1
|
Floresta G, Abbate V. Recent progress in the imaging of c-Met aberrant cancers with positron emission tomography. Med Res Rev 2022; 42:1588-1606. [PMID: 35292998 PMCID: PMC9314990 DOI: 10.1002/med.21885] [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: 07/09/2021] [Revised: 01/31/2022] [Accepted: 02/22/2022] [Indexed: 11/06/2022]
Abstract
Tyrosine-protein kinase Met-also known as c-Met or HGFR-is a membrane receptor protein with associated tyrosine kinase activity physiologically stimulated by its natural ligand, the hepatocyte growth factor (HGF), and is involved in different ways in cancer progression and tumourigenesis. Targeting c-Met with pharmaceuticals has been preclinically proved to have significant benefits for cancer treatment. Recently, evaluating the protein status during and before c-Met targeted therapy has been shown of relevant importance by different studies, demonstrating that there is a correlation between the status (e.g., aberrant activation and overexpression) of the HGFR with therapy response and clinical prognosis. Currently, clinical imaging based on positron emission tomography (PET) appears as one of the most promising tools for the in vivo real-time scanning of irregular alterations of the tyrosine-protein kinase Met and for the diagnosis of c-Met related cancers. In this study, we review the recent progress in the imaging of c-Met aberrant cancers with PET. Particular attention is directed on the development of PET probes with a range of different sizes (HGF, antibodies, anticalines, peptides, and small molecules), and radiolabeled with different radionuclides. The goal of this review is to report all the preclinical imaging studies based on PET imaging reported until now for in vivo diagnosis of c-Met in oncology to support the design of novel and more effective PET probes for in vivo evaluation of c-Met.
Collapse
Affiliation(s)
- Giuseppe Floresta
- Department of Analytical, Environmental and Forensic Sciences, Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - Vincenzo Abbate
- Department of Analytical, Environmental and Forensic Sciences, Institute of Pharmaceutical Sciences, King's College London, London, UK
| |
Collapse
|
2
|
Siegemund M, Oak P, Hansbauer EM, Allersdorfer A, Utschick K, Winter A, Grasmüller C, Galler G, Mayer JP, Weiche B, Prassler J, Kontermann RE, Rothe C. Pharmacokinetic Engineering of OX40-Blocking Anticalin Proteins Using Monomeric Plasma Half-Life Extension Domains. Front Pharmacol 2021; 12:759337. [PMID: 34759826 PMCID: PMC8573339 DOI: 10.3389/fphar.2021.759337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/04/2021] [Indexed: 12/26/2022] Open
Abstract
Anticalin® proteins have been proven as versatile clinical stage biotherapeutics. Due to their small size (∼20 kDa), they harbor a short intrinsic plasma half-life which can be extended, e.g., by fusion with IgG or Fc. However, for antagonism of co-immunostimulatory Tumor Necrosis Factor Receptor Superfamily (TNFRSF) members in therapy of autoimmune and inflammatory diseases, a monovalent, pharmacokinetically optimized Anticalin protein format that avoids receptor clustering and therefore potential activation is favored. We investigated the suitability of an affinity-improved streptococcal Albumin-Binding Domain (ABD) and the engineered Fab-selective Immunoglobulin-Binding Domain (IgBD) SpGC3Fab for plasma Half-Life Extension (HLE) of an OX40-specific Anticalin and bispecific Duocalin proteins, neutralizing OX40 and a second co-immunostimulatory TNFRSF member. The higher affinity of ABD fusion proteins to human serum albumin (HSA) and Mouse Serum Albumin (MSA), with a 4 to 5-order of magnitude lower KD compared with the binding affinity of IgBD fusions to human/mouse IgG, translated into longer terminal plasma half-lives (t1/2). Hence, the anti-OX40 Anticalin-ABD protein reached t1/2 values of ∼40 h in wild-type mice and 110 h in hSA/hFcRn double humanized mice, in contrast to ∼7 h observed for anti-OX40 Anticalin-IgBD in wild-type mice. The pharmacokinetics of an anti-OX40 Anticalin-Fc fusion protein was the longest in both models (t1/2 of 130 h and 146 h, respectively). Protein formats composed of two ABDs or IgBDs instead of one single HLE domain clearly showed longer presence in the circulation. Importantly, Anticalin-ABD and -IgBD fusions showed OX40 receptor binding and functional competition with OX40L-induced cellular reactivity in the presence of albumin or IgG, respectively. Our results suggest that fusion to ABD or IgBD can be a versatile platform to tune the plasma half-life of Anticalin proteins in response to therapeutic needs.
Collapse
Affiliation(s)
- Martin Siegemund
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Prajakta Oak
- Pieris Pharmaceuticals GmbH, Hallbergmoos, Germany
| | | | | | | | | | | | | | | | | | | | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | | |
Collapse
|
3
|
Huang R, Liu Y, Wang K, Wang Z, Zhang C, Zhang W, Zhao Z, Li G, Huang L, Chang Y, Zeng F, Jiang T, Hu H. High-sensitive clinical diagnostic method for PTPRZ1-MET and the characteristic protein structure contributing to ligand-independent MET activation. CNS Neurosci Ther 2021; 27:617-628. [PMID: 33645009 PMCID: PMC8025647 DOI: 10.1111/cns.13627] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 01/26/2021] [Accepted: 01/31/2021] [Indexed: 12/12/2022] Open
Abstract
Background PTPRZ1‐MET (ZM) is a critical genetic alteration driving the progression of lower‐grade glioma. Glioma patients harboring ZM could benefit from MET inhibitors. According to the remarkable role of ZM as a driver of glioma progression and indicator of MET inhibitor sensitivity, it is necessary to detect this alteration even when it presents in glioma with relatively fewer copies. Methods Herein, we proposed that ZM could be detected with a high‐sensitive method of reverse transcriptase PCR with 50 amplification cycles. Via this newly proposed detection method, we depicted the incidence preference of ZM fusion in a cohort of 485 glioma patients. To further explore the oncogenic nature of ZM, we predicated the protein structure alteration of MET kinase brought by its fusion partner. Results The incidence of ZM fusions was much higher than previous report. ZM fusions exhibited a striking preference in lower‐grade glioma and secondary glioblastoma. By contrast, none of patients with primary glioblastoma was detected harboring ZM fusion. In each of the four variants of ZM, the fusion partner segment of MET contained a remarkable coiled‐coil motif. In glioma cells expressing ZM, MET kinase could be activated in a ligand‐independent manner, which might be contributed by the special coiled‐coil structure brought by the fusion partner. Corresponding to the 3D structural analysis and cell line experiment, the ZM positive clinical specimens showed hyperactivations of MET signaling. Conclusions ZM fusions are critical drivers of glioma progression and effective target of MET inhibitor. Early detection could be performed with a high‐sensitive method of reverse transcriptase PCR. The hyperactivations of MET signaling driving glioma progression might be contributed by a ligand‐independent activation enabled by the protein structure modification of extracellular domain of MET in ZM fusions.
Collapse
Affiliation(s)
- Ruoyu Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Yanwei Liu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Kuanyu Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Zheng Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Chuanbao Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Wei Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Zheng Zhao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Lijie Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Yuanhao Chang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| |
Collapse
|
4
|
Abstract
INTRODUCTION Anticalin proteins are engineered versions of lipocalins that constitute a novel class of clinical-stage biopharmaceuticals. The lipocalins exhibit a central β-barrel with eight antiparallel β-strands and an α-helix attached to its side. Four structurally variable loops at the open end of the β-barrel form a pronounced binding pocket, which can be reshaped to generate specificities toward diverse disease-relevant molecular targets. AREAS COVERED This article reviews the current status of Anticalin engineering, from the basic principles to the development of Anticalins with high target affinity and specificity via combinatorial protein design and directed evolution, including examples of Anticalin-based drug candidates under preclinical and clinical development. EXPERT OPINION Combinatorial gene libraries together with powerful molecular selection techniques have enabled the expansion of the natural ligand specificities of lipocalins from small molecules to peptides and proteins. This biomolecular concept has been validated by structural analyses of a series of Anticalin•target complexes. Promising Anticalin lead candidates have reached different preclinical and clinical development stages in the areas of (immuno)oncology, metabolic, and respiratory diseases, as antidotes to treat intoxications and as novel antibiotics. Thus, Anticalins offer an alternative to antibodies with promising and potentially superior features as next-generation biologics.
Collapse
Affiliation(s)
| | - Elena Ilyukhina
- Chair of Biological Chemistry, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Arne Skerra
- Chair of Biological Chemistry, School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
5
|
Occupation of a thermoresistant-scaffold (αRep) at SP1-NC cleavage site disturbs the function of HIV-1 protease. Biosci Rep 2020; 40:225239. [PMID: 32519747 PMCID: PMC7313444 DOI: 10.1042/bsr20201131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 11/25/2022] Open
Abstract
HIV-1 nucleocapsid (NC) becomes an attractive target for the development of novel anti-HIV-1 agents. Discovering of non-antibody scaffolds that disrupt the function of NC will be a potential aspect for disturbing viral maturation process. Correspondingly, we explored the specific binding site of the thermoresistant-scaffold protein, αRep9A8 which formerly demonstrated the inhibitory effect on HIV-1 replication. The portion of Gag, CA21-SP1-NC has been used as a template for designing nine overlapping peptides (P4–P12). The P9 peptide showed the strongest binding activity followed by P8 and P12 respectively. The amino acid sequences on those peptides resemble the N-terminal domain of the NC proximity to the SP1-NC initial cleavage site and across the conserved CCHC zinc finger 1 (ZF1) of NC. The interaction KD between αRep9A8 with its target was 224.9 ± 57.4 nM. Consequently, αRep9A8 demonstrated the interference of the HIV-1 protease function by hindering a protease cleavage site. The released NC product from CA21-SP1-NC was diminished. The present study provided an additional information of αRep9A8 function in interfering of viral maturation processes resulting in the decremental efficiency of viral infectivity.
Collapse
|
6
|
Warnders FJ, Lub-de Hooge MN, de Vries EGE, Kosterink JGW. Influence of protein properties and protein modification on biodistribution and tumor uptake of anticancer antibodies, antibody derivatives, and non-Ig scaffolds. Med Res Rev 2018; 38:1837-1873. [PMID: 29635825 DOI: 10.1002/med.21498] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/30/2018] [Accepted: 03/02/2018] [Indexed: 12/11/2022]
Abstract
Newly developed protein drugs that target tumor-associated antigens are often modified in order to increase their therapeutic effect, tumor exposure, and safety profile. During the development of protein drugs, molecular imaging is increasingly used to provide additional information on their in vivo behavior. As a result, there are increasing numbers of studies that demonstrate the effect of protein modification on whole body distribution and tumor uptake of protein drugs. However, much still remains unclear about how to interpret obtained biodistribution data correctly. Consequently, there is a need for more insight in the correct way of interpreting preclinical and clinical imaging data. Summarizing the knowledge gained to date may facilitate this interpretation. This review therefore provides an overview of specific protein properties and modifications that can affect biodistribution and tumor uptake of anticancer antibodies, antibody fragments, and nonimmunoglobulin scaffolds. Protein properties that are discussed in this review are molecular size, target interaction, FcRn binding, and charge. Protein modifications that are discussed are radiolabeling, fluorescent labeling drug conjugation, glycosylation, humanization, albumin binding, and polyethylene glycolation.
Collapse
Affiliation(s)
- Frank-Jan Warnders
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jos G W Kosterink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,PharmacoTherapy, Epidemiology & Economy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
7
|
Hohlbaum AM, Gille H, Trentmann S, Kolodziejczyk M, Rattenstetter B, Laarakkers CM, Katzmann G, Christian HJ, Andersen N, Allersdorfer A, Olwill SA, Meibohm B, Audoly LP, Swinkels DW, van Swelm RPL. Sustained plasma hepcidin suppression and iron elevation by Anticalin-derived hepcidin antagonist in cynomolgus monkey. Br J Pharmacol 2018; 175:1054-1065. [PMID: 29329501 DOI: 10.1111/bph.14143] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Anaemia of chronic disease (ACD) has been linked to iron-restricted erythropoiesis imposed by high circulating levels of hepcidin, a 25 amino acid hepatocyte-derived peptide that controls systemic iron homeostasis. Here, we report the engineering of the human lipocalin-derived, small protein-based anticalin PRS-080 hepcidin antagonist with high affinity and selectivity. EXPERIMENTAL APPROACH Anticalin- and hepcidin-specific pharmacokinetic (PK)/pharmacodynamic modelling (PD) was used to design and select the suitable drug candidate based on t1/2 extension and duration of hepcidin suppression. The development of a novel free hepcidin assay enabled accurate analysis of bioactive hepcidin suppression and elucidation of the observed plasma iron levels after PRS-080-PEG30 administration in vivo. KEY RESULTS PRS-080 had a hepcidin-binding affinity of 0.07 nM and, after coupling to 30 kD PEG (PRS-080-PEG30), a t1/2 of 43 h in cynomolgus monkeys. Dose-dependent iron mobilization and hepcidin suppression were observed after a single i.v. dose of PRS-080-PEG30 in cynomolgus monkeys. Importantly, in these animals, suppression of free hepcidin and subsequent plasma iron elevation were sustained during repeated s.c. dosing. After repeated dosing and followed by a treatment-free interval, all iron parameters returned to pre-dose values. CONCLUSIONS AND IMPLICATIONS In conclusion, we developed a dose-dependent and safe approach for the direct suppression of hepcidin, resulting in prolonged iron mobilization to alleviate iron-restricted erythropoiesis that can address the root cause of ACD. PRS-080-PEG30 is currently in early clinical development.
Collapse
Affiliation(s)
| | | | | | | | | | - Coby M Laarakkers
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands.,Hepcidinanalysis.com, Nijmegen, The Netherlands
| | | | | | | | | | | | - Bernd Meibohm
- University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Dorine W Swinkels
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands.,Hepcidinanalysis.com, Nijmegen, The Netherlands
| | - Rachel P L van Swelm
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands.,Hepcidinanalysis.com, Nijmegen, The Netherlands
| |
Collapse
|
8
|
Masuda Y, Yamaguchi S, Suzuki C, Aburatani T, Nagano Y, Miyauchi R, Suzuki E, Yamamura N, Nagatomo K, Ishihara H, Okuno K, Nara F, Matschiner G, Hashimoto R, Takahashi T, Nishizawa T. Generation and Characterization of a Novel Small Biologic Alternative to Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Antibodies, DS-9001a, Albumin Binding Domain–Fused Anticalin Protein. J Pharmacol Exp Ther 2018; 365:368-378. [DOI: 10.1124/jpet.117.246652] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/15/2018] [Indexed: 01/06/2023] Open
|
9
|
Recent advances in the development of novel protein scaffolds based therapeutics. Int J Biol Macromol 2017; 102:630-641. [DOI: 10.1016/j.ijbiomac.2017.04.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022]
|
10
|
DiCara DM, Chirgadze DY, Pope AR, Karatt-Vellatt A, Winter A, Slavny P, van den Heuvel J, Parthiban K, Holland J, Packman LC, Mavria G, Hoffmann J, Birchmeier W, Gherardi E, McCafferty J. Characterization and structural determination of a new anti-MET function-blocking antibody with binding epitope distinct from the ligand binding domain. Sci Rep 2017; 7:9000. [PMID: 28827556 PMCID: PMC5567289 DOI: 10.1038/s41598-017-09460-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/25/2017] [Indexed: 12/18/2022] Open
Abstract
The growth and motility factor Hepatocyte Growth Factor/Scatter Factor (HGF/SF) and its receptor, the product of the MET proto-oncogene, promote invasion and metastasis of tumor cells and have been considered potential targets for cancer therapy. We generated a new Met-blocking antibody which binds outside the ligand-binding site, and determined the crystal structure of the Fab in complex with its target, which identifies the binding site as the Met Ig1 domain. The antibody, 107_A07, inhibited HGF/SF-induced cell migration and proliferation in vitro and inhibited growth of tumor xenografts in vivo. In biochemical assays, 107_A07 competes with both HGF/SF and its truncated splice variant NK1 for MET binding, despite the location of the antibody epitope on a domain (Ig1) not reported to bind NK1 or HGF/SF. Overlay of the Fab-MET crystal structure with the InternalinB-MET crystal structure shows that the 107_A07 Fab comes into close proximity with the HGF/SF-binding SEMA domain when MET is in the “compact”, InternalinB-bound conformation, but not when MET is in the “open” conformation. These findings provide further support for the importance of the “compact” conformation of the MET extracellular domain, and the relevance of this conformation to HGF/SF binding and signaling.
Collapse
Affiliation(s)
- Danielle M DiCara
- MRC Centre, Hills Road, Cambridge, CB2 2QH, UK.,Department of Oncology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK.,Genentech Inc., South San Francisco, 94080, USA
| | - Dimitri Y Chirgadze
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Anthony R Pope
- IONTAS Ltd, Babraham Institute, Babraham, Cambridgeshire, CB22 3AT, UK
| | | | - Anja Winter
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK.,Faculty of Natural Sciences, Keele University, Staffordshire, ST5 5BG, UK
| | - Peter Slavny
- IONTAS Ltd, Babraham Institute, Babraham, Cambridgeshire, CB22 3AT, UK
| | - Joop van den Heuvel
- Helmholtz Zentrum für Infektionsforschung, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Kothai Parthiban
- IONTAS Ltd, Babraham Institute, Babraham, Cambridgeshire, CB22 3AT, UK
| | - Jane Holland
- Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125, Berlin, Germany
| | - Len C Packman
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Georgia Mavria
- Leeds Institute of Cancer and Pathology, University of Leeds, St James' University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | - Jens Hoffmann
- Experimental Pharmacology & Oncology Berlin-Buch GmbH, Robert-Rössle-Str. 10, 13125, Berlin-Buch, Germany
| | - Walter Birchmeier
- Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125, Berlin, Germany
| | - Ermanno Gherardi
- MRC Centre, Hills Road, Cambridge, CB2 2QH, UK. .,Department of Oncology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK. .,Division of Immunology and General Pathology, Department of Molecular Medicine, 1 via A Ferrata, 27100, Pavia, Italy.
| | - John McCafferty
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK. .,IONTAS Ltd, Babraham Institute, Babraham, Cambridgeshire, CB22 3AT, UK.
| |
Collapse
|
11
|
Han Z, Wu Y, Wang K, Xiao Y, Cheng Z, Sun X, Shen B. Analysis of progress and challenges for various patterns of c-MET-targeted molecular imaging: a systematic review. EJNMMI Res 2017; 7:41. [PMID: 28485003 PMCID: PMC5422222 DOI: 10.1186/s13550-017-0286-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 04/17/2017] [Indexed: 01/27/2023] Open
Abstract
Background Mesenchymal–epithelial transition factor also named c-MET is a receptor tyrosine kinase for the hepatocyte growth factor that plays a pivotal role in tumorigenesis. c-MET-targeted therapies have been tested in preclinical models and patients, with significant benefits for cancer treatment. In recent years, many studies have shown that the expression level and activation status of c-MET are closely correlated to c-MET-targeted therapy response and clinical prognosis, thus highlighting the importance of evaluating the c-MET status during and prior to targeted therapy. Molecular imaging allows the monitoring of abnormal alterations of c-MET in real time and in vivo. Results In this review, we initially summarize the recent advances in c-MET-targeted molecular imaging, with a special focus on the development of imaging agents ranging in size from monoclonal antibody to small molecule. The aim of this review is to report the preclinical results and clinical application of all molecular imaging studies completed until now for in vivo detection of c-MET in cancer, in order to be beneficial to development of molecular probe and the combination of molecular imaging technologies for in vivo evaluation of c-MET. Various molecular probe targeted to c-MET possesses distinctive advantages and disadvantages. For example, antibody-based probes have high binding affinity but with long metabolic cycle as well as remarkable immunogenicity. Conclusions Although studies for c-MET-targeted molecular imaging have made many important advances, most of imaging agents specifically target to extracellular area of c-MET receptor; however, it is difficult to reflect entirely activation of c-MET. Therefore, small molecule probes based on tyrosine kinase inhibitors, which could target to intracellular area of c-MET without any immunogenicity, should be paid more attention.
Collapse
Affiliation(s)
- Zhaoguo Han
- Molecular Imaging Research Center, Harbin Medical University, 766Xiangan N street, Songbei District, Harbin, Heilongjiang, 150028, China.,TOF-PET/CT/MR center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yongyi Wu
- Molecular Imaging Research Center, Harbin Medical University, 766Xiangan N street, Songbei District, Harbin, Heilongjiang, 150028, China.,TOF-PET/CT/MR center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Kai Wang
- Molecular Imaging Research Center, Harbin Medical University, 766Xiangan N street, Songbei District, Harbin, Heilongjiang, 150028, China.,TOF-PET/CT/MR center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yadi Xiao
- Molecular Imaging Research Center, Harbin Medical University, 766Xiangan N street, Songbei District, Harbin, Heilongjiang, 150028, China.,TOF-PET/CT/MR center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Lucas Center, Room P089, 1201 Welch Rd, Stanford, CA, 94305-5484, USA.
| | - Xilin Sun
- Molecular Imaging Research Center, Harbin Medical University, 766Xiangan N street, Songbei District, Harbin, Heilongjiang, 150028, China. .,TOF-PET/CT/MR center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China. .,Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Lucas Center, Room P089, 1201 Welch Rd, Stanford, CA, 94305-5484, USA.
| | - Baozhong Shen
- Molecular Imaging Research Center, Harbin Medical University, 766Xiangan N street, Songbei District, Harbin, Heilongjiang, 150028, China. .,TOF-PET/CT/MR center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
12
|
Gille H, Hülsmeyer M, Trentmann S, Matschiner G, Christian HJ, Meyer T, Amirkhosravi A, Audoly LP, Hohlbaum AM, Skerra A. Functional characterization of a VEGF-A-targeting Anticalin, prototype of a novel therapeutic human protein class. Angiogenesis 2015; 19:79-94. [DOI: 10.1007/s10456-015-9490-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 11/01/2015] [Indexed: 10/22/2022]
|
13
|
Chen L, Li C, Zhu Y. The HGF inhibitory peptide HGP-1 displays promising in vitro and in vivo efficacy for targeted cancer therapy. Oncotarget 2015; 6:30088-101. [PMID: 26254225 PMCID: PMC4745783 DOI: 10.18632/oncotarget.3937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/28/2015] [Indexed: 12/15/2022] Open
Abstract
HGF/MET pathway mediates cancer initiation and development. Thus, inhibition on HGF-initiated MET signaling pathway would provide a new approach to cancer targeted therapeutics. In our study, we identified a targeting peptide candidate binding to HGF which was named HGF binding peptide-1 (HGP-1) via bacterial surface display methods coupled with fluorescence-activated cell sorting (FACS). HGP-1 showed the moderate affinity when determined with surface plasmon resonance (SPR) technique and high specificity in binding to HGF while assessed by fluorescence-based ELISA assay. The results from MTT and in vitro migration assay indicated that HGF-dependent cell proliferation and migration could be inhibited by HGP-1. In vivo administration of HGP-1 led to an effective inhibitory effect on tumor growth in A549 tumor xenograft models. Moreover, findings from Western Blots revealed that HGP-1 could down-regulated the phosphorylation levels of MET and ERK1/2 initiated by HGF, which suggested that HGP-1 could disrupt the activation of HGF/MET signaling to influence the cell activity. All the data highlighted the potential of HGP-1 to be a potent inhibitor for HGF/MET signaling.
Collapse
Affiliation(s)
- Lisha Chen
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.,Suzhou Institute of Nano-Tech and Nano-Bionics, CAS, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunlin Li
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.,Suzhou Institute of Nano-Tech and Nano-Bionics, CAS, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yimin Zhu
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
14
|
Vazquez-Lombardi R, Phan TG, Zimmermann C, Lowe D, Jermutus L, Christ D. Challenges and opportunities for non-antibody scaffold drugs. Drug Discov Today 2015; 20:1271-83. [PMID: 26360055 DOI: 10.1016/j.drudis.2015.09.004] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/06/2015] [Accepted: 09/01/2015] [Indexed: 12/22/2022]
Abstract
The first candidates from the promising class of small non-antibody protein scaffolds are now moving into clinical development and practice. Challenges remain, and scaffolds will need to be further tailored toward applications where they provide real advantages over established therapeutics to succeed in a rapidly evolving drug development landscape.
Collapse
Affiliation(s)
- Rodrigo Vazquez-Lombardi
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia
| | - Carsten Zimmermann
- University of San Diego, School of Business Administration, 5998 Alcala Park, San Diego, CA 92110, USA
| | - David Lowe
- MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK
| | - Lutz Jermutus
- MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK; Trinity Hall, University of Cambridge, Trinity Lane CB2 1TJ, UK.
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia.
| |
Collapse
|
15
|
Ueki R, Sando S. A DNA aptamer to c-Met inhibits cancer cell migration. Chem Commun (Camb) 2015; 50:13131-4. [PMID: 25223895 DOI: 10.1039/c4cc06016d] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The HGF/c-Met pathway is known to play a key role in cancer metastasis. CLN0003_SL1, a 50-mer DNA aptamer that binds to the c-Met protein, was found to inhibit HGF-induced c-Met activation. This aptamer could suppress cancer cell motility in vitro.
Collapse
Affiliation(s)
- R Ueki
- INAMORI Frontier Research Center, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | | |
Collapse
|
16
|
|
17
|
|
18
|
Gillespie DL, Aguirre MT, Ravichandran S, Leishman LL, Berrondo C, Gamboa JT, Wang L, King R, Wang X, Tan M, Malamas A, Lu ZR, Jensen RL. RNA interference targeting hypoxia-inducible factor 1α via a novel multifunctional surfactant attenuates glioma growth in an intracranial mouse model. J Neurosurg 2014; 122:331-41. [PMID: 25423275 DOI: 10.3171/2014.10.jns132363] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT High-grade gliomas are the most common form of adult brain cancer, and patients have a dismal survival rate despite aggressive therapeutic measures. Intratumoral hypoxia is thought to be a main contributor to tumorigenesis and angiogenesis of these tumors. Because hypoxia-inducible factor 1α (HIF-1α) is the major mediator of hypoxia-regulated cellular control, inhibition of this transcription factor may reduce glioblastoma growth. METHODS Using an orthotopic mouse model with U87-LucNeo cells, the authors used RNA interference to knock down HIF-1α in vivo. The small interfering RNA (siRNA) was packaged using a novel multifunctional surfactant, 1-(aminoethyl) iminobis[N-(oleicylcysteinylhistinyl-1-aminoethyl)propionamide] (EHCO), a nucleic acid carrier that facilitates cellular uptake and intracellular release of siRNA. Stereotactic injection was used to deliver siRNA locally through a guide-screw system, and delivery/uptake was verified by imaging of fluorescently labeled siRNA. Osmotic pumps were used for extended siRNA delivery to model a commonly used human intracranial drug-delivery technique, convection-enhanced delivery. RESULTS Mice receiving daily siRNA injections targeting HIF-1α had a 79% lower tumor volume after 50 days of treatment than the controls. Levels of the HIF-1 transcriptional targets vascular endothelial growth factor (VEGF), glucose transporter 1 (GLUT-1), c-MET, and carbonic anhydrase-IX (CA-IX) and markers for cell growth (MIB-1 and microvascular density) were also significantly lower. Altering the carrier EHCO by adding polyethylene glycol significantly increased the efficacy of drug delivery and subsequent survival. CONCLUSIONS Treating glioblastoma with siRNA targeting HIF-1α in vivo can significantly reduce tumor growth and increase survival in an intracranial mouse model, a finding that has direct clinical implications.
Collapse
|
19
|
Allonby O, El Zawily AM, Freywald T, Mousseau DD, Chlan J, Anderson D, Benmerah A, Sidhu V, Babu M, DeCoteau J, Freywald A. Ligand stimulation induces clathrin- and Rab5-dependent downregulation of the kinase-dead EphB6 receptor preceded by the disruption of EphB6-Hsp90 interaction. Cell Signal 2014; 26:2645-57. [PMID: 25152371 DOI: 10.1016/j.cellsig.2014.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/25/2014] [Accepted: 08/15/2014] [Indexed: 12/13/2022]
Abstract
Ligand-induced internalisation and subsequent downregulation of receptor tyrosine kinases (RTKs) serve to determine biological outputs of their signalling. Intrinsically kinase-deficient RTKs control a variety of biological responses, however, the mechanism of their downregulation is not well understood and its analysis is focused exclusively on the ErbB3 receptor. The Eph group of RTKs is represented by the EphA and EphB subclasses. Each bears one kinase-inactive member, EphA10 and EphB6, respectively, suggesting an important role for these molecules in the Eph signalling network. While EphB6 effects on cell behaviour have been assessed, the mechanism of its downregulation remains elusive. Our work reveals that EphB6 and its kinase-active relative, and signalling partner, EphB4, are downregulated in a similar manner in response to their common ligand, ephrin-B2. Following stimulation, both receptors are internalised through clathrin-coated pits and are degraded in lysosomes. Their targeting for lysosomal degradation relies on the activity of an early endosome regulator, the Rab5 GTPase, as this process is inhibited in the presence of a Rab5 dominant-negative mutant. EphB6 also interacts with the Hsp90 chaperone and EphB6 downregulation is preceded by their rapid dissociation. Moreover, the inhibition of Hsp90 results in EphB6 degradation, mimicking its ligand-induced downregulation. These processes appear to rely on overlapping mechanisms, since Hsp90 inhibition does not significantly enhance ligand-induced EphB6 elimination. Taken together, our observations define a novel mechanism for intrinsically kinase-deficient RTK downregulation and support an intriguing model, where Hsp90 dissociation acts as a trigger for ligand-induced receptor removal.
Collapse
Affiliation(s)
- Odette Allonby
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Amr M El Zawily
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Tanya Freywald
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Darrell D Mousseau
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Jennifer Chlan
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Deborah Anderson
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Cancer Research Unit, Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Alexandre Benmerah
- INSERM U1163, Laboratory of Inherited Kidney Diseases, 75015 Paris, France; Université Paris Descartes - Sorbonne Paris Cité, Institut Imagine, 75015 Paris, France.
| | - Vishaldeep Sidhu
- Department of Biochemistry, Research and Innovation Centre, University of Regina, Regina, SK,S4S 0A2, Canada.
| | - Mohan Babu
- Department of Biochemistry, Research and Innovation Centre, University of Regina, Regina, SK,S4S 0A2, Canada.
| | - John DeCoteau
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Andrew Freywald
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
20
|
Aprile G, Giampieri R, Bonotto M, Bittoni A, Ongaro E, Cardellino GG, Graziano F, Giuliani F, Fasola G, Cascinu S, Scartozzi M. The challenge of targeted therapies for gastric cancer patients: the beginning of a long journey. Expert Opin Investig Drugs 2014; 23:925-42. [PMID: 24806575 DOI: 10.1517/13543784.2014.912631] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Despite significant improvements in systemic chemotherapy over the last two decades, the prognosis of patients with advanced disease remains dismal. Collaborative, high-quality research and advances in high-throughput technologies have contributed to elucidate molecular pathways underpinning disease progression and have stimulated many clinical studies testing target therapies in the advanced disease setting. Although progress has been made thanks to trastuzumab in HER2 positive tumours, antiangiogenic drugs have produced conflicting results and EGFR-inhibitors have failed to show major improvements. AREAS COVERED While commenting on the results of many key Phase III randomized trials, the Authors discuss the most promising classes of novel targeted agents and present the current challenges toward a customized treatment. EXPERT OPINION Palliative chemotherapy became the worldwide standard of care for patients with advanced gastric cancers, producing significant life prolongation and improvement of life quality. Nevertheless, long-term outcomes of those patients remain poor. Because of the encouraging advancement in novel targeted therapies, such a disappointing scenario is now evolving. While results serve as a springboard for future research, more comprehensive efforts are needed to clarify the biological mechanisms underpinning cancer progression and help clinicians to develop new effective treatments.
Collapse
Affiliation(s)
- Giuseppe Aprile
- University and General Hospital, Department of Medical Oncology , Piazzale S Maria della Misericordia 1, 33100, Udine , Italy +39 432 559308 ; +39 432 559305 ;
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Terwisscha van Scheltinga AG, Lub-de Hooge MN, Hinner MJ, Verheijen RB, Allersdorfer A, Hülsmeyer M, Nagengast WB, Schröder CP, Kosterink JG, de Vries EG, Audoly L, Olwill SA. In Vivo Visualization of MET Tumor Expression and Anticalin Biodistribution with the MET-Specific Anticalin 89Zr-PRS-110 PET Tracer. J Nucl Med 2014; 55:665-71. [DOI: 10.2967/jnumed.113.124941] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
22
|
The apoptosis repressor with a CARD domain (ARC) gene is a direct hypoxia-inducible factor 1 target gene and promotes survival and proliferation of VHL-deficient renal cancer cells. Mol Cell Biol 2013; 34:739-51. [PMID: 24344197 DOI: 10.1128/mcb.00644-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The induction of hypoxia-inducible factors (HIFs) is essential for the adaptation of tumor cells to a low-oxygen environment. We found that the expression of the apoptosis inhibitor ARC (apoptosis repressor with a CARD domain) was induced by hypoxia in a variety of cancer cell types, and its induction is primarily HIF1 dependent. Chromatin immunoprecipitation (ChIP) and reporter assays also indicate that the ARC gene is regulated by direct binding of HIF1 to a hypoxia response element (HRE) located at bp -190 upstream of the transcription start site. HIFs play an essential role in the pathogenesis of renal cell carcinoma (RCC) under normoxic conditions, through the loss of the Von Hippel-Lindau gene (VHL). Accordingly, our results show that ARC is not expressed in normal renal tissue but is highly expressed in 65% of RCC tumors, which also express high levels of carbonic anhydrase IX (CAIX), a HIF1-dependent protein. Compared to controls, ARC-deficient RCCs exhibited decreased colony formation and increased apoptosis in vitro. In addition, loss of ARC resulted in a dramatic reduction of RCC tumor growth in SCID mice in vivo. Thus, HIF-mediated increased expression of ARC in RCC can explain how loss of VHL can promote survival early in tumor formation.
Collapse
|
23
|
Mross K, Richly H, Fischer R, Scharr D, Büchert M, Stern A, Gille H, Audoly LP, Scheulen ME. First-in-human phase I study of PRS-050 (Angiocal), an Anticalin targeting and antagonizing VEGF-A, in patients with advanced solid tumors. PLoS One 2013; 8:e83232. [PMID: 24349470 PMCID: PMC3862718 DOI: 10.1371/journal.pone.0083232] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/30/2013] [Indexed: 01/17/2023] Open
Abstract
Background To report the nonrandomized first-in-human phase I trial of PRS-050, a novel, rationally engineered Anticalin based on human tear lipocalin that targets and antagonizes vascular endothelial growth factor A (VEGF-A). Methods Patients with advanced solid tumors received PRS-050 at 0.1 mg/kg to 10 mg/kg by IV in successive dosing cohorts according to the 3+3 escalation scheme. The primary end point was safety. Results Twenty-six patients were enrolled; 25 were evaluable. Two patients experienced dose-limiting toxicity, comprising grade (G) 3 hypertension and G3 pyrexia, respectively. The maximum tolerated dose was not reached. Most commonly reported treatment-emergent adverse events (AEs) included chills (52%; G3, 4%), fatigue (52%; G3, 4%), hypertension (44%; G3, 16%), and nausea (40%, all G1/2). No anti–PRS-050 antibodies following multiple administration of the drug were detected. PRS-050 showed dose-proportional pharmacokinetics (PK), with a terminal half-life of approximately 6 days. Free VEGF-A was detectable at baseline in 9/25 patients, becoming rapidly undetectable after PRS-050 infusion for up to 3 weeks. VEGF-A/PRS-050 complex was detectable for up to 3 weeks at all dose levels, including in patients without detectable baseline-free VEGF-A. We also detected a significant reduction in circulating matrix metalloproteinase 2, suggesting this end point could be a pharmacodynamic (PD) marker of the drug’s activity. Conclusions PRS-050, a novel Anticalin with high affinity for VEGF-A, was well-tolerated when administered at the highest dose tested, 10 mg/kg. Based on target engagement and PK/PD data, the recommended phase II dose is 5 mg/kg every 2 weeks administered as a 120-minute infusion. Trial Registration ClinicalTrials.gov NCT01141257 http://clinicaltrials.gov/ct2/show/NCT01141257
Collapse
Affiliation(s)
- Klaus Mross
- Klinik für Tumorbiologie, Albert-Ludwigs Universität, Freiburg, Germany
- * E-mail:
| | - Heike Richly
- Department of Medical Oncology, West German Cancer Center, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Richard Fischer
- Department of Gastroenterology, University Medical Center, Freiburg, German
| | - Dirk Scharr
- Klinik für Tumorbiologie, Albert-Ludwigs Universität, Freiburg, Germany
| | - Martin Büchert
- Magnetic Resonance Development and Application Center, University Medical Center, Freiburg, Germany
| | | | | | | | - Max E. Scheulen
- Department of Medical Oncology, West German Cancer Center, University Hospital, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
24
|
Richter A, Eggenstein E, Skerra A. Anticalins: Exploiting a non-Ig scaffold with hypervariable loops for the engineering of binding proteins. FEBS Lett 2013; 588:213-8. [DOI: 10.1016/j.febslet.2013.11.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/01/2013] [Accepted: 11/04/2013] [Indexed: 01/28/2023]
|