1
|
Boyraz B, Tauber R, Dernedde J. Identification of an Immunoglobulin Paratope Binding to Keratan Sulfate and Expression of a Single-Chain Derivative for Imaging. Biomolecules 2025; 15:178. [PMID: 40001481 PMCID: PMC11852928 DOI: 10.3390/biom15020178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/06/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Keratan sulfate (KS) is a negatively charged carbohydrate linked to proteins. Several KS-bearing structural glycosaminoglycans participate to maintain the homeostasis of a functional extracellular matrix. Dysfunction of its biochemical composition and structure might therefore lead to pathological situations. For this reason, imaging of KS in tissues is an important diagnostic tool. Here, we describe the identification of the KS paratope derived from the ancestral anti-KS IgG mAb MZ15, as well as the engineering, functional recombinant expression in E. coli, and purification of an anti-KS single-chain variable fragment (ScFv). The ScFv enabled in vitro imaging of KS in cryosections of rat cornea by immunofluorescence microscopy comparable to the ancestral IgG MZ15.
Collapse
Affiliation(s)
- Burak Boyraz
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany;
- Fachbereich Biologie, Chemie, Pharmazie, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Rudolf Tauber
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Jens Dernedde
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany;
| |
Collapse
|
2
|
Moholkar DN, Kandimalla R, Gupta RC, Aqil F. Advances in lipid-based carriers for cancer therapeutics: Liposomes, exosomes and hybrid exosomes. Cancer Lett 2023; 565:216220. [PMID: 37209944 PMCID: PMC10325927 DOI: 10.1016/j.canlet.2023.216220] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/18/2023] [Accepted: 05/06/2023] [Indexed: 05/22/2023]
Abstract
Cancer has recently surpassed heart disease as the leading cause of deaths worldwide for the age group 45-65 and has been the primary focus for biomedical researchers. Presently, the drugs involved in the first-line cancer therapy are raising concerns due to high toxicity and lack of selectivity to cancer cells. There has been a significant increase in research with innovative nano formulations to entrap the therapeutic payload to enhance efficacy and eliminate or minimize toxic effects. Lipid-based carriers stand out due to their unique structural properties and biocompatible nature. The two main leaders of lipid-based drug carriers: long known liposomes and comparatively new exosomes have been well-researched. The similarity between the two lipid-based carriers is the vesicular structure with the core's capability to carry the payload. While liposomes utilize chemically derived and altered phospholipid components, the exosomes are naturally occurring vesicles with inherent lipids, proteins, and nucleic acids. More recently, researchers have focused on developing hybrid exosomes by fusing liposomes and exosomes. Combining these two types of vesicles may offer some advantages such as high drug loading, targeted cellular uptake, biocompatibility, controlled release, stability in harsh conditions and low immunogenicity.
Collapse
Affiliation(s)
- Disha N Moholkar
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Raghuram Kandimalla
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA; Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Ramesh C Gupta
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA; Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA.
| | - Farrukh Aqil
- Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA; Department of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
3
|
Lewis MR, Cutler CS, Jurisson SS. Targeted Antibodies and Peptides. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
4
|
Ultrasound-mediated blood-brain barrier opening enhances delivery of therapeutically relevant formats of a tau-specific antibody. Sci Rep 2019; 9:9255. [PMID: 31239479 PMCID: PMC6592925 DOI: 10.1038/s41598-019-45577-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/06/2019] [Indexed: 12/14/2022] Open
Abstract
The microtubule-associated protein tau is an attractive therapeutic target for the treatment of Alzheimer’s disease and related tauopathies as its aggregation strongly correlates with disease progression and is considered a key mediator of neuronal toxicity. Delivery of most therapeutics to the brain is, however, inefficient, due to their limited ability to cross the blood-brain barrier (BBB). Therapeutic ultrasound is an emerging non-invasive technology which transiently opens the BBB in a focused manner to allow peripherally delivered molecules to effectively enter the brain. In order to open a large area of the BBB, we developed a scanning ultrasound (SUS) approach by which ultrasound is applied in a sequential pattern across the whole brain. We have previously shown that delivery of an anti-tau antibody in a single-chain variable fragment (scFv) format to the brain is increased with SUS allowing for an enhanced therapeutic effect. Here we compared the delivery of an anti-tau antibody, RN2N, in an scFv, fragment antigen-binding (Fab) and full-sized immunoglobulin G (IgG) format, with and without sonication, into the brain of pR5 tau transgenic mice, a model of tauopathy. Our results revealed that the full-sized IgG reaches a higher concentration in the brain compared with the smaller formats by bypassing renal excretion. No differences in either the ultrasound-mediated uptake or distribution in the brain from the sonication site was observed across the different antibody formats, suggesting that ultrasound can be used to successfully increase the delivery of therapeutic molecules of various sizes into the brain for the treatment of neurological diseases.
Collapse
|
5
|
Sengupta S, Asha Krishnan M, Chattopadhyay S, Chelvam V. Comparison of prostate-specific membrane antigen ligands in clinical translation research for diagnosis of prostate cancer. Cancer Rep (Hoboken) 2019; 2:e1169. [PMID: 32721116 DOI: 10.1002/cnr2.1169] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/28/2019] [Accepted: 02/07/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA), overexpressed on prostate cancer (PCa), is a well-characterized cell surface protein to selectively diagnose PCa. PSMA's unique characteristics and its 1000-fold higher expression in PCa compared with other tissues renders it as a suitable biomarker for detection of PCa in its early stage. In this report, we critically analyze and recommend the requirements needed for the development of variety of PSMA-targeted molecular imaging agents based on antibodies, small molecule ligands, peptides, and aptamers. The targeting moieties are either conjugated to radionuclear isotopes or near-infrared agents for efficient diagnosis of PCa. RECENT FINDINGS From the analysis, it was found that several small molecule-derived PCa imaging agents are approved for clinical trials in Europe and the United States, and few are already in the clinical use for diagnosis of PCa. Even though 111In-labeled capromab pendetide was approved by the Food and Drug Administration (FDA) and other engineered antibodies are available for detection of PCa, but high production cost, low shelf life (less than 1 month at 4°C), possibility of human immuno reactions, and low blood clearance rate necessitated a need for developing new imaging agents, which are serum stable, cost-effective, and possesses longer shelf life (6 months), have fast clearance rate from nontargeted tissues during the diagnosis process. It is found that small molecule ligand-derived imaging agents possesses most of the desired properties expected for an ideal diagnostic agent when compared with other targeting moieties. CONCLUSION This report discusses in detail the homing moieties used in the development of targeted diagnostic tools for detection of PCa. The merits and demerits of monoclonal antibodies, small molecule ligands, peptides, and aptamers for imaging of PCa and intraoperative guided surgery are extensively analyzed. Among all, urea-based ligands were found to be most successful in preclinical and clinical trials and show a major promise for future commercialization.
Collapse
Affiliation(s)
- Sagnik Sengupta
- Discipline of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore, Indore, India
| | - Mena Asha Krishnan
- Discipline of Biosciences and Biomedical Engineering, School of Engineering, Indian Institute of Technology Indore, Indore, India
| | - Sudeshna Chattopadhyay
- Discipline of Biosciences and Biomedical Engineering, School of Engineering, Indian Institute of Technology Indore, Indore, India.,Discipline of Physics, School of Basic Sciences, Indian Institute of Technology Indore, Indore, India.,Discipline of Metallurgy Engineering and Material Science, School of Engineering, Indian Institute of Technology Indore, Indore, India
| | - Venkatesh Chelvam
- Discipline of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore, Indore, India.,Discipline of Biosciences and Biomedical Engineering, School of Engineering, Indian Institute of Technology Indore, Indore, India
| |
Collapse
|
6
|
Bumbaca B, Li Z, Shah DK. Pharmacokinetics of protein and peptide conjugates. Drug Metab Pharmacokinet 2019; 34:42-54. [PMID: 30573392 PMCID: PMC6378135 DOI: 10.1016/j.dmpk.2018.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/29/2018] [Accepted: 11/19/2018] [Indexed: 12/20/2022]
Abstract
Protein and peptide conjugates have become an important component of therapeutic and diagnostic medicine. These conjugates are primarily designed to improve pharmacokinetics (PK) of those therapeutic or imaging agents, which do not possess optimal disposition characteristics. In this review we have summarized preclinical and clinical PK of diverse protein and peptide conjugates, and have showcased how different conjugation approaches are used to obtain the desired PK. We have classified the conjugates into peptide conjugates, non-targeted protein conjugates, and targeted protein conjugates, and have highlighted diagnostic and therapeutic applications of these conjugates. In general, peptide conjugates demonstrate very short half-life and rapid renal elimination, and they are mainly designed to achieve high contrast ratio for imaging agents or to deliver therapeutic agents at sites not reachable by bulky or non-targeted proteins. Conjugates made from non-targeted proteins like albumin are designed to increase the half-life of rapidly eliminating therapeutic or imaging agents, and improve their delivery to tissues like solid tumors and inflamed joints. Targeted protein conjugates are mainly developed from antibodies, antibody derivatives, or endogenous proteins, and they are designed to improve the contrast ratio of imaging agents or therapeutic index of therapeutic agents, by enhancing their delivery to the site-of-action.
Collapse
Affiliation(s)
- Brandon Bumbaca
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA
| | - Zhe Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA.
| |
Collapse
|
7
|
Shah SQ, Gul-e-Raana, Uddin G. Imaging prostate cancer (PCa) with [99m
Tc(CO)3
]finasteride dithiocarbamate. J Labelled Comp Radiopharm 2018. [DOI: 10.1002/jlcr.3621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Syed Qaiser Shah
- Biochemistry Section, Institute of Chemical Sciences (ICS); University of Peshawar; Peshawar KPK Pakistan
| | - Gul-e-Raana
- Biochemistry Section, Institute of Chemical Sciences (ICS); University of Peshawar; Peshawar KPK Pakistan
| | - Ghias Uddin
- Organic Section, Institute of Chemical Sciences (ICS); University of Peshawar; Peshawar KPK Pakistan
| |
Collapse
|
8
|
Camelid Single-Domain Antibodies (VHHs) against Crotoxin: A Basis for Developing Modular Building Blocks for the Enhancement of Treatment or Diagnosis of Crotalic Envenoming. Toxins (Basel) 2018; 10:toxins10040142. [PMID: 29596324 PMCID: PMC5923308 DOI: 10.3390/toxins10040142] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/11/2018] [Accepted: 03/16/2018] [Indexed: 12/15/2022] Open
Abstract
Toxic effects triggered by crotalic envenoming are mainly related to crotoxin (CTX), composed of a phospholipase A2 (CB) and a subunit with no toxic activity (CA). Camelids produce immunoglobulins G devoid of light chains, in which the antigen recognition domain is called VHH. Given their unique characteristics, VHHs were selected using Phage Display against CTX from Crotalus durissus terrificus. After three rounds of biopanning, four sequence profiles for CB (KF498602, KF498603, KF498604, and KF498605) and one for CA (KF498606) were revealed. All clones presented the VHH hallmark in FR2 and a long CDR3, with the exception of KF498606. After expressing pET22b-VHHs in E. coli, approximately 2 to 6 mg of protein per liter of culture were obtained. When tested for cross-reactivity, VHHs presented specificity for the Crotalus genus and were capable of recognizing CB through Western blot. KF498602 and KF498604 showed thermostability, and displayed affinity constants for CTX in the micro or nanomolar range. They inhibited in vitro CTX PLA2 activity, and CB cytotoxicity. Furthermore, KF498604 inhibited the CTX-induced myotoxicity in mice by 78.8%. Molecular docking revealed that KF498604 interacts with the CA–CB interface of CTX, seeming to block substrate access. Selected VHHs may be alternatives for the crotalic envenoming treatment.
Collapse
|
9
|
Jiang X, Zhai J, Song D, Qu Q, Li M, Xing L, Miao S. Investigation of the structure of anti-human seminal plasma protein single-chain antibody and its association with linker peptide length. Mol Med Rep 2015; 12:4117-4122. [PMID: 26099852 PMCID: PMC4526083 DOI: 10.3892/mmr.2015.3980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 05/08/2015] [Indexed: 12/03/2022] Open
Abstract
To enhance the activity of seminoprotein single-chain variable fragment (γ-Sm-ScFv) antibodies, modulation of the length of the linker peptide, which connects the variable region of the heavy chain (VH) and the light chain (VL) of single-chain antibodies, was performed in the present study. Homologous modeling of single VH and VL were performed, respectively. Subsequently, modeling of the whole ScFv sequence, which was previously modified with added linkers of different lengths was also performed, and the (Gly4Ser)n peptide chain structure was used as the linker. The similarities between VH and VL prior to and following the addition of the linker were compared by applying the algorithm of protein similarity, based on spherical coordinates layering. In addition, changes in the fore and aft distance, and diffusion radius were calculated using a MATLAB tool, based on which changes in structural stability were analyzed. Finally, the single-chain antibody was assessed in a nude mouse model. When n=3 or n=6, the similarity between the original distance and VH and VL were the highest, and the fore and aft distance and diffusion radius were relatively close. In addition, the nude mouse model indicated that, when n=3 or n=6, the inhibitory rate of the single-chain antibody against tumor cells was significantly higher, compared with the other linker peptides of different lengths. The effect of structural changes of the linker peptides in the single-chain antibodies on the whole antibody molecule was examined at different levels using a combination of mathematical modeling, bioinformatics methods and biological experiments. The findings of the present study may provide a foundation for further investigation into the preparation of single-chain antibodies.
Collapse
Affiliation(s)
- Xin Jiang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jun Zhai
- Reproductive Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Dongkui Song
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qingshan Qu
- Department of Kidney Transplantation, People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| | - Ming Li
- Department of Kidney Transplantation, People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| | - Li Xing
- Department of Kidney Transplantation, People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| | - Shuzhai Miao
- Department of Kidney Transplantation, People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
10
|
Engineering venom's toxin-neutralizing antibody fragments and its therapeutic potential. Toxins (Basel) 2014; 6:2541-67. [PMID: 25153256 PMCID: PMC4147596 DOI: 10.3390/toxins6082541] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/16/2014] [Accepted: 08/04/2014] [Indexed: 11/24/2022] Open
Abstract
Serum therapy remains the only specific treatment against envenoming, but anti-venoms are still prepared by fragmentation of polyclonal antibodies isolated from hyper-immunized horse serum. Most of these anti-venoms are considered to be efficient, but their production is tedious, and their use may be associated with adverse effects. Recombinant antibodies and smaller functional units are now emerging as credible alternatives and constitute a source of still unexploited biomolecules capable of neutralizing venoms. This review will be a walk through the technologies that have recently been applied leading to novel antibody formats with better properties in terms of homogeneity, specific activity and possible safety.
Collapse
|
11
|
Xiong F, Xia L, Wang J, Wu B, Wang D, Yuan L, Cheng Y, Zhu H, Che X, Zhang Q, Zhao G, Wang Y. A high-affinity CDR-grafted antibody against influenza A H5N1 viruses recognizes a conserved epitope of H5 hemagglutinin. PLoS One 2014; 9:e88777. [PMID: 24558425 PMCID: PMC3928294 DOI: 10.1371/journal.pone.0088777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/15/2014] [Indexed: 01/19/2023] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5N1 virus infection is still a potential threat to public health worldwide. While vaccines and antiviral drugs are currently under development, neutralizing antibodies could offer an alternative strategy to prevent and treat H5N1 virus infection. In the present study, we had developed a humanized antibody against H5N1 viruses from mouse-derived hybridoma in order to minimize its immunogenicity for potential clinical application. The humanized antibody hH5M9 was generated by transferring the mouse complementarity determining region (CDR) residues together with four key framework region (FR) residues onto the FR of the human antibody. This humanized antibody exhibited high affinity and specificity comparable to the parental mouse or chimeric counterpart with broad and strong neutralization activity against all H5N1 clades and subclades except for Egypt clades investigated. Furthermore, through epitope mapping we identified a linear epitope on the top region of hemagglutinin (HA) that was H5N1 specific and conserved. Our results for the first time reported a humanized antibody against H5N1 viruses by CDR grafting method. With the expected lower immunogenicity, this humanized antibody was expected to be more efficacious than murine or human-mouse chimeric antibodies for future application in humans.
Collapse
Affiliation(s)
- Feifei Xiong
- School of Life Science and Technology, Tongji University, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Liliang Xia
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Jingfang Wang
- Shanghai Center for Systems Biomedicine, Shanghai Jiaotong University, Shanghai, China
| | - Biao Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Dengyu Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Longfang Yuan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Yating Cheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Hongying Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Xiaoyan Che
- Central Laboratory, Zhujiang Hospital, The Southern Medical University, Guangzhou, China
| | - Qinghua Zhang
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Guoping Zhao
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Ying Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
12
|
Figini M, Orlandi R. New Techniques for the Production of Therapeutic Recombinant Human Monoclonal Antibodies. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/bf03259294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
13
|
Murali R, Greene MI. Structure based antibody-like peptidomimetics. Pharmaceuticals (Basel) 2012; 5:209-35. [PMID: 24288089 PMCID: PMC3763629 DOI: 10.3390/ph5020209] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 01/17/2012] [Accepted: 01/19/2012] [Indexed: 12/22/2022] Open
Abstract
Biologics such as monoclonal antibodies (mAb) and soluble receptors represent new classes of therapeutic agents for treatment of several diseases. High affinity and high specificity biologics can be utilized for variety of clinical purposes. Monoclonal antibodies have been used as diagnostic agents when coupled with radionuclide, immune modulatory agents or in the treatment of cancers. Among other limitations of using large molecules for therapy the actual cost of biologics has become an issue. There is an effort among chemists and biologists to reduce the size of biologics which includes monoclonal antibodies and receptors without a reduction of biological efficacy. Single chain antibody, camel antibodies, Fv fragments are examples of this type of deconstructive process. Small high-affinity peptides have been identified using phage screening. Our laboratory used a structure-based approach to develop small-size peptidomimetics from the three-dimensional structure of proteins with immunoglobulin folds as exemplified by CD4 and antibodies. Peptides derived either from the receptor or their cognate ligand mimics the functions of the parental macromolecule. These constrained peptides not only provide a platform for developing small molecule drugs, but also provide insight into the atomic features of protein-protein interactions. A general overview of the reduction of monoclonal antibodies to small exocyclic peptide and its prospects as a useful diagnostic and as a drug in the treatment of cancer are discussed.
Collapse
Affiliation(s)
- Ramachandran Murali
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, D5091 Davis Building, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
- Department of Pathology and Laboratory of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark I. Greene
- Department of Pathology and Laboratory of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Koti M, Nagy E, Kaushik AK. A single point mutation in framework region 3 of heavy chain affects viral neutralization dynamics of single-chain Fv against bovine herpes virus type 1. Vaccine 2011; 29:7905-12. [DOI: 10.1016/j.vaccine.2011.08.077] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 07/20/2011] [Accepted: 08/15/2011] [Indexed: 10/17/2022]
|
15
|
Rajawat R, Narkar A, Damle A, Kumar GS, Mishra K. A Single-Chain Antibody Fragment Against Human Thyroglobulin: Construction and Evaluation of Immunoreactivity. Hybridoma (Larchmt) 2011; 30:253-9. [DOI: 10.1089/hyb.2010.0114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Rekha Rajawat
- Radiation Medicine Centre, BARC, Tata Memorial Hospital Annex, Parel, Mumbai, India
| | - Archana Narkar
- Radiation Medicine Centre, BARC, Tata Memorial Hospital Annex, Parel, Mumbai, India
| | - Archana Damle
- Radiation Medicine Centre, BARC, Tata Memorial Hospital Annex, Parel, Mumbai, India
| | | | - K.P. Mishra
- RB & HSD, BARC, Trombay, Mumbai, India; currently, United Research Center, Allahabad, India
| |
Collapse
|
16
|
Kasturirangan S, Li L, Emadi S, Boddapati S, Schulz P, Sierks MR. Nanobody specific for oligomeric β-amyloid stabilizes nontoxic form. Neurobiol Aging 2010; 33:1320-8. [PMID: 21067847 DOI: 10.1016/j.neurobiolaging.2010.09.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 09/09/2010] [Accepted: 09/20/2010] [Indexed: 01/05/2023]
Abstract
While accumulation and deposition of beta amyloid (Aβ) is a primary pathological feature of Alzheimer's disease (AD), increasing evidence has implicated small, soluble oligomeric aggregates of Aβ as the neurotoxic species in AD. Reagents that specifically recognize oligomeric morphologies of Aβ have potential diagnostic and therapeutic value. Using a novel biopanning technique that combines phage display technology and atomic force microscopy, we isolated the nanobody E1 against oligomeric Aβ. Here we show that E1 specifically recognizes a small oligomeric Aβ aggregate species distinct from the species recognized by the A4 nanobody previously reported by our group. While E1, like A4, blocks assembly of Aβ into larger oligomeric and fibrillar forms and prevents any Aβ induced toxicity toward neuronal cells, it does so by binding a small Aβ oligomeric species, directing its assembly toward a stable nontoxic conformation. The E1 nanobody selectively recognizes naturally occurring Aβ aggregates produced in human AD brain tissue indicating that a variety of morphologically distinct Aβ aggregate forms occur naturally and that a stable low-n nontoxic Aβ form exists that does not readily aggregate into larger forms. Because E1 catalyses the formation of a stable nontoxic low-n Aβ species it has potential value as a therapeutic reagent for AD which can be used in combination with other therapeutic approaches.
Collapse
|
17
|
Kuan CT, Srivastava N, McLendon RE, Marasco WA, Zalutsky MR, Bigner DD. Recombinant single-chain variable fragment antibodies against extracellular epitopes of human multidrug resistance protein MRP3 for targeting malignant gliomas. Int J Cancer 2010; 127:598-611. [PMID: 19937796 DOI: 10.1002/ijc.25062] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Multidrug resistance protein 3 (MRP3), a multidrug resistance protein identified by serial analysis of gene expression as a glioblastoma multiforme (GBM)-associated molecule, is highly expressed in GBM, but not in normal brain cells. Thus, MRP3 is a candidate for GBM immunotargeting, but to date, no monoclonal antibody has been isolated that can target an extracellular MRP3 epitope. By phage display, we have isolated 3 recombinant, fully human, single-chain Fv (scFv) antibodies, M25, M58 and M89, which specifically react with the extracellular N-terminus of human MRP3. In ELISA, these scFvs reacted only with the peptide used for screening and not with other MRP3-derived peptides. Flow cytometric analysis revealed that these scFv fragments bind specifically to viable human GBM cells displaying different MRP3 expression levels, but not to MRP3-null cells. Furthermore, these scFv antibodies failed to react with tumor cells overexpressing other MRP proteins, including MRP1, MRP2, MRP4 and MRP5. M25 and M58 also bound to viable neurospheres. Iodogen-labeled scFvs demonstrated a yield of 56-76%. The immunoreactive fractions of the radiolabeled M25, M58 and M89 scFvs were 32, 52 and 69%, respectively. M25 exhibited 20% internalization into D2159MG neurospheres, M58, 33% into D54MG cells and M89, 26% into D247MG. Immunohistochemical evaluation of human gliomas to determine the localization of MRP3 antigen using scFvs M25 and M58 showed a dense cytoplasmic and membranous staining pattern. These Fv-based recombinant antibodies, which possess superior tumor penetration capabilities and selectively target tumor cells that express MRP3, may potentially be used in immunotherapy and diagnosis for brain tumors and other cancers.
Collapse
Affiliation(s)
- Chien-Tsun Kuan
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Noninvasive molecular imaging approaches include nuclear, optical, magnetic resonance imaging, computed tomography, ultrasound, and photoacoustic imaging, which require accumulation of a signal delivered by a probe at the target site. Monoclonal antibodies are high affinity molecules that can be used for specific, high signal delivery to cell surface molecules. However, their long circulation time in blood makes them unsuitable as imaging probes. Efforts to improve antibodies pharmacokinetics without compromising affinity and specificity have been made through protein engineering. Antibody variants that differ in antigen binding sites and size have been generated and evaluated as imaging probes to target tissues of interest. Fast clearing fragments, such as single-chain variable fragment (scFv; 25 kDa), with 1 antigen-binding site (monovalent) demonstrated low accumulation in tumors because of the low exposure time to the target. Using scFv as building block to produce larger, bivalent fragments, such as scFv dimers (diabodies, 50 kDa) and scFv-fusion proteins (80 kDa minibodies and 105 kDa scFv-Fc), resulted in higher tumor accumulation because of their longer residence time in blood. Imaging studies with these fragments after radiolabeling have demonstrated excellent, high-contrast images in gamma cameras and positron emission tomography scanners. Several studies have also investigated antibody fragments conjugated to fluorescence (near infrared dyes), bioluminescence (luciferases), and quantum dots for optical imaging and iron oxides nanoparticles for magnetic resonance imaging. However, these studies indicate that there are several factors that influence successful targeting and imaging. These include stability of the antibody fragment, the labeling chemistry (direct or indirect), whether critical residues are modified, the number of antigen expressed on the cell, and whether the target has a rapid recycling rate or internalizes upon binding. The preclinical data presented are compelling, and it is evident that antibody-based molecular imaging tracers will play an important future role in the diagnosis and management of cancer and other diseases.
Collapse
Affiliation(s)
- Tove Olafsen
- UCLA Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA.
| | | |
Collapse
|
19
|
Kularatne SA, Zhou Z, Yang J, Post CB, Low PS. Design, synthesis, and preclinical evaluation of prostate-specific membrane antigen targeted (99m)Tc-radioimaging agents. Mol Pharm 2009; 6:790-800. [PMID: 19361232 DOI: 10.1021/mp9000712] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The high mortality and financial burden associated with prostate cancer can be partly attributed to a lack of sensitive screening methods for detection and staging of the disease. Guided by in silico docking studies using the crystal structure of PSMA, we designed and synthesized a series of PSMA-targeted (99m)Tc-chelate complexes for imaging PSMA-expressing human prostate cancer cells (LNCaP cell line). Of the six targeted radioimaging agents synthesized, three were found to bind LNCaP cells with low nanomolar affinity. Moreover, the same three PSMA-targeted imaging agents were shown to localize primarily to LNCaP tumor xenografts in nu/nu mice, with an average of 9.8 +/- 2.4% injected dose/g tissue accumulating in the tumor and only 0.11% injected dose/g tissue retained in the muscle at 4 h postinjection. Collectively, these high affinity, PSMA-specific radioimaging agents demonstrate significant potential for use in localizing prostate cancer masses, monitoring response to therapy, detecting prostate cancer recurrence following surgery, and selecting patients for subsequent PSMA-targeted chemotherapy.
Collapse
Affiliation(s)
- Sumith A Kularatne
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | |
Collapse
|
20
|
Kularatne SA, Wang K, Santhapuram HKR, Low PS. Prostate-specific membrane antigen targeted imaging and therapy of prostate cancer using a PSMA inhibitor as a homing ligand. Mol Pharm 2009; 6:780-9. [PMID: 19361233 DOI: 10.1021/mp900069d] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Prostate cancer (PCa) is a major cause of mortality and morbidity in Western society today. Current methods for detecting PCa are limited, leaving most early malignancies undiagnosed and sites of metastasis in advanced disease undetected. Major deficiencies also exist in the treatment of PCa, especially metastatic disease. In an effort to improve both detection and therapy of PCa, we have developed a PSMA-targeted ligand that delivers attached imaging and therapeutic agents selectively to PCa cells without targeting normal cells. The PSMA-targeted radioimaging agent (DUPA-(99m)Tc) was found to bind PSMA-positive human PCa cells (LNCaP cell line) with nanomolar affinity (K(D) = 14 nM). Imaging and biodistribution studies revealed that DUPA-(99m)Tc localizes primarily to LNCaP cell tumor xenografts in nu/nu mice (% injected dose/gram = 11.3 at 4 h postinjection; tumor-to-muscle ratio = 75:1). Two PSMA-targeted optical imaging agents (DUPA-FITC and DUPA-rhodamine B) were also shown to efficiently label PCa cells and to internalize and traffic to intracellular endosomes. A PSMA-targeted chemotherapeutic agent (DUPA-TubH) was demonstrated to kill PSMA-positive LNCaP cells in culture (IC(50) = 3 nM) and to eliminate established tumor xenografts in nu/nu mice with no detectable weight loss. Blockade of tumor targeting upon administration of excess PSMA inhibitor (PMPA) and the absence of targeting to PSMA-negative tumors confirmed the specificity of each of the above targeted reagents for PSMA. Tandem use of the imaging and therapeutic agents targeted to the same receptor could allow detection, staging, monitoring, and treatment of PCa with improved accuracy and efficacy.
Collapse
Affiliation(s)
- Sumith A Kularatne
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | |
Collapse
|
21
|
B3(Fab)-streptavidin Tetramer Has Higher Binding Avidity than B3(scFv)-streptavidin Tetramer. B KOREAN CHEM SOC 2009. [DOI: 10.5012/bkcs.2009.30.5.1101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Schirrmann T, Krauss J, Arndt MAE, Rybak SM, Dübel S. Targeted therapeutic RNases (ImmunoRNases). Expert Opin Biol Ther 2008; 9:79-95. [DOI: 10.1517/14712590802631862] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
23
|
High-level expression of a functional humanized single-chain variable fragment antibody against CD25 in Pichia pastoris. Appl Microbiol Biotechnol 2008; 81:33-41. [DOI: 10.1007/s00253-008-1568-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2008] [Revised: 06/04/2008] [Accepted: 06/06/2008] [Indexed: 10/21/2022]
|
24
|
Yang LJ, Hou YC, Bai YJ, Yao LB, Su CZ. Analysis of primary structure and modeling of spatial structure of single-chain variable region of antibody against human gastric cancer. Shijie Huaren Xiaohua Zazhi 2008; 16:2333-2336. [DOI: 10.11569/wcjd.v16.i21.2333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To analyze the sequence of heavy and light chain of the variable region (VH and VL) in the anti-human gastric cancer antibody, select a suitable linker peptide to construct single chain antibody, model the 3-D structure and predict the relationship between its structure and function.
METHODS: Using the sequences of VH and VL, which derived from the phage display library of antibodies against human gastric cancer, a suitable linker peptide was chosen to obtain the primary structure of the single-chain antibody with considering the C-terminal and N-terminal structural character. Furthermore, the 3-D theoretical structure was modeled with computer-guided homology modeling method. The stable spatial conformation of the single-chain variable region was determined with mechanism optimization and molecular dynamic simulation. The structural property and physical-chemical characters were analyzed using the distance geometry, surface electrostatic distribution and solvent accessible surface calculation.
RESULTS: The conformation of the successfully constructed single-chain antibody was stable. Except one amino acid conformation of connecting peptide, others' were reasonable. VL CDR1, CDR2 and VH CDR3 solution accessibility surface area distributed strongly, but VL CDR3, VH CDR1, and CDR2 were comparatively weak. VH CDR3 region had strong electronegativity.
CONCLUSION: The 3-D structure of the created single chain antibody is proved reasonable and reliable.
Collapse
|
25
|
JT George A, Epenetos AA. Section Review Biologicals & Immunologicals: Advances in antibody engineering. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543776.6.5.441] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
26
|
Yang LJ, Hou YC, Yao LB, Su CZ. Analysis of primary structure and modeling of spatial structure of heavy chain variable region of antibody against human gastric cancer. Shijie Huaren Xiaohua Zazhi 2008; 16:413-416. [DOI: 10.11569/wcjd.v16.i4.413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To confirm the primary structure of heavy chain variable regions (VH) of antibody against human gastric cancer based on the sequence analysis method and to model its three-dimensional structure using homology modeling method.
METHODS: The VH gene selected from the phage display library of antibodies against human gastric cancer was sequenced and analyzed. Its three-dimensional structure was modeled with computer homology modeling techniques and optimized using molecular mechanism method.
RESULTS: The sequence of VH was in agreement with the characteristics of the mouse antibody variable region. The FR and CDR were determined by Kabat analysis. The spatial structure of the VH was constructed and optimized with molecular mechanism method to obtain the stable 3-D structure.
CONCLUSION: The primary and three-dimensional structures of VH are reasonable and reliable and lay the theoretical foundation for further biological experiments.
Collapse
|
27
|
Abstract
Immunotherapy, based on mAbs specifically directed against cancer cells, is considered a precious strategy in the fight against cancer because of its selectivity and lack of multidrug resistant effects. However, there are obstacles to the complete success of current immunotherapy such as immune responses to nonhuman or even humanized antibodies and the large size of the antibodies, which hinders their diffusion into bulky tumors. Fully human, small immunoagents, capable of inhibiting tumor growth may overcome these problems and provide safe, highly selective and effective antitumor drugs. An attractive target for immunotherapy is ErbB2, a transmembrane tyrosine kinase receptor, overexpressed on tumor cells of different origin, with a key role in the development of malignancy. An anti-ErbB2 humanized monoclonal (Herceptin) is currently used with success for breast cancer therapy; however, it can engender cardiotoxicity and a high proportion of breast cancer patients are resistant to Herceptin treatment. Anti-ErbB2 immunoagents of human origin, with potentially no or very low immunogenicity have been engineered to assemble 'compact', i.e. reduced size, antibodies, one consisting of a human single-chain antibody fragment (scFv) fused to a human RNase to construct an immunoRNase and the other made up of two human scFv molecules fused to the Fc region of a human IgG1. By choosing a human antibody fragment as the immune moiety and a human RNase as the effector moiety, an immunoRNase would be both nonimmunogenic and nontoxic, as it becomes toxic only when the scFv promotes its internalization by target cells. The alternative strategy of compact antibodies was aimed at producing therapeutic agents with an increased half-life, prolonged tumor retention and the ability to recruit host effector functions. Moreover, the bivalency of compact antibodies can be exploited to construct bispecific antibodies, as well as for other therapeutic applications.
Collapse
Affiliation(s)
- Claudia De Lorenzo
- Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli Federico II, Naples, Italy.
| | | |
Collapse
|
28
|
Leamon CP, Jackman AL. Exploitation of the folate receptor in the management of cancer and inflammatory disease. VITAMINS AND HORMONES 2008; 79:203-33. [PMID: 18804696 DOI: 10.1016/s0083-6729(08)00407-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the last 25+ years, the folate receptor (FR) has emerged as an attractive tumor biomarker with the potential to be exploited for therapeutic purposes. Increasing evidence suggests that this endocytosing protein can functionally mediate the cellular uptake and retention of natural folates, certain antifolates, and folate-drug conjugates; the consequences of the latter two events could result in biological modulation, including (but not limited to) tumor-targeted cytotoxicity. Because its tissue expression profile appears to be somewhat limited to either tissues responsible for whole body retention of folates (e.g., kidney and placenta), or certain pathologic tissues (e.g., tumors or activated macrophages), the FR is believed to be a useful biological target for disease management. Indeed, recent years have been peppered with reports of novel FR-targeted therapies, and many have demonstrated impressive in vivo potency, particularly against tumor xenografts, without the undesirable toxicity that often accompanies nontargeted drug regimens. This chapter will provide essential details on the properties of the FR, including where it is expressed and how it has been successfully manipulated for therapeutic benefit.
Collapse
|
29
|
Wong JYC. Basic immunology of antibody targeted radiotherapy. Int J Radiat Oncol Biol Phys 2006; 66:S8-14. [PMID: 16979446 DOI: 10.1016/j.ijrobp.2005.05.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 05/31/2005] [Accepted: 05/31/2005] [Indexed: 10/24/2022]
Abstract
Antibody targeted radiotherapy brings an important new treatment modality to the radiation oncology clinic. Radiation dose to tumor and normal tissues are determined by a complex interplay of antibody, antigen, tumor, radionuclide, and host-related factors. A basic understanding of these immunologic and physiologic factors is important to optimally utilize this therapy in the clinic. Preclinical and clinical studies need to be continued to broaden our understanding and to develop new strategies to further improve the efficacy of this promising form of targeted therapy.
Collapse
Affiliation(s)
- Jeffrey Y C Wong
- Division of Radiation Oncology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA.
| |
Collapse
|
30
|
Sheikholvaezin A, Eriksson D, Riklund K, Stigbrand T. Construction and Purification of a Covalently Linked Divalent Tandem Single-Chain Fv Antibody Against Placental Alkaline Phosphatase. Hybridoma (Larchmt) 2006; 25:255-63. [PMID: 17044780 DOI: 10.1089/hyb.2006.25.255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Multivalency is a recognized means to increase the functional affinity of single-chain antibody fragments (scFvs) for optimizing tumor uptake at radioimmunotargeting. A unique divalent tandem single-chain Fv antibody [sc(Fv)2], based on the variable regions of the monoclonal antibody (MAb) H7, has now been generated. The antibody differs from other dimeric single-chain constructs in that a linker sequence (L) is introduced between the repeats of VL and VH domains (VL-L-VH-L-VL-L-VH). This construct was expressed as a His-tagged TrxA fusion protein in the Escherichia coli strain Origami B. Following cleavage with AcTev protease, the antibody was obtained in soluble and active form in E. coli and could be purified by Ni-NTA and cation-exchange chromatography. Purity and immunochemical properties were determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), enzyme-linked immunosorbent assay (ELISA), Western blot, and Biacore analyses. The [sc(Fv)2] displayed proper stability and could be purified to homogeneity. This antibody also maintained immunoreactivity at 42 degrees C with only slight decrease at 52 degrees C. The high affinity displayed by the original antibody H7, 6.7 x 10(9) M(-1), was only slightly decreased to 1.2 x 10(9) M(-1) as determined by Biacore. The generation of such a divalent single-chain Fv with a molecular weight around 60 kd would be of value for clinical applications such as radioimmunolocalization and radioimmunotherapy.
Collapse
Affiliation(s)
- Ali Sheikholvaezin
- Department of Clinical Microbiology, Immunology, Umeå University, Umeå, Sweden
| | | | | | | |
Collapse
|
31
|
Liu Y, Zhang W, Cheung LH, Niu T, Wu Q, Li C, Van Pelt CS, Rosenblum MG. The antimelanoma immunocytokine scFvMEL/TNF shows reduced toxicity and potent antitumor activity against human tumor xenografts. Neoplasia 2006; 8:384-93. [PMID: 16790087 PMCID: PMC1592449 DOI: 10.1593/neo.06121] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The immunocytokine scFvMEL/TNF, a fusion protein composed of human tumor necrosis factor (TNF) and a single-chain Fv antibody (scFv) scFvMEL targeting the melanoma gp240 antigen, demonstrates impressive cytotoxic effects against human melanoma cell lines in vitro. Pharmacokinetic studies of 125I-scFvMEL/TNF in BALB/c mice showed that the construct clears from the circulation with a terminal-phase half-life of 17.6 hours after intravenous administration. The maximum tolerated dose of scFvMEL/TNF in nude mice was 4 mg/kg, i.v., on a daily x5 schedule. There were no changes in gross pathology, clinical chemistry, or hematologic parameters in mice treated at doses of up to 3 mg/kg. Therapeutic studies at a dose of 2.5 mg/kg on athymic mice bearing established (approximately 50 mm3) human melanoma A375GFP xenograft tumors transfected with green fluorescent protein demonstrated potent tumor suppression and complete tumor regression of all lesions. There was no subsequent outgrowth of tumors from mice rendered tumor-free. These data show that scFvMEL/TNF can target melanoma cells in vivo and can result in pronounced antimelanoma effects after systemic administration. Toxicology studies indicate the relative safety of this agent at doses that are therapeutically effective and provide guidance to projected phase I starting doses on patients at this schedule.
Collapse
Affiliation(s)
- Yuying Liu
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Simmons DP, Abregu FA, Krishnan UV, Proll DF, Streltsov VA, Doughty L, Hattarki MK, Nuttall SD. Dimerisation strategies for shark IgNAR single domain antibody fragments. J Immunol Methods 2006; 315:171-84. [PMID: 16962608 DOI: 10.1016/j.jim.2006.07.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Revised: 07/07/2006] [Accepted: 07/25/2006] [Indexed: 11/23/2022]
Abstract
Immunoglobulin new antigen receptors (IgNARs) are unique single domain antibodies found in the serum of sharks. The individual variable (VNAR) domains bind antigen independently and are candidates for the smallest antibody-based immune recognition units (approximately 13 kDa). Here, we first isolated and sequenced the cDNA of a mature IgNAR antibody from the spotted wobbegong shark (Orectolobus maculatus) and confirmed the independent nature of the VNAR domains by dynamic light scattering. Second, we asked which of the reported antibody fragment dimerisation strategies could be applied to VNAR domains to produce small bivalent proteins with high functional affinity (avidity). In contrast to single chain Fv (scFv) fragments, separate IgNARs could not be linked into a tandem single chain format, with the resulting proteins exhibited only monovalent binding due solely to interaction of the N-terminal domain with antigen. Similarly, incorporation of C-terminal helix-turn-helix (dhlx) motifs, while resulting in efficiently dimerised protein, resulted in only a modest enhancement of affinity, probably due to an insufficiently long hinge region linking the antibody to the dhlx motif. Finally, generation of mutants containing half-cystine residues at the VNAR C-terminus produced dimeric recombinant proteins exhibiting high functional affinity for the target antigens, but at the cost of 50-fold decreased protein expression levels. This study demonstrates the potential for construction of bivalent or bispecific IgNAR-based binding reagents of relatively small size (approximately 26 kDa), equivalent to a monovalent antibody Fv fragment, for formulation into future diagnostic and therapeutic formats.
Collapse
|
33
|
Vollmers HP, Brändlein S. Natural IgM antibodies: the orphaned molecules in immune surveillance. Adv Drug Deliv Rev 2006; 58:755-65. [PMID: 16820243 DOI: 10.1016/j.addr.2005.08.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2005] [Accepted: 05/06/2006] [Indexed: 12/21/2022]
Abstract
Natural IgM antibodies are typical victims of prejudices which originated in the mid 80 s. Over the years, these molecules were considered as the pariahs among the immune competent molecules and their characteristic properties, like low affinity, cross-reactivity and pentameric structure, were assessed as useless, difficult, nebulous, etc. Today, mainly based on a few scientists' persistent work and the key discoveries on innate immune recognition, natural IgM antibodies are "back on stage". Their role in the immune response against bacteria, viruses, fungi and possibly modified self-components as well as in therapy and diagnosis of malignancies is accepted. All the so far negatively judged features are seen in a different light, e.g. low affinity seems to be good for function and does not exclude specificity, and cross-reactivity is no longer judged as unspecific, but instead as a very economic way of immune recognition. And at last, with the use of natural IgM antibodies, a new field of tumor-specific targets has been encountered, the carbo-neo-epitopes. Therefore, by having learned from nature, the renaissance of natural IgM antibodies opens a new area of cancer therapeutics and diagnostics.
Collapse
Affiliation(s)
- H Peter Vollmers
- Institute for Pathology, University Würzburg, Josef-Schneider-Str. 2, D-97080 Würzburg, Germany.
| | | |
Collapse
|
34
|
Shen YC, Wang XH, Wang XM, Chen ZL, Shen XP, Zhao CC, Li J. High efficient mammalian expression and secretion of a functional humanized single-chain Fv/human interleukin-2 molecules. World J Gastroenterol 2006; 12:3859-65. [PMID: 16804971 PMCID: PMC4087934 DOI: 10.3748/wjg.v12.i24.3859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct and produce a recombinant bispecific humanized single-chain Fv (sFv) /Interleukin-2 (IL-2) fusion protein by using mammalian cells.
METHODS: The sFv/IL-2 protein was genetically engineered, and transfected to mammalian cells to determine whether the mammalian protein folding machinery can produce and secrete active sFv/IL-2 with high efficiency.
RESULTS: The fusion protein was constructed and high efficiently expressed with yields up to 102 ± 4.2 mg/L in culture supernatant of the stably transfected 293 cell line. This recombinant fusion protein consisted of humanized variable heavy (VH) and light (VL) domains of monoclonal antibody (mAb) 520C9 directed against the human HER-2/neu (c-erbB2) proto-oncogene product p185, and human IL-2 connected by polypeptide linker. The fusion protein was shown to retain the immunostimulatory activities of IL-2 as measured by IL-2-dependent cell proliferation and cytotoxicity assays. In addition to its IL-2 activities, this fusion protein also possessed antigen-binding specificity against p185, as determined by indirect ELISA using p185 positive SKOV 3ip1 cells.
CONCLUSION: The large-scale preparation of the recombinant humanized sFv antibody/IL-2 fusion protein is performed with 293 cells. The recombinant humanized sFv antibody/IL-2 fusion protein may provide an effective means of targeting therapeutic doses of IL-2 to p185 positive tumors without increasing systemic toxicity or immunogenicity.
Collapse
Affiliation(s)
- Yue-Chun Shen
- Division of Cardiology, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, 48109, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Adams GP, Tai MS, McCartney JE, Marks JD, Stafford WF, Houston LL, Huston JS, Weiner LM. Avidity-mediated enhancement of in vivo tumor targeting by single-chain Fv dimers. Clin Cancer Res 2006; 12:1599-605. [PMID: 16533787 DOI: 10.1158/1078-0432.ccr-05-2217] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Radiolabeled single-chain Fv (sFv) molecules display highly specific tumor retention in the severe combined immunodeficient (SCID) mouse model; however, the absolute quantity of sFv retained in the tumors is diminished by the rapid renal elimination resulting from the small size of the sFv molecules (Mr 27,000) and by dissociation of the monovalent sFv from tumor-associated antigen. We previously reported significant improvement in tumor retention without a loss of targeting specificity on converting monovalent sFv into divalent [(sFv')2] dimers, linked by a disulfide bond between COOH-terminal cysteinyl peptides engineered into the sFv'. However, our data for enhanced dimer localization in tumors could not distinguish between the contributions of enhanced avidity and increased systemic retention associated with the larger size of 54 kDa [(sFv')2] dimers relative to 27-kDa sFv. In this investigation, we have compared tumor targeting of divalent anti-c-erbB-2/HER2/neu 741F8-1 (sFv')2 homodimers with monovalent 741F8/26-10 (sFv')2 heterodimers (Mr 54,000) and 741F8 sFv monomers (741F8 sFv has binding specificity for erbB-2/HER2/neu and 26-10 sFv specificity for digoxin and related cardiac glycosides). These studies allowed us to distinguish the dominant effect of valency over molecular weight in accounting for the superior tumor retention of 741F8-1 (sFv')2 homodimers. Each of the radioiodinated species was administered i.v. to SCID mice bearing SK-OV-3 human tumor xenografts and tumor localization at 24 hours post i.v. injection was determined for 125I-741F8-1 (sFv')2 (3.57 %ID/g), 125I-741F8/26-10 (sFv')2 (1.13 %ID/g), and 125I-741F8-1 sFv (1.25 %ID/g). These findings substantiate that the improved tumor retention of (sFv')2 homodimers over sFv monomers results from the availability of dual binding sites rather than from the slower systemic clearance of homodimers.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal, Humanized
- Antibody Affinity
- Antibody Specificity
- Dimerization
- Female
- Humans
- Immunoglobulin Fragments/administration & dosage
- Immunoglobulin Fragments/immunology
- Immunoglobulin Fragments/metabolism
- Mice
- Mice, Inbred ICR
- Mice, SCID
- Ovarian Neoplasms/diagnostic imaging
- Ovarian Neoplasms/metabolism
- Radionuclide Imaging
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacokinetics
- Tissue Distribution
- Transplantation, Heterologous
- Trastuzumab
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Gregory P Adams
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Corneillie TM, Whetstone PA, Meares CF. Irreversibly binding anti-metal chelate antibodies: Artificial receptors for pretargeting. J Inorg Biochem 2006; 100:882-90. [PMID: 16487590 DOI: 10.1016/j.jinorgbio.2006.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Revised: 12/14/2005] [Accepted: 01/02/2006] [Indexed: 10/25/2022]
Abstract
Antibodies against metal chelates may potentially be used in biomedical applications such as targeted imaging and therapy of cancer. Highly specific monoclonal antibodies can be developed, but their binding strength needs to be maximized for them to be of practical use. In general, the half-life for dissociation of an antibody-ligand complex is more than an order of magnitude lower than the half-lifetimes for decay of medically useful radiometal ions. Practically speaking, the metal chelate-based ligand will not be bound to its receptor long enough for all of the bound radiometal to decay. A novel approach to this problem is a combination of synthetic chemistry and site-directed mutagenesis, to position a mildly reactive group on the metal chelate adjacent to a complementary reactive group on the antibody when the complex is formed. The partners are chosen to be sufficiently unreactive so that they coexist with other molecules in living systems without undergoing reaction. When the antibody-chelate complex is formed the effective local concentrations of the two groups can be non-physically large, so that a permanent link is formed in the complex even though no reaction occurs when the partners are free in solution.
Collapse
Affiliation(s)
- Todd M Corneillie
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA
| | | | | |
Collapse
|
37
|
Karacay H, Brard PY, Sharkey RM, Chang CH, Rossi EA, McBride WJ, Ragland DR, Horak ID, Goldenberg DM. Therapeutic advantage of pretargeted radioimmunotherapy using a recombinant bispecific antibody in a human colon cancer xenograft. Clin Cancer Res 2006; 11:7879-85. [PMID: 16278412 DOI: 10.1158/1078-0432.ccr-05-1246] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To assess if pretargeting, using a combination of a recombinant bispecific antibody (bsMAb) that binds divalently to carcinoembryonic antigen (CEA) and monovalently to the hapten histamine-succinyl-glycine and a (90)Y-peptide, improves therapeutic efficacy in a human colon cancer-nude mouse xenograft compared with control animals given (90)Y-humanized anti-CEA immunoglobulin G (IgG). EXPERIMENTAL DESIGN Clearance and biodistribution were monitored by whole-body readings and necropsy. Animals were monitored for 34 weeks with a determination of residual disease and renal pathology in survivors. Hematologic toxicity was assessed separately in non-tumor-bearing NIH Swiss mice. RESULTS Hematologic toxicity was severe at doses of 100 to 200 microCi of (90)Y-IgG, yet mild in the pretargeted animals given 500 or 700 microCi of the (90)Y-peptide. Evidence of end-stage renal disease was found at 900 microCi of the pretargeted (90)Y-peptide whereas animals given 700 microCi showed only mild renal pathology, similar to that seen in control animals given (90)Y-IgG. Biodistribution data indicated that the average amount of tumor radioactivity by a 700-microCi dose of the pretargeted peptide over a 96-hour period was increased 2.5-fold (48 microCi/g) compared with 150 microCi of (90)Y-IgG (18.9 microCi/g). At these doses, survival (i.e., time to progression to 2.5 cm(3)) was significantly improved (P < 0.04) compared with (90)Y-IgG, with ablation of about one third of the tumors, whereas viable tumor was present in all of the (90)Y-IgG-treated animals. CONCLUSION Pretargeting increases the amount of radioactivity delivered to colorectal tumors sufficiently to improve the therapeutic index and responses as compared with conventional radioimmunotherapy.
Collapse
Affiliation(s)
- Habibe Karacay
- Center for Molecular Medicine and Immunology and the Garden State Cancer Center, Belleville, NJ 07109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Davern SM, Foote LJ, Lankford TK, Macy SD, Wall MD, Kennel SJ. Identification of an antilaminin-1 scFv that preferentially homes to vascular solid tumors. Cancer Biother Radiopharm 2006; 20:524-33. [PMID: 16248768 DOI: 10.1089/cbr.2005.20.524] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The tumor vasculature and extracellular matrix make attractive targets for distinguishing solid tumors from normal cells. In solid tumors, the processes of angiogenesis and metastasis potentially give rise to unique epitopes not usually accessible in homeostatic organs. Specific targeting of solid tumors for radioimmunotherapy requires that the targeting agent accumulate rapidly and at high levels at the tumor site. This study involved the selection of scFvs that recognize laminin-1 in vitro from the Tomlinson I and J phage display libraries. Selected, purified scFvs were radioiodinated and injected in tumor-bearing mice. One of these, scFv 15-9, exhibited preferential accumulation at subcutaneous tumors when compared to other antilaminin scFvs or to a control scFv. Autoradiographic analysis indicated that scFv15- 9 also displayed a higher vessel:parenchyma ratio than did two other antilaminin scFvs, scFv 15-6 and scFv 15-1, indicating a preferential accumulation of scFv 15-9 around vessel structures. Immunohistochemistry confirmed that scFv 15-9 accumulated at sites of endothelial cells lining vessel structures where significant levels of laminin were present. These data demonstrate that scFv 15-9 binds to a specific epitope on laminin and has potential for tumor endoradiotherapy in subcutaneous tumors.
Collapse
Affiliation(s)
- Sandra M Davern
- Oak Ridge National Laboratory, Building 45005, Bethel Valley Road, Oak Ridge, TN 37831, USA
| | | | | | | | | | | |
Collapse
|
39
|
Erlandsson A, Eriksson D, Johansson L, Riklund K, Stigbrand T, Sundström BE. In vivo clearing of idiotypic antibodies with antiidiotypic antibodies and their derivatives. Mol Immunol 2006; 43:599-606. [PMID: 15978666 DOI: 10.1016/j.molimm.2005.04.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Indexed: 11/20/2022]
Abstract
At immunolocalization of experimental tumors, idiotypic monoclonal antibodies, such as TS1 against cytokeratin 8, can be used to carry and deposit in vivo terapeutics in the tumor. These carriers also remain in the circulation and may cause negative side-effects in other tissues. In this report, several derivatives of the antiidiotypic antibody alphaTS1 were produced and tested for their clearing capacity of the idiotypic carrier antibody TS1. Intact monoclonal alphaTS1, scFv of a alphaTS1 and alphaTS1 Fab'2 and fragments were produced by recombinant technology or by cleavage with Ficin. The scFv was tailored by use of the variable domain genes of the light and heavy chain from the hybridoma clone in combination with a (Gly4Ser)3-linker, followed by expression in E. coli. When tested for clearing capacity, the intact divalent antiidiotypic IgG was found to be the most efficient. The divalent and the monovalent Fab fragment also demonstrated significant clearing, but lower than the intact antiidiotypic IgG. The alphaTS1 scFv antibody when injected separately was not found to clear the idiotype, but could do so when preincubated with the idiotype. Rapid excretion and in vivo instability of this low molecular weight antibody fragment may be the major reasons. Similar results were obtained when the system was reversed and the 131I-labeled antiidiotype IgG was cleared with the idiotype fragment. It is concluded that both intact antiidiotypic IgG, and Fab'2 fragments are able to clear the idiotypic antibodies. The experimental data support the conclusion that the Fc parts from both the idiotype and the antiidiotype may contribute to this elimination.
Collapse
Affiliation(s)
- Ann Erlandsson
- Department of Immunology, Umeå University, S-901 85 Umeå, Sweden
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Therapeutic antibodies are well established as an important class of drugs in modern medicine. The exquisite specificity and affinity for a specific target offered by antibodies has also encouraged their development as delivery vehicles for agents such as radionuclides to target tissues, for radioimmunoimaging and radioimmunotherapy. Specifically, in nuclear medicine, radionuclide-conjugated antibody molecules make it possible to image diseased loci with greater sensitivity than other imaging modalities such as magnetic resonance imaging. Furthermore, two radionuclide-conjugated antibodies have recently been approved for the therapy of non-Hodgkin's lymphoma. However, optimal implementation of antibodies has been limited by the extended circulation persistence that is characteristic of native antibodies, which is responsible for increased background activity in radioimmunoimaging applications and dose-related normal organ toxicities in radioimmunotherapy. In this article the current status of radiolabelled intact antibodies is reviewed, focusing on strategies to improve their pharmacokinetic properties to suit a desired application. Examples from the literature that represent different approaches to accomplishing this task in terms of their successes as well as limitations, and perspectives for the future are discussed.
Collapse
Affiliation(s)
- Vania Kenanova
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California-Los Angeles, 700 Westwood Plaza, Los Angeles, CA 90095, USA
| | | |
Collapse
|
41
|
Abstract
The past five years have witnessed the emergence of monoclonal antibodies as important therapeutics for cancer treatment. Lower toxicity for antibodies versus small molecules, the potential for increased efficacy by conjugation to radioisotopes and cellular toxins, or the ability to exploit immune cell functions have led to clinical performances on par or superior to conventional drug therapies. This review outlines the various immunoglobulin design strategies currently available, techniques used to reduce Ig antigenicity and toxicity, and points to consider during the manufacture of antibodies for use in clinical oncology.
Collapse
Affiliation(s)
- Jerome E Tanner
- TanTec Biosystems Inc., Dollard-Des-Ormeaux, Montreal, Quebec, Canada.
| |
Collapse
|
42
|
Yang X, Hu W, Li F, Xia H, Zhang Z. Gene cloning, bacterial expression, in vitro refolding, and characterization of a single-chain Fv antibody against PreS1(21-47) fragment of HBsAg. Protein Expr Purif 2005; 41:341-8. [PMID: 15866720 DOI: 10.1016/j.pep.2005.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Revised: 02/05/2005] [Indexed: 10/25/2022]
Abstract
The murine monoclonal antibody 125E11 is an IgG which recognizes PreS1(21-47) fragment of large hepatitis B surface antigen. It has been successfully used for clinical detection of HBV virion in serum of hepatitis B patients. In present study, the genes of variable region in heavy chain (VH) and light chain (VL) of 125E11 have been cloned. Sequence analysis of cloned VH gene and VL gene showed that they had general characterization of immunoglobin variable region genes. According to Kabat classification, VH gene and VL gene belong to VH10 family, subgroup IIID and Vkappa family subgroup I, respectively. An expression vector of 125E11 single-chain Fv antibody fusion protein, in which VH and VL peptide were connected by a flexible linker (Gly(4)Ser)(3), was constructed. The scFv fusion protein was highly expressed in Escherichia coli mainly in inclusion body form. Using urea and pH gradient gel filtration method, the refolding of scFv was efficiently achieved. The refolding efficiency reached about 11% and 2.7 mg refolded scFv was obtained from 1L of culture. The binding activity and specificity of 125E11 scFv against PreS1(21-47)-containing antigen were also analyzed.
Collapse
Affiliation(s)
- Xinxiu Yang
- Key Laboratory of Proteomics, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | |
Collapse
|
43
|
Oriuchi N, Higuchi T, Hanaoka H, Iida Y, Endo K. Current status of cancer therapy with radiolabeled monoclonal antibody. Ann Nucl Med 2005; 19:355-65. [PMID: 16164191 DOI: 10.1007/bf03027399] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Molecular targeting therapy has become a relevant therapeutic strategy for cancer. There are several monoclonal antibodies used for the treatment of malignant tumors. Radioimmunoconjugate is composed of antibody and radionuclide showing a synergistic effect of radiation and immunemediated cellular toxicity and thereby enabling increased efficacy and minimizing toxicity. Radioimmunotherapy using 131I- and 90Y-labeled anti-CD20 monoclonal antibodies is now indicated for the treatment of patients with CD20 antigen-expressing relapsed or refractory, low-grade or transformed non-Hodgkin's lymphoma (NHL), including patients who are refractory to anti-CD20 monoclonal antibody (rituximab) therapy in the United States. It has been exhibiting favorable anti-tumor efficacy in patients with NHL as compared with rituximab. Myelosuppression is the main side effect associated with the radioimmunotherapy but is usually reversible, and nonhematologic adverse reactions are mild to moderate. Following the impressive results of therapy using radiolabeled monoclonal antibodies for NHL, radioimmunotherapy for solid tumors has been examined; however, the results were unfavorable and did warrant further clinical trials as a single agent. Future studies on radioimmunotherapy for solid tumors should focus on the new strategies of targeting such as locoregional administration for intraperitoneal dissemination, and combination therapy with chemotherapy or cytostatic therapy. Although radioimmunotherapy for NHL has shown excellent results comparable to aggressive chemotherapy without severe adverse effects, additional clinical trials should be performed to define the proper role of radioimmunoconjugates as a relevant strategy for cure of NHL.
Collapse
Affiliation(s)
- Noboru Oriuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Japan.
| | | | | | | | | |
Collapse
|
44
|
Obata A, Kasamatsu S, Lewis JS, Furukawa T, Takamatsu S, Toyohara J, Asai T, Welch MJ, Adams SG, Saji H, Yonekura Y, Fujibayashi Y. Basic characterization of 64Cu-ATSM as a radiotherapy agent. Nucl Med Biol 2005; 32:21-8. [PMID: 15691658 DOI: 10.1016/j.nucmedbio.2004.08.012] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2004] [Revised: 07/29/2004] [Accepted: 08/20/2004] [Indexed: 01/06/2023]
Abstract
64Cu-diacetyl-bis(N4-methylthiosemicarbazone) (64Cu-ATSM) is a promising radiotherapy agent for the treatment of hypoxic tumors. In an attempt to elucidate the radiobiological basis of 64Cu-ATSM radiotherapy, we have investigated the cellular response patterns in vitro cell line models. Cells were incubated with 64Cu-ATSM, and the dose-response curves were obtained by performing a clonogenic survival assay. Radiation-induced damage in DNA was evaluated using the alkali comet assay and apoptotic cells were detected using Annexin V-FITC and propidium iodide staining methods. Washout rate and subcellular distribution of 64Cu in cells were investigated to further assess the effectiveness of 64Cu-ATSM therapy on a molecular basis. A direct comparison of subcellular localization of Cu-ATSM was made with the flow tracer analog Cu-pyruvladehyde-bis(N4-methylthiosemicarbazone). In this study, 64Cu-ATSM was shown to reduce the clonogenic survival rate of tumor cells in a dose-dependent manner. Under hypoxic conditions, cells took up 64Cu-ATSM and radioactive 64Cu was highly accumulated in the cells. In the 64Cu-ATSM-treated cells, DNA damage by the radiation emitted from 64Cu was detected, and inhibition of cell proliferation and induction of apoptosis was observed at 24 and 36 h after the treatment. The typical features of postmitotic apoptosis induced by radiation were observed following 64Cu-ATSM treatment. The majority of the 64Cu taken up into the cells remained in the postmitochondrial supernatant (the cellular residue after removal of the nuclei and mitochondria), which indicates that the beta- particle emitted from 64Cu may be as effective as the Auger electrons in 64Cu-ATSM therapy. These data allow us to postulate that 64Cu-ATSM will be able to attack the hypoxic tumor cells directly, as well as potentially affecting the peripheral nonhypoxic regions indirectly by the beta- particle decay of 64Cu.
Collapse
Affiliation(s)
- Atsushi Obata
- Biomedical Imaging Research Center, University of Fukui, Matsuoka, Fukui 910-1193, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lombardi A, Sperandei M, Cantale C, Giacomini P, Galeffi P. Functional expression of a single-chain antibody specific for the HER2 human oncogene in a bacterial reducing environment. Protein Expr Purif 2005; 44:10-5. [PMID: 16125411 DOI: 10.1016/j.pep.2005.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2005] [Revised: 05/16/2005] [Accepted: 05/17/2005] [Indexed: 11/15/2022]
Abstract
Recombinant antibody fragments represent useful tools for cancer diagnosis and therapy. Aberrant expression of the HER2 receptor is implicated in metastatic breast and ovary cancers, two malignancies with a high prevalence in young women. In this study, we focussed on a single-chain fragment of variable antibody regions specific for HER2 (scFv800E6) that can be expressed in a functional form in the cytoplasm of Escherichia coli. ScFv800E6 was extracted from bacterial cultures following induction at different temperatures and purified. The yield of both soluble and insoluble forms was measured. We found that scFv800E6 was functional when expressed in the soluble fraction in the bacteria cytosol. In addition, scFv800E6 extracted from inclusion bodies was easily refolded and largely recovered its functionality. Thus, scFv800E6 is intrinsically capable of efficient and functional folding in a reducing environment and represents one of the few described antibody fragments with a framework well adapted for cytoplasmic expression.
Collapse
Affiliation(s)
- Alessio Lombardi
- ENEA, BIOTEC-GEN Unit, CR Casaccia, Via Anguillarese 301, 00060 Rome, Italy
| | | | | | | | | |
Collapse
|
46
|
Emberson LM, Trivett AJ, Blower PJ, Nicholls PJ. Expression of an anti-CD33 single-chain antibody by Pichia pastoris. J Immunol Methods 2005; 305:135-51. [PMID: 16139294 DOI: 10.1016/j.jim.2005.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2004] [Revised: 03/10/2005] [Accepted: 04/06/2005] [Indexed: 11/23/2022]
Abstract
CD33 is a cell surface glycoprotein expressed on cells of myelomonocytic lineage, leukaemic cells, but not haematopoietic stem cells. By virtue of its expression pattern, CD33 has become a popular target for new immunotherapeutic approaches to treat acute myeloid leukaemia. The methylotrophic yeast Pichia pastoris strain KM71H was used to produce an anti-CD33 single chain variable fragment (scFv), with the intention of conjugation to a radioisotope, for therapeutic use. To direct secreted expression of the anti-CD33-scFv the alpha-mating factor secretory signal sequence (alpha-MF) was used, with constructs containing a complete (CS) and incomplete (INCS) cleavage site to accommodate the potential outcomes of dibasic endopeptidase, Kex2, and dipeptidyl amino peptidase, Ste13, processing. The anti-CD33-scFv was expressed in BMMY cultures using both constructs, with a final yield of 48 mg/l (CS) and 11 mg/l (INCS). N-terminal sequencing showed that the CS-scFv had not been cleaved by Ste13, leaving amino acids EAEA at the N-terminus. The INCS-scFv construct produced a mixture of 50% authentic scFv and 50% with 11 amino acids from the alpha-MF remaining at the N-terminus. Despite the aberrations in alpha-MF processing, the anti-CD33-scFv's produced from both constructs were found to be functional. Flow cytometry and Biacore analysis demonstrated binding to target antigen CD33 on the surface of human leukaemic cell line HL-60, and to recombinant soluble CD33 respectively.
Collapse
MESH Headings
- Antigens, CD/immunology
- Antigens, Differentiation, Myelomonocytic/immunology
- Binding Sites, Antibody/immunology
- Cloning, Molecular
- Flow Cytometry
- HL-60 Cells
- Humans
- Immunoglobulin Fragments/biosynthesis
- Immunoglobulin Fragments/genetics
- Immunoglobulin Fragments/immunology
- Immunoglobulin Variable Region/biosynthesis
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Immunotherapy/methods
- Jurkat Cells
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Pichia/genetics
- Pichia/immunology
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Sequence Analysis, Protein
- Sialic Acid Binding Ig-like Lectin 3
- Surface Plasmon Resonance
- Transformation, Genetic
Collapse
Affiliation(s)
- Louise M Emberson
- Department of Biosciences, University of Kent, Giles Lane, Canterbury, Kent, CT2 7NJ, UK
| | | | | | | |
Collapse
|
47
|
Abstract
One of the major obstacles in the development of bispecific antibodies (BsAb) has been the difficulty of producing the materials in sufficient quality and quantity by traditional technologies, such as the hybrid hybridoma and chemical conjugation methods. In contrast to the rapid and significant progress in the development of recombinant BsAb fragments (such as diabody and tandem single chain Fv), the successful design and production of full length IgG-like BsAb has been limited. Compared to smaller fragments, IgG-like BsAb have long serum half-life and are capable of supporting secondary immune functions, such as antibody-dependent cellular cytotoxicity and complement-mediated cytotoxicity. The development of IgG-like BsAb as therapeutic agents will depend heavily on our research progress in the design of recombinant BsAb constructs (or formats) and production efficiency. This review will focus on recent advances in various recombinant approaches to the engineering and production of IgG-like BsAb.
Collapse
Affiliation(s)
- Jonathan S Marvin
- Department of Antibody Technology, ImClone Systems Incorporated, New York, NY 10014, USA
| | | |
Collapse
|
48
|
Wittel UA, Jain M, Goel A, Baranowska-Kortylewicz J, Kurizaki T, Chauhan SC, Agrawal DK, Colcher D, Batra SK. Engineering and characterization of a divalent single-chain Fv angiotensin II fusion construct of the monoclonal antibody CC49. Biochem Biophys Res Commun 2005; 329:168-76. [PMID: 15721289 DOI: 10.1016/j.bbrc.2005.01.101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Indexed: 11/22/2022]
Abstract
For the therapy of solid tumors, co-administration of angiotensin II (AngII) results in an increased uptake of drugs into the tumor interstitium. We have engineered a dimeric sc(Fv)(2)-AngII fusion construct that combines the superior kinetics of covalent dimeric scFvs [sc(Fv)(2)], recognizing the pancarcinoma tumor-associated antigen 72 (TAG-72), with the advantageous intrinsic activity of AngII. The binding characteristics of the fusion construct were unaltered by the addition of the AngII sequence [affinity constant K(A) 1.18 x 10(7) and 8.42 x 10(6) M(-1) for sc(Fv)(2) and sc(Fv)(2)-AngII, respectively]. The binding of the fusion construct to the angiotensin receptor (AT(1)) was similar to AngII, and the arterial contraction was 16 +/- 1% of the response observed with norepinephrine. In animal studies, the radiolabeled sc(Fv)(2)-AngII construct exhibited similar uptake and a more homogeneous distribution within the tumor as compared to sc(Fv)(2).
Collapse
Affiliation(s)
- Uwe A Wittel
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lillo AM, Sun C, Gao C, Ditzel H, Parrish J, Gauss CM, Moss J, Felding-Habermann B, Wirsching P, Boger DL, Janda KD. A human single-chain antibody specific for integrin alpha3beta1 capable of cell internalization and delivery of antitumor agents. ACTA ACUST UNITED AC 2005; 11:897-906. [PMID: 15271348 DOI: 10.1016/j.chembiol.2004.04.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2004] [Revised: 03/26/2004] [Accepted: 04/07/2004] [Indexed: 11/26/2022]
Abstract
Selective antitumor chemotherapy can be achieved by using antibody-drug conjugates that recognize surface proteins upregulated in cancer cells. One such receptor is integrin alpha3beta1, which is overexpressed on malignant melanoma, prostate carcinoma, and glioma cells. We previously identified a human single-chain Fv antibody (scFv), denoted Pan10, specific for integrin alpha3beta1 that is internalized by human pancreatic cancer cells. Herein, we describe the chemical introduction of reactive thiol groups onto Pan10, the specific conjugation of the modified scFv to maleimide-derivatized analogs of the potent cytotoxic agent duocarmycin SA, and the properties of the resultant conjugates. Our findings provide evidence that Pan10-drug conjugates maintain the internalizing capacity of the parent scFv and are cytotoxic at nanomolar concentrations. Our Pan10-drug conjugates may be promising candidates for targeted chemotherapy of malignant diseases associated with overexpression of integrin alpha3beta1.
Collapse
Affiliation(s)
- Antonietta M Lillo
- Department of Chemistry, The Scripps Research Institute and The Skaggs Institute for Chemical Biology, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Boskovitz A, Wikstrand CJ, Kuan CT, Zalutsky MR, Reardon DA, Bigner DD. Monoclonal antibodies for brain tumour treatment. Expert Opin Biol Ther 2005; 4:1453-71. [PMID: 15335313 DOI: 10.1517/14712598.4.9.1453] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Conventional treatment of brain tumours includes surgical, radiotherapeutic and chemotherapeutic modalities. Nonetheless, the outcome of patients with brain tumours, in particular glioblastoma, remains poor. Immunotherapy with armed or unarmed monoclonal antibodies targeting tumour-specific antigens has emerged in the last two decades as a novel potential adjuvant treatment for all types of neoplasia. Many challenges to its implementation as a safe and viable therapy for brain tumours still need to be addressed; nevertheless, results from ongoing Phase I/II clinical trials are encouraging, as disease stabilisation and patient survival prolongation have been observed. Advances in preclinical and clinical research indicate that treatment of brain tumours with monoclonal antibodies can be increasingly adjusted to the characteristics of the targeted tumour and its environment. This aspect relies on the careful selection of the target antigen and corresponding specific monoclonal antibody, and antibody format (size, class, affinity), conjugation to the appropriate toxin or radioactive isotope (half-life, range), and proper compartmental administration.
Collapse
Affiliation(s)
- Abraham Boskovitz
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|