1
|
Tognolini M, Ferrari FR, Zappia A, Giorgio C. Ephrin receptor type-A2 (EphA2) targeting in cancer: a patent review (2018-present). Expert Opin Ther Pat 2024; 34:1009-1018. [PMID: 39259047 DOI: 10.1080/13543776.2024.2402382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/12/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION EphA2 is a tyrosine kinase receptor and is considered a promising target in cancer. Different approaches are used to target EphA2 receptor, and a lot of preclinical data demonstrate the potential exploitation of this receptor in clinical oncology for diagnosis and cancer therapy, including immunotherapy. AREAS COVERED In this review, we have summarized the recent patents involving the EphA2 targeting in cancer. For this aim, we used the patent database Patentscope covering the time period of 2018-present. Preclinical and clinical data of the inventions were considered when published on peer reviewed journals. Moreover, the clinicalTrial.gov identifiers (NCT numbers) were included when available. For an easier and more immediate reading, we classify the patents in different categories, considering the nature (aptamers, small molecules, antibodies, peptides, antigens and chimeric antigen receptors) of the inventions exploiting EphA2 in clinical oncology. EXPERT OPINION Despite the availability of a plethora of chemically diverse agents, there are no approved anticancer drugs targeting EphA2 yet. However, these intellectual properties, some of which supported by strong preclinical evidence, keep the hope that, after more than 30 years from its discovery, we will finally see the first EphA2 targeting agent approved in clinical oncology.
Collapse
Affiliation(s)
| | | | - Alfonso Zappia
- Food and Drug Department, University of Parma, Parma, Italy
| | | |
Collapse
|
2
|
Venu A, Zhang Y, Seong J, Hong Y, Lee WS, Min JJ. Engineering of an EPHA2-Targeted Monobody for the Detection of Colorectal Cancer. Cancer Genomics Proteomics 2024; 21:285-294. [PMID: 38670584 PMCID: PMC11059598 DOI: 10.21873/cgp.20447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/26/2024] [Accepted: 02/20/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND/AIM Colorectal cancer (CRC) is the third most common cancer worldwide, and is second only to lung cancer with respect to cancer-related deaths. Noninvasive molecular imaging using established markers is a new emerging method to diagnose CRC. The human ephrin receptor family type-A 2 (hEPHA2) oncoprotein is overexpressed at the early, but not late, stages of CRC. Previously, we reported development of an E1 monobody that is specific for hEPHA2-expressing cancer cells both in vitro and in vivo. Herein, we investigated the ability of the E1 monobody to detect hEPHA2 expressing colorectal tumors in a mouse model, as well as in CRC tissue. MATERIALS AND METHODS The expression of hEPHA2 on the surface of CRC cells was analyzed by western blotting and flow cytometry. The targeting efficacy of the E1 monobody for CRC cells was examined by flow cytometry, and immunofluorescence staining. E1 conjugated to the Renilla luciferase variant 8 (Rluc8) reporter protein was used for in vivo imaging in mice. Additionally, an enhanced green fluorescence protein (EGFP) conjugated E1 monobody was used to check the ability of the E1 monobody to target CRC tissue. RESULTS The E1 monobody bound efficiently to hEPHA2-expressing CRC cell lines, and E1 conjugated to the Rluc8 reporter protein targeted tumor tissues in mice transplanted with HCT116 CRC tumor cells. Finally, E1-EGFP stained tumor tissues from human CRC patients, showing a pattern similar to that of an anti-hEPHA2 antibody. CONCLUSION The E1 monobody has utility as an EPHA2 targeting agent for the detection of CRC.
Collapse
Affiliation(s)
- Akhil Venu
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeonnam, Republic of Korea
| | - Ying Zhang
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Jihyoun Seong
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Wan-Sik Lee
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea;
| | - Jung-Joon Min
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea;
| |
Collapse
|
3
|
Kobayashi T, Yamazaki K, Shinada J, Mizunuma M, Furukawa K, Chuman Y. Identification of Inhibitors of the Disease-Associated Protein Phosphatase Scp1 Using Antibody Mimetic Molecules. Int J Mol Sci 2024; 25:3737. [PMID: 38612548 PMCID: PMC11011526 DOI: 10.3390/ijms25073737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Protein phosphorylation is a prevalent translational modification, and its dysregulation has been implicated in various diseases, including cancer. Despite its significance, there is a lack of specific inhibitors of the FCP/SCP-type Ser/Thr protein phosphatase Scp1, characterized by high specificity and affinity. In this study, we focused on adnectin, an antibody-mimetic protein, aiming to identify Scp1-specific binding molecules with a broad binding surface that target the substrate-recognition site of Scp1. Biopanning of Scp1 was performed using an adnectin-presenting phage library with a randomized FG loop. We succeeded in identifying FG-1Adn, which showed high affinity and specificity for Scp1. Ala scanning analysis of the Scp1-binding sequence in relation to the FG-1 peptide revealed that hydrophobic residues, including aromatic amino acids, play important roles in Scp1 recognition. Furthermore, FG-1Adn was found to co-localize with Scp1 in cells, especially on the plasma membrane. In addition, Western blotting analysis showed that FG-1Adn increased the phosphorylation level of the target protein of Scp1 in cells, indicating that FG-1Adn can inhibit the function of Scp1. These results suggest that FG-1Adn can be used as a specific inhibitor of Scp1.
Collapse
Affiliation(s)
| | | | | | | | | | - Yoshiro Chuman
- Department of Chemistry, Faculty of Science, Niigata University, Niigata 950-2181, Japan; (T.K.); (K.Y.); (J.S.); (M.M.); (K.F.)
| |
Collapse
|
4
|
Campbell E, Luxton T, Kohl D, Goodchild SA, Walti C, Jeuken LJC. Chimeric Protein Switch Biosensors. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2024; 187:1-35. [PMID: 38273207 DOI: 10.1007/10_2023_241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Rapid detection of protein and small-molecule analytes is a valuable technique across multiple disciplines, but most in vitro testing of biological or environmental samples requires long, laborious processes and trained personnel in laboratory settings, leading to long wait times for results and high expenses. Fusion of recognition with reporter elements has been introduced to detection methods such as enzyme-linked immunoassays (ELISA), with enzyme-conjugated secondary antibodies removing one of the many incubation and wash steps. Chimeric protein switch biosensors go further and provide a platform for homogenous mix-and-read assays where long wash and incubation steps are eradicated from the process. Chimeric protein switch biosensors consist of an enzyme switch (the reporter) coupled to a recognition element, where binding of the analyte results in switching the activity of the reporter enzyme on or off. Several chimeric protein switch biosensors have successfully been developed for analytes ranging from small molecule drugs to large protein biomarkers. There are two main formats of chimeric protein switch biosensor developed, one-component and multi-component, and these formats exhibit unique advantages and disadvantages. Genetically fusing a recognition protein to the enzyme switch has many advantages in the production and performance of the biosensor. A range of immune and synthetic binding proteins have been developed as alternatives to antibodies, including antibody mimetics or antibody fragments. These are mainly small, easily manipulated proteins and can be genetically fused to a reporter for recombinant expression or manipulated to allow chemical fusion. Here, aspects of chimeric protein switch biosensors will be reviewed with a comparison of different classes of recognition elements and switching mechanisms.
Collapse
Affiliation(s)
- Emma Campbell
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Timothy Luxton
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Declan Kohl
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | | | - Christoph Walti
- School of Electronic and Electrical Engineering, University of Leeds, Leeds, UK
| | - Lars J C Jeuken
- School of Biomedical Sciences, University of Leeds, Leeds, UK.
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
5
|
Development of Antibody-like Proteins Targeting the Oncogenic Ser/Thr Protein Phosphatase PPM1D. Processes (Basel) 2022. [DOI: 10.3390/pr10081501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PPM1D, a protein Ser/Thr phosphatase, is overexpressed in various cancers and functions as an oncogenic protein by inactivating the p53 pathway. Therefore, molecules that bind PPM1D are expected to be useful anti-cancer agents. In this study, we constructed a phage display library based on the antibody-like small molecule protein adnectin and screened for PPM1D-specific binding molecules. We identified two adnectins, PMDB-1 and PMD-24, that bind PPM1D specific B-loop and PPM1D430 as targets, respectively. Specificity analyses of these recombinant proteins using other Ser/Thr protein phosphatases showed that these molecules bind to only PPM1D. Expression of PMDB-1 in breast cancer-derived MCF-7 cells overexpressing endogenous PPM1D stabilized p53, indicating that PMDB-1 functions as an inhibitor of PPM1D. Furthermore, MTT assay exhibited that MCF-7 cells expressing PMDB-1 showed inhibition of cell proliferation. These data suggest that the adnectin PMDB-1 identified in this study can be used as a lead compound for anti-cancer drugs targeting intracellular PPM1D.
Collapse
|
6
|
Zhang Y, Thangam R, You SH, Sultonova RD, Venu A, Min JJ, Hong Y. Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy. Cancers (Basel) 2021; 13:2801. [PMID: 34199835 PMCID: PMC8200062 DOI: 10.3390/cancers13112801] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 12/26/2022] Open
Abstract
Surface-exposed calreticulin (ecto-CRT) plays a crucial role in the phagocytic removal of apoptotic cells during immunotherapy. Ecto-CRT is an immunogenic signal induced in response to treatment with chemotherapeutic agents such as doxorubicin (DOX) and mitoxantrone (MTX), and two peptides (KLGFFKR (Integrin-α) and GQPMYGQPMY (CRT binding peptide 1, Hep-I)) are known to specifically bind CRT. To engineer CRT-specific monobodies as agents to detect immunogenic cell death (ICD), we fused these peptide sequences at the binding loops (BC and FG) of human fibronectin domain III (FN3). CRT-specific monobodies were purified from E. coli by affinity chromatography. Using these monobodies, ecto-CRT was evaluated in vitro, in cultured cancer cell lines (CT-26, MC-38, HeLa, and MDA-MB-231), or in mice after anticancer drug treatment. Monobodies with both peptide sequences (CRT3 and CRT4) showed higher binding to ecto-CRT than those with a single peptide sequence. The binding affinity of the Rluc8 fusion protein-engineered monobodies (CRT3-Rluc8 and CRT4-Rluc8) to CRT was about 8 nM, and the half-life in serum and tumor tissue was about 12 h. By flow cytometry and confocal immunofluorescence of cancer cell lines, and by in vivo optical bioluminescence imaging of tumor-bearing mice, CRT3-Rluc8 and CRT4-Rluc8 bound specifically to ecto-CRT and effectively detected pre-apoptotic cells after treatment with ICD-inducing agents (DOX and MTX) but not a non-ICD-inducing agent (gemcitabine). Using CRT-specific monobodies, it is possible to detect ecto-CRT induction in cancer cells in response to drug exposure. This technique may be used to predict the therapeutic efficiency of chemo- and immuno-therapeutics early during anticancer treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea; (Y.Z.); (R.T.); (S.-H.Y.); (R.D.S.); (A.V.)
| | - Ramar Thangam
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea; (Y.Z.); (R.T.); (S.-H.Y.); (R.D.S.); (A.V.)
- Department of Materials Science & Engineering, Korea University, Seoul 02841, Korea
| | - Sung-Hwan You
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea; (Y.Z.); (R.T.); (S.-H.Y.); (R.D.S.); (A.V.)
| | - Rukhsora D. Sultonova
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea; (Y.Z.); (R.T.); (S.-H.Y.); (R.D.S.); (A.V.)
| | - Akhil Venu
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea; (Y.Z.); (R.T.); (S.-H.Y.); (R.D.S.); (A.V.)
| | - Jung-Joon Min
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea; (Y.Z.); (R.T.); (S.-H.Y.); (R.D.S.); (A.V.)
- Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Yeongjin Hong
- Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, Korea; (Y.Z.); (R.T.); (S.-H.Y.); (R.D.S.); (A.V.)
- Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Korea
| |
Collapse
|
7
|
Miller CJ, McGinnis JE, Martinez MJ, Wang G, Zhou J, Simmons E, Amet T, Abdeen SJ, Van Huysse JW, Bowsher RR, Kay BK. FN3-based monobodies selective for the receptor binding domain of the SARS-CoV-2 spike protein. N Biotechnol 2021; 62:79-85. [PMID: 33556628 PMCID: PMC7863792 DOI: 10.1016/j.nbt.2021.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 01/19/2021] [Accepted: 01/31/2021] [Indexed: 12/17/2022]
Abstract
A phage library displaying 1010 variants of the fibronectin type III (FN3) domain was affinity selected with the biotinylated form of the receptor binding domain (RBD, residues 319-541) of the SARS-CoV-2 virus spike protein. Nine binding FN3 variants (i.e. monobodies) were recovered, representing four different primary structures. Soluble forms of the monobodies bound to several different preparations of the RBD and the S1 spike subunit, with affinities ranging from 3 to 14 nM as measured by bio-layer interferometry. Three of the four monobodies bound selectively to the RBD of SARS-CoV-2, with the fourth monobody showing slight cross-reactivity to the RBD of SARS-CoV-1 virus. Examination of binding to the spike fragments and its trimeric form revealed that the monobodies recognise at least three overlapping epitopes on the RBD of SARS-CoV-2. While pairwise tests failed to identify a monobody pair that could bind simultaneously to the RBD, one monobody could simultaneously bind to the RBD with the ectodomain of the cellular receptor angiotensin converting enzyme 2 (ACE2). All four monobodies successfully bound the RBD after overexpression in Chinese hamster ovary (CHO) cells as fusions to the Fc domain of human IgG1.
Collapse
Affiliation(s)
- Christina J Miller
- Tango Biosciences, Inc., 2201 W. Campbell Park Drive, Chicago, IL 60612 USA
| | | | - Michael J Martinez
- Tango Biosciences, Inc., 2201 W. Campbell Park Drive, Chicago, IL 60612 USA
| | - Guangli Wang
- Euprotein Inc., 675 US Highway 1, Suite 129, North Brunswick, NJ 08902 USA
| | - Jian Zhou
- LifeTein LLC, 100 Randolph Road, Suite 2D, Somerset, NJ 08873 USA
| | - Erica Simmons
- B2S Life Sciences, 97 East Monroe Street, Franklin, IN 46131 USA
| | - Tohti Amet
- B2S Life Sciences, 97 East Monroe Street, Franklin, IN 46131 USA
| | - Sanofar J Abdeen
- B2S Life Sciences, 97 East Monroe Street, Franklin, IN 46131 USA
| | | | - Ronald R Bowsher
- B2S Life Sciences, 97 East Monroe Street, Franklin, IN 46131 USA
| | - Brian K Kay
- Tango Biosciences, Inc., 2201 W. Campbell Park Drive, Chicago, IL 60612 USA.
| |
Collapse
|
8
|
Pyo A, You SH, Sik Kim H, Young Kim J, Min JJ, Kim DY, Hong Y. Production of 64Cu-labeled monobody for imaging of human EphA2-expressing tumors. Bioorg Med Chem Lett 2020; 30:127262. [PMID: 32527560 DOI: 10.1016/j.bmcl.2020.127262] [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: 03/02/2020] [Revised: 04/20/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
We previously reported on the monobody E1, which specifically targets the tumor marker hEphA2. In this study, we labeled NOTA-conjugated E1 with 64Cu (64Cu-NOTA-E1) and evaluated biologic characteristics. The uptake of 64Cu-NOTA-E1 in PC3 cells (a human prostate cancer cell line) with high expression of hEphA2 increased in a time-dependent manner. In PC3 xenograft mice, 64Cu-NOTA-E1 injected via the tail vein allowed visualization of tumors on positron emission tomography after 1 h and the highest uptake measured at 24 h post-injection. By contrast, the radioactivity of other tissues either did not increase or decreased over 24 h. This indicates that 64Cu-NOTA-E1 has high tumor uptake and retention, with rapid clearance, and low background values in other tissues. Therefore, 64Cu-NOTA-E1 should be suitable as a novel PET imaging agent for hEphA2-expressing tumors.
Collapse
Affiliation(s)
- Ayoung Pyo
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Sung-Hwan You
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Hyeon Sik Kim
- Medical Photonics Research Center, Korea Photonics Technology Institute, Gwangju, Republic of Korea
| | - Jung Young Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Dong-Yeon Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea.
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Hwasun, Republic of Korea.
| |
Collapse
|
9
|
Czerwińska M, Bilewicz A, Kruszewski M, Wegierek-Ciuk A, Lankoff A. Targeted Radionuclide Therapy of Prostate Cancer-From Basic Research to Clinical Perspectives. Molecules 2020; 25:E1743. [PMID: 32290196 PMCID: PMC7181060 DOI: 10.3390/molecules25071743] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed malignancy in men and the second leading cause of cancer-related deaths in Western civilization. Although localized prostate cancer can be treated effectively in different ways, almost all patients progress to the incurable metastatic castration-resistant prostate cancer. Due to the significant mortality and morbidity rate associated with the progression of this disease, there is an urgent need for new and targeted treatments. In this review, we summarize the recent advances in research on identification of prostate tissue-specific antigens for targeted therapy, generation of highly specific and selective molecules targeting these antigens, availability of therapeutic radionuclides for widespread medical applications, and recent achievements in the development of new-generation small-molecule inhibitors and antibody-based strategies for targeted prostate cancer therapy with alpha-, beta-, and Auger electron-emitting radionuclides.
Collapse
Affiliation(s)
- Malwina Czerwińska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
| | - Aleksander Bilewicz
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland;
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| | - Aneta Wegierek-Ciuk
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 24-406 Kielce, Poland;
| | - Anna Lankoff
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 24-406 Kielce, Poland;
| |
Collapse
|
10
|
Abstract
Bioconjugation of biologically useful proteins is in great demand (e.g., conjugation to biotins, metal chelators, and drug carriers to target specific tissues for both in vitro and in vivo use). These conjugates provide widespread opportunities for various biological and biomedical applications. Evolving state-of-the-art protein conjugation strategies have led to the development of many affinity ligands, including for cancer imaging and diagnosis. However, to achieve the desirable protein conjugates, there are many challenges that remain to be addressed in order to obtain a reproducible procedure for all proteins and ligands. These include a control over the protein modification and the efficiency of the conjugation while retaining the original biological protein affinity postmodification. Here we present detailed conjugation methods for the human fibronectin tenth type III domain (FN3) protein scaffold for use in preclinical PET imaging. More specifically, this chapter provides detailed methods to produce a FN3 and a FN3-chelator-conjugate, its labeling with the radionuclide 64-Cu, and its use for noninvasive PET imaging in mice.
Collapse
Affiliation(s)
- Arutselvan Natarajan
- Department of Radiology, James H. Clark Center, Stanford University, Stanford, CA, USA.
| | - Lotfi Abou-Elkacem
- Department of Radiology, James H. Clark Center, Stanford University, Stanford, CA, USA
| |
Collapse
|
11
|
Rigoglio NN, Rabelo ACS, Borghesi J, de Sá Schiavo Matias G, Fratini P, Prazeres PHDM, Pimentel CMMM, Birbrair A, Miglino MA. The Tumor Microenvironment: Focus on Extracellular Matrix. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1245:1-38. [PMID: 32266651 DOI: 10.1007/978-3-030-40146-7_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The extracellular matrix (ECM) regulates the development and maintains tissue homeostasis. The ECM is composed of a complex network of molecules presenting distinct biochemical properties to regulate cell growth, survival, motility, and differentiation. Among their components, proteoglycans (PGs) are considered one of the main components of ECM. Its composition, biomechanics, and anisotropy are exquisitely tuned to reflect the physiological state of the tissue. The loss of ECM's homeostasis is seen as one of the hallmarks of cancer and, typically, defines transitional events in tumor progression and metastasis. In this chapter, we discuss the types of proteoglycans and their roles in cancer. It has been observed that the amount of some ECM components is increased, while others are decreased, depending on the type of tumor. However, both conditions corroborate with tumor progression and malignancy. Therefore, ECM components have an increasingly important role in carcinogenesis and this leads us to believe that their understanding may be a key in the discovery of new anti-tumor therapies. In this book, the main ECM components will be discussed in more detail in each chapter.
Collapse
Affiliation(s)
- Nathia Nathaly Rigoglio
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Carolina Silveira Rabelo
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Jessica Borghesi
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Gustavo de Sá Schiavo Matias
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Paula Fratini
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | - Alexander Birbrair
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
12
|
Hilliard TS. The Impact of Mesothelin in the Ovarian Cancer Tumor Microenvironment. Cancers (Basel) 2018; 10:E277. [PMID: 30134520 PMCID: PMC6162689 DOI: 10.3390/cancers10090277] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 01/14/2023] Open
Abstract
Ovarian cancer is the deadliest gynecological disease among U.S. women. Poor 5-year survival rates (<30%) are due to presentation of most women at diagnosis with advanced stage disease with widely disseminated intraperitoneal metastasis. However, when diagnosed before metastatic propagation the overall 5-year survival rate is >90%. Metastasizing tumor cells grow rapidly and aggressively attach to the mesothelium of all organs within the peritoneal cavity, including the parietal peritoneum and the omentum, producing secondary lesions. In this review, the involvement of mesothelin (MSLN) in the tumor microenvironment is discussed. MSLN, a 40kDa glycoprotein that is overexpressed in many cancers including ovarian and mesotheliomas is suggested to play a role in cell survival, proliferation, tumor progression, and adherence. However, the biological function of MSLN is not fully understood as MSLN knockout mice do not present with an abnormal phenotype. Conversely, MSLN has been shown to bind to the ovarian cancer antigen, CA-125, and thought to play a role in the peritoneal diffusion of ovarian tumor cells. Although the cancer-specific expression of MSLN makes it a potential therapeutic target, more studies are needed to validate the role of MSLN in tumor metastasis.
Collapse
Affiliation(s)
- Tyvette S Hilliard
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA.
| |
Collapse
|
13
|
Gorman K, McGinnis J, Kay B. Generating FN3-Based Affinity Reagents Through Phage Display. ACTA ACUST UNITED AC 2018; 10:e39. [PMID: 29927113 DOI: 10.1002/cpch.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Antibodies are useful tools for detecting individual proteins in complex samples and for learning about their location, amount, binding partners, and function in cells. Unfortunately, generating antibodies is time consuming and laborious, and their affinity and/or specificity is often limited. This protocol offers a fast and inexpensive alternative to generate antibody surrogates through phage display of a library of fibronectin type III (FN3) monobody variants and affinity selection for binders. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Kevin Gorman
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Jennifer McGinnis
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Brian Kay
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
14
|
Sirois AR, Deny DA, Baierl SR, George KS, Moore SJ. Fn3 proteins engineered to recognize tumor biomarker mesothelin internalize upon binding. PLoS One 2018; 13:e0197029. [PMID: 29738555 PMCID: PMC5940182 DOI: 10.1371/journal.pone.0197029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/20/2018] [Indexed: 11/19/2022] Open
Abstract
Mesothelin is a cell surface protein that is overexpressed in numerous cancers, including breast, ovarian, lung, liver, and pancreatic tumors. Aberrant expression of mesothelin has been shown to promote tumor progression and metastasis through interaction with established tumor biomarker CA125. Therefore, molecules that specifically bind to mesothelin have potential therapeutic and diagnostic applications. However, no mesothelin-targeting molecules are currently approved for routine clinical use. While antibodies that target mesothelin are in development, some clinical applications may require a targeting molecule with an alternative protein fold. For example, non-antibody proteins are more suitable for molecular imaging and may facilitate diverse chemical conjugation strategies to create drug delivery complexes. In this work, we engineered variants of the fibronectin type III domain (Fn3) non-antibody protein scaffold to bind to mesothelin with high affinity, using directed evolution and yeast surface display. Lead engineered Fn3 variants were solubly produced and purified from bacterial culture at high yield. Upon specific binding to mesothelin on human cancer cell lines, the engineered Fn3 proteins internalized and co-localized to early endosomes. To our knowledge, this is the first report of non-antibody proteins engineered to bind mesothelin. The results validate that non-antibody proteins can be engineered to bind to tumor biomarker mesothelin, and encourage the continued development of engineered variants for applications such as targeted diagnostics and therapeutics.
Collapse
Affiliation(s)
- Allison R. Sirois
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Amherst, Massachusetts, United States of America
- Picker Engineering Program, Smith College, Northampton, Massachusetts, United States of America
| | - Daniela A. Deny
- Department of Biochemistry, Smith College, Northampton, Massachusetts, United States of America
| | - Samantha R. Baierl
- Picker Engineering Program, Smith College, Northampton, Massachusetts, United States of America
| | - Katia S. George
- Department of Biochemistry, Smith College, Northampton, Massachusetts, United States of America
| | - Sarah J. Moore
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Amherst, Massachusetts, United States of America
- Picker Engineering Program, Smith College, Northampton, Massachusetts, United States of America
- Department of Biological Sciences, Smith College, Northampton, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
15
|
Könning D, Kolmar H. Beyond antibody engineering: directed evolution of alternative binding scaffolds and enzymes using yeast surface display. Microb Cell Fact 2018; 17:32. [PMID: 29482656 PMCID: PMC6389260 DOI: 10.1186/s12934-018-0881-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/22/2018] [Indexed: 01/08/2023] Open
Abstract
Pioneered exactly 20 years ago, yeast surface display (YSD) continues to take a major role in protein engineering among the high-throughput display methodologies that have been developed to date. The classical yeast display technology relies on tethering an engineered protein to the cell wall by genetic fusion to one subunit of a dimeric yeast-mating agglutination receptor complex. This method enables an efficient genotype-phenotype linkage while exploiting the benefits of a eukaryotic expression machinery. Over the past two decades, a plethora of protein engineering efforts encompassing conventional antibody Fab and scFv fragments have been reported. In this review, we will focus on the versatility of YSD beyond conventional antibody engineering and, instead, place the focus on alternative scaffold proteins and enzymes which have successfully been tailored for purpose with regard to improving binding, activity or specificity.
Collapse
Affiliation(s)
- Doreen Könning
- Antibody-Drug Conjugates and Targeted NBE Therapeutics, Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, 64287 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, 64287 Darmstadt, Germany
| |
Collapse
|
16
|
Koriem KMM. Protective effect of natural products and hormones in colon cancer using metabolome: A physiological overview. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2017.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
17
|
Bedford R, Tiede C, Hughes R, Curd A, McPherson MJ, Peckham M, Tomlinson DC. Alternative reagents to antibodies in imaging applications. Biophys Rev 2017; 9:299-308. [PMID: 28752365 PMCID: PMC5578921 DOI: 10.1007/s12551-017-0278-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/06/2017] [Indexed: 12/21/2022] Open
Abstract
Antibodies have been indispensable tools in molecular biology, biochemistry and medical research. However, a number of issues surrounding validation, specificity and batch variation of commercially available antibodies have prompted research groups to develop novel non-antibody binding reagents. The ability to select highly specific monoclonal non-antibody binding proteins without the need for animals, the ease of production and the ability to site-directly label has enabled a wide variety of applications to be tested, including imaging. In this review, we discuss the success of a number of non-antibody reagents in imaging applications, including the recently reported Affimer.
Collapse
Affiliation(s)
- R Bedford
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - C Tiede
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - R Hughes
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - A Curd
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - M J McPherson
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
| | - Michelle Peckham
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK.
| | - Darren C Tomlinson
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
18
|
Kim MA, Yoon HS, Park SH, Kim DY, Pyo A, Kim HS, Min JJ, Hong Y. Engineering of monobody conjugates for human EphA2-specific optical imaging. PLoS One 2017; 12:e0180786. [PMID: 28686661 PMCID: PMC5501600 DOI: 10.1371/journal.pone.0180786] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 06/21/2017] [Indexed: 02/06/2023] Open
Abstract
In a previous study, we developed an E1 monobody specific for the tumor biomarker hEphA2 [PLoS ONE (2015) 10(7): e0132976]. E1 showed potential as a molecular probe for in vitro and in vivo targeting of cancers overexpressing hEphA2. In the present study, we constructed expression vectors for E1 conjugated to optical reporters such as Renilla luciferase variant 8 (Rluc8) or enhanced green fluorescent protein (EGFP) and purified such recombinant proteins by affinity chromatography in E. coli. E1-Rluc8 and E1-EGFP specifically bound to hEphA2 in human prostate cancer PC3 cells but not in human cervical cancer HeLa cells, which express hEphA2 at high and low levels, respectively. These recombinant proteins maintained >40% activity in mouse serum at 24 h. In vivo optical imaging for 24 h did not detect E1-EGFP signals, whereas E1-Rluc8 showed tumor-specific luminescence signals in PC3 but not in HeLa xenograft mice. E1-Rluc8 signals were detected at 4 h, peaked at 12 h, and were undetectable at 24 h. These results suggest the potential of E1-Rluc8 as an EphA2-specific optical imaging agent.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/biosynthesis
- Antibodies, Neoplasm/chemistry
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Female
- Genes, Reporter
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- HeLa Cells
- Heterografts
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/metabolism
- Luciferases/genetics
- Luciferases/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Nude
- Optical Imaging
- Organ Specificity
- Protein Engineering
- Receptor, EphA2/analysis
- Receptor, EphA2/genetics
- Receptor, EphA2/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Min-A Kim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Molecular Medicine (BK21Plus), Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hee Seung Yoon
- Department of Molecular Medicine (BK21Plus), Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Seung-Hwan Park
- Biological Resource Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Jeongeup, Republic of Korea
| | - Dong-Yeon Kim
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ayoung Pyo
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyeon Sik Kim
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- * E-mail:
| |
Collapse
|
19
|
Yu X, Yang YP, Dikici E, Deo SK, Daunert S. Beyond Antibodies as Binding Partners: The Role of Antibody Mimetics in Bioanalysis. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2017; 10:293-320. [PMID: 28375702 PMCID: PMC5895458 DOI: 10.1146/annurev-anchem-061516-045205] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The emergence of novel binding proteins or antibody mimetics capable of binding to ligand analytes in a manner analogous to that of the antigen-antibody interaction has spurred increased interest in the biotechnology and bioanalytical communities. The goal is to produce antibody mimetics designed to outperform antibodies with regard to binding affinities, cellular and tumor penetration, large-scale production, and temperature and pH stability. The generation of antibody mimetics with tailored characteristics involves the identification of a naturally occurring protein scaffold as a template that binds to a desired ligand. This scaffold is then engineered to create a superior binder by first creating a library that is then subjected to a series of selection steps. Antibody mimetics have been successfully used in the development of binding assays for the detection of analytes in biological samples, as well as in separation methods, cancer therapy, targeted drug delivery, and in vivo imaging. This review describes recent advances in the field of antibody mimetics and their applications in bioanalytical chemistry, specifically in diagnostics and other analytical methods.
Collapse
Affiliation(s)
- Xiaowen Yu
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| | - Yu-Ping Yang
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| | - Sapna K Deo
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136;
| |
Collapse
|
20
|
Leenheer D, ten Dijke P, Hipolito CJ. A current perspective on applications of macrocyclic-peptide-based high-affinity ligands. Biopolymers 2016; 106:889-900. [PMID: 27352774 PMCID: PMC5132055 DOI: 10.1002/bip.22900] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/24/2016] [Accepted: 05/31/2016] [Indexed: 01/05/2023]
Abstract
Monoclonal antibodies can bind with high affinity and high selectivity to their targets. As a tool in therapeutics or diagnostics, however, their large size (∼150 kDa) reduces penetration into tissue and prevents passive cellular uptake. To overcome these and other problems, minimized protein scaffolds have been chosen or engineered, with care taken to not compromise binding affinity or specificity. An alternate approach is to begin with a minimal non-antibody scaffold and select functional ligands from a de novo library. We will discuss the structure, production, applications, strengths, and weaknesses of several classes of antibody-derived ligands, that is, antibodies, intrabodies, and nanobodies, and nonantibody-derived ligands, that is, monobodies, affibodies, and macrocyclic peptides. In particular, this review is focussed on macrocyclic peptides produced by the Random non-standard Peptides Integrated Discovery (RaPID) system that are small in size (typically ∼2 kDa), but are able to perform tasks typically handled by larger proteinaceous ligands.
Collapse
Affiliation(s)
- Daniël Leenheer
- Ph.D. Program in Human Biology, School of Integrative and Global MajorsUniversity of TsukubaTsukubaIbarakiJapan
| | - Peter ten Dijke
- Leiden University Medical Center, Department of Molecular Cell BiologyLeidenSouth HollandThe Netherlands
- Cancer Signaling, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of TsukubaTsukubaIbarakiJapan
| | - Christopher John Hipolito
- Cancer Signaling, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of TsukubaTsukubaIbarakiJapan
| |
Collapse
|
21
|
Abou-Elkacem L, Wilson KE, Johnson SM, Chowdhury SM, Bachawal S, Hackel BJ, Tian L, Willmann JK. Ultrasound Molecular Imaging of the Breast Cancer Neovasculature using Engineered Fibronectin Scaffold Ligands: A Novel Class of Targeted Contrast Ultrasound Agent. Theranostics 2016; 6:1740-52. [PMID: 27570547 PMCID: PMC4997233 DOI: 10.7150/thno.15169] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/19/2016] [Indexed: 12/21/2022] Open
Abstract
Molecularly-targeted microbubbles (MBs) are increasingly being recognized as promising contrast agents for oncological molecular imaging with ultrasound. With the detection and validation of new molecular imaging targets, novel binding ligands are needed that bind to molecular imaging targets with high affinity and specificity. In this study we assessed a novel class of potentially clinically translatable MBs using an engineered 10th type III domain of human-fibronectin (MB-FN3VEGFR2) scaffold-ligand to image VEGFR2 on the neovasculature of cancer. The in vitro binding of MB-FN3VEGFR2 to a soluble VEGFR2 was assessed by flow-cytometry (FACS) and binding to VEGFR2-expressing cells was assessed by flow-chamber cell attachment studies under flow shear stress conditions. In vivo binding of MB-FN3VEGFR2 was tested in a transgenic mouse model (FVB/N Tg(MMTV/PyMT634Mul) of breast cancer and control litter mates with normal mammary glands. In vitro FACS and flow-chamber cell attachment studies showed significantly (P<0.01) higher binding to VEGFR2 using MB-FN3VEGFR2 than control agents. In vivo ultrasound molecular imaging (USMI) studies using MB-FN3VEGFR2 demonstrated specific binding to VEGFR2 and was significantly higher (P<0.01) in breast cancer compared to normal breast tissue. Ex vivo immunofluorescence-analysis showed significantly (P<0.01) increased VEGFR2-expression in breast cancer compared to normal mammary tissue. Our results suggest that MBs coupled to FN3-scaffolds can be designed and used for USMI of breast cancer neoangiogenesis. Due to their small size, stability, solubility, the lack of glycosylation and disulfide bonds, FN3-scaffolds can be recombinantly produced with the advantage of generating small, high affinity ligands in a cost efficient way for USMI.
Collapse
|
22
|
Huang R, Fang P, Hao Z, Kay BK. Directed Evolution of a Highly Specific FN3 Monobody to the SH3 Domain of Human Lyn Tyrosine Kinase. PLoS One 2016; 11:e0145872. [PMID: 26731115 PMCID: PMC4701441 DOI: 10.1371/journal.pone.0145872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 12/09/2015] [Indexed: 11/26/2022] Open
Abstract
Affinity reagents of high affinity and specificity are very useful for studying the subcellular locations and quantities of individual proteins. To generate high-quality affinity reagents for human Lyn tyrosine kinase, a phage display library of fibronectin type III (FN3) monobodies was affinity selected with a recombinant form of the Lyn SH3 domain. While a highly specific monobody, TA8, was initially isolated, we chose to improve its affinity through directed evolution. A secondary library of 1.2 × 109 variants was constructed and screened by affinity selection, yielding three variants, two of which have affinities of ~ 40 nM, a 130-fold increase over the original TA8 monobody. One of the variants, 2H7, displayed high specificity to the Lyn SH3 domain, as shown by ELISA and probing arrays of 150 SH3 domains. Furthermore, the 2H7 monobody was able to pull down endogenous Lyn from a lysate of Burkitt's lymphoma cells, thereby demonstrating its utility as an affinity reagent for detecting Lyn in a complex biological mixture.
Collapse
Affiliation(s)
- Renhua Huang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail: (RH); (BK)
| | - Pete Fang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Zengping Hao
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Brian K. Kay
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail: (RH); (BK)
| |
Collapse
|
23
|
Yun M, Park SH, Hong Y, Min JJ. C5-O-02Targeted Imaging and Theranostics with Peptides and Novel Protein Scaffolds, Repebody and Monobody. Microscopy (Oxf) 2015. [DOI: 10.1093/jmicro/dfv198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|