1
|
Rossotti MA, Trempe F, van Faassen H, Hussack G, Arbabi-Ghahroudi M. Isolation and Characterization of Single-Domain Antibodies from Immune Phage Display Libraries. Methods Mol Biol 2023; 2702:107-147. [PMID: 37679618 DOI: 10.1007/978-1-0716-3381-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Naturally occurring heavy chain antibodies (HCAbs) in Camelidae species were a surprise discovery in 1993 by Hamers et al. Since that time, antibody fragments derived from HCAbs have garnered considerable attention by researchers and biotechnology companies. Due to their biophysico-chemical advantages over conventional antibody fragments, camelid single-domain antibodies (sdAbs, VHHs, nanobodies) are being increasingly utilized as viable immunotherapeutic modalities. Currently there are multiple VHH-based therapeutic agents in different phases of clinical trials in various formats such as bi- and multivalent, bi- and multi-specific, CAR-T, and antibody-drug conjugates. The first approved VHH, a bivalent humanized VHH (caplacizumab), was approved for treating rare blood clotting disorders in 2018 by the EMA and the FDA in 2019. This was followed by the approval of an anti-BCMA VHH-based CAR-T cell product in 2022 (ciltacabtagene autoleucel; CARVYKTI™) and more recently a trivalent antitumor necrosis factor alpha-based VHH drug (ozoralizumab; Nanozora®) in Japan for the treatment of rheumatoid arthritis. In this chapter we provide protocols describing the latest developments in isolating antigen-specific VHHs including llama immunization, construction of phage-displayed libraries, phage panning and screening of the soluble VHHs by ELISA, affinity measurements by surface plasmon resonance, functional cell binding by flow cytometry, and additional validation by immunoprecipitation. We present and discuss comprehensive, step-by-step methods for isolating and characterization of antigen-specific VHHs. This includes protocols for expression, biotinylation, purification, and characterization of the isolated VHHs. To demonstrate the feasibility of the entire strategy, we present examples of VHHs previously isolated and characterized in our laboratory.
Collapse
Affiliation(s)
- Martin A Rossotti
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Frederic Trempe
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Henk van Faassen
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Greg Hussack
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Mehdi Arbabi-Ghahroudi
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
- Department of Biology, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
2
|
Alata W, Yogi A, Brunette E, Delaney CE, Faassen H, Hussack G, Iqbal U, Kemmerich K, Haqqani AS, Moreno MJ, Stanimirovic DB. Targeting insulin‐like growth factor‐1 receptor (IGF1R) for brain delivery of biologics. FASEB J 2022; 36:e22208. [DOI: 10.1096/fj.202101644r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/13/2022] [Accepted: 02/01/2022] [Indexed: 12/25/2022]
Affiliation(s)
- Wael Alata
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Alvaro Yogi
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Eric Brunette
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Christie E. Delaney
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Henk Faassen
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Greg Hussack
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Umar Iqbal
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Kristin Kemmerich
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Arsalan S. Haqqani
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Maria J. Moreno
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| | - Danica B. Stanimirovic
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa Ontario Canada
| |
Collapse
|
3
|
Trempe F, Rossotti MA, Maqbool T, MacKenzie CR, Arbabi-Ghahroudi M. Llama DNA Immunization and Isolation of Functional Single-Domain Antibody Binders. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2446:37-70. [PMID: 35157268 DOI: 10.1007/978-1-0716-2075-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genetic immunization is a simple, cost-effective, and powerful tool for inducing innate and adaptive immune responses to combat infectious diseases and difficult-to-treat illnesses. DNA immunization is increasingly used in the generation of monoclonal antibodies against targets for which pure proteins are unavailable or are difficult to express and purify (e.g., ion channels and receptors, transmembrane proteins, and emerging infectious pathogens). Genetic immunization has been successfully utilized in small inbred laboratory animals (mostly rodents); however, low immunogenicity of DNA/RNA injected into large mammals, including humans, is still a major challenge. Here, we provide a method for the genetic immunization of llamas, using a combination of biolistic transfection with a gene gun and intradermal injection with a DERMOJET® device, to elicit heavy-chain IgG responses against epidermal growth factor receptor (EGFR). We show the technique can be used to generate single-domain antibodies (VHHs) with nanomolar affinities to EGFR. We provide methods for gene gun bullet preparation, llama immunization, serology, phage-display library construction and panning, and VHH characterization.
Collapse
Affiliation(s)
- Frédéric Trempe
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Martin A Rossotti
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | | | - C Roger MacKenzie
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Mehdi Arbabi-Ghahroudi
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada. .,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada. .,Department of Biology, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
4
|
Huen J, Yan Z, Iwashkiw J, Dubey S, Gimenez MC, Ortiz ME, Patel SV, Jones MD, Riazi A, Terebiznik M, Babaei S, Shahinas D. A Novel Single Domain Antibody Targeting FliC Flagellin of Salmonella enterica for Effective Inhibition of Host Cell Invasion. Front Microbiol 2019; 10:2665. [PMID: 31849856 PMCID: PMC6901939 DOI: 10.3389/fmicb.2019.02665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 11/01/2019] [Indexed: 11/26/2022] Open
Abstract
The enteric pathogen, Salmonella enterica is a major cause of human gastroenteritis globally and with increasing bacterial resistance to antibiotics, alternative solutions are urgently needed. Single domain antibodies (sdAbs), the smallest antibody fragments that retain antigen binding specificity and affinity, are derived from variable heavy-chain only fragments (VHH) of camelid heavy-chain-only immunoglobulins. SdAbs typically contain a single disulfide bond simplifying recombinant protein production in microbial systems. These factors make sdAbs ideally suited for the development of effective anti-bacterial therapeutics. To this end, we generated an anti-Salmonella VHH library from which we screened for high affinity sdAbs. We present a novel sdAb (Abi-Se07) that targets the Salmonella virulence factor, FliC, required for bacterial motility and invasion of host cells. We demonstrate that Abi-Se07 bound FliC with a K D of 16.2 ± 0.1 nM. In addition, Abi-Se07 exhibited cross-serovar binding to whole cells of S. enterica serovar Typhimurium, Heidelberg, and Hadar. Abi-Se07 significantly inhibited bacterial motility and significantly reduced S. enterica colonization in a more native environment of chicken jejunum epithelium. Taken together, we have identified a novel anti-Salmonella sdAb and discuss future efforts toward therapeutic development.
Collapse
Affiliation(s)
- Jennifer Huen
- AbCelex Technologies Inc., Mississauga, ON, Canada
- Department of Biological Sciences, University of Toronto at Scarborough, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto at Scarborough, Toronto, ON, Canada
| | - Zhun Yan
- AbCelex Technologies Inc., Mississauga, ON, Canada
| | | | | | - Maria C. Gimenez
- Department of Biological Sciences, University of Toronto at Scarborough, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto at Scarborough, Toronto, ON, Canada
| | - Maria E. Ortiz
- Department of Biological Sciences, University of Toronto at Scarborough, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto at Scarborough, Toronto, ON, Canada
| | | | | | - Ali Riazi
- AbCelex Technologies Inc., Mississauga, ON, Canada
| | - Mauricio Terebiznik
- Department of Biological Sciences, University of Toronto at Scarborough, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto at Scarborough, Toronto, ON, Canada
| | - Saeid Babaei
- AbCelex Technologies Inc., Mississauga, ON, Canada
| | - Dea Shahinas
- AbCelex Technologies Inc., Mississauga, ON, Canada
| |
Collapse
|
5
|
Hussack G, Baral TN, Baardsnes J, van Faassen H, Raphael S, Henry KA, Zhang J, MacKenzie CR. A Novel Affinity Tag, ABTAG, and Its Application to the Affinity Screening of Single-Domain Antibodies Selected by Phage Display. Front Immunol 2017; 8:1406. [PMID: 29163485 PMCID: PMC5674936 DOI: 10.3389/fimmu.2017.01406] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/11/2017] [Indexed: 12/20/2022] Open
Abstract
ABTAG is a camelid single-domain antibody (sdAb) that binds to bovine serum albumin (BSA) with low picomolar affinity. In surface plasmon resonance (SPR) analyses using BSA surfaces, bound ABTAG can be completely dissociated from the BSA surfaces at low pH, over multiple cycles, without any reduction in the capacity of the BSA surfaces to bind ABTAG. A moderate throughput, SPR-based, antibody screening assay exploiting the unique features of ABTAG is described. Anti-carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) sdAbs were isolated from a phage-displayed sdAb library derived from the heavy chain antibody repertoire of a llama immunized with CEACAM6. Following one or two rounds of panning, enriched clones were expressed as ABTAG fusions in microtiter plate cultures. The sdAb-ABTAG fusions from culture supernatants were captured on BSA surfaces and CEACAM6 antigen was then bound to the captured molecules. The SPR screening method gives a read-out of relative expression levels of the fusion proteins and kinetic and affinity constants for CEACAM6 binding by the captured molecules. The library was also panned and screened by conventional methods and positive clones were subcloned and expressed for SPR analysis. Compared to conventional panning and screening, the SPR-based ABTAG method yielded a considerably higher diversity of binders, some with affinities that were three orders of magnitude higher affinity than those identified by conventional panning.
Collapse
Affiliation(s)
- Greg Hussack
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Toya Nath Baral
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, QC, Canada
| | - Henk van Faassen
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Shalini Raphael
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Kevin A Henry
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jianbing Zhang
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - C Roger MacKenzie
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| |
Collapse
|
6
|
Henry KA, Sulea T, van Faassen H, Hussack G, Purisima EO, MacKenzie CR, Arbabi-Ghahroudi M. A Rational Engineering Strategy for Designing Protein A-Binding Camelid Single-Domain Antibodies. PLoS One 2016; 11:e0163113. [PMID: 27631624 PMCID: PMC5025174 DOI: 10.1371/journal.pone.0163113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/04/2016] [Indexed: 12/21/2022] Open
Abstract
Staphylococcal protein A (SpA) and streptococcal protein G (SpG) affinity chromatography are the gold standards for purifying monoclonal antibodies (mAbs) in therapeutic applications. However, camelid VHH single-domain Abs (sdAbs or VHHs) are not bound by SpG and only sporadically bound by SpA. Currently, VHHs require affinity tag-based purification, which limits their therapeutic potential and adds considerable complexity and cost to their production. Here we describe a simple and rapid mutagenesis-based approach designed to confer SpA binding upon a priori non-SpA-binding VHHs. We show that SpA binding of VHHs is determined primarily by the same set of residues as in human mAbs, albeit with an unexpected degree of tolerance to substitutions at certain core and non-core positions and some limited dependence on at least one residue outside the SpA interface, and that SpA binding could be successfully introduced into five VHHs against three different targets with no adverse effects on expression yield or antigen binding. Next-generation sequencing of llama, alpaca and dromedary VHH repertoires suggested that species differences in SpA binding may result from frequency variation in specific deleterious polymorphisms, especially Ile57. Thus, the SpA binding phenotype of camelid VHHs can be easily modulated to take advantage of tag-less purification techniques, although the frequency with which this is required may depend on the source species.
Collapse
Affiliation(s)
- Kevin A. Henry
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, Canada, K1A 0R6
| | - Traian Sulea
- Human Health Therapeutics Portfolio, National Research Council Canada, 6100 Royalmount Avenue, Montreal, Quebec, Canada, H4P 2R2
| | - Henk van Faassen
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, Canada, K1A 0R6
| | - Greg Hussack
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, Canada, K1A 0R6
| | - Enrico O. Purisima
- Human Health Therapeutics Portfolio, National Research Council Canada, 6100 Royalmount Avenue, Montreal, Quebec, Canada, H4P 2R2
| | - C. Roger MacKenzie
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, Canada, K1A 0R6
- School of Environmental Sciences, University of Guelph, 50 Stone Road East, Guelph, Ontario, Canada, N1G 2W1
| | - Mehdi Arbabi-Ghahroudi
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, Canada, K1A 0R6
- School of Environmental Sciences, University of Guelph, 50 Stone Road East, Guelph, Ontario, Canada, N1G 2W1
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, Canada, K1S 5B6
- * E-mail:
| |
Collapse
|
7
|
Henry KA, Hussack G, Collins C, Zwaagstra JC, Tanha J, MacKenzie CR. Isolation of TGF-β-neutralizing single-domain antibodies of predetermined epitope specificity using next-generation DNA sequencing. Protein Eng Des Sel 2016; 29:439-443. [DOI: 10.1093/protein/gzw043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/20/2016] [Indexed: 11/14/2022] Open
|
8
|
Steeland S, Vandenbroucke RE, Libert C. Nanobodies as therapeutics: big opportunities for small antibodies. Drug Discov Today 2016; 21:1076-113. [DOI: 10.1016/j.drudis.2016.04.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/26/2016] [Accepted: 04/04/2016] [Indexed: 12/28/2022]
|
9
|
Krah S, Schröter C, Zielonka S, Empting M, Valldorf B, Kolmar H. Single-domain antibodies for biomedical applications. Immunopharmacol Immunotoxicol 2015; 38:21-8. [DOI: 10.3109/08923973.2015.1102934] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
10
|
Henry KA, Tanha J, Hussack G. Identification of cross-reactive single-domain antibodies against serum albumin using next-generation DNA sequencing. Protein Eng Des Sel 2015; 28:379-83. [PMID: 26319004 DOI: 10.1093/protein/gzv039] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
Antibodies that cross-react with multiple isoforms or homologue of a given protein are often desirable, especially in therapeutic applications. Here, we report the identification of several unique, clonally unrelated, single-domain antibodies (sdAbs) that bind to multiple serum albumin orthologues (human, rhesus, rat and mouse) with low-to-medium nanomolar affinity from a llama immunized only with human serum albumin. Using single-round panning of a phage-displayed sdAb library against serum albumins and next-generation DNA sequencing, we were able to predict patterns of sdAb reactivity to the albumins of different species with ∼90% accuracy. We expect this strategy to be generally applicable for identifying cross-reactive sdAbs, particularly when these exist at low frequency and/or are poorly enriched by panning. Moreover, the sdAbs identified here are of potential interest for serum half-life extension of biologics.
Collapse
Affiliation(s)
- Kevin A Henry
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, ON, Canada K1A 0R6
| | - Jamshid Tanha
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, ON, Canada K1A 0R6 School of Environmental Sciences, Ontario Agricultural College, University of Guelph, 50 Stone Road East, Guelph, ON, Canada N1G 2W1 Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 75 Laurier Avenue East, Ottawa, ON, Canada K1H 8M5
| | - Greg Hussack
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, ON, Canada K1A 0R6
| |
Collapse
|
11
|
Gene RW, Kumaran J, Aroche C, van Faassen H, Hall JC, MacKenzie CR, Arbabi-Ghahroudi M. High affinity anti-Internalin B VHH antibody fragments isolated from naturally and artificially immunized repertoires. J Immunol Methods 2015; 416:29-39. [DOI: 10.1016/j.jim.2014.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/22/2014] [Accepted: 10/22/2014] [Indexed: 11/25/2022]
|
12
|
Alturki NA, Henry KA, MacKenzie CR, Arbabi-Ghahroudi M. Isolation of Camelid Single-Domain Antibodies Against Native Proteins Using Recombinant Multivalent Peptide Ligands. Methods Mol Biol 2015; 1348:167-89. [PMID: 26424272 DOI: 10.1007/978-1-4939-2999-3_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Generation of antibodies against desired epitopes on folded proteins may be hampered by various characteristics of the target protein, including antigenic and immunogenic dominance of irrelevant epitopes and/or steric occlusion of the desired epitope. In such cases, peptides encompassing linear epitopes of the native protein represent attractive alternative reagents for immunization and screening. Peptide antigens are typically prepared by fusing or conjugating the peptide of interest to a carrier protein. The utility of such antigens depends on many factors including the peptide's amino acid sequence, display valency, display format (synthetic conjugate vs. recombinant fusion) and characteristics of the carrier. Here we provide detailed protocols for: (1) preparation of DNA constructs encoding peptides fused to verotoxin (VT) multimerization domain; (2) expression, purification, and characterization of the multivalent peptide-VT ligands; (3) concurrent panning of a non-immune phage-displayed camelid VHH library against the peptide-VT ligands and native protein; and (4) identification of VHHs enriched via panning using next-generation sequencing techniques. These methods are simple, rapid and can be easily adapted to yield custom peptide-VT ligands that appear to maintain the antigenic structures of the peptide. However, we caution that peptide sequences should be chosen with great care, taking into account structural, immunological, and biophysical information on the protein of interest.
Collapse
Affiliation(s)
- Norah A Alturki
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, ON, Canada, K1A 0R6.,College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Kevin A Henry
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, ON, Canada, K1A 0R6
| | - C Roger MacKenzie
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, ON, Canada, K1A 0R6.,School of Environmental Sciences, University of Guelph, Guelph, ON, Canada
| | - Mehdi Arbabi-Ghahroudi
- Human Health Therapeutics Portfolio, National Research Council Canada, 100 Sussex Drive, Ottawa, ON, Canada, K1A 0R6. .,School of Environmental Sciences, University of Guelph, Guelph, ON, Canada. .,Department of Biology, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
13
|
Critical roles of Clostridium difficile toxin B enzymatic activities in pathogenesis. Infect Immun 2014; 83:502-13. [PMID: 25404023 DOI: 10.1128/iai.02316-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
TcdB is one of the key virulence factors of Clostridium difficile that is responsible for causing serious and potentially fatal colitis. The toxin contains at least two enzymatic domains: an effector glucosyltransferase domain for inactivating host Rho GTPases and a cysteine protease domain for the delivery of the effector domain into host cytosol. Here, we describe a novel intrabody approach to examine the role of these enzymes of TcdB in cellular intoxication. By screening a single-domain heavy chain (V(H)H) library raised against TcdB, we identified two V(H)H antibodies, 7F and E3, that specifically inhibit TcdB cysteine protease and glucosyltransferase activities, respectively. Cytoplasmic expression of 7F intrabody in Vero cells inhibited TcdB autoprocessing and delayed cellular intoxication, whereas E3 intrabody completely blocked the cytopathic effects of TcdB holotoxin. These data also demonstrate for the first time that toxin autoprocessing occurs after cysteine protease and glucosyltransferase domains translocate into the cytosol of target cells. We further determined the role of the enzymatic activities of TcdB in in vivo toxicity using a sensitive systemic challenge model in mice. Consistent with these in vitro results, a cysteine protease noncleavable mutant, TcdB-L543A, delayed toxicity in mice, whereas glycosyltransferase-deficient TcdB demonstrated no toxicity up to 500-fold of the 50% lethal dose (LD50) when it was injected systemically. Thus, glucosyltransferase but not cysteine protease activity is critical for TcdB-mediated cytopathic effects and TcdB systemic toxicity, highlighting the importance of targeting toxin glucosyltransferase activity for future therapy.
Collapse
|
14
|
Kim DY, Hussack G, Kandalaft H, Tanha J. Mutational approaches to improve the biophysical properties of human single-domain antibodies. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1983-2001. [DOI: 10.1016/j.bbapap.2014.07.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/05/2014] [Accepted: 07/11/2014] [Indexed: 01/06/2023]
|
15
|
Hussack G, Riazi A, Ryan S, van Faassen H, MacKenzie R, Tanha J, Arbabi-Ghahroudi M. Protease-resistant single-domain antibodies inhibit Campylobacter jejuni motility. Protein Eng Des Sel 2014; 27:191-8. [PMID: 24742504 DOI: 10.1093/protein/gzu011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Camelid heavy-chain antibody variable domains (VHHs) are emerging as potential antimicrobial reagents. We have engineered a previously isolated VHH (FlagV1M), which binds Campylobacter jejuni flagella, for greater thermal and proteolytic stability. Mutants of FlagV1M were obtained from an error-prone polymerase chain reaction library that was panned in the presence of gastrointestinal (GI) proteases. Additional FlagV1M mutants were obtained through disulfide-bond engineering. Each approach produced VHHs with enhanced thermal stability and protease resistance. When the beneficial mutations from both approaches were combined, a hyperstabilized VHH was created with superior stability. The hyperstabilized VHH bound C. jejuni flagella with wild-type affinity and was capable of potently inhibiting C. jejuni motility in assays performed after sequential digestion with three major GI proteases, demonstrating the remarkable stability imparted to the VHH by combining our engineering approaches.
Collapse
Affiliation(s)
- Greg Hussack
- Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, ON, Canada K1A 0R6
| | - Ali Riazi
- AbCelex Technologies, Inc., Toronto, ON, Canada L4V 1T4
| | - Shannon Ryan
- Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, ON, Canada K1A 0R6
| | - Henk van Faassen
- Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, ON, Canada K1A 0R6
| | - Roger MacKenzie
- Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, ON, Canada K1A 0R6 School of Environmental Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1
| | - Jamshid Tanha
- Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, ON, Canada K1A 0R6 School of Environmental Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1 Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | - Mehdi Arbabi-Ghahroudi
- Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, ON, Canada K1A 0R6 School of Environmental Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1 Department of Biology, Carleton University, Ottawa, ON, Canada K1S 5B6
| |
Collapse
|
16
|
Riazi A, Strong PCR, Coleman R, Chen W, Hirama T, van Faassen H, Henry M, Logan SM, Szymanski CM, MacKenzie R, Ghahroudi MA. Pentavalent single-domain antibodies reduce Campylobacter jejuni motility and colonization in chickens. PLoS One 2013; 8:e83928. [PMID: 24391847 PMCID: PMC3877120 DOI: 10.1371/journal.pone.0083928] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 11/08/2013] [Indexed: 11/18/2022] Open
Abstract
Campylobacter jejuni is the leading cause of bacterial foodborne illness in the world, with symptoms ranging from acute diarrhea to severe neurological disorders. Contaminated poultry meat is a major source of C. jejuni infection, and therefore, strategies to reduce this organism in poultry, are expected to reduce the incidence of Campylobacter-associated diseases. We have investigated whether oral administration of C. jejuni-specific single-domain antibodies would reduce bacterial colonization levels in chickens. Llama single-domain antibodies specific for C. jejuni were isolated from a phage display library generated from the heavy chain IgG variable domain repertoire of a llama immunized with C. jejuni flagella. Two flagella-specific single-domain antibodies were pentamerized to yield high avidity antibodies capable of multivalent binding to the target antigen. When administered orally to C. jejuni-infected two-day old chicks, the pentabodies significantly reduced C. jejuni colonization in the ceca. In vitro, the motility of the bacteria was also reduced in the presence of the flagella-specific pentabodies, suggesting the mechanism of action is through either direct interference with flagellar motility or antibody-mediated aggregation. Fluorescent microscopy and Western blot analyses revealed specific binding of the anti-flagella pentabodies to the C. jejuni flagellin.
Collapse
Affiliation(s)
- Ali Riazi
- AbCelex Technologies Inc., Toronto, Ontario, Canada
| | - Philippa C. R. Strong
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Russell Coleman
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Wangxue Chen
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Tomoko Hirama
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Henk van Faassen
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Matthew Henry
- Dow AgroSciences, Indianapolis, Indiana, United States of America
| | - Susan M. Logan
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Christine M. Szymanski
- Centennial Centre for Interdisciplinary Science, Department of Biological Sciences and Alberta Glycomics Centre, The University of Alberta, Edmonton, Alberta, Canada
| | - Roger MacKenzie
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
- Centennial Centre for Interdisciplinary Science, Department of Biological Sciences and Alberta Glycomics Centre, The University of Alberta, Edmonton, Alberta, Canada
| | - Mehdi Arbabi Ghahroudi
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
- School of Environmental Sciences, University of Guelph, Guelph, Ontario, Canada
- Department of Biology, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
17
|
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: 46] [Impact Index Per Article: 3.8] [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
|
18
|
Tang Z, Feng M, Gao W, Phung Y, Chen W, Chaudhary A, St Croix B, Qian M, Dimitrov DS, Ho M. A human single-domain antibody elicits potent antitumor activity by targeting an epitope in mesothelin close to the cancer cell surface. Mol Cancer Ther 2013; 12:416-26. [PMID: 23371858 PMCID: PMC3624043 DOI: 10.1158/1535-7163.mct-12-0731] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Monoclonal antibodies against mesothelin are being evaluated for the treatment of mesothelioma and multiple forms of cancers, and show great promise for clinical development for solid cancers. Antibodies against mesothelin have been shown to act via immunotoxin-based inhibition of tumor growth and induction of antibody-dependent cell-mediated cytotoxicity (ADCC). However, complement-dependent cytotoxicity (CDC), considered an important additional mechanism of therapeutic antibodies against tumors, is inactive for such antibodies. Here, we used phage display antibody engineering technology and synthetic peptide screening to identify SD1, a human single-domain antibody to mesothelin. SD1 recognizes a conformational epitope at the C-terminal end (residues 539-588) of mesothelin close to the cell surface. To investigate SD1 as a potential therapeutic agent, we generated a recombinant human Fc (SD1-hFc) fusion protein. Interestingly, the SD1-hFc protein exhibits strong CDC activity, in addition to ADCC, against mesothelin-expressing tumor cells. Furthermore, it causes growth inhibition of human tumor xenografts in nude mice as a single agent. SD1 is the first human single-domain antibody targeting mesothelin-expressing tumors, shows potential as a cancer therapeutic candidate, and may improve current antibody therapy targeting mesothelin-expressing tumors.
Collapse
Affiliation(s)
- Zhewei Tang
- Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200062, China
- Antiody Therapy Section, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Mingqian Feng
- Antiody Therapy Section, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Wei Gao
- Antiody Therapy Section, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Yen Phung
- Antiody Therapy Section, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Weizao Chen
- Protein Interaction Group, CCR Nanobiology Program, Frederick National laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland 21702
| | - Amit Chaudhary
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, Frederick National laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland 21702
| | - Brad St Croix
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, Frederick National laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland 21702
| | - Min Qian
- Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Dimiter S. Dimitrov
- Protein Interaction Group, CCR Nanobiology Program, Frederick National laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland 21702
| | - Mitchell Ho
- Antiody Therapy Section, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
19
|
Abstract
In this chapter we describe in detail the current protocols that are used to express single-domain antibodies in bacteria. Bacteria are among the most common expression systems for expressing recombinant proteins. We present different approaches for carrying out periplasmic and cytoplasmic expression, as well as small-scale and large-scale expression. In addition, we discuss the advantages and possible drawbacks of each protocol. We present data related to expression vectors, expression conditions, methods of protein extraction and purification, and yield and purity analysis of sdAbs. We also highlight important points that need to be considered before sdAbs that have been expressed in bacteria are used either in vitro or in vivo.
Collapse
|
20
|
Kumaran J, Mackenzie CR, Arbabi-Ghahroudi M. Semiautomated panning of naive camelidae libraries and selection of single-domain antibodies against peptide antigens. Methods Mol Biol 2012; 911:105-24. [PMID: 22886248 DOI: 10.1007/978-1-61779-968-6_7] [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] [Indexed: 12/19/2022]
Abstract
With the identification of vast numbers of novel proteins through genomic and proteomic initiatives, the need for efficient processes to characterize and target them has increased. Antibodies are naturally designed molecules that can fulfill this need, and in vitro methodologies for isolating them from either immune or naïve sources have been extensively developed. However, access to pure protein antigens for screening purposes is a major hurdle due to the limitations associated with recombinant production of eukaryotic proteins. Consequently, rational peptide design based on proteomic methodologies such as protein modeling, secondary sequence prediction, and hydrophobicity/hydrophilicity prediction, in combination with other bioinformatics data, is being explored as a viable solution to isolate specific antibodies against difficult antigens. Single-domain antibodies are becoming the ideal antibody format due to their structural advantages and ease of production compared to conventional antibodies and antibody fragments derived from conventional antibodies. For screening purposes, phage display technology is a well-established technique. With this technique, a repertoire of antibody fragments can be displayed on the surface of filamentous phages (f1, fd, M13) followed by screening against various antigenic targets. Furthermore, the technique can be expanded to a high-throughput scale using a magnetic-based, in-solution panning protocol which allows for the screening of multiple target antigens simultaneously. In this chapter, we describe a semiautomated panning method to screen a naïve Camelidae library against rationally designed peptide antigens, followed by preliminary characterization of isolated binders.
Collapse
Affiliation(s)
- Jyothi Kumaran
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, ON, Canada
| | | | | |
Collapse
|
21
|
Abstract
Sales of monoclonal antibody (mAbs) therapies exceeded $ 40 billion in 2010 and are expected to reach $ 70 billion by 2015. The majority of the approved antibodies are targeting cancer and autoimmune diseases with the top 5 grossing antibodies populating these two areas. In addition over 100 monoclonal antibodies are in Phase II and III of clinical development and numerous others are in various pre-clinical and safety studies. Commercial production of monoclonal antibodies is one of the few biotechnology manufacturing areas that has undergone significant improvements and standardization over the last ten years. Platform technologies have been established based on the structural similarities of these molecules and the regulatory requirements. These improvements include better cell lines, advent of high-performing media free of animal-derived components, and advances in bioreactor and purification processes. In this chapter we will examine the progress made in antibody production as well as discuss the future of manufacturing for these molecules, including the emergence of single use technologies.
Collapse
|
22
|
Hussack G, Arbabi-Ghahroudi M, van Faassen H, Songer JG, Ng KKS, MacKenzie R, Tanha J. Neutralization of Clostridium difficile toxin A with single-domain antibodies targeting the cell receptor binding domain. J Biol Chem 2011; 286:8961-76. [PMID: 21216961 DOI: 10.1074/jbc.m110.198754] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile is a leading cause of nosocomial infection in North America and a considerable challenge to healthcare professionals in hospitals and nursing homes. The gram-positive bacterium produces two high molecular weight exotoxins, toxin A (TcdA) and toxin B (TcdB), which are the major virulence factors responsible for C. difficile-associated disease and are targets for C. difficile-associated disease therapy. Here, recombinant single-domain antibody fragments (V(H)Hs), which specifically target the cell receptor binding domains of TcdA or TcdB, were isolated from an immune llama phage display library and characterized. Four V(H)Hs (A4.2, A5.1, A20.1, and A26.8), all shown to recognize conformational epitopes, were potent neutralizers of the cytopathic effects of toxin A on fibroblast cells in an in vitro assay. The neutralizing potency was further enhanced when V(H)Hs were administered in paired or triplet combinations at the same overall V(H)H concentration, suggesting recognition of nonoverlapping TcdA epitopes. Biacore epitope mapping experiments revealed that some synergistic combinations consisted of V(H)Hs recognizing overlapping epitopes, an indication that factors other than mere epitope blocking are responsible for the increased neutralization. Further binding assays revealed TcdA-specific V(H)Hs neutralized toxin A by binding to sites other than the carbohydrate binding pocket of the toxin. With favorable characteristics such as high production yield, potent toxin neutralization, and intrinsic stability, these V(H)Hs are attractive systemic therapeutics but are more so as oral therapeutics in the destabilizing environment of the gastrointestinal tract.
Collapse
Affiliation(s)
- Greg Hussack
- Institute for Biological Sciences, National Research Council Canada, Ottawa, Ontario K1A 0R6, Canada
| | | | | | | | | | | | | |
Collapse
|