1
|
Knepper LE, Ankrom ET, Thévenin D. Enhancing Anti-Cancer Immune Response by Acidosis-Sensitive Nanobody Display. J Membr Biol 2024; 257:391-401. [PMID: 39254684 PMCID: PMC11584308 DOI: 10.1007/s00232-024-00322-3] [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: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024]
Abstract
One of the main challenges with many cancer immunotherapies is that biomarkers are needed for targeting. These biomarkers are often associated with tumors but are not specific to a particular tumor and can lead to damage in healthy tissues, resistance to treatment, or the need for customization for different types of cancer due to variations in targets. A promising alternative approach is to target the acidic microenvironment found in most solid tumor types. This can be achieved using the pH (Low) Insertion Peptide (pHLIP), which inserts selectively into cell membranes under acidic conditions, sparing healthy tissues. pHLIP has shown potential for imaging, drug delivery, and surface display. For instance, we previously used pHLIP to display epitopes on the surfaces of cancer cells, enabling antibody-mediated immune cell recruitment and selective killing of cancer cells. In this study, we further explored this concept by directly fusing an anti-CD16 nanobody, which activates natural killer (NK) cells, to pHLIP, eliminating the need for antibody recruitment. Our results demonstrated the insertion of pH-sensitive agents into cancer cells, activation of the CD16 receptor on effector cells, and successful targeting and destruction of cancer cells by high-affinity CD16+ NK cells in two cancer cell lines.
Collapse
Affiliation(s)
- Leah E Knepper
- Department of Chemistry, Lehigh University, Bethlehem, PA, 18015, USA
| | - Emily T Ankrom
- Department of Chemistry, Lehigh University, Bethlehem, PA, 18015, USA
| | - Damien Thévenin
- Department of Chemistry, Lehigh University, Bethlehem, PA, 18015, USA.
| |
Collapse
|
2
|
Knepper LE, Ankrom ET, Thévenin D. Enhancing Anti-Cancer Immune Response by Acidosis-sensitive Nanobody Display. RESEARCH SQUARE 2024:rs.3.rs-4750804. [PMID: 39184093 PMCID: PMC11343302 DOI: 10.21203/rs.3.rs-4750804/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
One of the main challenges with many cancer immuno-therapies is that they depend on biomarkers for targeting. These biomarkers are often associated with tumors but are not specific to a particular tumor, which can lead to damage in healthy tissues, resistance to treatment, and the need for customization for different types of cancer due to the variations in targets. A promising alternative approach is to target the acidic microenvironment found in most solid tumor types. This can be achieved using the pH (Low) Insertion Peptide (pHLIP), which inserts selectively into cell membranes in acidic conditions, sparing healthy tissues. pHLIP has shown potential for imaging, drug delivery, and surface display. For instance, we previously used pHLIP to display epitopes on the surfaces of cancer cells, enabling antibody-mediated immune cell recruitment and selective killing of cancer cells. In this study, we further this concept by directly fusing an anti-CD16 nanobody, which activates Natural Killer (NK) cells, to pHLIP, eliminating the need for antibody recruitment. Our results demonstrate pH-sensitive insertion into cancer cells, activation of the CD16 receptor on effector cells, and successful targeting and destruction of cancer cells by high-affinity CD16 + NK cells in two cancer cell lines.
Collapse
|
3
|
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
|
4
|
Altunay B, Morgenroth A, Beheshti M, Vogg A, Wong NCL, Ting HH, Biersack HJ, Stickeler E, Mottaghy FM. HER2-directed antibodies, affibodies and nanobodies as drug-delivery vehicles in breast cancer with a specific focus on radioimmunotherapy and radioimmunoimaging. Eur J Nucl Med Mol Imaging 2021; 48:1371-1389. [PMID: 33179151 PMCID: PMC8113197 DOI: 10.1007/s00259-020-05094-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE The aim of the present paper is to review the role of HER2 antibodies, affibodies and nanobodies as vehicles for imaging and therapy approaches in breast cancer, including a detailed look at recent clinical data from antibody drug conjugates and nanobodies as well as affibodies that are currently under development. RESULTS Clinical and preclinical studies have shown that the use of monoclonal antibodies in molecular imaging is impaired by slow blood clearance, associated with slow and low tumor uptake and with limited tumor penetration potential. Antibody fragments, such as nanobodies, on the other hand, can be radiolabelled with short-lived radioisotopes and provide high-contrast images within a few hours after injection, allowing early diagnosis and reduced radiation exposure of patients. Even in therapy, the small radioactively labeled nanobodies prove to be superior to radioactively labeled monoclonal antibodies due to their higher specificity and their ability to penetrate the tumor. CONCLUSION While monoclonal antibodies are well established drug delivery vehicles, the current literature on molecular imaging supports the notion that antibody fragments, such as affibodies or nanobodies, might be superior in this approach.
Collapse
Affiliation(s)
- Betül Altunay
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | - Mohsen Beheshti
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany
- Division of Molecular PET-Imaging and Theranostics , Paracelsus Medical University , Salzburg, 5020, Austria
| | - Andreas Vogg
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | | | - Hong Hoi Ting
- Nanomab Technology Limited, Shanghai, People's Republic of China
| | | | - Elmar Stickeler
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany
- Department of Gynecology and Obstetrics, RWTH Aachen, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany.
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany.
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), 6202, Maastricht, The Netherlands.
| |
Collapse
|
5
|
Wang W, Yuan J, Jiang C. Applications of nanobodies in plant science and biotechnology. PLANT MOLECULAR BIOLOGY 2021; 105:43-53. [PMID: 33037986 PMCID: PMC7547553 DOI: 10.1007/s11103-020-01082-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/05/2020] [Indexed: 05/15/2023]
Abstract
Present review summarizes the current applications of nanobodies in plant science and biotechnology, including plant expression of nanobodies, plant biotechnological applications, nanobody-based immunodetection, and nanobody-mediated resistance against plant pathogens. Nanobodies (Nbs) are variable domains of heavy chain-only antibodies (HCAbs) isolated from camelids. In spite of their single domain structure, nanobodies display many unique features, such as small size, high stability, and cryptic epitopes accessibility, which make them ideal for sophisticated applications in plants and animals. In this review, we summarize the current applications of nanobodies in plant science and biotechnology, focusing on nanobody expression in plants, plant biotechnological applications, determination of plant toxins and pathogens, and nanobody-mediated resistance against plant pathogens. Prospects and challenges of nanobody applications in plants are also discussed.
Collapse
Affiliation(s)
- Wenyi Wang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China.
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China.
| | - Jumao Yuan
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Changan Jiang
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
6
|
Pourjafar M, Samadi P, Saidijam M. MUC1 antibody-based therapeutics: the promise of cancer immunotherapy. Immunotherapy 2020; 12:1269-1286. [PMID: 33019839 DOI: 10.2217/imt-2020-0019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 09/15/2020] [Indexed: 01/06/2023] Open
Abstract
Antibody-based targeted therapies have been able to target cancers with enhanced specificity and high efficacy. In this regard, identifying cancer markers (antigens) that are only present (tumor-specific antigens) or have an increased expression (tumor-associated antigen) on the surface of cancer cells is a crucial step for targeted cancer treatment. Various cancer antigens have already been used for therapeutic and diagnostic purposes. MUC1 is one of the most important tumor markers with high levels of expression in various solid tumors which makes it as a potential target for antibody-based therapies. This review discusses preclinical and clinical results from various platforms based on monoclonal antibodies, nanobodies as well as bispecific antibodies against MUC1. We also highlight unmet challenges that must be overcome to generate more effective cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Mona Pourjafar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pouria Samadi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
7
|
Modarresi M, Javaran MJ, Shams-bakhsh M, Zeinali S, Behdani M, Mirzaee M. Transient expression of anti-VEFGR2 nanobody in Nicotiana tabacum and N. benthamiana. 3 Biotech 2018; 8:484. [PMID: 30467531 DOI: 10.1007/s13205-018-1500-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 11/01/2018] [Indexed: 12/31/2022] Open
Abstract
In human, the interaction between vascular endothelial growth factor (VEGF) and its receptor (VEGFR2) is critical for tumor angiogenesis. This is a vital process for cancer tumor growth and metastasis. Blocking VEGF/VEGFR2 conjugation by antibodies inhibits the neovascularization and tumor metastasis. This investigation designed to use a transient expression platform for production of recombinant anti-VEGFR2 nanobody in tobacco plants. At first, anti-VEGFR2-specific nanobody gene was cloned in a Turnip mosaic virus (TuMV)-based vector, and then, it was expressed in Nicotiana benthamiana and Nicotiana tabacum cv. Xanthi transiently. The expression of nanobody in tobacco plants were confirmed by reverse transcription-polymerase chain reaction (RT-PCR), dot blot, enzyme-linked immunosorbent assays (ELISA), and Western blot analysis. It was shown that tobacco plants could accumulate nanobody up to level 0.45% of total soluble protein (8.3 µg/100 mg of fresh leaf). This is the first report of the successful expression of the camelied anti-VEFGR2 nanobody gene in tobacco plants using a plant viral vector. This system provides a fast solution for production of pharmaceutical and commercial proteins such as anti-cancer nanobodies in tobacco plants.
Collapse
|
8
|
Nanobodies as novel therapeutic agents in envenomation. Biochim Biophys Acta Gen Subj 2018; 1862:2955-2965. [PMID: 30309831 DOI: 10.1016/j.bbagen.2018.08.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/27/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND An effective therapy against envenoming should be a priority in view of the high number scorpion stings and snakebites. Serum therapy is still widely applied to treat the envenomation victims; however this approach suffers from several shortcomings. The employment of monoclonal antibodies might be an outcome as these molecules are at the core of a variety of applications from protein structure determination to cancer treatment. The progress of activities in the twilight zone between genetic and antibody engineering have led to the development of a unique class of antibody fragments. These molecules possess several benefits and lack many possible disadvantages over classical antibodies. Within recombinant antibody formats, nanobodies or single domain antigen binding fragments derived from heavy chain only antibodies in camelids occupy a privileged position. SCOPE OF REVIEW In this paper we will briefly review the common methods of envenomation treatment and focus on details of various in vivo research activities that investigate the performance of recombinant, monoclonal nanobodies in venom neutralization. MAJOR CONCLUSIONS Nanobodies bind to their cognate target with high specificity and affinity, they can be produced in large quantities from microbial expression systems and are very robust even when challenged with harsh environmental conditions. Upon administering, they rapidly distribute throughout the body and seem to be well tolerated in humans posing low immunogenicity. GENERAL SIGNIFICANCE Scorpion and snake envenomation is a major issue in developing countries and nanobodies as a venom-neutralizing agent can be considered as a valuable and promising candidate in envenomation therapy.
Collapse
|
9
|
Tokuhara D. Challenges in developing mucosal vaccines and antibodies against infectious diarrhea in children. Pediatr Int 2018; 60:214-223. [PMID: 29290097 DOI: 10.1111/ped.13497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/14/2017] [Accepted: 12/26/2017] [Indexed: 12/24/2022]
Abstract
Infectious diarrhea in children can be life-threatening and imposes a large economic burden on healthcare systems, therefore more effective prophylactic and therapeutic drugs are needed urgently. Because most of the pathogens responsible for childhood diarrhea infect the gastrointestinal mucosa, providing protective immunity at the mucosal surface is an ideal way to control pathogen invasion and toxic activity. Mucosal (e.g. oral, nasal) vaccines are superior to systemic (subcutaneous or intramuscular) vaccination for conferring both mucosal and systemic pathogen-specific immune responses. Therefore, great efforts has been focused on the development of cost-effective mucosal vaccines for the past 50 years. Recent progress in plant genetic engineering has revolutionized the production of inexpensive and safe recombinant vaccine antigens. For example, rice plant biotechnology has facilitated the development of a cold-chain-free rice-based oral subunit vaccine against Vibrio cholerae. Furthermore, this technology has led to the creation of a rice-based oral antibody for prophylaxis and treatment of rotavirus gastroenteritis. This review summarizes current perspectives regarding the mucosal immune system and the development of mucosal vaccines and therapeutic antibodies, particularly rice-based products, and discusses future prospects regarding mucosal vaccines for children.
Collapse
Affiliation(s)
- Daisuke Tokuhara
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Abenoku, Osaka, Japan
| |
Collapse
|
10
|
Li Y, Zhou C, Li J, Liu J, Lin L, Li L, Cao D, Li Q, Wang Z. Single domain based bispecific antibody, Muc1-Bi-1, and its humanized form, Muc1-Bi-2, induce potent cancer cell killing in muc1 positive tumor cells. PLoS One 2018; 13:e0191024. [PMID: 29357376 PMCID: PMC5777659 DOI: 10.1371/journal.pone.0191024] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 12/26/2017] [Indexed: 12/31/2022] Open
Abstract
Muc1 is one of the most studied tumor antigens. However, antibodies or antibody-toxin conjugates against Muc1 have not shown significant efficacy for tumors with Muc1 overexpression. In this study, we employed bispecific antibody approach to target Muc1 positive tumor cells. A novel bispecific antibody, Muc1-Bi-1, was constructed by linking single domain antibodies, anti-Muc1-VHH and anti-CD16-VHH. Muc1-Bi-2, the humanized form of Muc1-Bi-1, was also constructed by grafting. Both Muc1-Bi bispecific antibodies can be efficiently expressed and purified from bacteria. In vitro, the Muc1-Bi bispecific antibodies can recruit Natural Killer (NK) cells to drive potent and specific cell killing of Muc1-overexpressing tumor cells. In xenograft model, the Muc1-Bi bispecific antibodies can suppress tumor growth in the presence of human peripheral blood mononuclear cells (PBMC). These data suggested that the single domain based Muc1-Bi may provide a valid strategy for targeting tumors with Muc1 overexpression.
Collapse
Affiliation(s)
- Yumei Li
- School of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Guangzhou, China
- Center for Cellular & Structural Biology, Sun Yat-sen University, Guangzhou, China
| | - Changhua Zhou
- School of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Guangzhou, China
- Center for Cellular & Structural Biology, Sun Yat-sen University, Guangzhou, China
| | - Jing Li
- School of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Guangzhou, China
- Center for Cellular & Structural Biology, Sun Yat-sen University, Guangzhou, China
| | - Jiayu Liu
- School of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Guangzhou, China
- Center for Cellular & Structural Biology, Sun Yat-sen University, Guangzhou, China
| | - Limin Lin
- School of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Guangzhou, China
- Center for Cellular & Structural Biology, Sun Yat-sen University, Guangzhou, China
| | - Li Li
- School of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Guangzhou, China
- Center for Cellular & Structural Biology, Sun Yat-sen University, Guangzhou, China
| | - Donglin Cao
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Qing Li
- School of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Guangzhou, China
- Center for Cellular & Structural Biology, Sun Yat-sen University, Guangzhou, China
- * E-mail: (QL); (ZW)
| | - Zhong Wang
- School of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Guangzhou, China
- Center for Cellular & Structural Biology, Sun Yat-sen University, Guangzhou, China
- * E-mail: (QL); (ZW)
| |
Collapse
|
11
|
Liu Y, Huang H. Expression of single-domain antibody in different systems. Appl Microbiol Biotechnol 2017; 102:539-551. [DOI: 10.1007/s00253-017-8644-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/09/2017] [Accepted: 11/12/2017] [Indexed: 10/18/2022]
|
12
|
Hu Y, Liu C, Muyldermans S. Nanobody-Based Delivery Systems for Diagnosis and Targeted Tumor Therapy. Front Immunol 2017; 8:1442. [PMID: 29163515 PMCID: PMC5673844 DOI: 10.3389/fimmu.2017.01442] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 10/17/2017] [Indexed: 01/01/2023] Open
Abstract
The development of innovative targeted therapeutic approaches are expected to surpass the efficacy of current forms of treatments and cause less damage to healthy cells surrounding the tumor site. Since the first development of targeting agents from hybridoma’s, monoclonal antibodies (mAbs) have been employed to inhibit tumor growth and proliferation directly or to deliver effector molecules to tumor cells. However, the full potential of such a delivery strategy is hampered by the size of mAbs, which will obstruct the targeted delivery system to access the tumor tissue. By serendipity, a new kind of functional homodimeric antibody format was discovered in camelidae, known as heavy-chain antibodies (HCAbs). The cloning of the variable domain of HCAbs produces an attractive minimal-sized alternative for mAbs, referred to as VHH or nanobodies (Nbs). Apart from their dimensions in the single digit nanometer range, the unique characteristics of Nbs combine a high stability and solubility, low immunogenicity and excellent affinity and specificity against all possible targets including tumor markers. This stimulated the development of tumor-targeted therapeutic strategies. Some autonomous Nbs have been shown to act as antagonistic drugs, but more importantly, the targeting capacity of Nbs has been exploited to create drug delivery systems. Obviously, Nb-based targeted cancer therapy is mainly focused toward extracellular tumor markers, since the membrane barrier prevents antibodies to reach the most promising intracellular tumor markers. Potential strategies, such as lentiviral vectors and bacterial type 3 secretion system, are proposed to deliver target-specific Nbs into tumor cells and to block tumor markers intracellularly. Simultaneously, Nbs have also been employed for in vivo molecular imaging to diagnose diseased tissues and to monitor the treatment effects. Here, we review the state of the art and focus on recent developments with Nbs as targeting moieties for drug delivery systems in cancer therapy and cancer imaging.
Collapse
Affiliation(s)
- Yaozhong Hu
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.,State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Changxiao Liu
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
13
|
Abstract
Ligand-induced activation of G protein-coupled receptors (GPCRs) is a key mechanism permitting communication between cells and organs. Enormous progress has recently elucidated the structural and dynamic features of GPCR transmembrane signaling. Nanobodies, the recombinant antigen-binding fragments of camelid heavy-chain-only antibodies, have emerged as important research tools to lock GPCRs in particular conformational states. Active-state stabilizing nanobodies have elucidated several agonist-bound structures of hormone-activated GPCRs and have provided insight into the dynamic character of receptors. Nanobodies have also been used to stabilize transient GPCR transmembrane signaling complexes, yielding the first structural insights into GPCR signal transduction across the cellular membrane. Beyond their in vitro uses, nanobodies have served as conformational biosensors in living systems and have provided novel ways to modulate GPCR function. Here, we highlight several examples of how nanobodies have enabled the study of GPCR function and give insights into potential future uses of these important tools.
Collapse
Affiliation(s)
- Aashish Manglik
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305; ,
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305; ,
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium;
- VIB Structural Biology Research Center, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| |
Collapse
|
14
|
Maffey L, Vega CG, Miño S, Garaicoechea L, Parreño V. Anti-VP6 VHH: An Experimental Treatment for Rotavirus A-Associated Disease. PLoS One 2016; 11:e0162351. [PMID: 27603013 PMCID: PMC5014449 DOI: 10.1371/journal.pone.0162351] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 08/22/2016] [Indexed: 01/10/2023] Open
Abstract
Species A Rotaviruses (RVA) remain a leading cause of mortality in children under 5 years of age. Current treatment options are limited. We assessed the efficacy of two VP6-specific llama-derived heavy chain antibody fragments (VHH) -2KD1 and 3B2- as an oral prophylactic and therapeutic treatment against RVA-induced diarrhea in a neonatal mouse model inoculated with virulent murine RVA (ECw, G16P[16]I7). Joint therapeutic administration of 2KD1+3B2 (200 μg/dose) successfully reduced diarrhea duration, RVA infection severity and virus shedding in feces. While the same dose of 2KD1 or 3B2 (200 μg) significantly reduced duration of RVA-induced diarrhea, 2KD1 was more effective in diminishing the severity of intestinal infection and RVA shedding in feces, perhaps because 2KD1 presented higher binding affinity for RVA particles than 3B2. Neither prophylactic nor therapeutic administration of the VHH interfered with the host's humoral immune response against RVA. When 2KD1 (200 μg) was administered after diarrhea development, it also significantly reduced RVA intestinal infection and fecal shedding. Host antibody responses against the oral VHH treatment were not detected, nor did viral escape mutants. Our findings show that oral administration of anti-VP6 VHH constitute, not only an effective prophylactic treatment against RVA-associated diarrhea, but also a safe therapeutic tool against RVA infection, even once diarrhea is present. Anti-VP6 VHH could be used complementary to ongoing vaccination, especially in populations that have shown lower immunization efficacy. These VHH could also be scaled-up to develop pediatric medication or functional food like infant milk formulas that might help treat RVA diarrhea.
Collapse
Affiliation(s)
- Lucía Maffey
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA Castelar, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas, CONICET, Buenos Aires, Argentina
| | - Celina G. Vega
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA Castelar, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas, CONICET, Buenos Aires, Argentina
| | - Samuel Miño
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA Castelar, Buenos Aires, Argentina
| | - Lorena Garaicoechea
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA Castelar, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas, CONICET, Buenos Aires, Argentina
| | - Viviana Parreño
- Instituto de Virología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, INTA Castelar, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas, CONICET, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
15
|
Characterization of single-domain antibodies against Foot and Mouth Disease Virus (FMDV) serotype O from a camelid and imaging of FMDV in baby hamster kidney-21 cells with single-domain antibody-quantum dots probes. BMC Vet Res 2015; 11:120. [PMID: 26001568 PMCID: PMC4489003 DOI: 10.1186/s12917-015-0437-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 05/12/2015] [Indexed: 01/17/2023] Open
Abstract
Background Foot-and-mouth disease (FMD) is a highly contagious disease that affects cloven-hoofed animals and causes significant economic losses to husbandry worldwide. The variable domain of heavy-chain antibodies (VHHs or single domain antibodies, sdAbs) are single-domain antigen-binding fragments derived from camelid heavy-chain antibodies. Results In this work, two sdAbs against FMD virus (FMDV) serotype O were selected from a camelid phage display immune library and expressed in Escherichia coli. The serotype specificity and affinity of the sdAbs were identified through enzyme-linked immunosorbent assay and surface plasmon resonance assay. Moreover, the sdAbs were conjugated with quantum dots to constitute probes for imaging FMD virions. Results demonstrated that the two sdAbs were specific for serotype O and shared no cross-reactivity with serotypes A and Asia 1. The equilibrium dissociation constant (KD) values of the two sdAbs ranged from 6.23 nM to 8.24 nM, which indicated high affinity to FMDV antigens. Co-localization with the sdAb-AF488 and sdAb-QD probes indicated the same location of FMDV virions in baby hamster kidney-21 (BHK-21) cells. Conclusions sdAb-QD probes are powerful tools to detect and image FMDV in BHK-21 cells. Electronic supplementary material The online version of this article (doi:10.1186/s12917-015-0437-2) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Wang J, Majkova Z, Bever CRS, Yang J, Gee SJ, Li J, Xu T, Hammock BD. One-step immunoassay for tetrabromobisphenol a using a camelid single domain antibody-alkaline phosphatase fusion protein. Anal Chem 2015; 87:4741-8. [PMID: 25849972 DOI: 10.1021/ac504735p] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Tetrabromobisphenol A (TBBPA) is a ubiquitous brominated flame retardant, showing widespread environmental and human exposures. A variable domain of the heavy chain antibody (VHH), naturally occurring in camelids, approaches the lower size limit of functional antigen-binding entities. The ease of genetic manipulation makes such VHHs a superior choice to use as an immunoreagent. In this study, a highly selective anti-TBBPA VHH T3-15 fused with alkaline phosphatase (AP) from E. coli was expressed, showing both an integrated TBBPA-binding capacity and enzymatic activity. A one-step immunoassay based on the fusion protein T3-15-AP was developed for TBBPA in 5% dimethyl sulfoxide (DMSO)/phosphate buffered saline (PBS, pH 7.4), with a half-maximum signal inhibition concentration (IC50) of 0.20 ng mL(-1). Compared to the parental VHH T3-15, T3-15-AP was able to bind to a wider variety of coating antigens and the assay sensitivity was slightly improved. Cross-reactivity of T3-15-AP with a set of important brominated analogues was negligible (<0.1%). Although T3-15-AP was susceptible to extreme heat (90 °C), much higher binding stability at ambient temperature was observed in the T3-15-AP-based assay for at least 70 days. A simple pretreatment method of diluting urine samples with DMSO was developed for a one-step assay. The recoveries of TBBPA from urine samples via this one-step assay ranged from 96.7% to 109.9% and correlated well with a high-performance liquid chromatography-tandem mass spectroscopy (HPLC-MS/MS) method. It is expected that the dimerized fusion protein, VHH-AP, will show promising applications in human exposure and environmental monitoring.
Collapse
Affiliation(s)
- Jia Wang
- †Beijing Key Laboratory of Biodiversity and Organic Farming, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, China.,§College of Food Science and Technology, Huazhong Agricultural Universiy, Wuhan 430070, China
| | - Zuzana Majkova
- ‡Department of Entomology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Candace R S Bever
- ‡Department of Entomology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Jun Yang
- ‡Department of Entomology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Shirley J Gee
- ‡Department of Entomology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Ji Li
- †Beijing Key Laboratory of Biodiversity and Organic Farming, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, China
| | - Ting Xu
- †Beijing Key Laboratory of Biodiversity and Organic Farming, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, China
| | - Bruce D Hammock
- ‡Department of Entomology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| |
Collapse
|
17
|
Ko K. Expression of recombinant vaccines and antibodies in plants. Monoclon Antib Immunodiagn Immunother 2015; 33:192-8. [PMID: 24937251 DOI: 10.1089/mab.2014.0049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Plants are able to perform post-translational maturations of therapeutic proteins required for their functional biological activity and suitable in vivo pharmacokinetics. Plants can be a low-cost, large-scale production platform of recombinant biopharmaceutical proteins such as vaccines and antibodies. Plants, however, lack mechanisms of processing authentic human N-glycosylation, which imposes a major limitation in their use as an expression system for therapeutic glycoproducts. Efforts have been made to circumvent plant-specific N-glycosylation, as well as to supplement the plant's endogenous system with human glycosyltransferases for non-immunogenic and humanized N-glycan production. Herein we review studies on the potential of plants to serve as production systems for therapeutic and prophylactic biopharmaceuticals. We have especially focused on recombinant vaccines and antibodies and new expression strategies to overcome the existing problems associated with their production in plants.
Collapse
Affiliation(s)
- Kisung Ko
- Department of Medicine, Therapeutic Protein Engineering Lab, College of Medicine, Chung-Ang University , Seoul, Korea
| |
Collapse
|
18
|
Single-domain antibodies targeting neuraminidase protect against an H5N1 influenza virus challenge. J Virol 2014; 88:8278-96. [PMID: 24829341 DOI: 10.1128/jvi.03178-13] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Influenza virus neuraminidase (NA) is an interesting target of small-molecule antiviral drugs. We isolated a set of H5N1 NA-specific single-domain antibodies (N1-VHHm) and evaluated their in vitro and in vivo antiviral potential. Two of them inhibited the NA activity and in vitro replication of clade 1 and 2 H5N1 viruses. We then generated bivalent derivatives of N1-VHHm by two methods. First, we made N1-VHHb by genetically joining two N1-VHHm moieties with a flexible linker. Second, bivalent N1-VHH-Fc proteins were obtained by genetic fusion of the N1-VHHm moiety with the crystallizable region of mouse IgG2a (Fc). The in vitro antiviral potency against H5N1 of both bivalent N1-VHHb formats was 30- to 240-fold higher than that of their monovalent counterparts, with 50% inhibitory concentrations in the low nanomolar range. Moreover, single-dose prophylactic treatment with bivalent N1-VHHb or N1-VHH-Fc protected BALB/c mice against a lethal challenge with H5N1 virus, including an oseltamivir-resistant H5N1 variant. Surprisingly, an N1-VHH-Fc fusion without in vitro NA-inhibitory or antiviral activity also protected mice against an H5N1 challenge. Virus escape selection experiments indicated that one amino acid residue close to the catalytic site is required for N1-VHHm binding. We conclude that single-domain antibodies directed against influenza virus NA protect against H5N1 virus infection, and when engineered with a conventional Fc domain, they can do so in the absence of detectable NA-inhibitory activity. IMPORTANCE Highly pathogenic H5N1 viruses are a zoonotic threat. Outbreaks of avian influenza caused by these viruses occur in many parts of the world and are associated with tremendous economic loss, and these viruses can cause very severe disease in humans. In such cases, small-molecule inhibitors of the viral NA are among the few treatment options for patients. However, treatment with such drugs often results in the emergence of resistant viruses. Here we show that single-domain antibody fragments that are specific for NA can bind and inhibit H5N1 viruses in vitro and can protect laboratory mice against a challenge with an H5N1 virus, including an oseltamivir-resistant virus. In addition, plant-produced VHH fused to a conventional Fc domain can protect in vivo even in the absence of NA-inhibitory activity. Thus, NA of influenza virus can be effectively targeted by single-domain antibody fragments, which are amenable to further engineering.
Collapse
|
19
|
De Meyer T, Muyldermans S, Depicker A. Nanobody-based products as research and diagnostic tools. Trends Biotechnol 2014; 32:263-70. [PMID: 24698358 DOI: 10.1016/j.tibtech.2014.03.001] [Citation(s) in RCA: 320] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/17/2014] [Accepted: 03/05/2014] [Indexed: 01/25/2023]
Abstract
Since the serendipitous discovery 20 years ago of bona fide camelid heavy-chain antibodies, their single-domain antigen-binding fragments, known as VHHs or nanobodies, have received a progressively growing interest. As a result of the beneficial properties of these stable recombinant entities, they are currently highly valued proteins for multiple applications, including fundamental research, diagnostics, and therapeutics. Today, with the original patents expiring, even more academic and industrial groups are expected to explore innovative VHH applications. Here, we provide a thorough overview of novel implementations of VHHs as research and diagnostic tools, and of the recently evaluated production platforms for several VHHs and VHH-derived antibody formats.
Collapse
Affiliation(s)
- Thomas De Meyer
- Department of Plant Systems Biology, VIB, 9052 Gent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Gent, Belgium
| | - Serge Muyldermans
- Structural Biology Research Center, VIB, 1050 Brussel, Belgium; Research Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussel, Belgium
| | - Ann Depicker
- Department of Plant Systems Biology, VIB, 9052 Gent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Gent, Belgium.
| |
Collapse
|
20
|
Baral TN, MacKenzie R, Arbabi Ghahroudi M. Single-domain antibodies and their utility. ACTA ACUST UNITED AC 2013; 103:2.17.1-2.17.57. [PMID: 24510545 DOI: 10.1002/0471142735.im0217s103] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Engineered monoclonal antibody fragments have gained market attention due to their versatility and tailor-made potential and are now considered to be an important part of future immunobiotherapeutics. Single-domain antibodies (sdAbs), also known as nanobodies, are derived from VHHs [variable domains (V) of heavy-chain-only antibodies (HCAb)] of camelid heavy-chain antibodies. These nature-made sdAbs are well suited for various applications due to their favorable characteristics such as small size, ease of genetic manipulation, high affinity and solubility, overall stability, resistance to harsh conditions (e.g., low pH, high temperature), and low immunogenicity. Most importantly, sdAbs have the feature of penetrating into cavities and recognizing hidden epitopes normally inaccessible to conventional antibodies, mainly due to their protruding CDR3/H3 loops. In this unit, we will present and discuss comprehensive and step-by-step protocols routinely practiced in our laboratory for isolating sdAbs from immunized llamas (or other members of the Camelidae family) against target antigens using phage-display technology. Expression, purification, and characterization of the isolated sdAbs will then be described, followed by presentation of several examples of applications of sdAbs previously characterized in our laboratory and elsewhere.
Collapse
Affiliation(s)
- Toya Nath Baral
- Human Health Therapeutics, Life Sciences Division, National Research Council Canada, Ottawa, Ontario, Canada
| | - Roger MacKenzie
- Human Health Therapeutics, Life Sciences Division, National Research Council Canada, Ottawa, Ontario, Canada.,University of Guelph, Guelph, Ontario, Canada
| | - Mehdi Arbabi Ghahroudi
- Human Health Therapeutics, Life Sciences Division, National Research Council Canada, Ottawa, Ontario, Canada.,University of Guelph, Guelph, Ontario, Canada.,Department of Biology, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
21
|
De Buck S, Nolf J, De Meyer T, Virdi V, De Wilde K, Van Lerberge E, Van Droogenbroeck B, Depicker A. Fusion of an Fc chain to a VHH boosts the accumulation levels in Arabidopsis seeds. PLANT BIOTECHNOLOGY JOURNAL 2013; 11:1006-16. [PMID: 23915060 DOI: 10.1111/pbi.12094] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 05/18/2023]
Abstract
Nanobodies® (VHHs) provide powerful tools in therapeutic and biotechnological applications. Nevertheless, for some applications, bivalent antibodies perform much better, and for this, an Fc chain can be fused to the VHH domain, resulting in a bivalent homodimeric VHH-Fc complex. However, the production of bivalent antibodies in Escherichia coli is rather inefficient. Therefore, we compared the production of VHH7 and VHH7-Fc as antibodies of interest in Arabidopsis seeds for detecting prostate-specific antigen (PSA), a well-known biomarker for prostate cancer in the early stages of tumour development. The influence of the signal sequence (camel versus plant) and that of the Fc chain origin (human, mouse or pig) were evaluated. The accumulation levels of VHHs were very low, with a maximum of 0.13% VHH of total soluble protein (TSP) in homozygous T3 seeds, while VHH-Fc accumulation levels were at least 10- to 100-fold higher, with a maximum of 16.25% VHH-Fc of TSP. Both the camel and plant signal peptides were efficiently cleaved off and did not affect the accumulation levels. However, the Fc chain origin strongly affected the degree of proteolysis, but only had a slight influence on the accumulation level. Analysis of the mRNA levels suggested that the low amount of VHHs produced in Arabidopsis seeds was not due to a failure in transcription, but rather to translation inefficiency, protein instability and/or degradation. Most importantly, the plant-produced VHH7 and VHH7-Fc antibodies were functional in detecting PSA and could thus be used for diagnostic applications.
Collapse
Affiliation(s)
- Sylvie De Buck
- Department of Plant Systems Biology, VIB, Gent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Gent, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Tokuhara D, Álvarez B, Mejima M, Hiroiwa T, Takahashi Y, Kurokawa S, Kuroda M, Oyama M, Kozuka-Hata H, Nochi T, Sagara H, Aladin F, Marcotte H, Frenken LGJ, Iturriza-Gómara M, Kiyono H, Hammarström L, Yuki Y. Rice-based oral antibody fragment prophylaxis and therapy against rotavirus infection. J Clin Invest 2013; 123:3829-38. [PMID: 23925294 DOI: 10.1172/jci70266] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 06/13/2013] [Indexed: 12/30/2022] Open
Abstract
Rotavirus-induced diarrhea is a life-threatening disease in immunocompromised individuals and in children in developing countries. We have developed a system for prophylaxis and therapy against rotavirus disease using transgenic rice expressing the neutralizing variable domain of a rotavirus-specific llama heavy-chain antibody fragment (MucoRice-ARP1). MucoRice-ARP1 was produced at high levels in rice seeds using an overexpression system and RNAi technology to suppress the production of major rice endogenous storage proteins. Orally administered MucoRice-ARP1 markedly decreased the viral load in immunocompetent and immunodeficient mice. The antibody retained in vitro neutralizing activity after long-term storage (>1 yr) and boiling and conferred protection in mice even after heat treatment at 94°C for 30 minutes. High-yield, water-soluble, and purification-free MucoRice-ARP1 thus forms the basis for orally administered prophylaxis and therapy against rotavirus infections.
Collapse
Affiliation(s)
- Daisuke Tokuhara
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Sera of camelids contain both conventional heterotetrameric antibodies and unique functional heavy (H)-chain antibodies (HCAbs). The H chain of these homodimeric antibodies consists of one antigen-binding domain, the VHH, and two constant domains. HCAbs fail to incorporate light (L) chains owing to the deletion of the first constant domain and a reshaped surface at the VHH side, which normally associates with L chains in conventional antibodies. The genetic elements composing HCAbs have been identified, but the in vivo generation of these antibodies from their dedicated genes into antigen-specific and affinity-matured bona fide antibodies remains largely underinvestigated. However, the facile identification of antigen-specific VHHs and their beneficial biochemical and economic properties (size, affinity, specificity, stability, production cost) supported by multiple crystal structures have encouraged antibody engineering of these single-domain antibodies for use as a research tool and in biotechnology and medicine.
Collapse
Affiliation(s)
- Serge Muyldermans
- Research Group Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium.
| |
Collapse
|
24
|
Cui L, Peng H, Zhang R, Chen Y, Zhao L, Tang K. Recombinant hHscFv-RC-RNase protein derived from transgenic tobacco acts as a bifunctional molecular complex against hepatocellular carcinoma. Biotechnol Appl Biochem 2012; 59:323-9. [PMID: 23586908 DOI: 10.1002/bab.1039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 08/09/2012] [Indexed: 01/03/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common clinical primary malignant tumor; however, efficient drugs for the treatment of HCC are still lacking at the present time. To develop a new approach for liver cancer therapy, we designed a chimeric gene (his-HR) encoding a single-chain variable fragment of human HAb25 (hHscFv) fused to a cytotoxic ribonuclease from Rana catesbeiana (RC-RNase) and expressed the corresponding fusion protein in transgenic tobacco (Nicotiana tabacum). Eleven positive transgenic plant lines were identified from 204 regenerated tobacco plants by PCR and Southern blot analysis, and the immunocompetence of the recombinant his-HR protein was confirmed by Western blotting. The expression levels of his-HR protein ranged from 0.75 to 1.99 µg/g in the fresh tobacco leaves. To characterize the bifunction of the expressed his-HR protein in tobacco, binding specificity and cell toxicity to several cell lines were examined by the indirect immunocytochemical streptavidin-biotin complex method and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide assay. Data indicated that the his-HR protein had stronger specific binding affinity to HepG2 (human liver HCC cell line) than to the other tumor cell lines and normal liver cell line, and the capacity to kill the HCC cell lines SMMC7721 and HepG2 with an half maximal inhibiting concentration of 2.0 and 2.4 nM, respectively. The results suggest that recombinant bifunctional his-HR protein derived from transgenic plants may provide a novel strategy to treat HCC in the future.
Collapse
Affiliation(s)
- Lijie Cui
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
25
|
Rahbarizadeh F, Ahmadvand D, Sharifzadeh Z. Nanobody; an old concept and new vehicle for immunotargeting. Immunol Invest 2011; 40:299-338. [PMID: 21244216 DOI: 10.3109/08820139.2010.542228] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The use of antibodies in cancer therapy has come a long way since the day Paul Ehrlich described the concept and Kohler and Milstein devised the hybridoma technology to bring this theory to reality. The synthesis of murine monoclonal antibodies (mAbs) was the first success in this field, leading to the invention of chimerization, the production of variable fragments (Fv) with the progression to domain antibodies (dAb) and later humanization technologies to maximize the clinical utility of murine mAbs. It was just by chance that dAbs were found to exist in ?heavy chain? immunoglobulins from Camelidae family and cartilaginous fish. These unique antibody fragments interact with antigen by virtue of only one single variable domain, referred to as VHH or nanobody. Several characteristics make nanobody use superior to the abovementioned antibodies. They are non-immunogenic and show high thermal and chemical stability. There are several reports of raising specific nanobodies against enzymes, haptens, pathogens, toxins and tumor markers, which are outlined in this paper. All these characteristics make them strong candidates as targeting agents for cancer therapy.
Collapse
Affiliation(s)
- Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | | | | |
Collapse
|
26
|
Conrad U, Plagmann I, Malchow S, Sack M, Floss DM, Kruglov AA, Nedospasov SA, Rose-John S, Scheller J. ELPylated anti-human TNF therapeutic single-domain antibodies for prevention of lethal septic shock. PLANT BIOTECHNOLOGY JOURNAL 2011; 9:22-31. [PMID: 20444206 DOI: 10.1111/j.1467-7652.2010.00523.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Tumour necrosis factor (TNF) is a major pro-inflammatory cytokine involved in multiple inflammatory diseases. The detrimental activity of TNF can be blocked by various antagonists, and commercial therapeutics based upon this principle have been approved for treatment of diseases including rheumatoid arthritis, Crohn's disease and psoriasis. In a search for new, improved anti-inflammatory therapeutics we have designed a single-domain monoclonal antibody (V(H) H), which recognizes TNF. The antibody component (TNF-V(H) H) is based upon an anti-human TNF Camelidae heavy-chain monoclonal antibody, which was linked to an elastin-like polypeptide (ELP). We demonstrate that ELP fusion to the TNF-V(H) H enhances accumulation of the fusion protein during biomanufacturing in transgenic tobacco plants. With this study, we show for the first time that this plant-derived anti-human TNF-V(H) H antibody was biologically active in vivo. Therefore, therapeutic application of TNF-V(H) H-ELP fusion protein was tested in humanized TNF mice and was shown to be effective in preventing death caused by septic shock. The in vivo persistence of the ELPylated antibody was ∼24 fold longer than that of non-ELPylated TNF-V(H) H.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Disease Models, Animal
- Elastin
- Escherichia coli
- Galactose
- Gene Expression
- Humans
- L Cells
- Lipopolysaccharides
- Mice
- Peptides
- Plants, Genetically Modified
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/isolation & purification
- Recombinant Fusion Proteins/therapeutic use
- Shock, Septic/chemically induced
- Shock, Septic/immunology
- Shock, Septic/prevention & control
- Nicotiana/genetics
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- Udo Conrad
- Institute of Plant Genetics and Crop Plant Research (IPK), Phytoantibodies, Gatersleben, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Obembe OO, Popoola JO, Leelavathi S, Reddy SV. Advances in plant molecular farming. Biotechnol Adv 2010; 29:210-22. [PMID: 21115109 DOI: 10.1016/j.biotechadv.2010.11.004] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 11/12/2010] [Accepted: 11/12/2010] [Indexed: 01/01/2023]
Abstract
Plant molecular farming (PMF) is a new branch of plant biotechnology, where plants are engineered to produce recombinant pharmaceutical and industrial proteins in large quantities. As an emerging subdivision of the biopharmaceutical industry, PMF is still trying to gain comparable social acceptance as the already established production systems that produce these high valued proteins in microbial, yeast, or mammalian expression systems. This article reviews the various cost-effective technologies and strategies, which are being developed to improve yield and quality of the plant-derived pharmaceuticals, thereby making plant-based production system suitable alternatives to the existing systems. It also attempts to overview the different novel plant-derived pharmaceuticals and non-pharmaceutical protein products that are at various stages of clinical development or commercialization. It then discusses the biosafety and regulatory issues, which are crucial (if strictly adhered to) to eliminating potential health and environmental risks, which in turn is necessary to earning favorable public perception, thus ensuring the success of the industry.
Collapse
Affiliation(s)
- Olawole O Obembe
- Department of Biological Sciences, Covenant University, PMB 1023 Ota, Ogun State, Nigeria.
| | | | | | | |
Collapse
|
28
|
Abstract
The overexpression and aberrant glycosylation of MUC1 is associated with a wide variety of cancers, making it an ideal target for immunotherapeutic strategies. This review highlights the main avenues of research in this field, focusing on adenocarcinomas, from the preclinical to clinical; the problems and possible solutions associated with each approach; and speculates on the direction of MUC1 immunotherapeutic research over the next 5-10 years.
Collapse
Affiliation(s)
- Richard E Beatson
- Breast Cancer Biology Group, King's College London, Guy's Hospital, London SE1 9RT, UK
| | | | | |
Collapse
|
29
|
Teh YHA, Kavanagh TA. High-level expression of Camelid nanobodies in Nicotiana benthamiana. Transgenic Res 2010; 19:575-86. [PMID: 19862637 DOI: 10.1007/s11248-009-9338-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Accepted: 10/13/2009] [Indexed: 11/30/2022]
Abstract
Nanobodies (or VHHs) are single-domain antigen-binding fragments derived from Camelid heavy chain-only antibodies. Their small size, monomeric behaviour, high stability and solubility, and ability to bind epitopes not accessible to conventional antibodies make them especially suitable for many therapeutic and biotechnological applications. Here we describe high-level expression, in Nicotiana benthamiana, of three versions of an anti-hen egg white lysozyme (HEWL) nanobody which include the original VHH from an immunized library (cAbLys3), a codon-optimized derivative, and a codon-optimized hybrid nanobody comprising the CDRs of cAbLys3 grafted onto an alternative 'universal' nanobody framework. His6- and StrepII-tagged derivatives of each nanobody were targeted for accumulation in the cytoplasm, chloroplast and apoplast using different pre-sequences. When targeted to the apoplast, intact functional nanobodies accumulated at an exceptionally high level (up to 30% total leaf protein), demonstrating the great potential of plants as a nanobody production system.
Collapse
Affiliation(s)
- Yi-Hui Audrey Teh
- Plant Molecular Genetics Laboratory, Smurfit Institute of Genetics, Trinity College, Dublin 2, Ireland
| | | |
Collapse
|
30
|
Karg SR, Kallio PT. The production of biopharmaceuticals in plant systems. Biotechnol Adv 2009; 27:879-894. [PMID: 19647060 DOI: 10.1016/j.biotechadv.2009.07.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 07/15/2009] [Accepted: 07/17/2009] [Indexed: 12/20/2022]
Abstract
Biopharmaceuticals present the fastest growing segment in the pharmaceutical industry, with an ever widening scope of applications. Whole plants as well as contained plant cell culture systems are being explored for their potential as cheap, safe, and scalable production hosts. The first plant-derived biopharmaceuticals have now reached the clinic. Many biopharmaceuticals are glycoproteins; as the Golgi N-glycosylation machinery of plants differs from the mammalian machinery, the N-glycoforms introduced on plant-produced proteins need to be taken into consideration. Potent systems have been developed to change the plant N-glycoforms to a desired or even superior form compared to the native mammalian N-glycoforms. This review describes the current status of biopharmaceutical production in plants for industrial applications. The recent advances and tools which have been utilized to generate glycoengineered plants are also summarized and compared with the relevant mammalian systems whenever applicable.
Collapse
Affiliation(s)
- Saskia R Karg
- Institute of Microbiology, ETH Zurich, Wolfgang-Pauli Strasse 10, CH-8093 Zürich, Switzerland.
| | - Pauli T Kallio
- Institute of Microbiology, ETH Zurich, Wolfgang-Pauli Strasse 10, CH-8093 Zürich, Switzerland.
| |
Collapse
|
31
|
Behar G, Chames P, Teulon I, Cornillon A, Alshoukr F, Roquet F, Pugnière M, Teillaud JL, Gruaz-Guyon A, Pèlegrin A, Baty D. Llama single-domain antibodies directed against nonconventional epitopes of tumor-associated carcinoembryonic antigen absent from nonspecific cross-reacting antigen. FEBS J 2009; 276:3881-93. [DOI: 10.1111/j.1742-4658.2009.07101.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
32
|
SPECT imaging with 99mTc-labeled EGFR-specific nanobody for in vivo monitoring of EGFR expression. Mol Imaging Biol 2008; 10:167-75. [PMID: 18297364 DOI: 10.1007/s11307-008-0133-8] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 11/30/2007] [Accepted: 12/14/2007] [Indexed: 01/19/2023]
Abstract
PURPOSE Overexpression of the epidermal growth factor receptor (EGFR) occurs with high incidence in various carcinomas. The oncogenic expression of the receptor has been exploited for immunoglobulin-based diagnostics and therapeutics. We describe the use of a llama single-domain antibody fragment, termed Nanobody, for the in vivo radioimmunodetection of EGFR overexpressing tumors using single photon emission computed tomography (SPECT) in mice. METHODS Fluorescence-activated cell sorting (FACS) analysis was performed to evaluate the specificity and selectivity of 8B6 Nanobody to bind EGFR on EGFR overexpressing cells. The Nanobody was then labeled with (99m)Tc via its C-terminal histidine tail. Uptake in normal organs and tissues was assessed by ex vivo analysis. In vivo tumor targeting of (99m)Tc-8B6 Nanobody was evaluated via pinhole SPECT in mice bearing xenografts of tumor cells with either high (A431) or moderate (DU145) overexpression of EGFR. RESULTS FACS analysis indicated that the 8B6 Nanobody only recognizes cells overexpressing EGFR. In vivo blood clearance of (99m)Tc-8B6 Nanobody is relatively fast (half-life, 1.5 h) and mainly via the kidneys. At 3 h postinjection, total kidney accumulation is high (46.6+/-0.9%IA) compared to total liver uptake (18.9+/-0.6%IA). Pinhole SPECT imaging of mice bearing A431 xenografts showed higher average tumor uptake (5.2+/-0.5%IA/cm(3)) of (99m)Tc-8B6 Nanobody compared to DU145 xenografts (1.8+/-0.3%IA/cm(3), p<0.001). CONCLUSION The EGFR-binding Nanobody investigated in this study shows high specificity and selectivity towards EGFR overexpressing cells. Pinhole SPECT analysis with (99m)Tc-8B6 Nanobody enabled in vivo discrimination between tumors with high and moderate EGFR overexpression. The favorable biodistribution further corroborates the suitability of Nanobodies for in vivo tumor imaging.
Collapse
|