1
|
Jung KO, Youn H, Kim SH, Kim YH, Kang KW, Chung JK. A new fluorescence/PET probe for targeting intracellular human telomerase reverse transcriptase (hTERT) using Tat peptide-conjugated IgM. Biochem Biophys Res Commun 2016; 477:483-9. [PMID: 27317485 DOI: 10.1016/j.bbrc.2016.06.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
Abstract
Despite an increasing need for methods to visualize intracellular proteins in vivo, the majority of antibody-based imaging methods available can only detect membrane proteins. The human telomerase reverse transcriptase (hTERT) is an intracellular target of great interest because of its high expression in several types of cancer. In this study, we developed a new probe for hTERT using the Tat peptide. An hTERT antibody (IgG or IgM) was conjugated with the Tat peptide, a fluorescence dye and (64)Cu. HT29 (hTERT+) and U2OS (hTERT-) were used to visualize the intracellular hTERT. The hTERT was detected by RT-PCR and western blot. Fluorescence signals for hTERT were obtained by confocal microscopy, live cell imaging, and analyzed by Tissue-FAXS. In nude mice, tumors were visualized using the fluorescence imaging devices Maestro™ and PETBOX. In RT-PCR and western blot, the expression of hTERT was detected in HT29 cells, but not in U2OS cells. Fluorescence signals were clearly observed in HT29 cells and in U2OS cells after 1 h of treatment, but signals were only detected in HT29 cells after 24 h. Confocal microscopy showed that 9.65% of U2OS and 78.54% of HT29 cells had positive hTERT signals. 3D animation images showed that the probe could target intranuclear hTERT in the nucleus. In mice models, fluorescence and PET imaging showed that hTERT in HT29 tumors could be efficiently visualized. In summary, we developed a new method to visualize intracellular and intranuclear proteins both in vitro and in vivo.
Collapse
Affiliation(s)
- Kyung Oh Jung
- Department of Nuclear Medicine, Seoul National University College of Medicine, South Korea; Biomedical Sciences, Seoul National University College of Medicine, South Korea; Cancer Research Institute, Seoul National University College of Medicine, South Korea; Tumor Microenvironment Global Core Research Center, Seoul National University, South Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, South Korea; Cancer Research Institute, Seoul National University College of Medicine, South Korea; Tumor Microenvironment Global Core Research Center, Seoul National University, South Korea; Cancer Imaging Center, Seoul National University Hospital, Seoul, South Korea.
| | - Seung Hoo Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, South Korea; Cancer Research Institute, Seoul National University College of Medicine, South Korea
| | - Young-Hwa Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, South Korea; Biomedical Sciences, Seoul National University College of Medicine, South Korea; Cancer Research Institute, Seoul National University College of Medicine, South Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, South Korea; Cancer Research Institute, Seoul National University College of Medicine, South Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, South Korea; Biomedical Sciences, Seoul National University College of Medicine, South Korea; Cancer Research Institute, Seoul National University College of Medicine, South Korea; Tumor Microenvironment Global Core Research Center, Seoul National University, South Korea.
| |
Collapse
|
2
|
Beidler CB, Petrovan RJ, Conner EM, Boyles JS, Yang DD, Harlan SM, Chu S, Ellis B, Datta-Mannan A, Johnson RL, Stauber A, Witcher DR, Breyer MD, Heuer JG. Generation and activity of a humanized monoclonal antibody that selectively neutralizes the epidermal growth factor receptor ligands transforming growth factor-α and epiregulin. J Pharmacol Exp Ther 2014; 349:330-43. [PMID: 24518034 DOI: 10.1124/jpet.113.210765] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
At least seven distinct epidermal growth factor (EGF) ligands bind to and activate the EGF receptor (EGFR). This activation plays an important role in the embryo and in the maintenance of adult tissues. Importantly, pharmacologic EGFR inhibition also plays a critical role in the pathophysiology of diverse disease states, especially cancer. The roles of specific EGFR ligands are poorly defined in these disease states. Accumulating evidence suggests a role for transforming growth factor α (TGFα) in skin, lung, and kidney disease. To explore the role of Tgfa, we generated a monoclonal antibody (mAb41) that binds to and neutralizes human Tgfa with high affinity (KD = 36.5 pM). The antibody also binds human epiregulin (Ereg) (KD = 346.6 pM) and inhibits ligand induced myofibroblast cell proliferation (IC50 values of 0.52 and 1.12 nM for human Tgfa and Ereg, respectively). In vivo, a single administration of the antibody to pregnant mice (30 mg/kg s.c. at day 14 after plug) or weekly administration to neonate mice (20 mg/kg s.c. for 4 weeks) phenocopy Tgfa knockout mice with curly whiskers, stunted growth, and expansion of the hypertrophic zone of growth plate cartilage. Humanization of this monoclonal antibody to a human IgG4 antibody (LY3016859) enables clinical development. Importantly, administration of the humanized antibody to cynomolgus monkeys is absent of the skin toxicity observed with current EGFR inhibitors used clinically and no other pathologies were noted, indicating that neutralization of Tgfa could provide a relatively safe profile as it advances in clinical development.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Animals, Newborn
- Antibodies, Monoclonal, Humanized/metabolism
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Neutralizing/metabolism
- Antibodies, Neutralizing/pharmacology
- Cell Line
- Cell Proliferation/drug effects
- Epidermal Growth Factor/metabolism
- Epiregulin
- ErbB Receptors/metabolism
- Humans
- Immunoglobulin G/immunology
- Macaca fascicularis
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Myofibroblasts/cytology
- Myofibroblasts/drug effects
- Myofibroblasts/metabolism
- Protein Binding
- Transforming Growth Factor alpha/genetics
- Transforming Growth Factor alpha/metabolism
Collapse
|
3
|
Hoet RM, Cohen EH, Kent RB, Rookey K, Schoonbroodt S, Hogan S, Rem L, Frans N, Daukandt M, Pieters H, van Hegelsom R, Neer NCV, Nastri HG, Rondon IJ, Leeds JA, Hufton SE, Huang L, Kashin I, Devlin M, Kuang G, Steukers M, Viswanathan M, Nixon AE, Sexton DJ, Hoogenboom HR, Ladner RC. Generation of high-affinity human antibodies by combining donor-derived and synthetic complementarity-determining-region diversity. Nat Biotechnol 2005; 23:344-8. [PMID: 15723048 DOI: 10.1038/nbt1067] [Citation(s) in RCA: 218] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Accepted: 01/05/2005] [Indexed: 11/09/2022]
Abstract
Combinatorial libraries of rearranged hypervariable V(H) and V(L) sequences from nonimmunized human donors contain antigen specificities, including anti-self reactivities, created by random pairing of V(H)s and V(L)s. Somatic hypermutation of immunoglobulin genes, however, is critical in the generation of high-affinity antibodies in vivo and occurs only after immunization. Thus, in combinatorial phage display libraries from nonimmunized donors, high-affinity antibodies are rarely found. Lengthy in vitro affinity maturation is often needed to improve antibodies from such libraries. We report the construction of human Fab libraries having a unique combination of immunoglobulin sequences captured from human donors and synthetic diversity in key antigen contact sites in heavy-chain complementarity-determining regions 1 and 2. The success of this strategy is demonstrated by identifying many monovalent Fabs against multiple therapeutic targets that show higher affinities than approved therapeutic antibodies. This very often circumvents the need for affinity maturation, accelerating discovery of antibody drug candidates.
Collapse
|
4
|
Vasserot AP, Dickinson CD, Tang Y, Huse WD, Manchester KS, Watkins JD. Optimization of protein therapeutics by directed evolution. Drug Discov Today 2003; 8:118-26. [PMID: 12568781 DOI: 10.1016/s1359-6446(02)02590-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Directed evolution is a broadly applicable technology platform that is ideally suited to address the need for protein optimization and to fully exploit the therapeutic potential of biologics. The approach takes advantage of the remarkable structural and functional plasticity of proteins and permits the rapid remodeling of biologics into new entities with improved functions. The ability to ameliorate virtually any characteristic of a protein can translate into significant clinical benefits, including decreased immunogenicity, higher potency, greater efficacy and improved safety profile, and can considerably increase the probability of successfully developing and commercializing biotherapeutics.
Collapse
Affiliation(s)
- Alain P Vasserot
- Applied Molecular Evolution (AME), 3520 Dunhill Street 92121, San Diego, CA, USA.
| | | | | | | | | | | |
Collapse
|