or endogenous intracellular antigens within living cells. Such binders can be applied in various formats, e.g. as chromobodies for live cell microscopy or as biosensors to decipher complex intracellular signaling pathways. In addition, protein knockouts can be achieved by target-specific Nbs, while modulating Nbs have the potential as future therapeutics. The development of fine-tunable and switchable Nb-based systems that simultaneously provide spatial and temporal control has recently taken the application of these binders to the next level.
Collapse
Affiliation(s)
- Teresa R Wagner
- Pharmaceutical Biotechnology, Eberhard Karls University, Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University, Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
33
|
Tang Q, Owens RJ, Naismith JH. Structural Biology of Nanobodies against the Spike Protein of SARS-CoV-2. Viruses 2021; 13:v13112214. [PMID: 34835020 PMCID: PMC8625641 DOI: 10.3390/v13112214] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 12/28/2022] Open
Abstract
Nanobodies are 130 amino acid single-domain antibodies (VHH) derived from the unique heavy-chain-only subclass of Camelid immunogloblins. Their small molecular size, facile expression, high affinity and stability have combined to make them unique targeting reagents with numerous applications in the biomedical sciences. The first nanobody agent has now entered the clinic as a treatment against a blood disorder. The spread of the SARS-CoV-2 virus has seen the global scientific endeavour work to accelerate the development of technologies to try to defeat a pandemic that has now killed over four million people. In a remarkably short period of time, multiple studies have reported nanobodies directed against the viral Spike protein. Several agents have been tested in culture and demonstrate potent neutralisation of the virus or pseudovirus. A few agents have completed animal trials with very encouraging results showing their potential for treating infection. Here, we discuss the structural features that guide the nanobody recognition of the receptor binding domain of the Spike protein of SARS-CoV-2.
Collapse
Affiliation(s)
- Qilong Tang
- Structural Biology, The Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot OX11 0FA, UK;
- The Wellcome Centre for Human Genetics, Division of Structural Biology, University of Oxford, Headington, Oxford OX3 7BN, UK
| | - Raymond J. Owens
- Structural Biology, The Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot OX11 0FA, UK;
- The Wellcome Centre for Human Genetics, Division of Structural Biology, University of Oxford, Headington, Oxford OX3 7BN, UK
- Correspondence: (R.J.O.); (J.H.N.)
| | - James H. Naismith
- Structural Biology, The Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot OX11 0FA, UK;
- The Wellcome Centre for Human Genetics, Division of Structural Biology, University of Oxford, Headington, Oxford OX3 7BN, UK
- Correspondence: (R.J.O.); (J.H.N.)
| |
Collapse
|
34
|
Nanobody-based CTLA4 inhibitors for immune checkpoint blockade therapy of canine cancer patients. Sci Rep 2021; 11:20763. [PMID: 34675296 PMCID: PMC8531395 DOI: 10.1038/s41598-021-00325-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/06/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer is the leading cause of death in the geriatric dog population. Currently, the use of immune checkpoint inhibitors (ICIs) such as anti-CTLA4 antibodies has markedly improved the prognosis of several cancers in their advanced stages. However, ICIs targeting CTLA4 blockade to treat canine cancer patients are yet to define. In this study, we sought to develop, characterize and assess whether chimeric heavy chain only antibodies (cHcAbs) against CTLA4 are viable therapeutic candidates for the treatment of canine cancers. Anti-CTLA4 nanobodies (Nbs) were identified from a yeast nanobody (Nb) library using magnetic-assisted cell sorting (MACS) and flow cytometry. cHcAbs were engineered by genetically fusing the DNA sequences coding for anti-CTLA4 Nbs with the Fc domain of the subclass B of canine IgG. Recombinant cHcAbs were purified from ExpiCHO-S cells. Stable cell lines expressing canine CTLA4 and FcγRI were used to elucidate the binding ability and specificity of cHcAbs. PBMCs isolated from healthy dogs were used to evaluate the ability of cHcAbs to activate canine PBMCs (cPBMCs). Novel Nbs were identified using the extracellular domain of canine CTLA4 protein to screen a fully synthetic yeast nanobody library. Purified Nbs bind specifically to natïve canine CTLA4. We report that chimeric HcAbs, which were engineered by fusing the anti-CTLA4 Nbs and Fc region of subclass B of canine IgG, were half the size of a conventional mAb and formed dimers. The chimeric HcAbs specifically binds both with canine CTLA4 and Fcγ receptors. As the binding of Nbs overlapped with the MYPPPY motif of canine CTLA4, these Nbs were expected to sterically disrupt the interaction of canine CTLA4 to B-7s. Like their human counterpart, canine CTLA4 was expressed on helper T cells and a small subset of cytotoxic T cells. Canine Tregs also constitutively expressed CTLA4, and stimulation with PMA/Ionomycin dramatically increased expression of CTLA4 on the cell surface. Stimulation of cPBMCs in the presence of agonistic anti-CD3 Ab and cHcAb6 significantly increased the expression of IFN-γ as compared to the isotype control. This study identifies a novel nanobody-based CTLA4 inhibitor for the treatment of canine cancer patients.
Collapse
|
35
|
Trashin S, Morales-Yánez F, Thiruvottriyur Shanmugam S, Paredis L, Carrión EN, Sariego I, Muyldermans S, Polman K, Gorun SM, De Wael K. Nanobody-Based Immunosensor Detection Enhanced by Photocatalytic-Electrochemical Redox Cycling. Anal Chem 2021; 93:13606-13614. [PMID: 34585567 DOI: 10.1021/acs.analchem.1c02876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Detection of antigenic biomarkers present in trace amounts is of crucial importance for medical diagnosis. A parasitic disease, human toxocariasis, lacks an adequate diagnostic method despite its worldwide occurrence. The currently used serology tests may stay positive even years after a possibly unnoticed infection, whereas the direct detection of a re-infection or a still active infection remains a diagnostic challenge due to the low concentration of circulating parasitic antigens. We report a time-efficient sandwich immunosensor using small recombinant single-domain antibodies (nanobodies) derived from camelid heavy-chain antibodies specific to Toxocara canis antigens. An enhanced sensitivity to pg/mL levels is achieved by using a redox cycle consisting of a photocatalytic oxidation and electrochemical reduction steps. The photocatalytic oxidation is achieved by a photosensitizer generating singlet oxygen (1O2) that, in turn, readily reacts with p-nitrophenol enzymatically produced under alkaline conditions. The photooxidation produces benzoquinone that is electrochemically reduced to hydroquinone, generating an amperometric response. The light-driven process could be easily separated from the background, thus making amperometric detection more reliable. The proposed method for detection of the toxocariasis antigen marker shows superior performances compared to other detection schemes with the same nanobodies and outperforms by at least two orders of magnitude the assays based on regular antibodies, thus suggesting new opportunities for electrochemical immunoassays of challenging low levels of antigens.
Collapse
Affiliation(s)
| | - Francisco Morales-Yánez
- Department of Biomedical Sciences, Institute of Tropical Medicine, 2000 Antwerp, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | | | - Linda Paredis
- Department of Biomedical Sciences, Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Erik N Carrión
- Department of Chemistry and Biochemistry and the Center for Functional Materials, Seton Hall University, South Orange, New Jersey 07079, United States
| | - Idalia Sariego
- Department of Parasitology, Institute of Tropical Medicine Pedro Kouri, 17100 Havana, Cuba
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Katja Polman
- Department of Biomedical Sciences, Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Sergiu M Gorun
- Department of Chemistry and Biochemistry and the Center for Functional Materials, Seton Hall University, South Orange, New Jersey 07079, United States
| | | |
Collapse
|
36
|
Scaled preparation of extracellular vesicles from conditioned media. Adv Drug Deliv Rev 2021; 177:113940. [PMID: 34419502 DOI: 10.1016/j.addr.2021.113940] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/13/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) especially of mesenchymal stem/stomal cells (MSCs) are increasingly considered as biotherapeutic agents for a variety of different diseases. For translating them effectively into the clinics, scalable production processes fulfilling good manufacturing practice (GMP) are needed. Like for other biotherapeutic agents, the manufacturing of EV products can be subdivided in the upstream and downstream processing and the subsequent quality control, each of them containing several unit operations. During upstream processing (USP), cells are isolated, stored (cell banking) and expanded; furthermore, EV-containing conditioned media are produced. During downstream processing (DSP), conditioned media (CM) are processed to obtain concentrated and purified EV products. CM are either stored until DSP or are directly processed. As first unit operation in DSP, clarification removes remaining cells, debris and other larger impurities. The key operations of each EV DSP is volume-reduction combined with purification of the concentrated EVs. Most of the EV preparation methods used in conventional research labs including differential centrifugation procedures are limited in their scalability. Consequently, it is a major challenge in the therapeutic EV field to identify appropriate EV concentration and purification methods allowing scale up. As EVs share several features with enveloped viruses, that are used for more than two decades in the clinics now, several principles can be adopted to EV manufacturing. Here, we introduce and discuss volume reducing and purification methods frequently used for viruses and analyze their value for the manufacturing of EV-based therapeutics.
Collapse
|
37
|
Huo J, Mikolajek H, Le Bas A, Clark JJ, Sharma P, Kipar A, Dormon J, Norman C, Weckener M, Clare DK, Harrison PJ, Tree JA, Buttigieg KR, Salguero FJ, Watson R, Knott D, Carnell O, Ngabo D, Elmore MJ, Fotheringham S, Harding A, Moynié L, Ward PN, Dumoux M, Prince T, Hall Y, Hiscox JA, Owen A, James W, Carroll MW, Stewart JP, Naismith JH, Owens RJ. A potent SARS-CoV-2 neutralising nanobody shows therapeutic efficacy in the Syrian golden hamster model of COVID-19. Nat Commun 2021; 12:5469. [PMID: 34552091 PMCID: PMC8458290 DOI: 10.1038/s41467-021-25480-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/12/2021] [Indexed: 12/18/2022] Open
Abstract
SARS-CoV-2 remains a global threat to human health particularly as escape mutants emerge. There is an unmet need for effective treatments against COVID-19 for which neutralizing single domain antibodies (nanobodies) have significant potential. Their small size and stability mean that nanobodies are compatible with respiratory administration. We report four nanobodies (C5, H3, C1, F2) engineered as homotrimers with pmolar affinity for the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. Crystal structures show C5 and H3 overlap the ACE2 epitope, whilst C1 and F2 bind to a different epitope. Cryo Electron Microscopy shows C5 binding results in an all down arrangement of the Spike protein. C1, H3 and C5 all neutralize the Victoria strain, and the highly transmissible Alpha (B.1.1.7 first identified in Kent, UK) strain and C1 also neutralizes the Beta (B.1.35, first identified in South Africa). Administration of C5-trimer via the respiratory route showed potent therapeutic efficacy in the Syrian hamster model of COVID-19 and separately, effective prophylaxis. The molecule was similarly potent by intraperitoneal injection.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/pharmacology
- Cryoelectron Microscopy
- Crystallography, X-Ray
- Disease Models, Animal
- Dose-Response Relationship, Immunologic
- Epitopes/chemistry
- Epitopes/metabolism
- Female
- Male
- Mesocricetus
- Neutralization Tests
- SARS-CoV-2/drug effects
- Single-Domain Antibodies/administration & dosage
- Single-Domain Antibodies/immunology
- Single-Domain Antibodies/metabolism
- Single-Domain Antibodies/pharmacology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/metabolism
- COVID-19 Drug Treatment
Collapse
Affiliation(s)
- Jiandong Huo
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK
- Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Protein Production UK, The Rosalind Franklin Institute - Diamond Light Source, The Research Complex at Harwell, Science Campus, Didcot, UK
| | | | - Audrey Le Bas
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK
- Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Protein Production UK, The Rosalind Franklin Institute - Diamond Light Source, The Research Complex at Harwell, Science Campus, Didcot, UK
| | - Jordan J Clark
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Parul Sharma
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Anja Kipar
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Joshua Dormon
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK
- Protein Production UK, The Rosalind Franklin Institute - Diamond Light Source, The Research Complex at Harwell, Science Campus, Didcot, UK
| | - Chelsea Norman
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK
- Protein Production UK, The Rosalind Franklin Institute - Diamond Light Source, The Research Complex at Harwell, Science Campus, Didcot, UK
| | - Miriam Weckener
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK
| | - Daniel K Clare
- Diamond Light Source Ltd, Harwell Science Campus, Didcot, UK
| | - Peter J Harrison
- Protein Production UK, The Rosalind Franklin Institute - Diamond Light Source, The Research Complex at Harwell, Science Campus, Didcot, UK
- Diamond Light Source Ltd, Harwell Science Campus, Didcot, UK
| | - Julia A Tree
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | - Karen R Buttigieg
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | | | - Robert Watson
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | - Daniel Knott
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | - Oliver Carnell
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | - Didier Ngabo
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | - Michael J Elmore
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | - Susan Fotheringham
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | - Adam Harding
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Lucile Moynié
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK
| | - Philip N Ward
- Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Protein Production UK, The Rosalind Franklin Institute - Diamond Light Source, The Research Complex at Harwell, Science Campus, Didcot, UK
| | - Maud Dumoux
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK
| | - Tessa Prince
- Diamond Light Source Ltd, Harwell Science Campus, Didcot, UK
| | - Yper Hall
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
| | - Julian A Hiscox
- Diamond Light Source Ltd, Harwell Science Campus, Didcot, UK
- Department of Preventive Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Infectious Diseases Horizontal Technology Centre (ID HTC), A*STAR, Singapore, Singapore
| | - Andrew Owen
- Department of Pharmacology and Therapeutics, Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, UK
| | - William James
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Miles W Carroll
- National Infection Service, Public Health England, Porton Down, Salisbury, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - James P Stewart
- Diamond Light Source Ltd, Harwell Science Campus, Didcot, UK
- Department of Preventive Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Infectious Disease, University of Georgia, Georgia, USA
| | - James H Naismith
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK.
- Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
- Protein Production UK, The Rosalind Franklin Institute - Diamond Light Source, The Research Complex at Harwell, Science Campus, Didcot, UK.
| | - Raymond J Owens
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK.
- Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
- Protein Production UK, The Rosalind Franklin Institute - Diamond Light Source, The Research Complex at Harwell, Science Campus, Didcot, UK.
| |
Collapse
|
38
|
Dietsch F, Nominé Y, Stoessel A, Kostmann C, Bonhoure A, Chatton B, Donzeau M. Small p53 derived peptide suitable for robust nanobodies dimerization. J Immunol Methods 2021; 498:113144. [PMID: 34481824 DOI: 10.1016/j.jim.2021.113144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 11/15/2022]
Abstract
Bivalent VHHs have been shown to display better functional affinity compared with their monovalent counterparts. Bivalency can be achieved either by inserting a hinge region between both VHHs units or by using modules that lead to dimerization. In this report, a small self-associating peptide originating from the tetramerization domain of p53 was developed as a tool for devicing nanobody dimerization. This E3 peptide was evaluated for the dimerization of an anti-eGFP nanobody (nano-eGFP-E3) whose activity was compared to a bivalent anti-eGFP constructed in tandem using GS rich linker. The benefit of bivalency in terms of avidity and specificity was assessed in different in vitro and in cellulo assays. In ELISA and SPR, the dimeric and tandem formats were nearly equivalent in terms of gain of avidity compared to the monovalent counterpart. However, in cellulo, the nano-eGFP-E3 construct showed its superiority over the tandem format in terms of specificity with a highest and better ratio signal-to-noise. All together, the E3 peptide provides a universal suitable tool for the construction of dimeric biomolecules, in particular antibody fragments with improved functional affinity.
Collapse
Affiliation(s)
- Frank Dietsch
- Université de Strasbourg, UMR7242 Biotechnologie et Signalisation Cellulaire, Ecole Supérieure de Biotechnologie Strasbourg, F-67412 Illkirch, France
| | - Yves Nominé
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, F-67412 Illkirch, France
| | - Audrey Stoessel
- Université de Strasbourg, UMR7242 Biotechnologie et Signalisation Cellulaire, Ecole Supérieure de Biotechnologie Strasbourg, F-67412 Illkirch, France
| | - Camille Kostmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, F-67412 Illkirch, France
| | - Anna Bonhoure
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, F-67412 Illkirch, France
| | - Bruno Chatton
- Université de Strasbourg, UMR7242 Biotechnologie et Signalisation Cellulaire, Ecole Supérieure de Biotechnologie Strasbourg, F-67412 Illkirch, France
| | - Mariel Donzeau
- Université de Strasbourg, UMR7242 Biotechnologie et Signalisation Cellulaire, Ecole Supérieure de Biotechnologie Strasbourg, F-67412 Illkirch, France.
| |
Collapse
|
39
|
Nanobody multimerization strategy to enhance the sensitivity of competitive ELISA for detection of ochratoxin A in coffee samples. Food Control 2021. [DOI: 10.1016/j.foodcont.2021.108167] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
40
|
Panikar SS, Banu N, Haramati J, Del Toro-Arreola S, Riera Leal A, Salas P. Nanobodies as efficient drug-carriers: Progress and trends in chemotherapy. J Control Release 2021; 334:389-412. [PMID: 33964364 DOI: 10.1016/j.jconrel.2021.05.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 01/24/2023]
Abstract
Nanobodies (Nb) have a promising future as a part of next generation chemodrug delivery systems. Nb, or VHH, are small (15 kDa) monomeric antibody fragments consisting of the antigen binding region of heavy chain antibodies. Heavy chain antibodies are naturally produced by camelids, however the structure of their VHH regions can be readily reproduced in industrial expression systems, such as bacteria or yeast. Due to their small size, high solubility, remarkable stability, manipulatable characteristics, excellent in vivo tissue penetration, conjugation advantages, and ease of production, Nb have many advantages when compared against their antibody precursors. In this review, we discuss the generation and selection of Nbs via phage display libraries for easy screening, and the conjugation techniques involved in creating target-specific nanocarriers. Furthermore, we provide a comprehensive overview of recent developments and perspectives in the field of Nb drug conjugates (NDCs) and Nb-based drug vehicles (NDv) with respect to antitumor therapeutics.
Collapse
Affiliation(s)
- Sandeep Surendra Panikar
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autonoma de México (UNAM), Apartado Postal 1-1010, Queretaro, Queretaro 76000, Mexico.
| | - Nehla Banu
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Jesse Haramati
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Susana Del Toro-Arreola
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Annie Riera Leal
- UC Davis Institute for Regenerative Cures, Department of Dermatology, University of California, Davis, 2921 Stockton Blvd, Rm 1630, Sacramento, CA 95817, USA
| | - Pedro Salas
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autonoma de México (UNAM), Apartado Postal 1-1010, Queretaro, Queretaro 76000, Mexico
| |
Collapse
|
41
|
Anderson G, Liu JL, Esparza TJ, Voelker BT, Hofmann ER, Goldman ER. Single-Domain Antibodies for the Detection of SARS-CoV-2 Nucleocapsid Protein. Anal Chem 2021; 93:7283-7291. [PMID: 33955213 PMCID: PMC8117401 DOI: 10.1021/acs.analchem.1c00677] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/22/2021] [Indexed: 12/19/2022]
Abstract
The goal of this work was to develop recombinantly expressed variable domains derived from camelid heavy-chain antibodies known as single-domain antibodies (sdAbs) directed against the SARS-CoV-2 nucleocapsid protein for incorporation into detection assays. To achieve this, a llama was immunized using a recombinant SARS-CoV-2 nucleocapsid protein and an immune phage-display library of variable domains was developed. The sdAbs selected from this library segregated into five distinct sequence families. Three of these families bind to unique epitopes with high affinity, low nM to sub-nM KD, as determined by surface plasmon resonance. To further enhance the utility of these sdAbs for the detection of nucleocapsid protein, homobivalent and heterobivalent genetic fusion constructs of the three high-affinity sdAbs were prepared. The effectiveness of the sdAbs for the detection of nucleocapsid protein was evaluated using MagPlex fluid array assays, a multiplexed immunoassay on color-coded magnetic microspheres. Using the optimal bivalent pair, one immobilized on the microsphere and the other serving as the biotinylated recognition reagent, a detection limit as low as 50 pg/mL of recombinant nucleocapsid and of killed virus down to 1.28 × 103 pfu/mL was achieved. The sdAbs described here represent immune reagents that can be tailored to be optimized for a number of detection platforms and may one day aid in the detection of SARS-CoV-2 to assist in controlling the current pandemic.
Collapse
Affiliation(s)
- George
P. Anderson
- Center
for Biomolecular Science and Engineering, US Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, District of Columbia 20375, United States
| | - Jinny L. Liu
- Center
for Biomolecular Science and Engineering, US Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, District of Columbia 20375, United States
| | - Thomas J. Esparza
- Laboratory
of Functional and Molecular Imaging, The
National Institute of Neurological Disorders and Stroke Intramural
Research Program, Bethesda, Maryland 20892, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland 20892, United States
| | - Bruce T. Voelker
- Chemical
Biological Center, U.S. Army Combat Capabilities
Development Command, 8198 Blackhawk Road, Aberdeen Proving Ground, Maryland 21010, United States
| | - E. Randal Hofmann
- Chemical
Biological Center, U.S. Army Combat Capabilities
Development Command, 8198 Blackhawk Road, Aberdeen Proving Ground, Maryland 21010, United States
- EXCET,
Inc., 6225 Brandon Avenue
#360, Springfield, Virginia 22150, United States
| | - Ellen R. Goldman
- Center
for Biomolecular Science and Engineering, US Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, District of Columbia 20375, United States
| |
Collapse
|
42
|
Ma J, Mo Y, Tang M, Shen J, Qi Y, Zhao W, Huang Y, Xu Y, Qian C. Bispecific Antibodies: From Research to Clinical Application. Front Immunol 2021; 12:626616. [PMID: 34025638 PMCID: PMC8131538 DOI: 10.3389/fimmu.2021.626616] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (BsAbs) are antibodies with two binding sites directed at two different antigens or two different epitopes on the same antigen. The clinical therapeutic effects of BsAbs are superior to those of monoclonal antibodies (MoAbs), with broad applications for tumor immunotherapy as well as for the treatment of other diseases. Recently, with progress in antibody or protein engineering and recombinant DNA technology, various platforms for generating different types of BsAbs based on novel strategies, for various uses, have been established. More than 30 mature commercial technology platforms have been used to create and develop BsAbs based on the heterologous recombination of heavy chains and matching of light chains. The detailed mechanisms of clinical/therapeutic action have been demonstrated with these different types of BsAbs. Three kinds of BsAbs have received market approval, and more than 110 types of BsAbs are at various stages of clinical trials. In this paper, we elaborate on the classic platforms, mechanisms, and applications of BsAbs. We hope that this review can stimulate new ideas for the development of BsAbs and improve current clinical strategies.
Collapse
Affiliation(s)
- Jiabing Ma
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yicheng Mo
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Menglin Tang
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Junjie Shen
- IND Center, Chongqing Precision Biotech Co., Ltd., Chongqing, China
| | - Yanan Qi
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Wenxu Zhao
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Yi Huang
- IND Center, Chongqing Precision Biotech Co., Ltd., Chongqing, China
| | - Yanmin Xu
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Cheng Qian
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
43
|
Valenzuela Nieto G, Jara R, Watterson D, Modhiran N, Amarilla AA, Himelreichs J, Khromykh AA, Salinas-Rebolledo C, Pinto T, Cheuquemilla Y, Margolles Y, López González Del Rey N, Miranda-Chacon Z, Cuevas A, Berking A, Deride C, González-Moraga S, Mancilla H, Maturana D, Langer A, Toledo JP, Müller A, Uberti B, Krall P, Ehrenfeld P, Blesa J, Chana-Cuevas P, Rehren G, Schwefel D, Fernandez LÁ, Rojas-Fernandez A. Potent neutralization of clinical isolates of SARS-CoV-2 D614 and G614 variants by a monomeric, sub-nanomolar affinity nanobody. Sci Rep 2021; 11:3318. [PMID: 33558635 PMCID: PMC7870875 DOI: 10.1038/s41598-021-82833-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Despite unprecedented global efforts to rapidly develop SARS-CoV-2 treatments, in order to reduce the burden placed on health systems, the situation remains critical. Effective diagnosis, treatment, and prophylactic measures are urgently required to meet global demand: recombinant antibodies fulfill these requirements and have marked clinical potential. Here, we describe the fast-tracked development of an alpaca Nanobody specific for the receptor-binding-domain (RBD) of the SARS-CoV-2 Spike protein with potential therapeutic applicability. We present a rapid method for nanobody isolation that includes an optimized immunization regimen coupled with VHH library E. coli surface display, which allows single-step selection of Nanobodies using a simple density gradient centrifugation of the bacterial library. The selected single and monomeric Nanobody, W25, binds to the SARS-CoV-2 S RBD with sub-nanomolar affinity and efficiently competes with ACE-2 receptor binding. Furthermore, W25 potently neutralizes SARS-CoV-2 wild type and the D614G variant with IC50 values in the nanomolar range, demonstrating its potential as antiviral agent.
Collapse
Affiliation(s)
| | - Ronald Jara
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Watterson
- School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, Australia
- The Australian Institute for Biotechnology and Nanotechnology, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, Australia
- The Australian Institute for Biotechnology and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Johanna Himelreichs
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Alexander A Khromykh
- School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | | | - Teresa Pinto
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Yorka Cheuquemilla
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Berking Biotechnology, Valdivia, Chile
| | - Yago Margolles
- Department of Microbial Biotechnology, National Biotechnology Center, Superior Council of Scientific Research, Madrid, Spain
| | | | - Zaray Miranda-Chacon
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Alexei Cuevas
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | | | - Camila Deride
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Institute of Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | | | - Héctor Mancilla
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Maturana
- NanoTemper Technologies GmbH, Floessergasse 4, 81369, Munich, Germany
| | - Andreas Langer
- NanoTemper Technologies GmbH, Floessergasse 4, 81369, Munich, Germany
| | - Juan Pablo Toledo
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Ananda Müller
- Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
- Institute of Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Benjamín Uberti
- Institute of Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Paola Krall
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Department of Pediatrics and Children's Surgery Oriente, Universidad de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, 28938, Madrid, Spain
| | - Pedro Chana-Cuevas
- CETRAM & Faculty of Medical Science, Universidad de Santiago de Chile, Santiago, Chile
| | - German Rehren
- Technology Transfer and Licensing Office, Universidad Austral de Chile, Valdivia, Chile
| | - David Schwefel
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Luis Ángel Fernandez
- Department of Microbial Biotechnology, National Biotechnology Center, Superior Council of Scientific Research, Madrid, Spain
| | - Alejandro Rojas-Fernandez
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.
- Berking Biotechnology, Valdivia, Chile.
- Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile.
- Institute of Philosophy and Complexity Sciences, Santiago, Chile.
| |
Collapse
|
44
|
Neuroscience and Neuroimmunology Solutions for Osteoarthritis Pain: Biological Drugs, Growth Factors, Peptides and Monoclonal Antibodies Targeting Peripheral Nerves. NEUROSCI 2021. [DOI: 10.3390/neurosci2010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Neuroscience is a vast discipline that deals with the anatomy, biochemistry, molecular biology, physiology and pathophysiology of central and peripheral nerves. Advances made through basic, translational, and clinical research in the field of neuroscience have great potential for long-lasting and beneficial impacts on human and animal health. The emerging field of biological therapy is intersecting with the disciplines of neuroscience, orthopaedics and rheumatology, creating new horizons for interdisciplinary and applied research. Biological drugs, growth factors, therapeutic peptides and monoclonal antibodies are being developed and tested for the treatment of painful arthritic and rheumatic diseases. This concise communication focuses on the solutions provided by the fields of neuroscience and neuroimmunology for real-world clinical problems in the field of orthopaedics and rheumatology, focusing on synovial joint pain and the emerging biological treatments that specifically target pathways implicated in osteoarthritis pain in peripheral nerves.
Collapse
|
45
|
Bracken CJ, Lim SA, Solomon P, Rettko NJ, Nguyen DP, Zha BS, Schaefer K, Byrnes JR, Zhou J, Lui I, Liu J, Pance K, Zhou XX, Leung KK, Wells JA. Bi-paratopic and multivalent VH domains block ACE2 binding and neutralize SARS-CoV-2. Nat Chem Biol 2021; 17:113-121. [PMID: 33082574 PMCID: PMC8356808 DOI: 10.1038/s41589-020-00679-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/25/2020] [Indexed: 11/14/2022]
Abstract
Neutralizing agents against SARS-CoV-2 are urgently needed for the treatment and prophylaxis of COVID-19. Here, we present a strategy to rapidly identify and assemble synthetic human variable heavy (VH) domains toward neutralizing epitopes. We constructed a VH-phage library and targeted the angiotensin-converting enzyme 2 (ACE2) binding interface of the SARS-CoV-2 Spike receptor-binding domain (Spike-RBD). Using a masked selection approach, we identified VH binders to two non-overlapping epitopes and further assembled these into multivalent and bi-paratopic formats. These VH constructs showed increased affinity to Spike (up to 600-fold) and neutralization potency (up to 1,400-fold) on pseudotyped SARS-CoV-2 virus when compared to standalone VH domains. The most potent binder, a trivalent VH, neutralized authentic SARS-CoV-2 with a half-maximal inhibitory concentration (IC50) of 4.0 nM (180 ng ml-1). A cryo-EM structure of the trivalent VH bound to Spike shows each VH domain engaging an RBD at the ACE2 binding site, confirming our original design strategy.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/antagonists & inhibitors
- Angiotensin-Converting Enzyme 2/chemistry
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/immunology
- Animals
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/chemistry
- Antibodies, Viral/genetics
- Antibodies, Viral/immunology
- Binding Sites, Antibody/genetics
- Binding Sites, Antibody/immunology
- Chlorocebus aethiops
- Cryoelectron Microscopy
- HEK293 Cells
- Humans
- Models, Molecular
- Peptide Library
- Protein Binding
- Protein Conformation, alpha-Helical
- Protein Conformation, beta-Strand
- Protein Interaction Domains and Motifs
- SARS-CoV-2
- Single-Chain Antibodies/chemistry
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Spike Glycoprotein, Coronavirus/antagonists & inhibitors
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Vero Cells
Collapse
Affiliation(s)
- Colton J Bracken
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Shion A Lim
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Paige Solomon
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Nicholas J Rettko
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Duy P Nguyen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Lyell Immunopharma Inc., Seattle, WA, USA
| | - Beth Shoshana Zha
- Department of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Kaitlin Schaefer
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - James R Byrnes
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Jie Zhou
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Irene Lui
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Jia Liu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Merck & Co., South San Francisco, CA, USA
| | - Katarina Pance
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Xin X Zhou
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin K Leung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA.
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
46
|
Selection and Characterization of Single-Domain Antibodies for Detection of Lassa Nucleoprotein. Antibodies (Basel) 2020; 9:antib9040071. [PMID: 33348599 PMCID: PMC7768477 DOI: 10.3390/antib9040071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 11/26/2022] Open
Abstract
Lassa virus is the etiologic agent of Lassa fever, an acute and often fatal illness endemic to West Africa. It is important to develop new reagents applicable either for the specific diagnosis or as improved therapeutics for the treatment of Lassa fever. Here, we describe the development and initial testing of llama-derived single-domain antibodies that are specific for the Lassa virus nucleoprotein. Four sequence families based on complementarity-determining region (CDR) homology were identified by phage-based enzyme-linked immunosorbent assays, however, the highest affinity clones all belonged to the same sequence family which possess a second disulfide bond between Framework 2 and CDR3. The affinity and thermal stability were evaluated for each clone. A MagPlex-based homogeneous sandwich immunoassay for Lassa virus-like particles was also demonstrated to show their potential for further development as diagnostic reagents.
Collapse
|
47
|
Huang S, van Duijnhoven SMJ, Sijts AJAM, van Elsas A. Bispecific antibodies targeting dual tumor-associated antigens in cancer therapy. J Cancer Res Clin Oncol 2020; 146:3111-3122. [PMID: 32989604 PMCID: PMC7679314 DOI: 10.1007/s00432-020-03404-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Bispecific antibodies (BsAbs) have emerged as a leading drug class for cancer therapy and are becoming increasingly of interest for therapeutic applications. As of April 2020, over 123 BsAbs are under clinical evaluation for use in oncology (including the two marketed BsAbs Blinatumomab and Catumaxomab). The majority (82 of 123) of BsAbs under clinical evaluation can be categorized as bispecific immune cell engager whereas a second less well-discussed subclass of BsAbs targets two tumor-associated antigens (TAAs). In this review, we summarize the clinical development of dual TAAs targeting BsAbs and provide an overview of critical considerations when designing dual TAA targeting BsAbs. METHODS Herein the relevant literature and clinical trials published in English until April 1st 2020 were searched using PubMed and ClinicalTrials.gov database. BsAbs were considered to be active in clinic if their clinical trials were not terminated, withdrawn or completed before 2018 without reporting results. Data missed by searching ClinicalTrials.gov was manually curated. RESULTS Dual TAAs targeting BsAbs offer several advantages including increased tumor selectivity, potential to concurrently modulate two functional pathways in the tumor cell and may yield improved payload delivery. CONCLUSIONS Dual TAAs targeting BsAbs represent a valuable class of biologics and early stage clinical studies have demonstrated promising anti-tumor efficacy in both hematologic malignancies and solid tumors.
Collapse
Affiliation(s)
- Shuyu Huang
- Aduro Biotech Europe, Oss, The Netherlands
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | | | - Alice J A M Sijts
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
48
|
Mir MA, Mehraj U, Sheikh BA, Hamdani SS. Nanobodies: The "Magic Bullets" in therapeutics, drug delivery and diagnostics. Hum Antibodies 2020; 28:29-51. [PMID: 31322555 DOI: 10.3233/hab-190390] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Antibodies represent a well-established class of clinical diagnostics for medical applications as well as essential research and biotechnological tools. Although both polyclonal and monoclonal antibodies are indispensable reagents in basic research and diagnostics but both of them have their limitations. Hence, there is urgent need to develop strategies aimed at production of alternative scaffolds and recombinant antibodies of smaller dimensions that could be easily produced, selected and manipulated. Unlike conventional antibodies, members of Camelidae and sharks produce antibodies composed only of heavy chains with small size, high solubility, thermal stability, refolding capacity and good tissue penetration in vivo. The discovery of these naturally occurring antibodies having only heavy-chain in Camelidae family and their further development into small recombinant nanobodies represents an attractive alternative in drug delivery, diagnostics and imaging. Nanobody derivatives are soluble, stable, versatile, have unique refolding capacities, reduced aggregation tendencies and high-target binding capabilities. They can be genetically customized to target enzymes, transmembrane proteins or molecular interactions. Their ability to recognize recessed antigenic sites has been attributed to their smaller size and the ability of the extended CDR3 loop to quickly penetrate into such epitopes. With the advent of molecular engineering and phage display technology, they can be of potential use in molecular imaging, drug delivery and therapeutics for several major diseases. In this review we present the recent advances in nanobodies for modulating immune functions, for targeting cancers, viruses, toxins and microbes as well as their utility as diagnostic and biosensor tools.
Collapse
|
49
|
Muyldermans S. A guide to: generation and design of nanobodies. FEBS J 2020; 288:2084-2102. [PMID: 32780549 PMCID: PMC8048825 DOI: 10.1111/febs.15515] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/03/2020] [Accepted: 08/07/2020] [Indexed: 01/09/2023]
Abstract
A nanobody (Nb) is a registered trademark of Ablynx, referring to the single antigen-binding domain of heavy chain-only antibodies (HCAbs) that are circulating in Camelidae. Nbs are produced recombinantly in micro-organisms and employed as research tools or for diagnostic and therapeutic applications. They were - and still are - also named single-domain antibodies (sdAbs) or variable domain of the heavy chain of HCAbs (VHH). A variety of methods are currently in use for the fast and efficient generation of target-specific Nbs. Such Nbs are produced at low cost and associate with high affinity to their cognate antigen. They are robust, strictly monomeric and easy to tailor into more complex entities to meet the requirements of their application. Here, we review the various sources and different strategies that have been developed to identify rapidly, target-specific Nbs. We further discuss a variety of engineering technologies that have been explored to broaden the application range of Nbs and summarise those applications where designed Nbs might offer a marked advantage over other affinity reagents.
Collapse
Affiliation(s)
- Serge Muyldermans
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, Belgium.,Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, China
| |
Collapse
|
50
|
Bracken CJ, Lim SA, Solomon P, Rettko NJ, Nguyen DP, Zha BS, Schaefer K, Byrnes JR, Zhou J, Lui I, Liu J, Pance K, Zhou XX, Leung KK, Wells JA. Bi-paratopic and multivalent human VH domains neutralize SARS-CoV-2 by targeting distinct epitopes within the ACE2 binding interface of Spike. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.08.08.242511. [PMID: 32817948 PMCID: PMC7430580 DOI: 10.1101/2020.08.08.242511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Neutralizing agents against SARS-CoV-2 are urgently needed for treatment and prophylaxis of COVID-19. Here, we present a strategy to rapidly identify and assemble synthetic human variable heavy (VH) domain binders with high affinity toward neutralizing epitopes without the need for high-resolution structural information. We constructed a VH-phage library and targeted a known neutralizing site, the angiotensin-converting enzyme 2 (ACE2) binding interface of the trimeric SARS-CoV-2 Spike receptor-binding domain (Spike-RBD). Using a masked selection approach, we identified 85 unique VH binders to two non-overlapping epitopes within the ACE2 binding site on Spike-RBD. This enabled us to systematically link these VH domains into multivalent and bi-paratopic formats. These multivalent and bi-paratopic VH constructs showed a marked increase in affinity to Spike (up to 600-fold) and neutralization potency (up to 1400-fold) on pseudotyped SARS-CoV-2 virus when compared to the standalone VH domains. The most potent binder, a trivalent VH, neutralized authentic SARS-CoV-2 with half-minimal inhibitory concentration (IC 50 ) of 4.0 nM (180 ng/mL). A cryo-EM structure of the trivalent VH bound to Spike shows each VH domain bound an RBD at the ACE2 binding site, explaining its increased neutralization potency and confirming our original design strategy. Our results demonstrate that targeted selection and engineering campaigns using a VH-phage library can enable rapid assembly of highly avid and potent molecules towards therapeutically important protein interfaces.
Collapse
Affiliation(s)
- Colton J. Bracken
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Shion A. Lim
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Paige Solomon
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Nicholas J. Rettko
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Duy P. Nguyen
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Beth Shoshana Zha
- Department of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kaitlin Schaefer
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - James R. Byrnes
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Jie Zhou
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Irene Lui
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Jia Liu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Katarina Pance
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - QCRG Structural Biology Consortium
- Quantitative Biosciences Institute (QBI) Coronavirus Research Group Structural Biology Consortium, University of California San Francisco, San Francisco, CA 94158, USA
| | - Xin X. Zhou
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - Kevin K. Leung
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
| | - James A. Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, CA
| |
Collapse
|