1
|
Ito K, Matsuda Y, Mine A, Miyairi K, Kikuchi Y, Konishi A. Bacterially Secretable Single-Chain Tandem Macrocyclic Peptides for High Affinity and Inhibitory Activity. Chembiochem 2023; 24:e202200599. [PMID: 36409290 DOI: 10.1002/cbic.202200599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/21/2022] [Indexed: 11/23/2022]
Abstract
The inhibition of protein-protein interactions (PPIs) is an effective approach for therapy. Owing to their large binding surface areas to target proteins, macrocyclic peptides are suitable molecules for PPI inhibition. In this study, we developed single-chain tandem macrocyclic peptides (STaMPtides) that inhibits the vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2). They were artificially designed to comprise two different VEGFR2-binding macrocyclic peptides linked in tandem by peptide linkers and secreted by Corynebacterium glutamicum. Most potent VEGFR2-inhibitory STaMPtides with length-optimized linkers exhibited >1000 times stronger inhibitory activity than their parental monomeric peptides, possibly due to the avidity effect of heterodimerization. Our approach of using STaMPtides for PPI inhibition may be used to inhibit other extracellular factors, such as growth factors and cytokines.
Collapse
Affiliation(s)
- Kenichiro Ito
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki, 210-8681, Kanagawa, Japan
| | - Yoshihiko Matsuda
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki, 210-8681, Kanagawa, Japan
| | - Ayako Mine
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki, 210-8681, Kanagawa, Japan
| | - Kyohei Miyairi
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki, 210-8681, Kanagawa, Japan
| | - Yoshimi Kikuchi
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki, 210-8681, Kanagawa, Japan
| | - Atsushi Konishi
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki, 210-8681, Kanagawa, Japan
| |
Collapse
|
2
|
Ito K, Matsuda Y, Mine A, Shikida N, Takahashi K, Miyairi K, Shimbo K, Kikuchi Y, Konishi A. Single-chain tandem macrocyclic peptides as a scaffold for growth factor and cytokine mimetics. Commun Biol 2022; 5:56. [PMID: 35031676 PMCID: PMC8760323 DOI: 10.1038/s42003-022-03015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/28/2021] [Indexed: 11/28/2022] Open
Abstract
Mimetics of growth factors and cytokines are promising tools for culturing large numbers of cells and manufacturing regenerative medicine products. In this study, we report single-chain tandem macrocyclic peptides (STaMPtides) as mimetics in a new multivalent peptide format. STaMPtides, which contain two or more macrocyclic peptides with a disulfide-closed backbone and peptide linkers, are successfully secreted into the supernatant by Corynebacterium glutamicum-based secretion technology. Without post-secretion modification steps, such as macrocyclization or enzymatic treatment, bacterially secreted STaMPtides form disulfide bonds, as designed; are biologically active; and show agonistic activities against respective target receptors. We also demonstrate, by cell-based assays, the potential of STaMPtides, which mimic growth factors and cytokines, in cell culture. The STaMPtide technology can be applied to the design, screening, and production of growth factor and cytokine mimetics.
Collapse
Affiliation(s)
- Kenichiro Ito
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan.
| | - Yoshihiko Matsuda
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Ayako Mine
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Natsuki Shikida
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Kazutoshi Takahashi
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Kyohei Miyairi
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Kazutaka Shimbo
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Yoshimi Kikuchi
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Atsushi Konishi
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-8681, Japan
| |
Collapse
|
3
|
The Role of VEGF Receptors as Molecular Target in Nuclear Medicine for Cancer Diagnosis and Combination Therapy. Cancers (Basel) 2021; 13:cancers13051072. [PMID: 33802353 PMCID: PMC7959315 DOI: 10.3390/cancers13051072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary The rapid development of diagnostic and therapeutic methods of the cancer treatment causes that these diseases are becoming better known and the fight against them is more and more effective. Substantial contribution in this development has nuclear medicine that enables very early cancer diagnosis and early start of the so-called targeted therapy. This therapeutic concept compared to the currently used chemotherapy, causes much fewer undesirable side effects, due to targeting a specific lesion in the body. This review article discusses the possible applications of radionuclide-labelled tracers (peptides, antibodies or synthetic organic molecules) that can visualise cancer cells through pathological blood vessel system in close tumour microenvironment. Hence, at a very early step of oncological disease, targeted therapy can involve in tumour formation and growth. Abstract One approach to anticancer treatment is targeted anti-angiogenic therapy (AAT) based on prevention of blood vessel formation around the developing cancer cells. It is known that vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptors (VEGFRs) play a pivotal role in angiogenesis process; hence, application of angiogenesis inhibitors can be an effective approach in anticancer combination therapeutic strategies. Currently, several types of molecules have been utilised in targeted VEGF/VEGFR anticancer therapy, including human VEGF ligands themselves and their derivatives, anti-VEGF or anti-VEGFR monoclonal antibodies, VEGF binding peptides and small molecular inhibitors of VEGFR tyrosine kinases. These molecules labelled with diagnostic or therapeutic radionuclides can become, respectively, diagnostic or therapeutic receptor radiopharmaceuticals. In targeted anti-angiogenic therapy, diagnostic radioagents play a unique role, allowing the determination of the emerging tumour, to monitor the course of treatment, to predict the treatment outcomes and, first of all, to refer patients for AAT. This review provides an overview of design, synthesis and study of radiolabelled VEGF/VEGFR targeting and imaging agents to date. Additionally, we will briefly discuss their physicochemical properties and possible application in combination targeted radionuclide tumour therapy.
Collapse
|
4
|
Mandal A, Patel P, Pal D, Mitra AK. Multi-Layered Nanomicelles as Self-Assembled Nanocarrier Systems for Ocular Peptide Delivery. AAPS PharmSciTech 2019; 20:66. [PMID: 30627825 DOI: 10.1208/s12249-018-1267-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/04/2018] [Indexed: 12/31/2022] Open
Abstract
Despite the great potential of peptides as therapeutics, there is an unmet challenge in sustaining delivery of sufficient amounts in their native forms. This manuscript describes a novel nanocarrier capable of delivering functional small peptides in its native form. Self-assembling multi-layered nanomicelles composed of two polymers, polyoxyethylene hydrogenated castor oil 40 (HCO-40) and octoxynol 40 (OC-40), were designed to combine hydrophilic interaction and solvent-induced encapsulation of peptides and proteins. The polymers are employed to encapsulate peptide or protein in the core of the organo-nanomicelles which are further encapsulated with another layer of the same polymers to form an aqueous stable nanomicellar solution. The size of the multi-layered nanomicelles ranges from ~ 16 to 20 nm with zeta potential close to neutral (~ - 2.44 to 0.39 mV). In vitro release studies revealed that octreotide-loaded multi-layered nanomicelles released octreotide at much slower rate in simulated tear fluid (STF) (~ 27 days) compared to PBST (~ 11 days) in its native form. MTT assay demonstrated negligible toxicity of the multi-layered nanomicelles at lower concentrations in human retinal pigment epithelial (HRPE, D407), human conjunctival epithelial (CCL 20.2), and rhesus choroid-retinal endothelial (RF/6A) cells. This work demonstrates an efficient small peptide delivery platform with significant advantages over existing approaches, as it does not require modification of the peptide, is biodegradable, and has a small size and high loading capacity.
Collapse
|
5
|
Milbradt J, Sonntag E, Wagner S, Strojan H, Wangen C, Lenac Rovis T, Lisnic B, Jonjic S, Sticht H, Britt WJ, Schlötzer-Schrehardt U, Marschall M. Human Cytomegalovirus Nuclear Capsids Associate with the Core Nuclear Egress Complex and the Viral Protein Kinase pUL97. Viruses 2018; 10:v10010035. [PMID: 29342872 PMCID: PMC5795448 DOI: 10.3390/v10010035] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/05/2018] [Accepted: 01/10/2018] [Indexed: 02/07/2023] Open
Abstract
The nuclear phase of herpesvirus replication is regulated through the formation of regulatory multi-component protein complexes. Viral genomic replication is followed by nuclear capsid assembly, DNA encapsidation and nuclear egress. The latter has been studied intensely pointing to the formation of a viral core nuclear egress complex (NEC) that recruits a multimeric assembly of viral and cellular factors for the reorganization of the nuclear envelope. To date, the mechanism of the association of human cytomegalovirus (HCMV) capsids with the NEC, which in turn initiates the specific steps of nuclear capsid budding, remains undefined. Here, we provide electron microscopy-based data demonstrating the association of both nuclear capsids and NEC proteins at nuclear lamina budding sites. Specifically, immunogold labelling of the core NEC constituent pUL53 and NEC-associated viral kinase pUL97 suggested an intranuclear NEC-capsid interaction. Staining patterns with phospho-specific lamin A/C antibodies are compatible with earlier postulates of targeted capsid egress at lamina-depleted areas. Important data were provided by co-immunoprecipitation and in vitro kinase analyses using lysates from HCMV-infected cells, nuclear fractions, or infectious virions. Data strongly suggest that nuclear capsids interact with pUL53 and pUL97. Combined, the findings support a refined concept of HCMV nuclear trafficking and NEC-capsid interaction.
Collapse
Affiliation(s)
- Jens Milbradt
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany.
| | - Eric Sonntag
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany.
| | - Sabrina Wagner
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany.
| | - Hanife Strojan
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany.
| | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany.
| | - Tihana Lenac Rovis
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia.
| | - Berislav Lisnic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia.
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia.
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany.
| | - William J Britt
- Departments of Pediatrics and Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany.
| |
Collapse
|
6
|
Liu R, Li X, Xiao W, Lam KS. Tumor-targeting peptides from combinatorial libraries. Adv Drug Deliv Rev 2017; 110-111:13-37. [PMID: 27210583 DOI: 10.1016/j.addr.2016.05.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 02/07/2023]
Abstract
Cancer is one of the major and leading causes of death worldwide. Two of the greatest challenges in fighting cancer are early detection and effective treatments with no or minimum side effects. Widespread use of targeted therapies and molecular imaging in clinics requires high affinity, tumor-specific agents as effective targeting vehicles to deliver therapeutics and imaging probes to the primary or metastatic tumor sites. Combinatorial libraries such as phage-display and one-bead one-compound (OBOC) peptide libraries are powerful approaches in discovering tumor-targeting peptides. This review gives an overview of different combinatorial library technologies that have been used for the discovery of tumor-targeting peptides. Examples of tumor-targeting peptides identified from each combinatorial library method will be discussed. Published tumor-targeting peptide ligands and their applications will also be summarized by the combinatorial library methods and their corresponding binding receptors.
Collapse
Affiliation(s)
- Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; Division of Hematology & Oncology, Department of Internal Medicine, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
7
|
Three-dimensional ultrasound molecular imaging of angiogenesis in colon cancer using a clinical matrix array ultrasound transducer. Invest Radiol 2015; 50:322-9. [PMID: 25575176 DOI: 10.1097/rli.0000000000000128] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES We sought to assess the feasibility and reproducibility of 3-dimensional ultrasound molecular imaging (USMI) of vascular endothelial growth factor receptor 2 (VEGFR2) expression in tumor angiogenesis using a clinical matrix array transducer and a clinical grade VEGFR2-targeted contrast agent in a murine model of human colon cancer. MATERIALS AND METHODS Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care. Mice with human colon cancer xenografts (n = 33) were imaged with a clinical ultrasound system and transducer (Philips iU22; X6-1) after intravenous injection of either clinical grade VEGFR2-targeted microbubbles or nontargeted control microbubbles. Nineteen mice were scanned twice to assess imaging reproducibility. Fourteen mice were scanned both before and 24 hours after treatment with either bevacizumab (n = 7) or saline only (n = 7). Three-dimensional USMI data sets were retrospectively reconstructed into multiple consecutive 1-mm-thick USMI data sets to simulate 2-dimensional imaging. Vascular VEGFR2 expression was assessed ex vivo using immunofluorescence. RESULTS Three-dimensional USMI was highly reproducible using both VEGFR2-targeted microbubbles and nontargeted control microbubbles (intraclass correlation coefficient, 0.83). The VEGFR2-targeted USMI signal significantly (P = 0.02) decreased by 57% after antiangiogenic treatment compared with the control group, which correlated well with ex vivo VEGFR2 expression on immunofluorescence (ρ = 0.93, P = 0.003). If only central 1-mm tumor planes were analyzed to assess antiangiogenic treatment response, the USMI signal change was significantly (P = 0.006) overestimated by an average of 27% (range, 2%-73%) compared with 3-dimensional USMI. CONCLUSIONS Three-dimensional USMI is feasible and highly reproducible and allows accurate assessment and monitoring of VEGFR2 expression in tumor angiogenesis in a murine model of human colon cancer.
Collapse
|
8
|
Jagoda EM, Bhattacharyya S, Kalen J, Riffle L, Leeder A, Histed S, Williams M, Wong KJ, Xu B, Szajek LP, Elbuluk O, Cecchi F, Raffensperger K, Golla M, Bottaro DP, Choyke P. Imaging the Met Receptor Tyrosine Kinase (Met) and Assessing Tumor Responses to a Met Tyrosine Kinase Inhibitor in Human Xenograft Mouse Models with a [
99m
Tc] (AH-113018) or CY 5** (AH-112543) Labeled Peptide. Mol Imaging 2015. [DOI: 10.2310/7290.2015.00023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Elaine M. Jagoda
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Sibaprasad Bhattacharyya
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Joseph Kalen
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Lisa Riffle
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Avrum Leeder
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Stephanie Histed
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Mark Williams
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Karen J. Wong
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Biying Xu
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Lawrence P. Szajek
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Osama Elbuluk
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Fabiola Cecchi
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Kristen Raffensperger
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Meghana Golla
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Donald P. Bottaro
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| | - Peter Choyke
- From the Molecular Imaging Program, National Cancer Institute (NCI), Bethesda, MD; ADRD, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Small Animal Imaging Program, NCI, Leidos Biomedical Research, Inc. (formerly SAIC-Frederick, Inc.), Frederick, MD; Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Rockville, MD; PET Department, Clinical Center, NIH,
| |
Collapse
|
9
|
Payen T, Dizeux A, Baldini C, Le Guillou-Buffello D, Lamuraglia M, Comperat E, Lucidarme O, Bridal SL. VEGFR2-Targeted Contrast-Enhanced Ultrasound to Distinguish between Two Anti-Angiogenic Treatments. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:2202-2211. [PMID: 25980323 DOI: 10.1016/j.ultrasmedbio.2015.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/01/2015] [Accepted: 04/21/2015] [Indexed: 06/04/2023]
Abstract
The aim of this study was to evaluate the capacity of BR55, an ultrasound contrast agent specifically targeting vascular endothelial growth factor receptor 2 (VEGFR2), to distinguish the specific anti-VEGFR2 therapy effect of sunitinib from other anti-angiogenic effects of a therapy (imatinib) that does not directly inhibit VEGFR2. Sunitinib, imatinib and placebo were administered daily for 11 d (264 h) to 45 BalbC mice bearing ectopic CT26 murine colorectal carcinomas. During the course of therapy, B-mode ultrasound, contrast-enhanced ultrasound and VEGFR2-targeted contrast-enhanced ultrasound were performed to assess tumor morphology, vascularization and VEGFR2 expression, respectively. The angiogenic effects on these three aspects were characterized using tumor volume, contrast-enhanced area and differential targeted enhancement. Necrosis, microvasculature and expression of VEGFR2 were also determined by histology and immunostaining. B-Mode imaging revealed that tumor growth was significantly decreased in sunitinib-treated mice at day 11 (p < 0.05), whereas imatinib did not affect growth. Functional evaluation revealed that the contrast-enhanced area decreased significantly (p < 0.02) and by similar amounts under both anti-angiogenic treatments by day 8 (192 h): -23% for imatinib and -21% for sunitinib. No significant decrease was observed in the placebo group. Targeted contrast-enhanced imaging revealed lower differential targeted enhancement, that is, lower levels of VEGFR2 expression, in sunitinib-treated mice relative to placebo-treated mice from 24 h (p < 0.05) and relative to both placebo- and imatinib-treated mice from 48 h (p < 0.05). Histologic assessment of tumors after the final imaging indicated that necrotic area was significantly higher for the sunitinib group (21%) than for the placebo (8%, p < 0.001) and imatinib (11%, p < 0.05) groups. VEGFR2-targeted ultrasound was able to sensitively differentiate the anti-VEGFR2 effect from the reduced area of tumor with functional flow produced by both anti-angiogenic agents. BR55 molecular imaging was, thus, able both to detect early therapeutic response to sunitinib in CT26 tumors as soon as 24 h after the beginning of the treatment and to provide early discrimination (48 h) between tumor response during anti-angiogenic therapy targeting VEGFR2 expression and response during anti-angiogenic therapy not directly acting on this receptor.
Collapse
Affiliation(s)
- Thomas Payen
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France
| | - Alexandre Dizeux
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France
| | - Capucine Baldini
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France
| | | | - Michele Lamuraglia
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France; Medical Oncology Department, Hopital Louis-Mourier, AP-HP, Colombes, France
| | - Eva Comperat
- Anatomic Pathology Department, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Olivier Lucidarme
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, AP-HP, Laboratoire d'Imagerie Biomédicale, Paris, France
| | - S Lori Bridal
- Laboratoire d'Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Paris, France.
| |
Collapse
|
10
|
Maaß F, Wüstehube-Lausch J, Dickgießer S, Valldorf B, Reinwarth M, Schmoldt HU, Daneschdar M, Avrutina O, Sahin U, Kolmar H. Cystine-knot peptides targeting cancer-relevant human cytotoxic T lymphocyte-associated antigen 4 (CTLA-4). J Pept Sci 2015; 21:651-60. [PMID: 25964162 DOI: 10.1002/psc.2782] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/15/2015] [Accepted: 03/16/2015] [Indexed: 11/09/2022]
Abstract
Cystine-knot peptides sharing a common fold but displaying a notably large diversity within the primary structure of flanking loops have shown great potential as scaffolds for the development of therapeutic and diagnostic agents. In this study, we demonstrated that the cystine-knot peptide MCoTI-II, a trypsin inhibitor from Momordica cochinchinensis, can be engineered to bind to cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), an inhibitory receptor expressed by T lymphocytes, that has emerged as a target for the treatment of metastatic melanoma. Directed evolution was used to convert a cystine-knot trypsin inhibitor into a CTLA-4 binder by screening a library of variants using yeast surface display. A set of cystine-knot peptides possessing dissociation constants in the micromolar range was obtained; the most potent variant was synthesized chemically. Successive conjugation with neutravidin, fusion to antibody Fc domain or the oligomerization domain of C4b binding protein resulted in oligovalent variants that possessed enhanced (up to 400-fold) dissociation constants in the nanomolar range. Our data indicate that display of multiple knottin peptides on an oligomeric scaffold protein is a valid strategy to improve their functional affinity with ramifications for applications in diagnostics and therapy.
Collapse
Affiliation(s)
- Franziska Maaß
- Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | | | - Stephan Dickgießer
- Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Bernhard Valldorf
- Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Michael Reinwarth
- Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | | | | | - Olga Avrutina
- Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | | | - Harald Kolmar
- Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
11
|
Subtype-specific binding peptides enhance the therapeutic efficacy of nanomedicine in the treatment of ovarian cancer. Cancer Lett 2015; 360:39-47. [PMID: 25661733 DOI: 10.1016/j.canlet.2015.01.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/31/2014] [Accepted: 01/29/2015] [Indexed: 01/15/2023]
Abstract
Currently, epithelial ovarian cancer is viewed as a heterogeneous disease with five major histological subtypes. Clear cell carcinoma represents a specific histological subtype of epithelial ovarian cancer that demonstrates more aggressive clinical behavior and drug resistance compared with other subtypes. Nevertheless, clear cell carcinoma is treated in the same manner as the other subtypes without any particular consideration to its unique clinical characteristics. To improve the therapeutic efficacy of the current liposomal doxorubicin approach for the treatment of clear cell carcinoma, we aimed to develop a novel peptide-conjugated liposomal doxorubicin to actively target this subtype. Two phage clones (OC-6 and OC-26) that specifically bound to clear cell carcinoma were isolated from a phage peptide display library after biopanning procedures. The peptide sequences were translated and aligned (OCSP-6 for OC-6, and OCSP-26 for OC-26, respectively). Peptide-conjugated nanoparticles demonstrated better tumor endocytosis and time-dependent gradual increase of intracellular drug uptake than non-targeting liposomal nanoparticles. Furthermore, peptide-conjugated liposomal doxorubicin better controlled tumors than did non-targeting liposomal doxorubicin. The current work may pave a new way for the development of drugs that target each subtype of epithelial ovarian cancer in the future.
Collapse
|
12
|
Jagoda EM, Bhattacharyya S, Kalen J, Riffle L, Leeder A, Histed S, Williams M, Wong KJ, Xu B, Szajek LP, Elbuluk O, Cecchi F, Raffensperger K, Golla M, Bottaro DP, Choyke P. Imaging the Met Receptor Tyrosine Kinase (Met) and Assessing Tumor Responses to a Met Tyrosine Kinase Inhibitor in Human Xenograft Mouse Models with a [99mTc] (AH-113018) or Cy 5** (AH-112543) Labeled Peptide. Mol Imaging 2015; 14:499-515. [PMID: 26461980 PMCID: PMC7709139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023] Open
Abstract
Developing an imaging agent targeting the hepatocyte growth factor receptor protein (Met) status of cancerous lesions would aid in the diagnosis and monitoring of Met-targeted tyrosine kinase inhibitors (TKIs). A peptide targeting Met labeled with [(99m)Tc] had high affinity in vitro (Kd = 3.3 nM) and detected relative changes in Met in human cancer cell lines. In vivo [(99m)Tc]-Met peptide (AH-113018) was retained in Met-expressing tumors, and high-expressing Met tumors (MKN-45) were easily visualized and quantitated using single-photon emission computed tomography or optical imaging. In further studies, MKN-45 mouse xenografts treated with PHA 665752 (Met TKI) or vehicle were monitored weekly for tumor responses by [(99m)Tc]-Met peptide imaging and measurement of tumor volumes. Tumor uptake of [(99m)Tc]-Met peptide was significantly decreased as early as 1 week after PHA 665752 treatment, corresponding to decreases in tumor volumes. These results were comparable to Cy5**-Met peptide (AH-112543) fluorescence imaging using the same treatment model. [(99m)Tc] or Cy5**-Met peptide tumor uptake was further validated by histologic (necrosis, apoptosis) and immunoassay (total Met, p Met, and plasma shed Met) assessments in imaged and nonimaged cohorts. These data suggest that [(99m)Tc] or Cy5**-Met peptide imaging may have clinical diagnostic, prognostic, and therapeutic monitoring applications.
Collapse
|
13
|
Reich LL, Dutta S, Keating AE. SORTCERY-A High-Throughput Method to Affinity Rank Peptide Ligands. J Mol Biol 2014; 427:2135-50. [PMID: 25311858 DOI: 10.1016/j.jmb.2014.09.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 08/13/2014] [Accepted: 09/24/2014] [Indexed: 10/24/2022]
Abstract
Uncovering the relationships between peptide and protein sequences and binding properties is critical for successfully predicting, re-designing and inhibiting protein-protein interactions. Systematically collected data that link protein sequence to binding are valuable for elucidating determinants of protein interaction but are rare in the literature because such data are experimentally difficult to generate. Here we describe SORTCERY, a high-throughput method that we have used to rank hundreds of yeast-displayed peptides according to their affinities for a target interaction partner. The procedure involves fluorescence-activated cell sorting of a library, deep sequencing of sorted pools and downstream computational analysis. We have developed theoretical models and statistical tools that assist in planning these stages. We demonstrate SORTCERY's utility by ranking 1026 BH3 (Bcl-2 homology 3) peptides with respect to their affinities for the anti-apoptotic protein Bcl-xL. Our results are in striking agreement with measured affinities for 19 individual peptides with dissociation constants ranging from 0.1 to 60nM. High-resolution ranking can be used to improve our understanding of sequence-function relationships and to support the development of computational models for predicting and designing novel interactions.
Collapse
Affiliation(s)
- Lothar Luther Reich
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Sanjib Dutta
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Amy E Keating
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
14
|
Shrivastava A, von Wronski M, Tweedle MF, Nunn AD. Identification of ideal peptides for heterovalent ligands. Methods Mol Biol 2014; 1088:97-105. [PMID: 24146399 DOI: 10.1007/978-1-62703-673-3_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Recent use of hetero-dimerization to improve the affinity of peptide ligands has made peptides an attractive alternative to small molecules and proteins. Using this strategy, we have developed peptides with affinities comparable to antibodies and specificities often better than small molecules or antibodies. These peptides can be used as a delivery vehicle for drugs or diagnostics, especially in the case of tumor targeting cytotoxic drugs or targeted diagnostics. We describe here an assay to identify an ideal pair of peptides suitable for heterovalent ligands.
Collapse
Affiliation(s)
- Ajay Shrivastava
- Ernst Felder Laboratories, Bracco Research USA, Princeton, NJ, USA
| | | | | | | |
Collapse
|
15
|
Kenniston JA, Conley GP, Sexton DJ, Nixon AE. A homogeneous fluorescence anisotropy assay for measuring transglutaminase 2 activity. Anal Biochem 2013; 436:13-5. [DOI: 10.1016/j.ab.2013.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 12/28/2012] [Accepted: 01/12/2013] [Indexed: 11/16/2022]
|
16
|
Fischer G, Schirrmacher R, Wängler B, Wängler C. Radiolabeled Heterobivalent Peptidic Ligands: an Approach with High Future Potential for in vivo Imaging and Therapy of Malignant Diseases. ChemMedChem 2013; 8:883-90. [DOI: 10.1002/cmdc.201300081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Indexed: 12/19/2022]
|
17
|
Kawakami T, Ishizawa T, Fujino T, Reid PC, Suga H, Murakami H. In vitro selection of multiple libraries created by genetic code reprogramming to discover macrocyclic peptides that antagonize VEGFR2 activity in living cells. ACS Chem Biol 2013; 8:1205-14. [PMID: 23517428 DOI: 10.1021/cb300697h] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We report the in vitro selection of thioether-macrocyclized peptides against vascular endothelial growth factor receptor 2 (VEGFR2) from multiple, highly diverse peptide libraries constructed utilizing genetic code reprogramming. The macrocyclic peptide libraries consisted of combinations of four types of amino acid linkers for cyclization and two types of elongator amino acid compositions, including four backbone-modified non-proteinogenic amino acids. Affinity selection from these libraries, using our recently developed TRAP (Transcription-translation coupled with Association of Puromycin-linker) display, yielded multiple anti-VEGFR2 macrocyclic peptide leads. Further antagonizing activity-based screening of the chemically synthesized lead peptides identified a potent macrocyclic peptide that inhibited VEGF-induced VEGFR2 autophosphorylation, proliferation, and angiogenesis of living vascular endothelial cells. The TRAP display-based selection from multiple, highly diverse peptide libraries followed by activity-based screening of selected peptides is a powerful strategy for discovering biologically active peptides targeted to various biomolecules.
Collapse
Affiliation(s)
- Takashi Kawakami
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo,
153-8902, Japan
| | - Takahiro Ishizawa
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo,
153-8902, Japan
| | - Tomoshige Fujino
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo,
153-8902, Japan
| | - Patrick C. Reid
- PeptiDream Inc., 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505,
Japan
| | - Hiroaki Suga
- Department of Chemistry,
Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, 113-0033, Tokyo, Japan
| | - Hiroshi Murakami
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo,
153-8902, Japan
| |
Collapse
|
18
|
Bachawal SV, Jensen KC, Lutz AM, Gambhir SS, Tranquart F, Tian L, Willmann JK. Earlier detection of breast cancer with ultrasound molecular imaging in a transgenic mouse model. Cancer Res 2013; 73:1689-98. [PMID: 23328585 DOI: 10.1158/0008-5472.can-12-3391] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
While there is an increasing role of ultrasound for breast cancer screening in patients with dense breast, conventional anatomical ultrasound lacks sensitivity and specificity for early breast cancer detection. In this study, we assessed the potential of ultrasound molecular imaging using clinically translatable vascular endothelial growth factor receptor type 2 (VEGFR2)-targeted microbubbles (MB(VEGFR2)) to improve the diagnostic accuracy of ultrasound in earlier detection of breast cancer and ductal carcinoma in situ (DCIS) in a transgenic mouse model [FVB/N-Tg(MMTV-PyMT)634Mul]. In vivo binding specificity studies (n = 26 tumors) showed that ultrasound imaging signal was significantly higher (P < 0.001) using MB(VEGFR2) than nontargeted microbubbles and imaging signal significantly decreased (P < 0.001) by blocking antibodies. Ultrasound molecular imaging signal significantly increased (P < 0.001) when breast tissue (n = 315 glands) progressed from normal [1.65 ± 0.17 arbitrary units (a.u.)] to hyperplasia (4.21 ± 1.16), DCIS (15.95 ± 1.31), and invasive cancer (78.1 ± 6.31) and highly correlated with ex vivo VEGFR2 expression [R(2) = 0.84; 95% confidence interval (CI), 0.72-0.91; P < 0.001]. At an imaging signal threshold of 4.6 a.u., ultrasound molecular imaging differentiated benign from malignant entities with a sensitivity of 84% (95% CI, 78-88) and specificity of 89% (95% CI, 81-94). In a prospective screening trail (n = 63 glands), diagnostic performance of detecting DCIS and breast cancer was assessed and two independent readers correctly diagnosed malignant disease in more than 95% of cases and highly agreed between each other [intraclass correlation coefficient (ICC) = 0.98; 95% CI, 97-99]. These results suggest that VEGFR2-targeted ultrasound molecular imaging allows highly accurate detection of DCIS and breast cancer in transgenic mice and may be a promising approach for early breast cancer detection in women.
Collapse
Affiliation(s)
- Sunitha V Bachawal
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Shrivastava A, Wang SH, Raju N, Gierach I, Ding H, Tweedle MF. Heterobivalent dual-target probe for targeting GRP and Y1 receptors on tumor cells. Bioorg Med Chem Lett 2012; 23:687-92. [PMID: 23265893 DOI: 10.1016/j.bmcl.2012.11.110] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 11/20/2012] [Accepted: 11/26/2012] [Indexed: 11/19/2022]
Abstract
Receptor targeting ligands for imaging and/or therapy of cancer are limited by heterogeneity of receptor expression by tumor cells, both inter-patient and intra-patient. It is often more important for imaging agents to identify local and distant spread of disease than it is to identify a specific receptor presence. Two natural hormone peptide receptors, GRPR and Y1, are specifically interesting because expression of GRPR, Y1 or both is up-regulated in most breast cancers. We describe here the design and development of a new heterobivalent peptide ligand, truncated bombesin (t-BBN)/BVD15-DO3A, for dual-targeting of GRPR and Y1, and validation of its dual binding capability. Such a probe should be useful in imaging cells, tissues and tumors that are GRPR and/or Y1 positive and should target radioisotopes, for example, (68)Ga and/or (177)Lu, to more tumors cells than single GRPR or Y1 targeted probes. A GRP targeting ligand, J-G-Abz4-QWAVGHLM-NH(2) (J-G-Abz4-t-BBN), and an Y1 targeting ligand, INP-K[ε-J-(α-DO3A-ε-DGa)-K]-YRLRY-NH(2)([ε-J-(α-DO3A-ε-DGa)-K]-BVD-15), were synthesized and coupled to produce the heterobivalent ligand, t-BBN/BVD15-DO3A. Competitive displacement binding assays using t-BBN/BVD15-DO3A against (125)I-Tyr(4)-BBN yielded an IC(50) value of 18 ± 0.7 nM for GRPR in T-47D cells, a human breast cancer cell line. A similar assay using t-BBN/BVD15-DO3A against porcine (125)I-NPY showed IC(50) values of 80 ± 11 nM for Y1 receptor in MCF7 cells, another human breast cancer cell line. In conclusion, it is possible to construct a single DO3A chelate containing probe that can target both GRPR and Y1 on human tumor cells.
Collapse
Affiliation(s)
- Ajay Shrivastava
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
20
|
Frinking PJA, Tardy I, Théraulaz M, Arditi M, Powers J, Pochon S, Tranquart F. Effects of acoustic radiation force on the binding efficiency of BR55, a VEGFR2-specific ultrasound contrast agent. ULTRASOUND IN MEDICINE & BIOLOGY 2012; 38:1460-1469. [PMID: 22579540 DOI: 10.1016/j.ultrasmedbio.2012.03.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 03/08/2012] [Accepted: 03/25/2012] [Indexed: 05/31/2023]
Abstract
This work describes an in vivo study analyzing the effect of acoustic radiation force (ARF) on the binding of BR55 VEGFR2-specific contrast-agent microbubbles in a model of prostatic adenocarcinoma in rat. A commercial ultrasound system was modified by implementing high duty-cycle 3.5-MHz center frequency ARF bursts in a scanning configuration. This enabled comparing the effects of ARF on binding in tumor and healthy tissue effectively in the same field of view. Bubble binding was established by measuring late-phase enhancement in amplitude modulation (AM) contrast-specific imaging mode (4 MHz, 150 kPa) 10 min after agent injection when the unbound bubbles were cleared from the circulation. Optimal experimental conditions, such as agent concentration (0.4 × 10(8)-1.6 × 10(8) bubbles/kg), acoustic pressure amplitude (26-51 kPa) and duty-cycle (20%-95%) of the ARF bursts, were evaluated in their ability to enhance binding in tumor without significantly increasing binding in healthy tissue. Using the optimal conditions (38 kPa peak-negative pressure, 95% duty cycle), ARF-assisted binding of BR55 improved significantly in tumor (by a factor of 7) at a lower agent dose compared with binding without ARF, and it had an insignificant effect on binding in healthy tissue. Thus, the high binding specificity of BR55 microbubbles for targeting VEGFR2 present at sites of active angiogenesis was confirmed by this study. Therefore, it is believed that based on the results obtained in this work, ultrasound molecular imaging using target-specific contrast-agent microbubbles should preferably be performed in combination with ARF.
Collapse
Affiliation(s)
- Peter J A Frinking
- Bracco Suisse SA, Geneva Research Center and Manufacturing Site, Plan-les-Ouates/Geneva, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
21
|
Zhang XX, Eden HS, Chen X. Peptides in cancer nanomedicine: drug carriers, targeting ligands and protease substrates. J Control Release 2012; 159:2-13. [PMID: 22056916 PMCID: PMC3288222 DOI: 10.1016/j.jconrel.2011.10.023] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 10/18/2011] [Indexed: 01/22/2023]
Abstract
Peptides are attracting increasing attention as therapeutic agents, as the technologies for peptide development and manufacture continue to mature. Concurrently, with booming research in nanotechnology for biomedical applications, peptides have been studied as an important class of components in nanomedicine, and they have been used either alone or in combination with nanomaterials of every reported composition. Peptides possess many advantages, such as smallness, ease of synthesis and modification, and good biocompatibility. Their functions in cancer nanomedicine, discussed in this review, include serving as drug carriers, as targeting ligands, and as protease-responsive substrates for drug delivery.
Collapse
Affiliation(s)
- Xiao-Xiang Zhang
- Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
22
|
High throughput profiling of serum phosphoproteins/peptides using the SELDI-TOF-MS platform. Methods Mol Biol 2012; 818:199-216. [PMID: 22083825 DOI: 10.1007/978-1-61779-418-6_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Protein phosphorylation is a dynamic post-translational modification that plays a critical role in the regulation of a wide spectrum of biological events and cellular functions including signal transduction, gene expression, cell proliferation, and apoptosis. Determination of the sites and magnitudes of protein phosphorylation has been an essential step in the analysis of the control of many biological systems. A high throughput analysis of phosphorylation of proteins would provide a simple, logical, and useful tool for a functional dissection and prediction of biological functions and signaling pathways in association with these important molecular events. We have developed a functional proteomics technique using the ProteinChip array-based SELDI-TOF-MS analysis for high throughput profiling of phosphoproteins/phosphopeptides in human serum for the early detection and diagnosis as well as for the molecular staging of human cancer. The methodology and experimental approach consists of five steps: (1) generation of a total peptide pool of serum proteins by a global trypsin digestion; (2) rapid isolation of phosphopeptides from the total serum peptide pool by an affinity selection, purification, and enrichment using a novel automated micro-bioprocessing system with phospho-antibody-conjugated paramagnetic beads and a hybrid magnet plate; (3) high throughput phosphopeptide analysis on ProteinChip arrays by automated SELDI-TOF-MS; and (4) bioinformatics and statistical methods for data analysis. This method with appropriate modifications may be equally applicable to serine-, threonine- and tyrosine-phosphorylated proteins and for selectively isolating, profiling, and identifying phosphopeptides present in a highly complex phosphor-peptide mixture prepared from various human specimens such as cells, tissue samples, and serum and other body fluids.
Collapse
|
23
|
Gupta N, Belcher PE, Johnston SA, Diehnelt CW. Engineering a Synthetic Ligand for Tumor Necrosis Factor–Alpha. Bioconjug Chem 2011; 22:1473-8. [DOI: 10.1021/bc200091c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Nidhi Gupta
- Center for Innovations in Medicine, The Biodesign Institute at Arizona State University, Tempe, Arizona 85287, United States
| | - Paul E. Belcher
- Center for Innovations in Medicine, The Biodesign Institute at Arizona State University, Tempe, Arizona 85287, United States
| | - Stephen Albert Johnston
- Center for Innovations in Medicine, The Biodesign Institute at Arizona State University, Tempe, Arizona 85287, United States
| | - Chris W. Diehnelt
- Center for Innovations in Medicine, The Biodesign Institute at Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
24
|
Josan JS, Handl HL, Sankaranarayanan R, Xu L, Lynch RM, Vagner J, Mash EA, Hruby VJ, Gillies RJ. Cell-specific targeting by heterobivalent ligands. Bioconjug Chem 2011; 22:1270-8. [PMID: 21639139 DOI: 10.1021/bc1004284] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Current cancer therapies exploit either differential metabolism or targeting to specific individual gene products that are overexpressed in aberrant cells. The work described herein proposes an alternative approach--to specifically target combinations of cell-surface receptors using heteromultivalent ligands ("receptor combination approach"). As a proof-of-concept that functionally unrelated receptors can be noncovalently cross-linked with high avidity and specificity, a series of heterobivalent ligands (htBVLs) were constructed from analogues of the melanocortin peptide ligand ([Nle(4), dPhe(7)]-α-MSH) and the cholecystokinin peptide ligand (CCK-8). Binding of these ligands to cells expressing the human Melanocortin-4 receptor and the Cholecystokinin-2 receptor was analyzed. The MSH(7) and CCK(6) were tethered with linkers of varying rigidity and length, constructed from natural and/or synthetic building blocks. Modeling data suggest that a linker length of 20-50 Å is needed to simultaneously bind these two different G-protein coupled receptors (GPCRs). These ligands exhibited up to 24-fold enhancement in binding affinity to cells that expressed both (bivalent binding), compared to cells with only one (monovalent binding) of the cognate receptors. The htBVLs had up to 50-fold higher affinity than that of a monomeric CCK ligand, i.e., Ac-CCK(6)-NH(2). Cell-surface targeting of these two cell types with labeled heteromultivalent ligand demonstrated high avidity and specificity, thereby validating the receptor combination approach. This ability to noncovalently cross-link heterologous receptors and target individual cells using a receptor combination approach opens up new possibilities for specific cell targeting in vivo for therapy or imaging.
Collapse
Affiliation(s)
- Jatinder S Josan
- Department of Chemistry & Biochemistry, 1306 E. University Blvd., The University of Arizona, Tucson, Arizona 85721, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
OBJECTIVES To evaluate BR55, a new VEGFR2-specific ultrasound contrast agent, for imaging prostate tumors in an orthotopic model in the rat. MATERIALS AND METHODS Rat prostate adenocarcinoma were established by injection of G Dunning R-3327 tumor cells in one lobe of the prostate of Copenhagen rats. Imaging experiments were performed with BR55, SonoVue, and streptavidin-functionalized microbubbles coupled with an anti-vascular endothelial growth factor receptor 2 (VEGFR2) antibody using a clinical ultrasound scanner. Contrast enhancement in the tumor and healthy prostate was followed over time by intermittent imaging at low acoustic power. Signal quantification and statistical analysis were performed in the tumor and healthy tissue to compare the behavior of the 3 contrast agents. Immunohistochemistry was performed on the prostate and tumor specimen to determine the expression of VEGFR2. RESULTS Comparable contrast enhancement was observed in tumors at peak intensity for BR55 and SonoVue. Then, once unbound microbubbles had cleared from the circulation, a strong enhancement of the tumor was obtained with BR55, whereas no significant microbubble accumulation was detected in the healthy prostate tissue. SonoVue microbubbles were rapidly eliminated, and no significant binding was observed in the tumor. The tumor to prostate ratio calculated after signal quantification was about 20 for the 3 doses of BR55 tested. The enhancement obtained with BR55 in the tumor was not significantly different from the one observed with antibody-coupled streptavidin microbubbles. Intense staining for VEGFR2 was detected in the tumor vessels by immunohistochemistry. CONCLUSIONS This study showed that BR55 binding to prostate tumors resulted in a strong enhancement of the lesions as early as a few minutes after contrast injection, whereas minimal nonspecific accumulation occurred in the healthy part of the gland. BR55, like SonoVue, provide information on tissue perfusion during the early vascular phase, but BR55 binding to the tumoral endothelium allows to gain additional information by highlighting the sites of active angiogenesis. The late phase enhancement of the tumor should be particularly valuable for prostate cancer detection and for biopsy guidance.
Collapse
|
26
|
Molecular and functional ultrasound imaging in differently aggressive breast cancer xenografts using two novel ultrasound contrast agents (BR55 and BR38). Eur Radiol 2011; 21:1988-95. [DOI: 10.1007/s00330-011-2138-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 03/21/2011] [Accepted: 04/07/2011] [Indexed: 10/18/2022]
|
27
|
Empting M, Avrutina O, Meusinger R, Fabritz S, Reinwarth M, Biesalski M, Voigt S, Buntkowsky G, Kolmar H. "Triazole bridge": disulfide-bond replacement by ruthenium-catalyzed formation of 1,5-disubstituted 1,2,3-triazoles. Angew Chem Int Ed Engl 2011; 50:5207-11. [PMID: 21544910 DOI: 10.1002/anie.201008142] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 02/21/2011] [Indexed: 12/20/2022]
Affiliation(s)
- Martin Empting
- Clemens-Schöpf-Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Petersenstrasse 22, 64287 Darmstadt, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Empting M, Avrutina O, Meusinger R, Fabritz S, Reinwarth M, Biesalski M, Voigt S, Buntkowsky G, Kolmar H. “Triazolbrücke”: ein Disulfidbrückenersatz durch Ruthenium- katalysierte Bildung von 1,5-disubstituierten 1,2,3-Triazolen. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201008142] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
29
|
PYSZ MARYBETHA, WILLMANN JÜRGENK. Targeted contrast-enhanced ultrasound: an emerging technology in abdominal and pelvic imaging. Gastroenterology 2011; 140:785-90. [PMID: 21255573 PMCID: PMC4162392 DOI: 10.1053/j.gastro.2011.01.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
30
|
Pysz MA, Foygel K, Rosenberg J, Gambhir SS, Schneider M, Willmann JK. Antiangiogenic cancer therapy: monitoring with molecular US and a clinically translatable contrast agent (BR55). Radiology 2010; 256:519-27. [PMID: 20515975 DOI: 10.1148/radiol.10091858] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To develop and test human kinase insert domain receptor (KDR)-targeted microbubbles (MBs) (MB(KDR)) for imaging KDR at the molecular level and for monitoring antiangiogenic therapy in a human colon cancer xenograft tumor model in mice. MATERIALS AND METHODS Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care. A heterodimeric peptide that binds to human KDR with low nanomolar affinity (K(D) = 0.5 nmol/L) was coupled onto the surface of perfluorobutane-containing lipid-shelled MBs (MB(KDR)). Binding specificity of MB(KDR) to human KDR and cross-reactivity with murine vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) were tested in cell culture under flow shear stress conditions (at 100 sec(-1)). In vivo binding specificity of MB(KDR) to VEGFR2 was tested in human LS174T colon cancer xenografts in mice with a 40-MHz ultrasonographic (US) transducer. Targeted contrast material-enhanced US imaging signal by using MB(KDR) was longitudinally measured during 6 days in tumors with (n = 6) and without (n = 6) antiangiogenic treatment (anti-VEGF antibody). Ex vivo VEGFR2 staining and microvessel density analysis were performed. Significant differences were evaluated (t, Mann-Whitney, or Wilcoxon test). RESULTS Cell culture experiments showed four times greater binding specificity of MB(KDR) to human KDR and cross-reactivity to murine VEGFR2 (P < or = .01). In vivo imaging signal was more than three times higher (P = .01) with MB(KDR) compared with control MBs and decreased significantly (approximately fourfold lower, P = .03) following in vivo receptor blocking with anti-VEGFR2 antibody. One day after initiation of antiangiogenic therapy, imaging signal was significantly decreased (approximately 46% lower, P = .02) in treated versus untreated tumors; it remained significantly lower (range, 46%-84% decreased; P = .038) during the following 5 days. Microvessel density was significantly reduced (P = .04) in treated (mean, 7.3 microvessels per square millimeter +/- 4.7 [standard deviation]) versus untreated tumors (mean, 22.0 microvessels per square millimeter +/- 9.4); VEGFR2 expression was significantly decreased (>50% lower, P = .03) in treated tumors. CONCLUSION Human MB(KDR) allow in vivo imaging and longitudinal monitoring of VEGFR2 expression in human colon cancer xenografts.
Collapse
Affiliation(s)
- Marybeth A Pysz
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
31
|
Diehnelt CW, Shah M, Gupta N, Belcher PE, Greving MP, Stafford P, Johnston SA. Discovery of high-affinity protein binding ligands--backwards. PLoS One 2010; 5:e10728. [PMID: 20502719 PMCID: PMC2873402 DOI: 10.1371/journal.pone.0010728] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 04/13/2010] [Indexed: 12/31/2022] Open
Abstract
Background There is a pressing need for high-affinity protein binding ligands for all proteins in the human and other proteomes. Numerous groups are working to develop protein binding ligands but most approaches develop ligands using the same strategy in which a large library of structured ligands is screened against a protein target to identify a high-affinity ligand for the target. While this methodology generates high-affinity ligands for the target, it is generally an iterative process that can be difficult to adapt for the generation of ligands for large numbers of proteins. Methodology/Principal Findings We have developed a class of peptide-based protein ligands, called synbodies, which allow this process to be run backwards – i.e. make a synbody and then screen it against a library of proteins to discover the target. By screening a synbody against an array of 8,000 human proteins, we can identify which protein in the library binds the synbody with high affinity. We used this method to develop a high-affinity synbody that specifically binds AKT1 with a Kd<5 nM. It was found that the peptides that compose the synbody bind AKT1 with low micromolar affinity, implying that the affinity and specificity is a product of the bivalent interaction of the synbody with AKT1. We developed a synbody for another protein, ABL1 using the same method. Conclusions/Significance This method delivered a high-affinity ligand for a target protein in a single discovery step. This is in contrast to other techniques that require subsequent rounds of mutational improvement to yield nanomolar ligands. As this technique is easily scalable, we believe that it could be possible to develop ligands to all the proteins in any proteome using this approach.
Collapse
Affiliation(s)
- Chris W. Diehnelt
- Center for Innovations in Medicine, Arizona State University, Tempe, Arizona, United States of America
| | - Miti Shah
- Center for Innovations in Medicine, Arizona State University, Tempe, Arizona, United States of America
| | - Nidhi Gupta
- Center for Innovations in Medicine, Arizona State University, Tempe, Arizona, United States of America
| | - Paul E. Belcher
- Center for Innovations in Medicine, Arizona State University, Tempe, Arizona, United States of America
| | - Matthew P. Greving
- Center for BioOptical Nanotechnology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Phillip Stafford
- Center for Innovations in Medicine, Arizona State University, Tempe, Arizona, United States of America
| | - Stephen Albert Johnston
- Center for Innovations in Medicine, Arizona State University, Tempe, Arizona, United States of America
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- * E-mail:
| |
Collapse
|
32
|
BR55: a lipopeptide-based VEGFR2-targeted ultrasound contrast agent for molecular imaging of angiogenesis. Invest Radiol 2010; 45:89-95. [PMID: 20027118 DOI: 10.1097/rli.0b013e3181c5927c] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES BR55, an ultrasound contrast agent functionalized with a heterodimer peptide targeting the vascular endothelial growth factor receptor 2 (VEGFR2), was evaluated in vitro and in vivo, demonstrating its potential for specific tumor detection. MATERIALS AND METHODS The targeted contrast agent was prepared by incorporation of a biospecific lipopeptide into the microbubble membrane. Experiments were performed in vitro to demonstrate the binding capacities of BR55 microbubbles on immobilized receptor proteins and on various endothelial or transfected cells expressing VEGFR2. The performance of BR55 microbubbles was compared with that of streptavidin-conjugated microbubbles targeted to the same receptor by coupling them to a biotinylated antibody. The specificity of BR55 binding to human and mouse endothelial cells was determined in competition experiments with the free lipopeptide, vascular endothelial growth factor (VEGF), or a VEGFR2-specific antibody. Molecular ultrasound imaging of VEGFR2 was performed in an orthotopic breast tumor model in rats using a nondestructive, contrast-specific imaging mode. RESULTS BR55 was shown to bind specifically to the immobilized recombinant VEGFR2 under flow (dynamic conditions). BR55 accumulation on the target over time was similar to that of microbubbles bearing a specific antibody. BR55 avidly bound to cells expressing VEGFR2, and the pattern of microbubble distribution was correlated with the pattern of receptor expression determined by immunocytochemistry. The binding of targeted microbubbles on cells was competed off by an excess of free lipopeptide, the natural ligand (VEGF) and by a VEGFR2-specific antibody (P < 0.001). Although selected for the human receptor, the VEGFR2-binding lipopeptide was also shown to recognize the rodent receptor. Tumor perfusion was assessed during the vascular phase of BR55, and then the malignant lesion was highlighted by specific accumulation of the targeted microbubbles on tumoral endothelium. The presence of VEGFR2 was confirmed by immunofluorescence staining of tumor cryosections. CONCLUSIONS VEGFR2-targeted ultrasound contrast agents such as BR55 will likely prove useful in human for the early detection of tumors as well as for the assessment of response to specific treatments.
Collapse
|
33
|
McDonnell KA, Low SC, Hoehn T, Donnelly R, Palmieri H, Fraley C, Sakorafas P, Mezo AR. Synthesis and structure-activity relationships of dimeric peptide antagonists of the human immunoglobulin G-human neonatal Fc receptor (IgG-FcRn) interaction. J Med Chem 2010; 53:1587-96. [PMID: 20092334 DOI: 10.1021/jm901128z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The neonatal Fc receptor, FcRn, regulates the half-life of IgG in vivo and may be a target in the treatment of autoimmune disease. Monomeric peptide antagonists of the human IgG-human FcRn interaction were dimerized using three different synthetic methodologies: thiol/alkyl halide coupling of unprotected peptides, reductive alkylation of unprotected peptides, and on-resin amide bond formation with protected peptides. It was found that dimerization of monomeric peptides increased the in vitro activity of the peptide monomers more than 200-fold. Human IgG catabolism experiments in human FcRn transgenic mice were used to assess the in vivo activity of peptide dimers that possessed different linkers, cyclizations, and affinities for FcRn. Overall, it was found that the linker joining two monomeric peptides had only a minor effect on the in vitro potency but that in vitro potency was predictive of in vivo activity.
Collapse
Affiliation(s)
- Kevin A McDonnell
- Syntonix Pharmaceuticals, Inc., a Subsidiary of Biogen Idec, 9 Fourth Avenue, Waltham, Massachusetts 02451, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Yan Y, Chen X. Peptide heterodimers for molecular imaging. Amino Acids 2010; 41:1081-92. [PMID: 20232091 DOI: 10.1007/s00726-010-0546-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 02/24/2010] [Indexed: 12/19/2022]
Abstract
One main issue with peptide-based molecular imaging probes is their relatively low tumor affinity and short retention time. To improve peptide binding affinity, multivalency approach has been introduced. Traditionally, this approach involves the use of peptide homodimers or homomultimers in which peptide ligands of the same type are constructed with suitable linkers. Recently, a new approach using peptide heterodimers has emerged as a promising method for targeting multi-receptor over-expressed tumor cells. Significant affinity enhancements have been observed with peptide heterodimers compared with their parent peptide monomers. In a peptide heterodimer, two different peptide ligands capable of targeting two different receptors are covalently linked. The binding modes of peptide heterodimers can be monovalent or bivalent depending on whether simultaneous binding of two ligands can be achieved. Increased local ligand concentration and improved binding kinetics contribute to enhanced binding in both monovalent- and bivalent binding modes, while multivalency effect also plays an important role in bivalent binding mode. As many tumors overexpress multiple receptors, more peptide heterodimer-based molecular imaging probes are expected to be developed in future. This review article will discuss the peptide homodimers and heterodimers for molecular imaging with special emphasis on peptide heterodimers.
Collapse
Affiliation(s)
- Yongjun Yan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
35
|
Kenrick SA, Daugherty PS. Bacterial display enables efficient and quantitative peptide affinity maturation. Protein Eng Des Sel 2010; 23:9-17. [PMID: 19903738 DOI: 10.1093/protein/gzp065] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A quantitative screening method was developed to enable isolation and affinity maturation of peptide ligands specific for a given target from peptide libraries displayed on the outer surface of Escherichia coli using multi-parameter flow cytometry. From a large, random 15-mer peptide library, screening identified a core motif of W-E/D-W-E/D that conferred binding to vascular endothelial growth factor (VEGF). One cycle of affinity maturation resulted in the identification of several families of VEGF-binding peptides having distinct consensus sequences, from which a preferred disulfide constraint emerged. In the second affinity maturation cycle, high affinity peptides were favored by the addition of a decoy protein that bound an adjacent epitope on the display scaffold. The decoy apparently reduced rebinding or avidity effects, and the resulting peptides exhibited consensus at 12 of 19 amino acid positions. Peptides identified and affinity matured using bacterial display were remarkably similar to the best affinity matured using phage display and exhibited comparable dissociation constants (within 2-fold; K(D) = 4.7 x 10(-7) M). Screening of bacterial-displayed peptide libraries using cytometry enabled optimization of screening conditions to favor affinity and specificity and rapid clonal characterization. Bacterial display thus provides a new quantitative tool for the discovery and evolutionary optimization of protein-specific peptide ligands.
Collapse
Affiliation(s)
- Sophia A Kenrick
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | | |
Collapse
|
36
|
Pillai R, Marinelli ER, Fan H, Nanjappan P, Song B, von Wronski MA, Cherkaoui S, Tardy I, Pochon S, Schneider M, Nunn AD, Swenson RE. A phospholipid-PEG2000 conjugate of a vascular endothelial growth factor receptor 2 (VEGFR2)-targeting heterodimer peptide for contrast-enhanced ultrasound imaging of angiogenesis. Bioconjug Chem 2010; 21:556-62. [PMID: 20170116 DOI: 10.1021/bc9005688] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The transition of a targeted ultrasound contrast agent from animal imaging to testing in clinical studies requires considerable chemical development. The nature of the construct changes from an agent that is chemically attached to microbubbles to one where the targeting group is coupled to a phospholipid, for direct incorporation to the bubble surface. We provide an efficient method to attach a heterodimeric peptide to a pegylated phospholipid and show that the resulting construct retains nanomolar affinity for its target, vascular endothelial growth factor receptor 2 (VEGFR2), for both the human (kinase insert domain-containing receptor - KDR) and the mouse (fetal liver kinase 1 - Flk-1) receptors. The purified phospholipid-PEG-peptide isolated from TFA-based eluents is not stable with respect to hydrolysis of the fatty ester moieties. This leads to the time-dependent formation of the lysophospholipid and the phosphoglycerylamide derived from the degradation of the product. Purification of the product using neutral eluent systems provides a stable product. Methods to prepare the lysophospholipid (hydrolysis product) are also included. Biacore binding data demonstrated the retention of binding of the lipopeptide to the KDR receptor. The phospholipid-PEG2000-peptide is smoothly incorporated into gas-filled microbubbles and provides imaging of angiogenesis in a rat tumor model.
Collapse
Affiliation(s)
- R Pillai
- The Ernst Felder Laboratories, Bracco Research USA, 305 College Road East, Princeton, New Jersey 08540, and Bracco Research SA, Route de la Galaise, 31, CH-1228 Plan-les-Ouates, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kessel C, Kreuz W, Klich K, Becker-Peters K, Vorpahl F, Dietrich U, Klingebiel T, Königs C. Multimerization of peptide mimotopes for blocking of factor VIII neutralizing antibodies. ChemMedChem 2009; 4:1364-70. [PMID: 19533722 DOI: 10.1002/cmdc.200900023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
About 30 % of patients with severe hemophilia A develop neutralizing antibodies (inhibitors) to coagulation factor VIII (FVIII) upon treatment with exogenous factor preparations. Two peptides, C6 (NPVENMMDRDSQ) and H10 (QSPWQTWFTRAL), that mimic putative inhibitor epitopes (mimotopes), were previously selected by phage display screening of plasma samples from patients with inhibitors. Synthetic peptide mimotopes inhibited IgG binding to FVIII (IC(50): 30-50 microM). This effect was increased by an equimolar combination of both mimotopes. Mimotopes were fused to the C-terminal multimerization domain of the C4bp alpha-chain and expressed as multimers in 293T cells. Multimerized mimotopes showed improved binding to anti-FVIII IgG and prolonged in vitro half-life relative to synthetic peptides. The two mimotopes were combined in heteromultimers by co-transfection of 293T cells with respective vectors, resulting in bi-specific molecules that almost completely blocked polyclonal antibody binding to FVIII (IC(50): 2-3 microM). This strategy is capable of functionally improving synthetic peptides by multimerization and could provide a basis for novel therapeutic approaches for patients with hemophilia A and inhibitors.
Collapse
Affiliation(s)
- Christoph Kessel
- Department of Pediatrics III, JW Goethe University, 60596 Frankfurt am Main, Germany
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Xu L, Vagner J, Josan J, Lynch RM, Morse DL, Baggett B, Han H, Mash EA, Hruby VJ, Gillies RJ. Enhanced targeting with heterobivalent ligands. Mol Cancer Ther 2009; 8:2356-65. [PMID: 19671749 DOI: 10.1158/1535-7163.mct-08-1183] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A novel approach to specifically target tumor cells for detection and treatment is the proposed use of heteromultivalent ligands, which are designed to interact with, and noncovalently crosslink, multiple different cell surface receptors. Although enhanced binding has been shown for synthetic homomultivalent ligands, proof of cross-linking requires the use of ligands with two or more different binding moieties. As proof-of-concept, we have examined the binding of synthetic heterobivalent ligands to cell lines that were engineered to coexpress two different G-protein-coupled human receptors, i.e., the human melanocortin 4 receptor (MC4R) expressed in combination with either the human delta-opioid receptor (deltaOR) or the human cholecystokinin-2 receptor (CCK2R). Expression levels of these receptors were characterized by time-resolved fluorescence saturation binding assays using Europium-labeled ligands; Eu-DPLCE, Eu-NDP-alpha-MSH, and Eu-CCK8 for the deltaOR, MC4R, and CCK2R, respectively. Heterobivalent ligands were synthesized to contain a MC4R agonist connected via chemical linkers to either a deltaOR or a CCK2R agonist. In both cell systems, the heterobivalent constructs bound with much higher affinity to cells expressing both receptors, compared with cells with single receptors or to cells where one of the receptors was competitively blocked. These results indicate that synthetic heterobivalent ligands can noncovalently crosslink two unrelated cell surface receptors, making feasible the targeting of receptor combinations. The in vitro cell models described herein will lead to the development of multivalent ligands for target combinations identified in human cancers.
Collapse
Affiliation(s)
- Liping Xu
- Department of Pharmaceutical Analytics, Pharmaceutical Institute, University of Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Radiotherapeutic drugs and medical imaging agents, although used for different purposes, both benefit from precise targeting. When systemically administered, either would be most useful if designed to find and bind only a tumor, single type of cell, or unique molecular assembly thereon. In this Account, we examine the use of small peptides, natural and synthetic, to create biochemically specific molecular imaging agents and radiotherapeutic pharmaceuticals, discussing three distinct examples. In one project, a small natural peptide known to target members of the bombesin family of receptors was chemically attached to a strong, versatile metal chelator, DO3A, through a series of small-molecule linkers. The linkers powerfully affected not only binding strength for the bombesin receptors, tissue distribution, and tumor uptake in vivo but also receptor subtype specificity. When the assembly is combined with an active metal ion for human trials, the versatility of the DO3A (dodecanetriacetate) chelate affords choices in selecting the metal ion for different purposes: lutetium for a combination radiotherapeutic and diagnostic agent, (177)Lu-AMBA, and gallium for a positron emission tomography (PET) imaging agent, (68)Ga-AMBA. We also created small (approximately 5-kDa) bivalent peptides, each composed of different chemically linked peptides derived from phage display. The monomer peptides bound to the same target protein, VEGF-R2, a primary target of vascular endothelial growth factor (VEGF), the angiogenesis-stimulating protein. Several families of the monomer peptides did not compete with one another for the binding site on VEGF-R2. Their combination into fully synthetic hetero-bivalent molecules yielded subnanomolar K(d) values and greater than 100-fold improvements over homo-bivalent molecules. Biological activity was evident in the hetero-bivalents, whereas none or very little existed in homo-bivalents, monomers, and monomer mixtures. In ultrasound imaging, tiny bubbles (2 microm in diameter) filled with inert gas can be used as effective contrast agents. By coating the shell of such bubbles with the peptide TKPPR (a tuftsin antagonist), we created contrast agents that bound unexpectedly to cultured endothelial cells expressing angiogenesis targets; the binding was attributable to a previously unnoticed and powerful multivalency effect. TKPPR binds specifically to neuropilin-1 (NP-1), a VEGF co-receptor, but only when multimerized is it avid. Tuftsin, a small peptide derived from immunoglobulin G (IgG) that binds to macrophages during inflammation, has been studied for over 30 years; the receptor has never been cloned. Our results led to new conclusions about tuftsin, NP-1, and the purpose, heretofore unknown, of exon 8 in VEGF, which appears to be involved in NP-1 binding. Our disparate projects demonstrate that small-peptide targeted molecules can be very versatile in drug discovery in combination with classical medicinal chemistry. In particular, multivalent interactions can lead to unpredictable and useful biochemical information, as well as new drug candidates.
Collapse
Affiliation(s)
- Michael F. Tweedle
- Departments of Radiology, Chemistry, and Pharmacy; The Ohio State University; Biomedical Research Tower; 460 West 12th Avenue, Columbus, Ohio 43215
| |
Collapse
|
40
|
Zhao P, Grabinski T, Gao C, Skinner RS, Giambernardi T, Su Y, Hudson E, Resau J, Gross M, Vande Woude GF, Hay R, Cao B. Identification of a met-binding peptide from a phage display library. Clin Cancer Res 2007; 13:6049-55. [PMID: 17947467 DOI: 10.1158/1078-0432.ccr-07-0035] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Aberrant c-Met expression has been implicated in most types of human cancer. We are developing Met-directed imaging and therapeutic agents. EXPERIMENTAL DESIGN To seek peptides that bind specifically to receptor Met, the Met-expressing cell lines S114 and SK-LMS-1 were used for biopanning with a random peptide phage display library. Competition ELISA, fluorescence-activated cell sorting analysis, an internalization assay, and a cell proliferation assay were used to characterize a Met-binding peptide in vitro. To evaluate the utility of the peptide as a diagnostic agent in vivo, 125I-labeled peptide was injected i.v. into nude mice bearing s.c. xenografts of the Met-expressing and hepatocyte growth factor (HGF)/scatter factor-expressing SK-LMS-1/HGF, and total body scintigrams were obtained between 1 and 24 h postinjection. RESULTS One Met-binding peptide (YLFSVHWPPLKA), designated Met-pep1, reacts with Met on the cell surface and competes with HGF/scatter factor binding to Met in a dose-dependent manner. Met-pep1 is internalized by Met-expressing cells after receptor binding. Met-pep1 inhibits human leiomyosarcoma SK-LMS-1 cell proliferation in vitro. In SK-LMS-1 mouse xenografts, tumor-associated activity was imaged as early as 1 h postinjection and remained visible in some animals as late as 24 h postinjection. CONCLUSIONS Met-pep1 specifically interacts with Met: it is internalized by Met-expressing cells and inhibits tumor cell proliferation in vitro; it is a potential diagnostic agent for tumor imaging.
Collapse
Affiliation(s)
- Ping Zhao
- Laboratories of Antibody Technology, Van Andel Research Institute, Grand Rapids, Michigan and Nuclear Medicine Service, Department of Veterans Affairs Healthcare System, Ann Arbor, Michigan 49503, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Wang H, Cai W, Chen K, Li ZB, Kashefi A, He L, Chen X. A new PET tracer specific for vascular endothelial growth factor receptor 2. Eur J Nucl Med Mol Imaging 2007; 34:2001-10. [PMID: 17694307 DOI: 10.1007/s00259-007-0524-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Accepted: 06/30/2007] [Indexed: 12/20/2022]
Abstract
PURPOSE Noninvasive positron emission tomography (PET) imaging of vascular endothelial growth factor receptor 2 (VEGFR-2) expression could be a valuable tool for evaluation of patients with a variety of malignancies, and particularly for monitoring those undergoing antiangiogenic therapies that block VEGF/VEGFR-2 function. The aim of this study was to develop a VEGFR-2-specific PET tracer. METHODS The D63AE64AE67A mutant of VEGF(121) (VEGF(DEE)) was generated by recombinant DNA technology. VEGF(121) and VEGF(DEE) were purified and conjugated with DOTA for (64)Cu labeling. The DOTA conjugates were tested in vitro for VEGFR-2 specificity and functional activity. In vivo tumor targeting efficacy and pharmacokinetics of (64)Cu-labeled VEGF(121) and VEGF(DEE) were compared using an orthotopic 4T1 murine breast tumor model. Blocking experiments, biodistribution studies, and immunofluorescence staining were carried out to confirm the noninvasive imaging results. RESULTS Cell binding assay demonstrated that VEGF(DEE) had about 20-fold lower VEGFR-1 binding affinity and only slightly lower VEGFR-2 binding affinity as compared with VEGF(121). MicroPET imaging studies revealed that both (64)Cu-DOTA-VEGF(121) and (64)Cu-DOTA-VEGF(DEE) had rapid and prominent activity accumulation in VEGFR-2-expressing 4T1 tumors. The renal uptake of (64)Cu-DOTA-VEGF(DEE) was significantly lower than that of (64)Cu-DOTA-VEGF(121) as rodent kidneys expressed high levels of VEGFR-1 based on immunofluorescence staining. Blocking experiments and biodistribution studies confirmed the VEGFR specificity of (64)Cu-DOTA-VEGF(DEE). CONCLUSION We have developed a VEGFR-2-specific PET tracer, (64)Cu-DOTA-VEGF(DEE). It has comparable tumor targeting efficacy to (64)Cu-DOTA-VEGF(121) but much reduced renal toxicity. This tracer may be translated into the clinic for imaging tumor angiogenesis and monitoring antiangiogenic treatment efficacy.
Collapse
Affiliation(s)
- Hui Wang
- The Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA 94305-5484, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Sato AK, Viswanathan M, Kent RB, Wood CR. Therapeutic peptides: technological advances driving peptides into development. Curr Opin Biotechnol 2006; 17:638-42. [PMID: 17049837 DOI: 10.1016/j.copbio.2006.10.002] [Citation(s) in RCA: 253] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2006] [Revised: 08/23/2006] [Accepted: 10/06/2006] [Indexed: 12/31/2022]
Abstract
As potential therapeutics, peptides offer several advantages over small molecules (increased specificity) and antibodies (small size). Nevertheless, a number of key issues have hampered their use as drug candidates. A series of new technologies have recently been developed that allow peptides to be viable drug candidates in areas usually restricted to protein therapeutics, such as monoclonal antibodies. These include the development of various types of peptide-conjugates that have lower rates of clearance and hence the potential to increase the exposure of peptide drug candidates in chronic diseases. Structural additions have also been made to peptides, including the use of unnatural amino acids, mainchain modifications and other novel substitutions, which have helped to improve peptide stability and further their therapeutic potential.
Collapse
Affiliation(s)
- Aaron K Sato
- Dyax Corporation, 300 Technology Square, Cambridge, MA 02139, USA
| | | | | | | |
Collapse
|
43
|
von Wronski MA, Raju N, Pillai R, Bogdan NJ, Marinelli ER, Nanjappan P, Ramalingam K, Arunachalam T, Eaton S, Linder KE, Yan F, Pochon S, Tweedle MF, Nunn AD. Tuftsin Binds Neuropilin-1 through a Sequence Similar to That Encoded by Exon 8 of Vascular Endothelial Growth Factor. J Biol Chem 2006; 281:5702-10. [PMID: 16371354 DOI: 10.1074/jbc.m511941200] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tuftsin, Thr-Lys-Pro-Arg (TKPR), is an immunostimulatory peptide with reported nervous system effects as well. We unexpectedly found that tuftsin and a higher affinity antagonist, TKPPR, bind selectively to neuropilin-1 and block vascular endothelial growth factor (VEGF) binding to that receptor. Dimeric and tetrameric forms of TKPPR had greatly increased affinity for neuropilin-1 based on competition binding experiments. On endothelial cells tetrameric TKPPR inhibited the VEGF(165)-induced autophosphorylation of vascular endothelial growth factor receptor-2 (VEGFR-2) even though it did not directly inhibit VEGF binding to VEGFR-2. Homology between exon 8 of VEGF and TKPPR suggests that the sequence coded for by exon 8 may stabilize VEGF binding to neuropilin-1 to facilitate signaling through VEGFR-2. Given the overlap between processes involving neuropilin-1 and tuftsin, we propose that at least some of the previously reported effects of tuftsin are mediated through neuropilin-1.
Collapse
Affiliation(s)
- Mathew A von Wronski
- Ernst Felder Laboratories, Bracco Research USA, 305 College Road East, Princeton, NJ 08540, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
We identified 1113 articles (103 reviews, 1010 primary research articles) published in 2005 that describe experiments performed using commercially available optical biosensors. While this number of publications is impressive, we find that the quality of the biosensor work in these articles is often pretty poor. It is a little disappointing that there appears to be only a small set of researchers who know how to properly perform, analyze, and present biosensor data. To help focus the field, we spotlight work published by 10 research groups that exemplify the quality of data one should expect to see from a biosensor experiment. Also, in an effort to raise awareness of the common problems in the biosensor field, we provide side-by-side examples of good and bad data sets from the 2005 literature.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
45
|
Tweedle MF. Adventures in multivalency, the Harry S. Fischer memorial lecture CMR 2005; Evian, France. CONTRAST MEDIA & MOLECULAR IMAGING 2006; 1:2-9. [PMID: 17193594 DOI: 10.1002/cmmi.91] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This review discusses multivalency in the context of drug discovery, specifically the discovery of new diagnostic imaging and related agents. The aim is to draw attention to the powerful role that multivalency plays throughout research involving molecular biology, in general, and much of biochemically targeted contrast agent research, in particular. Two examples from the author's laboratory are described. We created small (approximately 5 kDa) peptide 'dimers' composed of two different, chemically linked peptides. The monomer peptides both bound to the same target protein with K(d) approximately 100 s nM, while the heterodimers had sub-nM K(d) values. Biological activity was evident in the heterodimers where none or very little existed in homodimers, monomers or monomer mixtures. Two different tyrosine kinases (KDR and C-Met) and four peptide families produced consistent results: multivalent heterodimers were uniquely different. The second example begins with making two micron ultrasound bubbles coated with the peptide, TKPPR (a Tuftsin antagonist) as a negative control for bubbles targeted with angiogenesis target-binding peptides. Unexpected binding of a 'negative' control, (TKPPR)-targeted bubble to endothelial cells expressing angiogenesis targets, led to the surprising result that TKPPR, only when multimerized, binds avidly, specifically and actively to neuropilin-1, a VEGF co-receptor. VEGF is the primary stimulator of angiogenesis. Tuftsin is a small peptide (TKPR) derived from IgG that binds to macrophages during inflammation, and has been studied for over 30 years. The receptor has never been cloned. The results led to new conclusions about Tuftsin, neuropilin-1 and the purpose, up to now unknown, of exon 8 in VEGF. Multivalency can be used rationally to solve practical problems in drug discovery. When targeting larger structures, multivalency is frequently unavoidable, and can lead to unpredictable and useful biochemical information, as well as to new drug candidates.
Collapse
Affiliation(s)
- Michael F Tweedle
- Ernst Felder Laboratories, Bracco Research USA Inc., 305 College Rd East, Princeton, NJ 08540, USA.
| |
Collapse
|
46
|
Pillai R, Marinelli ER, Swenson RE. A flexible method for preparation of peptide homo- and heterodimers functionalized with affinity probes, chelating ligands, and latent conjugating groups. Biopolymers 2006; 84:576-85. [PMID: 16845666 DOI: 10.1002/bip.20570] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dimerization of peptides can provide high binding entities to serve as targeted diagnostics or therapeutics. We developed methods for the preparation of homo- and heterodimer peptides bearing functional molecules (affinity probes, chelating ligands, or latent conjugating moieties). Monomer peptides, optionally bearing spacer groups, are tethered using a bifunctional linker, (di-succinimidyl glutarate, DSG) to provide the dimers. Protected or unprotected peptides can be employed for dimer assembly. Multiple lysine N(epsilon)-amino groups are controlled using the (4,4-dimethyl-2,6-dioxocyclohex-1-ylidene)-3-methylbutyl (ivDde) protecting group. Functional molecules are optionally incorporated into the component peptides or into the assembled dimer. The methods are efficient and scaleable.
Collapse
Affiliation(s)
- Radhakrishna Pillai
- The Ernst Felder Laboratories, Bracco Research USA, Inc., 305 College Road East, Princeton, NJ 08540-6608, USA.
| | | | | |
Collapse
|