1
|
Nagy Á, Ulmert D, Zedan W, Storey CM, Park J, Geres S, Lückerath K, Sjöström K, Westin H, Peekhaus N, Thorek DL, Karlström AE, Altai M. Impact of site-specific conjugation strategies on the pharmacokinetics of antibody conjugated radiotherapeutics. Eur J Med Chem 2024; 280:116927. [PMID: 39378827 DOI: 10.1016/j.ejmech.2024.116927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/21/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
Antibody radionuclide conjugates are an emerging modality for targeted imaging and potent therapy of disseminated disease. Coupling of radionuclides to monoclonal antibodies (mAbs) is typically achieved by applying non-site-specific labelling techniques. With the ambition of reducing variability, increasing labelling efficacy and stability, several site-specific conjugation strategies have been developed in recent years for toxin- and fluorophore-mAb conjugates. In this study, we studied two site-specific labelling strategies for the conjugation of the macrocyclic chelating agent, DOTA, to the anti-Leucine Rich Repeat Containing 15 (LRRC15) mAb DUNP19. Specifically, one approach utilized a DOTA-bearing peptide (FcIII) with a strong affinity for the fragment crystallizable (Fc) domain of the human IgG1 of DUNP19 (DUNP19LF-FcIII-DOTASS), while the other leveraged a chemo-enzymatic technique to substitute the N-linked bi-antennary oligosaccharides in the human IgG1 Fc domain with DOTA (DUNP19LF-gly-DOTASS). To assess if these methods impact the antibody's binding properties and targeting efficacy, comparative in vitro and in vivo studies of the generated DUNP19-conjugates were performed. While the LRRC15 binding of both radioimmunoconjugates remained intact, the conjugation methods had different impacts on their abilities to interact with FcRn and FcγRs. In vitro assessments of DUNP19LF-FcIII-DOTASS and DUNP19LF-gly-DOTASS demonstrated markedly decreased affinity for FcRn and FcγRIIIa (CD16), respectively. DUNP19LF-FcIII-DOTASS demonstrated increased blood and tissue kinetics in vivo, confirming loss of FcRn binding. While the ablated FcγR interaction of DUNP19LF-gly-DOTASS had no immediate impact on in vivo biodistribution, reduced immunotherapeutic effect can be expected in future studies as a result of reduced NK-cells interaction. In conclusion, our findings underscore the necessity for meticulous consideration and evaluation of mAb labelling strategies, extending beyond mere conjugation efficiency and radiolabeling yields. Notably, site-specific labelling methods were found to significantly influence the immunological impact of Fc interactions. Therefore, it is of paramount importance to consider the intended diagnostic or therapeutic application of the construct and to adopt conjugation strategies that ensure the preservation of critical pharmacological properties and functionality of the antibody in use.
Collapse
Affiliation(s)
- Ábel Nagy
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - David Ulmert
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Wahed Zedan
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Claire M Storey
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Julie Park
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Susanne Geres
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, DKTK, Essen, Germany
| | | | | | - Norbert Peekhaus
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Daniel Lj Thorek
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA; Oncologic Imaging Program, Siteman Cancer Center, St. Louis, Missouri, USA
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - Mohamed Altai
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden.
| |
Collapse
|
2
|
Tagawa H, Saeki R, Yamamoto C, Tanito K, Tanaka C, Munekawa S, Nii T, Kishimura A, Murakami H, Mori T, Katayama Y. The effect of Fc region affinity of protein-based antibody-recruiting molecules on antibody-dependent cellular cytotoxicity. RSC Adv 2024; 14:22860-22866. [PMID: 39040702 PMCID: PMC11262565 DOI: 10.1039/d4ra03391d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/06/2024] [Indexed: 07/24/2024] Open
Abstract
Previously, we reported anticancer molecules, Fc-binding antibody-recruiting molecules (Fc-ARMs), which crosslink proteins on cancer cells with endogenous immunoglobulin Gs (IgGs) via their Fc region. The mobilized IgGs on cancer cells can accommodate natural killer cells to induce antibody-dependent cellular cytotoxicity (ADCC). Because previous Fc-ARMs utilized Fc-binding peptides, their affinity to IgGs is weak, which resulted in the limited induction capability of ADCC. Previous Fc-ARMs also unitized small molecular ligands to cancer cells, which limited their universal applicability to any cancer cells. A recent study reported that protein-based Fc-ARMs might overcome the issues associated with non-proteinous Fc-ARMs. Here, we examined the universality of a protein-based Fc-ARM by replacing its tumor-binding domain with a human epidermal growth factor receptor 2 (HER2)-specific affibody (ZHER2:342). We also examined the requirement of its Fc-binding domain affinity. We found that the Fc-ARMs accepted an affibody as a tumor-binding domain to induce ADCC. Furthermore, the required residence time of the complex between Fc-ARM and IgG was ∼102 min, which was comparable to that when monoclonal antibodies bind to their specific antigens. However, we found that the extent of ADCC induced by Fc-ARM was lower than that of conventional IgG-mediated ADCC, indicating that further enhancement of the affinity of the antibody-binding terminus and tumor-binding terminus of Fc-ARM may be needed to achieve ADCC equivalent to that of conventional IgG-mediated ADCC.
Collapse
Affiliation(s)
- Hiroshi Tagawa
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Riku Saeki
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Chihaya Yamamoto
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Kenta Tanito
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Chihiro Tanaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Furo-cho, Chikusa-ku Nagoya 464-8603 Japan
| | - Shoki Munekawa
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Teruki Nii
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Akihiro Kishimura
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Center for Future Chemistry, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- International Research Center for Molecular Systems, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Hiroshi Murakami
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Furo-cho, Chikusa-ku Nagoya 464-8603 Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University Furo-cho, Chikusa-ku Nagoya 464-8603 Japan
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Center for Future Chemistry, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Center for Future Chemistry, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| |
Collapse
|
3
|
Amash A, Volkers G, Farber P, Griffin D, Davison KS, Goodman A, Tonikian R, Yamniuk A, Barnhart B, Jacobs T. Developability considerations for bispecific and multispecific antibodies. MAbs 2024; 16:2394229. [PMID: 39189686 DOI: 10.1080/19420862.2024.2394229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024] Open
Abstract
Bispecific antibodies (bsAb) and multispecific antibodies (msAb) encompass a diverse variety of formats that can concurrently bind multiple epitopes, unlocking mechanisms to address previously difficult-to-treat or incurable diseases. Early assessment of candidate developability enables demotion of antibodies with low potential and promotion of the most promising candidates for further development. Protein-based therapies have a stringent set of developability requirements in order to be competitive (e.g. high-concentration formulation, and long half-life) and their assessment requires a robust toolkit of methods, few of which are validated for interrogating bsAbs/msAbs. Important considerations when assessing the developability of bsAbs/msAbs include their molecular format, likelihood for immunogenicity, specificity, stability, and potential for high-volume production. Here, we summarize the critical aspects of developability assessment, and provide guidance on how to develop a comprehensive plan tailored to a given bsAb/msAb.
Collapse
Affiliation(s)
- Alaa Amash
- AbCellera Biologics Inc, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | - Tim Jacobs
- AbCellera Biologics Inc, Vancouver, BC, Canada
| |
Collapse
|
4
|
Delaney S, Nagy Á, Karlström AE, Zeglis BM. Site-Specific Photoaffinity Bioconjugation for the Creation of 89Zr-Labeled Radioimmunoconjugates. Mol Imaging Biol 2023; 25:1104-1114. [PMID: 37052759 PMCID: PMC10570397 DOI: 10.1007/s11307-023-01818-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/14/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023]
Abstract
PURPOSE Site-specific approaches to bioconjugation produce well-defined and homogeneous immunoconjugates with potential for superior in vivo behavior compared to analogs synthesized using traditional, stochastic methods. The possibility of incorporating photoaffinity chemistry into a site-specific bioconjugation strategy is particularly enticing, as it could simplify and accelerate the preparation of homogeneous immunoconjugates for the clinic. In this investigation, we report the synthesis, in vitro characterization, and in vivo evaluation of a site-specifically modified, 89Zr-labeled radioimmunoconjugate created via the reaction between an mAb and an Fc-binding protein bearing a photoactivatable 4-benzoylphenylalanine residue. PROCEDURES A variant of the Fc-binding Z domain of protein A containing a photoactivatable, 4-benzoylphenylalanine residue - Z(35BPA) - was modified with desferrioxamine (DFO), combined with the A33 antigen-targeting mAb huA33, and irradiated with UV light. The resulting immunoconjugate - DFOZ(35BPA)-huA33 - was purified and characterized via SDS-PAGE, MALDI-ToF mass spectrometry, surface plasmon resonance, and flow cytometry. The radiolabeling of DFOZ(35BPA)-huA33 was optimized to produce [89Zr]Zr-DFOZ(35BPA)-huA33, and the immunoreactivity of the radioimmunoconjugate was determined with SW1222 human colorectal cancer cells. Finally, the in vivo performance of [89Zr]Zr-DFOZ(35BPA)-huA33 in mice bearing subcutaneous SW1222 xenografts was interrogated via PET imaging and biodistribution experiments and compared to that of a stochastically labeled control radioimmunoconjugate, [89Zr]Zr-DFO-huA33. RESULTS HuA33 was site-specifically modified with Z(35BPA)-DFO, producing an immunoconjugate with on average 1 DFO/mAb, high in vitro stability, and high affinity for its target. [89Zr]Zr-DFOZ(35BPA)-huA33 was synthesized in 95% radiochemical yield and exhibited a specific activity of 2 mCi/mg and an immunoreactive fraction of ~ 0.85. PET imaging and biodistribution experiments revealed that high concentrations of the radioimmunoconjugate accumulated in tumor tissue (i.e., ~ 40%ID/g at 120 h p.i.) but also that the Z(35BPA)-bearing immunoPET probe produced higher uptake in the liver, spleen, and kidneys than its stochastically modified cousin, [89Zr]Zr-DFO-huA33. CONCLUSIONS Photoaffinity chemistry and an Fc-binding variant of the Z domain were successfully leveraged to create a novel site-specific strategy for the synthesis of radioimmunoconjugates. The probe synthesized using this method - DFOZ(35BPA)-huA33 - was well-defined and homogeneous, and the resulting radioimmunoconjugate ([89Zr]Zr-DFOZ(35BPA)-huA33) boasted high specific activity, stability, and immunoreactivity. While the site-specifically modified radioimmunoconjugate produced high activity concentrations in tumor tissue, it also yielded higher uptake in healthy organs than a stochastically modified analog, suggesting that optimization of this system is necessary prior to clinical translation.
Collapse
Affiliation(s)
- Samantha Delaney
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ábel Nagy
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Brian M Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA.
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
5
|
Abstract
Therapeutic fusion proteins are a class of hybrid constructs that combine distinct biomolecules into a single platform with the additive effects of the components. The ability to fuse two unrelated proteins provides a means to localize mechanisms to better treat a range of diseases. Fusion proteins can be designed to impart diverse functions, including increasing half-life, providing targeting, and enabling sustained signaling. Of these, half-life extenders, which are fused to a therapeutic protein to increase exposure, are the most established group of fusion proteins, with many clinical successes. Rapid advances in antibody and antibody-derivative technology have enabled the fusion of targeting domains with therapeutic proteins. An emerging group of therapeutic fusion proteins has two separate active functions. Although most research for therapeutic fusion proteins focuses on cancer, prior successes provide a foundation for studies into other diseases as well. The exponential emergence of biopharmaceuticals gives precedence for increased research into therapeutic fusion proteins for a multitude of diseases.
Collapse
Affiliation(s)
- Morgan C Marsh
- Department of Molecular Pharmaceutics, University of Utah, 30 South 2000 East, Room 301, Salt Lake City, Utah, 84112, USA
| | - Shawn C Owen
- Department of Molecular Pharmaceutics, University of Utah, 30 South 2000 East, Room 301, Salt Lake City, Utah, 84112, USA.
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, 84112, USA.
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84112, USA.
| |
Collapse
|
6
|
Wong JYK, Ekanayake AI, Kharchenko S, Kirberger SE, Qiu R, Kelich P, Sarkar S, Li J, Fernandez KX, Alvizo-Paez ER, Miao J, Kalhor-Monfared S, John JD, Kang H, Choi H, Nuss JM, Vederas JC, Lin YS, Macauley MS, Vukovic L, Pomerantz WCK, Derda R. Genetically encoded discovery of perfluoroaryl macrocycles that bind to albumin and exhibit extended circulation in vivo. Nat Commun 2023; 14:5654. [PMID: 37704629 PMCID: PMC10499988 DOI: 10.1038/s41467-023-41427-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 08/17/2023] [Indexed: 09/15/2023] Open
Abstract
Peptide-based therapeutics have gained attention as promising therapeutic modalities, however, their prevalent drawback is poor circulation half-life in vivo. In this paper, we report the selection of albumin-binding macrocyclic peptides from genetically encoded libraries of peptides modified by perfluoroaryl-cysteine SNAr chemistry, with decafluoro-diphenylsulfone (DFS). Testing of the binding of the selected peptides to albumin identified SICRFFC as the lead sequence. We replaced DFS with isosteric pentafluorophenyl sulfide (PFS) and the PFS-SICRFFCGG exhibited KD = 4-6 µM towards human serum albumin. When injected in mice, the concentration of the PFS-SICRFFCGG in plasma was indistinguishable from the reference peptide, SA-21. More importantly, a conjugate of PFS-SICRFFCGG and peptide apelin-17 analogue (N3-PEG6-NMe17A2) showed retention in circulation similar to SA-21; in contrast, apelin-17 analogue was cleared from the circulation after 2 min. The PFS-SICRFFC is the smallest known peptide macrocycle with a significant affinity for human albumin and substantial in vivo circulation half-life. It is a productive starting point for future development of compact macrocycles with extended half-life in vivo.
Collapse
Affiliation(s)
- Jeffrey Y K Wong
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Arunika I Ekanayake
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Serhii Kharchenko
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Steven E Kirberger
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ryan Qiu
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Payam Kelich
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Susmita Sarkar
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Jiaqian Li
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Edgar R Alvizo-Paez
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Jiayuan Miao
- Department of Chemistry, Tufts University, Medford, MA, 02155, USA
| | | | - J Dwyer John
- Ferring Research Institute, San Diego, CA, 92121, USA
| | - Hongsuk Kang
- Quantum Intelligence Corp., 31F, One IFC, 10 Gukjegeumyung-ro, Yeongdeungpo-gu-Seoul, Republic of Korea
| | - Hwanho Choi
- Quantum Intelligence Corp., 31F, One IFC, 10 Gukjegeumyung-ro, Yeongdeungpo-gu-Seoul, Republic of Korea
| | - John M Nuss
- Ferring Research Institute, San Diego, CA, 92121, USA
| | - John C Vederas
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University, Medford, MA, 02155, USA
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Lela Vukovic
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX, 79968, USA
| | | | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada.
| |
Collapse
|
7
|
Liu C, Li J, Hu Q, Xu X, Zhang X. Generation of a CD70-Specific Fusion Nanobody with IgG Recruiting Capacity for Tumor Killing. Int J Nanomedicine 2023; 18:3325-3338. [PMID: 37361386 PMCID: PMC10289098 DOI: 10.2147/ijn.s410281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Purpose Due to its competitive advantages such as small size, high stability, easy production, and good tissue penetration compared with monoclonal antibodies (mAb), nanobodies (Nbs) were considered the next generation of therapeutics. However, the absence of Fc fragments and Fc-triggered immune effectors limits their clinical applications. In order to overcome these limitations, we develop a novel approach by attaching an IgG binding domain (IgBD) to Nbs for recruiting endogenous IgG and recovering the immune effectors for tumor killing. Material and Methods We linked a Streptococcal Protein G-derived IgBD, termed C3Fab, at the C-terminus of a CD70-specific Nb 3B6 to construct an endogenous IgG recruitment antibody (termed EIR). The recombinant Nb3B6-C3Fab was expressed in E. coli BL21 (DE3) and purified by nickel affinity chromatography. We further evaluated the binding, recruitment of IgG, and the serum half-life of Nb3B6-C3Fab. The tumor-killing effects on CD70 positive cells mediated by antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity were also detected. Results We successfully constructed a IgBD fused Nb3B6-C3Fab with high affinity for CD70 and mouse IgG (mIgG). Nb3B6-C3Fab can specifically bind to CD70 positive tumor cells and recruit mIgG on the cell surface. Ligating of Nb3B6 with C3Fab increased its serum half-life in mice almost 39-fold from 0.96 h to 37.67 h. Moreover, we demonstrated remarkable cytotoxicity of Nb3B6-C3Fab to CD70 positive tumor cells via C3Fab by immune effector cells. Conclusion Our study demonstrates that IgBD fusion endows Nbs with the ability for endogenous IgG recruitment and half-life promotion. Linking IgBD to Nbs is an effective strategy to recovering immune effectors for tumor killing.
Collapse
Affiliation(s)
- Chang Liu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, People’s Republic of China
| | - Jiangwei Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, People’s Republic of China
| | - Qianqian Hu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, People’s Republic of China
| | - Xinlan Xu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, People’s Republic of China
| | - Xin Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, People’s Republic of China
| |
Collapse
|
8
|
IgG Fc Affinity Ligands and Their Applications in Antibody-Involved Drug Delivery: A Brief Review. Pharmaceutics 2023; 15:pharmaceutics15010187. [PMID: 36678816 PMCID: PMC9862274 DOI: 10.3390/pharmaceutics15010187] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Antibodies are not only an important class of biotherapeutic drugs, but also are targeting moieties for achieving active targeting drug delivery. Meanwhile, the rapidly increasing application of antibodies and Fc-fusion proteins has inspired the emerging development of downstream processing technologies. Thus, IgG Fc affinity ligands have come into being and have been widely exploited in antibody purification strategies. Given the high binding affinity and specificity to IgGs, binding stability in physiological medium conditions, and favorable toxicity and immunogenicity profiles, Fc affinity ligands are gradually applied to antibody delivery, non-covalent antibody-drug conjugates or antibody-mediated active-targeted drug delivery systems. In this review, we will briefly introduce IgG affinity ligands that are widely used at present and summarize their diverse applications in the field of antibody-involved drug delivery. The challenges and outlook of these systems are also discussed.
Collapse
|
9
|
Lu H, Jin Y, Yang H, Tao Z, Chen J, Chen S, Feng Y, Xin H, Lu X. A trimeric immunoglobin G-binding domain outperforms recombinant protein G and protein L as a ligand for fragment antigen-binding purification. J Chromatogr A 2022; 1681:463464. [DOI: 10.1016/j.chroma.2022.463464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022]
|
10
|
Hwang J, Jung Y, Moon S, Yu S, Oh H, Kim S, Kim KW, Yoon JH, Chun J, Kim SJ, Chung WJ, Kweon DH. Nanodisc-Mediated Conversion of Virustatic Antiviral Antibody to Disrupt Virus Envelope in Infected Cells. SMALL METHODS 2022; 6:e2101516. [PMID: 35107214 DOI: 10.1002/smtd.202101516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Indexed: 06/14/2023]
Abstract
Many antibody-based antivirals, including broadly neutralizing antibodies (bnAbs) against various influenza virus strains, suffer from limited potency. A booster of the antiviral activity of an antibody is expected to facilitate development of antiviral therapeutics. In this study, a nanodisc (ND), a discoidal lipid bilayer encircled by membrane scaffold proteins, is engineered to provide virucidal properties to antibodies, thereby augmenting their antiviral activity. NDs carrying the Fc-binding peptide sequence form an antibody-ND complex (ANC), which can co-endocytose into cells infected with influenza virus. ANC efficiently inhibits endosome escape of viral RNA by dual complimentary mode of action. While the antibody moiety in an ANC inhibits hemagglutinin-mediated membrane fusion, its ND moiety destroys the viral envelope using free hemagglutinins that are not captured by antibodies. Providing virus-infected host cells with the ability to self-eliminate by the synergistic effect of ANC components dramatically amplifies the antiviral efficacy of a bnAb against influenza virus. When the efficacy of ANC is assessed in mouse models, administration of ANCs dramatically reduces morbidity and mortality compared to bnAb alone. This study is the first to demonstrate the novel nanoparticle ANC and its role in combating viral infections, suggesting that ANC is a versatile platform applicable to various viruses.
Collapse
Affiliation(s)
- Jaehyeon Hwang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Younghun Jung
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seokoh Moon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seokhyeon Yu
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyunseok Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Soomin Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeong Won Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jeong Hyeon Yoon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihwan Chun
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sang Jick Kim
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea
| | - Woo-Jae Chung
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
11
|
Lü JM, Liang Z, Liu D, Zhan B, Yao Q, Chen C. Two Antibody-Guided Lactic-co-Glycolic Acid-Polyethylenimine (LGA-PEI) Nanoparticle Delivery Systems for Therapeutic Nucleic Acids. Pharmaceuticals (Basel) 2021; 14:841. [PMID: 34577541 PMCID: PMC8470087 DOI: 10.3390/ph14090841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/14/2021] [Accepted: 08/23/2021] [Indexed: 01/10/2023] Open
Abstract
We previously reported a new polymer, lactic-co-glycolic acid-polyethylenimine (LGA-PEI), as an improved nanoparticle (NP) delivery for therapeutic nucleic acids (TNAs). Here, we further developed two antibody (Ab)-conjugated LGA-PEI NP technologies for active-targeting delivery of TNAs. LGA-PEI was covalently conjugated with a single-chain variable fragment antibody (scFv) against mesothelin (MSLN), a biomarker for pancreatic cancer (PC), or a special Ab fragment crystallizable region-binding peptide (FcBP), which binds to any full Ab (IgG). TNAs used in the current study included tumor suppressor microRNA mimics (miR-198 and miR-520h) and non-coding RNA X-inactive specific transcript (XIST) fragments; green fluorescence protein gene (GFP plasmid DNA) was also used as an example of plasmid DNA. MSLN scFv-LGA-PEI NPs with TNAs significantly improved their binding and internalization in PC cells with high expression of MSLN in vitro and in vivo. Anti-epidermal growth factor receptor (EGFR) monoclonal Ab (Cetuximab) binding to FcBP-LGA-PEI showed active-targeting delivery of TNAs to EGFR-expressing PC cells.
Collapse
Affiliation(s)
- Jian-Ming Lü
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
| | - Zhengdong Liang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
| | - Dongliang Liu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
| | - Bin Zhan
- National School of Tropical Medicine and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA;
| | - Qizhi Yao
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Changyi Chen
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
| |
Collapse
|
12
|
von Witting E, Hober S, Kanje S. Affinity-Based Methods for Site-Specific Conjugation of Antibodies. Bioconjug Chem 2021; 32:1515-1524. [PMID: 34369763 PMCID: PMC8377709 DOI: 10.1021/acs.bioconjchem.1c00313] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Conjugation of various reagents to antibodies has long been an elegant way to combine the superior binding features of the antibody with other desired but non-natural functions. Applications range from labels for detection in different analytical assays to the creation of new drugs by conjugation to molecules which improves the pharmaceutical effect. In many of these applications, it has been proven advantageous to control both the site and the stoichiometry of the conjugation to achieve a homogeneous product with predictable, and often also improved, characteristics. For this purpose, many research groups have, during the latest decade, reported novel methods and techniques, based on small molecules, peptides, and proteins with inherent affinity for the antibody, for site-specific conjugation of antibodies. This review provides a comprehensive overview of these methods and their applications and also describes a historical perspective of the field.
Collapse
Affiliation(s)
- Emma von Witting
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, AlbaNova University Centre, SE-114 19, Stockholm, Sweden
| | - Sophia Hober
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, AlbaNova University Centre, SE-114 19, Stockholm, Sweden
| | - Sara Kanje
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, AlbaNova University Centre, SE-114 19, Stockholm, Sweden
| |
Collapse
|
13
|
Sasaki K, Harada M, Yoshikawa T, Tagawa H, Harada Y, Yonemitsu Y, Ryujin T, Kishimura A, Mori T, Katayama Y. Fc-Binding Antibody-Recruiting Molecules Targeting Prostate-Specific Membrane Antigen: Defucosylation of Antibody for Efficacy Improvement*. Chembiochem 2020; 22:496-500. [PMID: 32969164 DOI: 10.1002/cbic.202000577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/13/2020] [Indexed: 12/16/2022]
Abstract
Synthetic small molecules that redirect endogenous antibodies to target cells are promising drug candidates because they overcome the potential shortcomings of therapeutic antibodies, such as immunogenicity and the need for intravenous delivery. Previously, we reported a novel class of bispecific molecules targeting the antibody Fc region and folate receptor, named Fc-binding antibody-recruiting molecules (Fc-ARMs). Fc-ARMs can theoretically recruit most endogenous antibodies, inducing antibody-dependent cell-mediated cytotoxicity (ADCC) to eliminate cancer cells. Herein, we describe new Fc-ARMs that target prostate cancer (Fc-ARM-Ps). Fc-ARM-Ps recruited antibodies to cancer cells expressing prostate-specific membrane antigen but did so with lower efficiency compared with Fc-ARMs targeting the folate receptor. Upon recruitment by Fc-ARM-P, defucosylated antibodies efficiently activated natural killer cells and induced ADCC, whereas antibodies with intact N-glycans did not. The results suggest that the affinity between recruited antibodies and CD16a, a type of Fc receptor expressed on immune cells, could be a key factor controlling immune activation in the Fc-ARM strategy.
Collapse
Affiliation(s)
- Koichi Sasaki
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishiku, Fukuoka, 819-0395, Japan.,Present address: Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Osaka, 599-8531, Japan
| | - Minori Harada
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Takuma Yoshikawa
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishiku, Fukuoka, 819-0395, Japan
| | - Hiroshi Tagawa
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yui Harada
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yoshikazu Yonemitsu
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takaaki Ryujin
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Akihiro Kishimura
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishiku, Fukuoka, 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan.,International Research Center for Molecular Systems, Kyushu University, Fukuoka, 819-0395, Japan
| | - Takeshi Mori
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishiku, Fukuoka, 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yoshiki Katayama
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishiku, Fukuoka, 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan.,International Research Center for Molecular Systems, Kyushu University, Fukuoka, 819-0395, Japan.,Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan, 32023, Taiwan
| |
Collapse
|
14
|
Jendryczko K, Chudzian J, Skinder N, Opaliński Ł, Rzeszótko J, Wiedlocha A, Otlewski J, Szlachcic A. FGF2-Derived PeptibodyF2-MMAE Conjugate for Targeted Delivery of Cytotoxic Drugs into Cancer Cells Overexpressing FGFR1. Cancers (Basel) 2020; 12:E2992. [PMID: 33076489 PMCID: PMC7602595 DOI: 10.3390/cancers12102992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 10/10/2020] [Indexed: 12/23/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are emerging targets for directed cancer therapy. Presented here is a new FGFR1-targeting conjugate, the peptibodyF2, which employs peptibody, a fusion of peptide and the Fc fragment of human IgG as a selective targeting agent and drug carrier. Short peptide based on FGF2 sequence was used to construct a FGFR1-targeting peptibody. We have shown that this peptide ensures specific delivery of peptibodyF2 into FGFR1-expressing cells. In order to use peptibodyF2 as a delivery vehicle for cytotoxic drugs, we have conjugated it with MMAE, a drug widely used in antibody-drug conjugates for targeted therapy. Resulting conjugate shows high and specific cytotoxicity towards FGFR1-positive cells, i.e., squamous cell lung carcinoma NCI-H520, while remaining non-toxic for FGFR1-negative cells. Such peptibody-drug conjugate can serve as a basis for development of therapy for tumors with overexpressed or malfunctioning FGFRs.
Collapse
Affiliation(s)
- Karolina Jendryczko
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Julia Chudzian
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Natalia Skinder
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Łukasz Opaliński
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Jakub Rzeszótko
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Antoni Wiedlocha
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway;
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 01163 Warsaw, Poland
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Anna Szlachcic
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| |
Collapse
|
15
|
Ligand-Mediated Targeting of Cytokine Interleukin-27 Enhances Its Bioactivity In Vivo. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:739-751. [PMID: 32346551 PMCID: PMC7177159 DOI: 10.1016/j.omtm.2020.03.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
We have examined the role of a novel targeted cytokine, interleukin-27 (IL-27), modified at the C terminus with a dual targeting and therapeutic heptapeptide, in treating prostate cancer. IL-27 has shown promise in halting tumor growth and mediating tumor regression in several cancer models, including prostate cancer. We describe our findings on the effects of targeted IL-27 gene delivery on prostate cancer cells in vitro and in vivo and how the targeting enhances bioactivity of the IL-27 cytokine. We applied the IL-27 gene delivery protocol utilizing sonoporation (sonodelivery) with the goal of reducing prostate tumor growth in an immunocompetent TC2R C57/BL6 model. The reduction in tumor growth and effector cellular profiles implicate targeted IL-27 as more effective than an untargeted version of IL-27 in promoting bioactivity, as assessed by STAT1 and IFN-γ reporter genes. Moreover, enhanced antitumor effects and significantly higher accumulation of natural killer T (NKT) and CD8 effector cells in the tumors were observed. These results support a novel IL-27-based targeting strategy that is promising since it shows improved therapeutic efficacy while utilizing simple and effective sonodelivery methods.
Collapse
|
16
|
Lu S, Liu G, Chen T, Wang W, Hu J, Tang D, Peng X. Lentivirus-Mediated hFGF21 Stable Expression in Liver of Diabetic Rats Model and Its Antidiabetic Effect Observation. Hum Gene Ther 2020; 31:472-484. [PMID: 32027183 DOI: 10.1089/hum.2019.322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The incidence of type 2 diabetes mellitus (T2DM) has been increasing annually, which is a serious threat to human health. Fibroblast growth factor 21 (FGF21) is one of the most popular targets for the treatment of diabetes because it effectively improves glycolipid metabolism. In our experiment, human FGF21 (hFGF21) was injected and stably expressed in the liver tissues of a rat T2DM model with lentivirus system. Based on clinical and histopathological examinations, islet cells were protected and liver tissue lesions were repaired for >4 months. Glucose metabolism and histopathology were controlled perfectly when hFGF21 was stably expressed in partial liver of T2DM rats. The results showed that the liver tissue cell apoptosis was reduced, the lipid droplet content was decreased, the oxidative stress indexes were improved, the glycogen content was increased, and the islet cells were increased too. Besides, insulin sensitivity and glycogen synthesis-related genes expression were increased, but cell apoptosis-related genes caspase3 and NFκB expression were decreased. The effectiveness of results suggested that injecting hFGF21 to rats liver could effectively treat T2DM.
Collapse
Affiliation(s)
- Shuaiyao Lu
- Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, China
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Diseases, Kunming, China
| | - Guanglong Liu
- The First People's Hospital of Yunnan Province, Kunming, China
| | - Tianxing Chen
- The First People's Hospital of Yunnan Province, Kunming, China
| | - Wanpu Wang
- The First People's Hospital of Yunnan Province, Kunming, China
| | - Jingwen Hu
- Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, China
| | - Donghong Tang
- Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, China
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Diseases, Kunming, China
| | - Xiaozhong Peng
- Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, China
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Diseases, Kunming, China
| |
Collapse
|
17
|
Figueiredo Neto M, Liu S, Salameh JW, Yokota H, Figueiredo ML. Interleukin-27 Gene Delivery Targeting IL-6Rα-Expressing Cells as a Stress Response Therapy. Int J Mol Sci 2020; 21:E1108. [PMID: 32046108 PMCID: PMC7038084 DOI: 10.3390/ijms21031108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/28/2020] [Accepted: 02/06/2020] [Indexed: 01/11/2023] Open
Abstract
Interleukin-27 (IL-27) has shown promise in halting tumor growth and mediating tumor regression in several models, including prostate cancer. We describe our findings on the effects of IL-27 on the gene expression changes of TC2R prostate adenocarcinoma cells. We utilized RNAseq to assess profile differences between empty vector control, vector delivering IL-27 modified at its C-terminus with a non-specific peptide, and IL-27 modified at the C-terminus with a peptide targeting the IL-6-Rα. The targeted IL-27 had higher bioactivity and activity in vivo in a recent study by our group, but the mechanisms underlying this effect had not been characterized in detail at the gene expression level on tumor cells. In the present work, we sought to examine potential mechanisms for targeted IL-27 enhanced activity directly on tumor cells. The targeted IL-27 appeared to modulate several changes that would be consistent with an anti-tumor effect, including upregulation in the Interferon (IFN) and Interferon regulatory factor (IRF), oxidative phosphorylation, Janus kinase/Signal transducers and activators of transcription (JAK/STAT), and eukaryotic initiation factor 2 (EIF2) signaling. Of these signaling changes predicted by ingenuity pathway analyses (IPA), the novel form also with the highest significance (-log(Benjamini-Hochberg (B-H)) p-value) was the EIF2 signaling upregulation. We validated this predicted change by assaying for eukaryotic initiation factor 2 alpha (eIF2α), or phosphorylated eIF2α (p-eIF2α), and caspase-3 levels. We detected an increase in the phosphorylated form of eIF2α and in the cleaved caspase-3 fraction, indicating that the EIF2 signaling pathway was upregulated in these prostate tumor cells following targeted IL-27 gene delivery. This approach of targeting cytokines to enhance their activity against cancer cells is a novel approach to help augment IL-27's bioactivity and efficacy against prostate tumors and could be extended to other conditions where it could help interfere with the EIF2α pathway and promote caspase-3 activation.
Collapse
Affiliation(s)
- Manoel Figueiredo Neto
- Department of Basic Medical Sciences and Interdisciplinary Biomedical Sciences Program, Purdue University, 625 Harrison Street, LYNN 2177, West Lafayette, IN 47907, USA; (M.F.N.); (J.W.S.)
| | - Shengzhi Liu
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (S.L.); (H.Y.)
| | - Janelle Wes Salameh
- Department of Basic Medical Sciences and Interdisciplinary Biomedical Sciences Program, Purdue University, 625 Harrison Street, LYNN 2177, West Lafayette, IN 47907, USA; (M.F.N.); (J.W.S.)
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (S.L.); (H.Y.)
| | - Marxa Leão Figueiredo
- Department of Basic Medical Sciences and Interdisciplinary Biomedical Sciences Program, Purdue University, 625 Harrison Street, LYNN 2177, West Lafayette, IN 47907, USA; (M.F.N.); (J.W.S.)
- Purdue Center for Cancer Research and Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
18
|
Bi W, Xu W, Cheng L, Xue J, Wang Q, Yu F, Xia S, Wang Q, Li G, Qin C, Lu L, Su L, Jiang S. IgG Fc-binding motif-conjugated HIV-1 fusion inhibitor exhibits improved potency and in vivo half-life: Potential application in combination with broad neutralizing antibodies. PLoS Pathog 2019; 15:e1008082. [PMID: 31805154 PMCID: PMC6894747 DOI: 10.1371/journal.ppat.1008082] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/16/2019] [Indexed: 12/23/2022] Open
Abstract
The clinical application of conventional peptide drugs, such as the HIV-1 fusion inhibitor enfuvirtide, is limited by their short half-life in vivo. To overcome this limitation, we developed a new strategy to extend the in vivo half-life of a short HIV-1 fusion inhibitory peptide, CP24, by fusing it with the human IgG Fc-binding peptide (IBP). The newly engineered peptide IBP-CP24 exhibited potent and broad anti-HIV-1 activity with IC50 values ranging from 0.2 to 173.7 nM for inhibiting a broad spectrum of HIV-1 strains with different subtypes and tropisms, including those resistant to enfuvirtide. Most importantly, its half-life in the plasma of rhesus monkeys was 46.1 h, about 26- and 14-fold longer than that of CP24 (t1/2 = 1.7 h) and enfuvirtide (t1/2 = 3 h), respectively. IBP-CP24 intravenously administered in rhesus monkeys could not induce significant IBP-CP24-specific antibody response and it showed no obvious in vitro or in vivo toxicity. In the prophylactic study, humanized mice pretreated with IBP-CP24 were protected from HIV-1 infection. As a therapeutic treatment, coadministration of IBP-CP24 and normal human IgG to humanized mice with chronic HIV-1 infection resulted in a significant decrease of plasma viremia. Combining IBP-CP24 with a broad neutralizing antibody (bNAb) targeting CD4-binding site (CD4bs) in gp120 or a membrane proximal external region (MPER) in gp41 exhibited synergistic effect, resulting in significant dose-reduction of the bNAb and IBP-CP24. These results suggest that IBP-CP24 has the potential to be further developed as a new HIV-1 fusion inhibitor-based, long-acting anti-HIV drug that can be used alone or in combination with a bNAb for treatment and prevention of HIV-1 infection.
Collapse
Affiliation(s)
- Wenwen Bi
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Liang Cheng
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jing Xue
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Re-emerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Qian Wang
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Fei Yu
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shuai Xia
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qi Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Guangming Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Chuan Qin
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Re-emerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- * E-mail: (LL); (LS); (SJ)
| | - Lishan Su
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail: (LL); (LS); (SJ)
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
- * E-mail: (LL); (LS); (SJ)
| |
Collapse
|
19
|
Zorzi A, Linciano S, Angelini A. Non-covalent albumin-binding ligands for extending the circulating half-life of small biotherapeutics. MEDCHEMCOMM 2019; 10:1068-1081. [PMID: 31391879 PMCID: PMC6644573 DOI: 10.1039/c9md00018f] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 06/01/2019] [Indexed: 12/13/2022]
Abstract
Peptides and small protein scaffolds are gaining increasing interest as therapeutics. Similarly to full-length antibodies, they can bind a target with a high binding affinity and specificity while remaining small enough to diffuse into tissues. However, despite their numerous advantages, small biotherapeutics often suffer from a relatively short circulating half-life, thus requiring frequent applications that ultimately restrict their ease of use and user compliance. To overcome this limitation, a large variety of half-life extension strategies have been developed in the last decades. Linkage to ligands that non-covalently bind to albumin, the most abundant serum protein with a circulating half-life of ∼19 days in humans, represents one of the most successful approaches for the generation of long-lasting biotherapeutics with improved pharmacokinetic properties and superior efficacy in the clinic.
Collapse
Affiliation(s)
- Alessandro Zorzi
- Institute of Chemical Sciences and Engineering , School of Basic Sciences , Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne CH-1015 , Switzerland
| | - Sara Linciano
- Department of Molecular Sciences and Nanosystems , Ca' Foscari University of Venice , Via Torino 155 , Venezia Mestre 30172 , Italy
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems , Ca' Foscari University of Venice , Via Torino 155 , Venezia Mestre 30172 , Italy
- European Centre for Living Technologies (ECLT) , San Marco 2940 , Venice 30124 , Italy .
| |
Collapse
|
20
|
Nielsen TB, Thomsen RP, Mortensen MR, Kjems J, Nielsen PF, Nielsen TE, Kodal ALB, Cló E, Gothelf KV. Peptide‐Directed DNA‐Templated Protein Labelling for The Assembly of a Pseudo‐IgM. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201903134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Thorbjørn B. Nielsen
- Research ChemistryNovo Nordisk A/S Novo Nordisk Park 2760 Måløv Denmark
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO)Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| | - Rasmus P. Thomsen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO)Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
- Department of Molecular Biology and Genetics, and Interdisciplinary Nanoscience Centre (iNANO) Denmark
| | - Michael R. Mortensen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO)Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, and Interdisciplinary Nanoscience Centre (iNANO) Denmark
| | - Per Franklin Nielsen
- Analysis and MS Characterisation 2Novo Nordisk A/S Novo Nordisk Park 2760 Måløv Denmark
| | - Thomas E. Nielsen
- Research ChemistryNovo Nordisk A/S Novo Nordisk Park 2760 Måløv Denmark
| | | | - Emiliano Cló
- Research ChemistryNovo Nordisk A/S Novo Nordisk Park 2760 Måløv Denmark
| | - Kurt V. Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO)Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| |
Collapse
|
21
|
Nielsen TB, Thomsen RP, Mortensen MR, Kjems J, Nielsen PF, Nielsen TE, Kodal ALB, Cló E, Gothelf KV. Peptide-Directed DNA-Templated Protein Labelling for The Assembly of a Pseudo-IgM. Angew Chem Int Ed Engl 2019; 58:9068-9072. [PMID: 30995340 DOI: 10.1002/anie.201903134] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/15/2019] [Indexed: 12/30/2022]
Abstract
The development of methods for conjugation of DNA to proteins is of high relevance for the integration of protein function and DNA structures. Here, we demonstrate that protein-binding peptides can direct a DNA-templated reaction, selectively furnishing DNA-protein conjugates with one DNA label. Quantitative conversion of oligonucleotides is achieved at low stoichiometries and the reaction can be performed in complex biological matrixes, such as cell lysates. Further, we have used a star-like pentameric DNA nanostructure to assemble five DNA-Rituximab conjugates, made by our reported method, into a pseudo-IgM antibody structure that was subsequently characterized by negative-stain transmission electron microscopy (nsTEM) analysis.
Collapse
Affiliation(s)
- Thorbjørn B Nielsen
- Research Chemistry, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark.,Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Rasmus P Thomsen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark.,Department of Molecular Biology and Genetics, and Interdisciplinary Nanoscience Centre (iNANO), Denmark
| | - Michael R Mortensen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, and Interdisciplinary Nanoscience Centre (iNANO), Denmark
| | - Per Franklin Nielsen
- Analysis and MS Characterisation 2, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark
| | - Thomas E Nielsen
- Research Chemistry, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark
| | - Anne Louise B Kodal
- Research Chemistry, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark
| | - Emiliano Cló
- Research Chemistry, Novo Nordisk A/S, Novo Nordisk Park, 2760, Måløv, Denmark
| | - Kurt V Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| |
Collapse
|
22
|
Zhou F, Kroetsch A, Nguyen VP, Huang X, Ogoke O, Parashurama N, Park S. High-Affinity Antibody Detection with a Bivalent Circularized Peptide Containing Antibody-Binding Domains. Biotechnol J 2019; 14:e1800647. [PMID: 30810268 DOI: 10.1002/biot.201800647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/27/2019] [Indexed: 11/05/2022]
Abstract
Direct chemical labeling of antibody produces molecules with poorly defined modifications. Use of a small antibody-binding protein as an adapter can simplify antibody functionalization by forming a specific antibody-bound complex and introducing site-specific modifications. To stabilize a noncovalent antibody complex that may be used without chemical crosslinking, a bivalent antibody-binding protein is engineered with an improved affinity of interaction by joining two Z domains with a conformationally flexible linker. The linker is essential for the increase in affinity because it allows simultaneous binding of both domains. The molecule is further circularized using a split intein, creating a novel adapter protein ("lasso"), which binds human immunoglobulin G1 (IgG1) with K D = 0.53 n m and a dissociation rate that is 55- to 84-fold slower than Z. The lasso contains a unique cysteine for conjugation with a reporter and may be engineered to introduce other functional groups, including a biotin tag and protease recognition sequences. When used in enzyme-linked immunosorbent assay (ELISA), the lasso generates a stronger reporter signal compared to a secondary antibody and lowers the limit of detection by 12-fold. The small size of the lasso and a long half-life of dissociation make the peptide a useful tool in antibody detection and immobilization.
Collapse
Affiliation(s)
- Fangyu Zhou
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY, 14260, USA
| | - Andrew Kroetsch
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY, 14260, USA
| | - Vyncent P Nguyen
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY, 14260, USA
| | - Xiao Huang
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY, 14260, USA
| | - Ogechi Ogoke
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY, 14260, USA
| | - Natesh Parashurama
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY, 14260, USA
| | - Sheldon Park
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY, 14260, USA
| |
Collapse
|
23
|
Rattanapisit K, Srifa S, Kaewpungsup P, Pavasant P, Phoolcharoen W. Plant-produced recombinant Osteopontin-Fc fusion protein enhanced osteogenesis. ACTA ACUST UNITED AC 2019; 21:e00312. [PMID: 30847284 PMCID: PMC6389792 DOI: 10.1016/j.btre.2019.e00312] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 01/27/2023]
Abstract
Osteopontin (OPN) plays an important role in the bone regeneration process. The plant-produced OPN-Fc increases the protein expression level and facilitates the purification of the recombinant protein. The plant-produced OPN-Fc can stimulate the expression of osteogenic related genes and the calcium deposition in hPDL cells. The plant-produced OPN-Fc has potential application in tissue engineering in the future.
Osteopontin (OPN) plays an important role in the bone regeneration process. Previous investigation showed that recombinant human OPN was able to express in Nicotiana benthamiana leaves and induced the osteogenic related genes. Nevertheless, the purification of OPN from plant proteins with Ni affinity chromatography was still not effective enough. To improve the quality of protein expression and purification in plants, we constructed an Fc-based form of OPN. The complete OPN protein was fused to the human IgG1 Fc domain. Here, we showed that the plant-produced OPN-Fc increases the protein expression level and facilitates the purification of the recombinant protein. Our result showed that the plant-produced OPN-Fc can stimulate the expression of osteogenic related genes such as DMP1, OSX, and Wnt3a and also the calcium deposition in hPDL cells. These findings suggest that the plant-produced OPN-Fc has potential application in tissue engineering in the future.
Collapse
Affiliation(s)
- Kaewta Rattanapisit
- Research Unit for Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
| | - Suchada Srifa
- Research Unit of Mineralized Tissue, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Pornjira Kaewpungsup
- Research Unit of Mineralized Tissue, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Research Unit of Mineralized Tissue, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Waranyoo Phoolcharoen
- Research Unit for Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand.,Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
24
|
Park J, Lee Y, Ko BJ, Yoo TH. Peptide-Directed Photo-Cross-Linking for Site-Specific Conjugation of IgG. Bioconjug Chem 2018; 29:3240-3244. [PMID: 30179444 DOI: 10.1021/acs.bioconjchem.8b00515] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Conjugation of antibody has expanded its applications in therapeutics and diagnostics, and various methods have been developed based on chemical or enzymatic reactions. However, the majority of them have focused on synthetic molecules such as small molecules, nucleic acids, or synthetic materials, but site-specific conjugation of antibody with protein cargo has rarely been demonstrated. In this Communication, we report a PEptide-DIrected Photo-cross-linking (PEDIP) reaction for site-specific conjugation of IgG with protein using an Fc-binding peptide and a photoreactive amino acid analogue, and demonstrate this method by developing an immunotoxin composed of a Her2-targeting IgG (trastuzumab) and an engineered Pseudomonas exotoxin A (PE24). The ADP-ribosylation of eukaryotic elongation factor-2 by the bacterial toxin inhibits the ribosomal translation of protein, and the trastuzumab-PE24 conjugate exhibited the cytotoxicity toward Her2-overexpressing cell lines. The PEDIP reaction can also be applied for many other types of cargo with slight modifications of the method.
Collapse
Affiliation(s)
| | | | - Byoung Joon Ko
- New Drug Development Center , Osong Medical Innovative Foundation , 123 Osongsaengmyeong-ro , Osong-eup, Cheongju , 28160 , Korea
| | | |
Collapse
|
25
|
Yang H, Feng Y, Cai H, Jia D, Li H, Tao Z, Zhong Y, Li Z, Shi Q, Wan L, Li L, Lu X. Endogenous IgG-based affinity-controlled release of TRAIL exerts superior antitumor effects. Am J Cancer Res 2018; 8:2459-2476. [PMID: 29721092 PMCID: PMC5928902 DOI: 10.7150/thno.23880] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/08/2018] [Indexed: 02/05/2023] Open
Abstract
The inefficiency of recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based clinical regimens has been dominantly attributed to the short half-life of TRAIL. Affinity-controlled release using endogenous long-acting proteins, such as IgG and albumin, as carriers is extremely attractive for improving the pharmacokinetics of TRAIL. Up to now, it is unclear whether IgG-binding is efficient for affinity-controlled release of TRAIL. Methods: An IgG-binding affibody, IgBD, was genetically fused to the N-terminus of TRAIL to produce IgBD-TRAIL.The IgG-binding ability, cytotoxicity, serum half-life, and in vivo antitumor effect of IgBD-TRAIL were compared with that of TRAIL. In addition, an albumin-binding affibody, ABD, was fused to TRAIL to produce ABD-TRAIL. The cytototoxicity, serum half-life, and antitumor effect of IgBD-TRAIL and ABD-TRAIL were compared. Results: IgBD fusion endowed TRAIL with high affinity (nM) for IgG without interference with its cytotoxicity. The serum half-life of IgBD-TRAIL is 50-60 times longer than that of TRAIL and the tumor uptake of IgBD-TRAIL at 8-24 h post-injection was 4-7-fold that of TRAIL. In vivo antitumor effect of IgBD-TRAIL was at least 10 times greater than that of TRAIL. Owing to the high affinity (nM) for albumin, the serum half-life of ABD-TRAIL was 80-90 times greater than that of TRAIL. However, after binding to albumin, the cytotoxicity of ABD-TRAIL was reduced more than 10 times. In contrast, binding to IgG had little impact on the cytotoxicity of IgBD-TRAIL. Consequently, intravenously injected IgBD-TRAIL showed antitumor effects superior to those of ABD-TRAIL. Conclusions: Endogenous long-acting proteins, particularly IgG-based affinity-controlled release, prolonged the serum half-life as well as significantly enhanced the antitumor effect of TRAIL. IgBD-mediated endogenous IgG binding might be a novel approach for the affinity-controlled release of other protein drugs.
Collapse
|
26
|
Cavaco M, Castanho MARB, Neves V. Peptibodies: An elegant solution for a long-standing problem. Biopolymers 2017; 110. [PMID: 29266205 DOI: 10.1002/bip.23095] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/20/2017] [Accepted: 11/24/2017] [Indexed: 01/09/2023]
Abstract
Chimeric proteins composed of a biologically active peptide and a fragment crystallizable (Fc) domain of immunoglobulin G (IgG) are known as peptibodies. They present an extended half-life due to neonatal Fc receptor (FcRn) salvage pathway, a decreased renal clearance rate owing to its increased size (≈70 kDa) and, depending on the peptide used in the design of the peptibody, an active-targeting moiety. Also, the peptides therapeutic activity is boosted by the number of peptides in the fusion protein (at least two peptides) and to some peptides' alterations. Peptibodies are mainly obtained through recombinant DNA technology. However, to improve peptide properties, "unnatural" changes have been introduced to the original peptides' sequence, for instance, the incorporation of D- or non-natural amino acid residues or even cyclization thus, limiting the application of genetic engineering in the production of peptibodies, since these peptides must be obtained via chemical synthesis. This constrains prompted the development of new methods for conjugation of peptides to Fc domains. Another challenge, subject of intense research, relates to the large-scale production of such peptibodies using these new techniques, which can be minimized by their proved value. To date, two peptibodies, romiplostim and dulaglutide, have been approved and stay as the standard of care in their areas of action. Furthermore, a considerable number of peptibodies are currently in preclinical and clinical development.
Collapse
Affiliation(s)
- Marco Cavaco
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
| | - Miguel A R B Castanho
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
| | - Vera Neves
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
| |
Collapse
|
27
|
Fc-Binding Ligands of Immunoglobulin G: An Overview of High Affinity Proteins and Peptides. MATERIALS 2016; 9:ma9120994. [PMID: 28774114 PMCID: PMC5456964 DOI: 10.3390/ma9120994] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 11/26/2016] [Accepted: 11/29/2016] [Indexed: 01/20/2023]
Abstract
The rapidly increasing application of antibodies has inspired the development of several novel methods to isolate and target antibodies using smart biomaterials that mimic the binding of Fc-receptors to antibodies. The Fc-binding domain of antibodies is the primary binding site for e.g., effector proteins and secondary antibodies, whereas antigens bind to the Fab region. Protein A, G, and L, surface proteins expressed by pathogenic bacteria, are well known to bind immunoglobulin and have been widely exploited in antibody purification strategies. Several difficulties are encountered when bacterial proteins are used in antibody research and application. One of the major obstacles hampering the use of bacterial proteins is sample contamination with trace amounts of these proteins, which can invoke an immune response in the host. Many research groups actively develop synthetic ligands that are able to selectively and strongly bind to antibodies. Among the reported ligands, peptides that bind to the Fc-domain of antibodies are attractive tools in antibody research. Besides their use as high affinity ligands in antibody purification chromatography, Fc-binding peptides are applied e.g., to localize antibodies on nanomaterials and to increase the half-life of proteins in serum. In this review, recent developments of Fc-binding peptides are presented and their binding characteristics and diverse applications are discussed.
Collapse
|
28
|
Deci MB, Ferguson SW, Liu M, Peterson DC, Koduvayur SP, Nguyen J. Utilizing clathrin triskelions as carriers for spatially controlled multi-protein display. Biomaterials 2016; 108:120-8. [PMID: 27627809 DOI: 10.1016/j.biomaterials.2016.08.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/20/2016] [Accepted: 08/26/2016] [Indexed: 10/21/2022]
Abstract
The simultaneous and spatially controlled display of different proteins on nanocarriers is a desirable property not often achieved in practice. Here, we report the use of clathrin triskelions as a versatile platform for functional protein display. We hypothesized that site-specific molecular epitope recognition would allow for effective and ordered protein attachment to clathrin triskelions. Clathrin binding peptides (CBPs) were genetically fused to mCherry and green fluorescent protein (GFP), expressed, and loaded onto clathrin triskelions by site-specific binding. Attachment was confirmed by surface plasmon resonance. mCherry fusion proteins modified with various CBPs displayed binding affinities between 470 nM and 287 μM for the clathrin triskelions. Simultaneous attachment of GFP-Wbox and mCherry-Cbox fusion constructs to the clathrin terminal domain was verified by Förster resonance energy transfer. The circulating half-lives, area under the curve, and the terminal half-lives of GFP and mCherry were significantly increased when attached to clathrin triskelions. Clathrin triskelion technology is useful for the development of versatile and multifunctional carriers for spatially controlled protein or peptide display with tremendous potential in nanotechnology, drug delivery, vaccine development, and targeted therapeutic applications.
Collapse
Affiliation(s)
- Michael B Deci
- Department of Pharmaceutical Sciences, School of Pharmacy, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Scott W Ferguson
- Department of Pharmaceutical Sciences, School of Pharmacy, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Maixian Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Damian C Peterson
- Department of Pharmaceutical Sciences, School of Pharmacy, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Sujatha P Koduvayur
- Department of Electrical and Computer Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Juliane Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA.
| |
Collapse
|
29
|
Qiu Y, Lv W, Xu M, Xu Y. Single chain antibody fragments with pH dependent binding to FcRn enabled prolonged circulation of therapeutic peptide in vivo. J Control Release 2016; 229:37-47. [DOI: 10.1016/j.jconrel.2016.03.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 03/07/2016] [Accepted: 03/12/2016] [Indexed: 12/25/2022]
|
30
|
Martins JP, Kennedy PJ, Santos HA, Barrias C, Sarmento B. A comprehensive review of the neonatal Fc receptor and its application in drug delivery. Pharmacol Ther 2016; 161:22-39. [PMID: 27016466 DOI: 10.1016/j.pharmthera.2016.03.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Advances in the understanding of neonatal Fc receptor (FcRn) biology and function have demonstrated that this receptor, primarily identified for the transfer of passive immunity from mother infant, is involved in several biological and immunological processes. In fact, FcRn is responsible for the long half-life of IgG and albumin in the serum, by creating an intracellular protein reservoir, which is protected from lysosomal degradation and, importantly, trafficked across the cell. Such discovery has led researchers to hypothesize the role for this unique receptor in the controlled delivery of therapeutic agents. A great amount of FcRn-based strategies are already under extensive investigation, in which FcRn reveals to have profound impact on the biodistribution and half-life extension of therapeutic agents. This review summarizes the main findings on FcRn biology, function and distribution throughout different tissues, together with the main advances on the FcRn-based therapeutic opportunities and model systems, which indicate that this receptor is a potential target for therapeutic regimen modification.
Collapse
Affiliation(s)
- João Pedro Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal
| | - Patrick J Kennedy
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal; Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI -00014 Helsinki, Finland
| | - Cristina Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde and Instituto Universitário de Ciências da Saúde, 4585-116 Gandra, Portugal.
| |
Collapse
|
31
|
Unverdorben F, Hutt M, Seifert O, Kontermann RE. A Fab-Selective Immunoglobulin-Binding Domain from Streptococcal Protein G with Improved Half-Life Extension Properties. PLoS One 2015; 10:e0139838. [PMID: 26430884 PMCID: PMC4592230 DOI: 10.1371/journal.pone.0139838] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/17/2015] [Indexed: 12/02/2022] Open
Abstract
Background Half-life extension strategies have gained increasing interest to improve the pharmacokinetic and pharmacodynamic properties of protein therapeutics. Recently, we established an immunoglobulin-binding domain (IgBD) from streptococcal protein G (SpGC3) as module for half-life extension. SpGC3 is capable of binding to the Fc region as well as the CH1 domain of Fab arms under neutral and acidic conditions. Methodology/Principal Findings Using site-directed mutagenesis, we generated a Fab-selective mutant (SpGC3Fab) to avoid possible interference with the FcRn-mediated recycling process and improved its affinity for mouse and human IgG by site-directed mutagenesis and phage display selections. In mice, this affinity-improved mutant (SpGC3FabRR) conferred prolonged plasma half-lives compared with SpGC3Fab when fused to small recombinant antibody fragments, such as single-chain Fv (scFv) and bispecific single-chain diabody (scDb). Hence, the SpGC3FabRR domain seems to be a suitable fusion partner for the half-life extension of small recombinant therapeutics. Conclusions/Significance The half-life extension properties of SpGC3 can be retained by restricting binding to the Fab fragment of serum immunoglobulins and can be improved by increasing binding activity. The modified SpGC3 module should be suitable to extend the half-life of therapeutic proteins and, thus to improve therapeutic activity.
Collapse
Affiliation(s)
- Felix Unverdorben
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Meike Hutt
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
32
|
Hossain MA, Haugaard-Kedström LM, Rosengren KJ, Bathgate RAD, Wade JD. Chemically synthesized dicarba H2 relaxin analogues retain strong RXFP1 receptor activity but show an unexpected loss of in vitro serum stability. Org Biomol Chem 2015; 13:10895-903. [PMID: 26368576 DOI: 10.1039/c5ob01539a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peptides and proteins are now acknowledged as viable alternatives to small molecules as potential therapeutic agents. A primary limitation to their more widespread acceptance is their generally short in vivo half-lives due to serum enzyme susceptibility and rapid renal clearance. Numerous chemical approaches to address this concern have been undertaken in recent years. The replacement of disulfide bonds with non-reducible elements has been demonstrated to be one effective means by eliminating the deleterious effect of serum reductases. In particular, substitution with dicarba bonds via ring closure metathesis has been increasingly applied to many bioactive cystine-rich peptides. We used this approach for the replacement of the A-chain intramolecular disulfide bond of human relaxin 2 (H2 relaxin), an insulin-like peptide that has important regulatory roles in cardiovascular and connective tissue homeostasis that has led to successful Phase IIIa clinical trials for the treatment of acute heart failure. Use of efficient solid phase synthesis of the two peptide chains was followed by on-resin ring closure metathesis and formation of the dicarba bond within the A-chain and then by off-resin combination with the B-chain via sequential directed inter-chain disulfide bond formation. After purification and comprehensive chemical characterization, the two isomeric synthetic H2 relaxin analogues were shown to retain near-equipotent RXFP1 receptor binding and activation propensity. Unexpectedly, the in vitro serum stability of the analogues was greatly reduced compared with the native peptide. Circular dichroism spectroscopy studies showed subtle differences in the secondary structures between dicarba analogues and H2 relaxin suggesting that, although the overall fold is retained, it may be destabilized which could account for rapid degradation of dicarba analogues in serum. Caution is therefore recommended when using ring closure metathesis as a general approach to enhance peptide stability.
Collapse
Affiliation(s)
- Mohammed Akhter Hossain
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia.
| | | | | | | | | |
Collapse
|
33
|
The neonatal Fc receptor, FcRn, as a target for drug delivery and therapy. Adv Drug Deliv Rev 2015; 91:109-24. [PMID: 25703189 DOI: 10.1016/j.addr.2015.02.005] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/22/2022]
Abstract
Immunoglobulin G (IgG)-based drugs are arguably the most successful class of protein therapeutics due in part to their remarkably long blood circulation. This arises from IgG interaction with the neonatal Fc receptor, FcRn. FcRn is the central regulator of IgG and albumin homeostasis throughout life and is increasingly being recognized as an important player in autoimmune disease, mucosal immunity, and tumor immune surveillance. Various engineering approaches that hijack or disrupt the FcRn-mediated transport pathway have been devised to develop long-lasting and non-invasive protein therapeutics, protein subunit vaccines, and therapeutics for treatment of autoimmune and infectious disease. In this review, we highlight the diverse biological functions of FcRn, emerging therapeutic opportunities, as well as the associated challenges of targeting FcRn for drug delivery and disease therapy.
Collapse
|
34
|
Abstract
The purpose of making a "biobetter" biologic is to improve on the salient characteristics of a known biologic for which there is, minimally, clinical proof of concept or, maximally, marketed product data. There already are several examples in which second-generation or biobetter biologics have been generated by improving the pharmacokinetic properties of an innovative drug, including Neulasta(®) [a PEGylated, longer-half-life version of Neupogen(®) (filgrastim)] and Aranesp(®) [a longer-half-life version of Epogen(®) (epoetin-α)]. This review describes the use of protein fusion technologies such as Fc fusion proteins, fusion to human serum albumin, fusion to carboxy-terminal peptide, and other polypeptide fusion approaches to make biobetter drugs with more desirable pharmacokinetic profiles.
Collapse
Affiliation(s)
- William R Strohl
- Janssen BioTherapeutics, Janssen Research and Development, LLC, Pharmaceutical Companies of Johnson & Johnson, SH31-21757, 1400 Welsh and McKean Roads, PO Box 776, Spring House, PA, 19477, USA,
| |
Collapse
|
35
|
Doki T, Takano T, Koyama Y, Hohdatsu T. Identification of the peptide derived from S1 domain that inhibits type I and type II feline infectious peritonitis virus infection. Virus Res 2015; 204:13-20. [PMID: 25896976 PMCID: PMC7114445 DOI: 10.1016/j.virusres.2015.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/03/2015] [Accepted: 04/08/2015] [Indexed: 12/25/2022]
Abstract
Feline infectious peritonitis (FIP) is a coronavirus-induced fatal disease in cats. We synthesized peptides derived from the S1 domain of the type I FIPV S protein. We investigated inhibitory effects of peptides on FIPV infection. 5 peptides significantly inhibited type I FIPV. 2 of 5 peptides significantly inhibited not only type I, but also type II FIPV.
Feline infectious peritonitis virus (FIPV) can cause a lethal disease in cats, feline infectious peritonitis (FIP). A therapeutic drug that is effective against FIP has not yet been developed. Peptides based on viral protein amino acid sequences have recently been attracting attention as new antiviral drugs. In the present study, we synthesized 30 overlapping peptides based on the amino acid sequence of the S1 domain of the type I FIPV strain KU-2 S protein, and investigated their inhibitory effects on FIPV infection. To evaluate the inhibitory effects on type I FIPV infection of these peptides, we investigated a method to increase the infection efficiency of poorly replicative type I FIPV. The efficiency of type I FIPV infection was increased by diluting the virus with medium containing a polycation. Of the 30 peptides, I-S1-8 (S461-S480), I-S1-9 (S471-S490), I-S1-10 (S481-S500), I-S1-16 (S541-S560), and I-S1-22 (S601-S620) significantly decreased the infectivity of FIPV strain KU-2 while I-S1-9 and I-S1-16 exhibited marked inhibitory effects on FIPV infection. The inhibitory effects on FIPV infection of these 2 peptides on other type I and type II FIPV strains, feline herpesvirus (FHV), and feline calicivirus (FCV) were also examined. These 2 peptides specifically inhibited type I and type II FIPV, but did FHV or FCV infection. In conclusion, the possibility of peptides derived from the S protein of type I FIPV strain KU-2 as anti-FIPV agents effective not only for type I, but also type II FIPV was demonstrated in vitro.
Collapse
Affiliation(s)
- Tomoyoshi Doki
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan.
| | - Tomomi Takano
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan.
| | - Yusuke Koyama
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan.
| | - Tsutomu Hohdatsu
- Laboratory of Veterinary Infectious Disease, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan.
| |
Collapse
|