1
|
Bozorgchami N, Ahmadzadeh M, Hatamabadi D, Yazdani A, Shahhosseini S, Mohit E. Preparation, Characterization, and Radiolabeling of Anti-HER2 scFv With Technetium Tricarbonyl and Stability Studies. J Labelled Comp Radiopharm 2024; 67:168-179. [PMID: 38485465 DOI: 10.1002/jlcr.4090] [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: 12/23/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 05/14/2024]
Abstract
Breast cancer is the most common diagnosed cancer, and the second cause of cancer death among women, worldwide. HER2 overexpression occurred in approximately 15% to 20% of breast cancers. Invasive biopsy method has been used for detection of HER2 overexpression. HER2-targeted imaging via an appropriate radionuclide is a promising method for sensitive and accurate identification of HER2+ primary and metastatic lesions. 99mTc-anti-HER2 scFv can specifically target malignancies and be used for diagnosis of the cancer type and metastasis as well as treatment of breast cancer. We radiolabeled anti-HER2 scFv that was expressed in Escherichia coli and purified through Ni-NTA resin under native condition with 99mTc-tricarbonyl formed from boranocarbonate. HER2-based ELISA, BCA, TLC, and HPLC were used in this study. In the current study, anti-HER2 scFv was lyophilized before radiolabeling. It was found that freeze-drying did not change the binding activity of anti-HER2 scFv to HER2. Results demonstrated direct anti-HER2 scFv radiolabeling by 99mTc-tricarbonyl to hexahistidine sequence (His-tag) without any changes in biological activity and radiochemical purity of around 98%. Stability analysis revealed that 99mTc-anti-HER2 scFv is stable for at least 24 h in PBS buffer, normal saline, human plasma proteins, and histidine solution.
Collapse
Affiliation(s)
- Negar Bozorgchami
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Ahmadzadeh
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Food and Drug Laboratory Research Center, Food and Drug Administration, The Ministry of Health and Medical Education, Tehran, Iran
| | - Dara Hatamabadi
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Yazdani
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soraya Shahhosseini
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Pijeira MSO, Nunes PSG, Chaviano SL, Diaz AMA, DaSilva JN, Ricci-Junior E, Alencar LMR, Chen X, Santos-Oliveira R. Medicinal (Radio) Chemistry: Building Radiopharmaceuticals for the Future. Curr Med Chem 2024; 31:5481-5534. [PMID: 37594105 DOI: 10.2174/0929867331666230818092634] [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: 03/10/2023] [Revised: 05/30/2023] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Radiopharmaceuticals are increasingly playing a leading role in diagnosing, monitoring, and treating disease. In comparison with conventional pharmaceuticals, the development of radiopharmaceuticals does follow the principles of medicinal chemistry in the context of imaging-altered physiological processes. The design of a novel radiopharmaceutical has several steps similar to conventional drug discovery and some particularity. In the present work, we revisited the insights of medicinal chemistry in the current radiopharmaceutical development giving examples in oncology, neurology, and cardiology. In this regard, we overviewed the literature on radiopharmaceutical development to study overexpressed targets such as prostate-specific membrane antigen and fibroblast activation protein in cancer; β-amyloid plaques and tau protein in brain disorders; and angiotensin II type 1 receptor in cardiac disease. The work addresses concepts in the field of radiopharmacy with a special focus on the potential use of radiopharmaceuticals for nuclear imaging and theranostics.
Collapse
Affiliation(s)
- Martha Sahylí Ortega Pijeira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
| | - Paulo Sérgio Gonçalves Nunes
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas SP13083-970, Brazil
| | - Samila Leon Chaviano
- Laboratoire de Biomatériaux pour l'Imagerie Médicale, Axe Médicine Régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Aida M Abreu Diaz
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean N DaSilva
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Eduardo Ricci-Junior
- Laboratório de Desenvolvimento Galênico, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Laboratory of Biophysics and Nanosystems, Federal University of Maranhão, Av. dos Portugueses, 1966, Vila Bacanga, São Luís MA65080-805, Brazil
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore 117597, Singapore
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro State University, Rio de Janeiro 23070200, Brazil
| |
Collapse
|
3
|
Bidkar AP, Wang S, Bobba KN, Chan E, Bidlingmaier S, Egusa EA, Peter R, Ali U, Meher N, Wadhwa A, Dhrona S, Dasari C, Beckford-Vera D, Su Y, Tang R, Zhang L, He J, Wilson DM, Aggarwal R, VanBrocklin HF, Seo Y, Chou J, Liu B, Flavell RR. Treatment of Prostate Cancer with CD46-targeted 225Ac Alpha Particle Radioimmunotherapy. Clin Cancer Res 2023; 29:1916-1928. [PMID: 36917693 PMCID: PMC10183825 DOI: 10.1158/1078-0432.ccr-22-3291] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/19/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023]
Abstract
PURPOSE Radiopharmaceutical therapy is changing the standard of care in prostate cancer and other malignancies. We previously reported high CD46 expression in prostate cancer and developed an antibody-drug conjugate and immunoPET agent based on the YS5 antibody, which targets a tumor-selective CD46 epitope. Here, we present the preparation, preclinical efficacy, and toxicity evaluation of [225Ac]DOTA-YS5, a radioimmunotherapy agent based on the YS5 antibody. EXPERIMENTAL DESIGN [225Ac]DOTA-YS5 was developed, and its therapeutic efficiency was tested on cell-derived (22Rv1, DU145), and patient-derived (LTL-545, LTL484) prostate cancer xenograft models. Biodistribution studies were carried out on 22Rv1 tumor xenograft models to confirm the targeting efficacy. Toxicity analysis of the [225Ac]DOTA-YS5 was carried out on nu/nu mice to study short-term (acute) and long-term (chronic) toxicity. RESULTS Biodistribution study shows that [225Ac]DOTA-YS5 agent delivers high levels of radiation to the tumor tissue (11.64% ± 1.37%ID/g, 28.58% ± 10.88%ID/g, 29.35% ± 7.76%ID/g, and 31.78% ± 5.89%ID/g at 24, 96, 168, and 408 hours, respectively), compared with the healthy organs. [225Ac]DOTA-YS5 suppressed tumor size and prolonged survival in cell line-derived and patient-derived xenograft models. Toxicity analysis revealed that the 0.5 μCi activity levels showed toxicity to the kidneys, likely due to redistribution of daughter isotope 213Bi. CONCLUSIONS [225Ac]DOTA-YS5 suppressed the growth of cell-derived and patient-derived xenografts, including prostate-specific membrane antigen-positive and prostate-specific membrane antigen-deficient models. Overall, this preclinical study confirms that [225Ac]DOTA-YS5 is a highly effective treatment and suggests feasibility for clinical translation of CD46-targeted radioligand therapy in prostate cancer.
Collapse
Affiliation(s)
- Anil P. Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Emily Chan
- Department of Pathology, University of California, San Francisco, California
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Emily A. Egusa
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Robin Peter
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- Department of Nuclear Engineering, University of California, Berkeley, California
| | - Umama Ali
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Chandrashekhar Dasari
- Department of Surgery, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California
| | - Denis Beckford-Vera
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Ryan Tang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Li Zhang
- Department of Medicine and the Department of Epidemiology and Biostatistics, University of California, Berkeley, California
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Rahul Aggarwal
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Jonathan Chou
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California
| |
Collapse
|
4
|
Li J, Huang T, Hua J, Wang Q, Su Y, Chen P, Bidlingmaier S, Li A, Xie Z, Bidkar AP, Shen S, Shi W, Seo Y, Flavell RR, Gioeli D, Dreicer R, Li H, Liu B, He J. CD46 targeted 212Pb alpha particle radioimmunotherapy for prostate cancer treatment. J Exp Clin Cancer Res 2023; 42:61. [PMID: 36906664 PMCID: PMC10007843 DOI: 10.1186/s13046-023-02636-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/01/2023] [Indexed: 03/13/2023] Open
Abstract
We recently identified CD46 as a novel prostate cancer cell surface antigen that shows lineage independent expression in both adenocarcinoma and small cell neuroendocrine subtypes of metastatic castration resistant prostate cancer (mCRPC), discovered an internalizing human monoclonal antibody YS5 that binds to a tumor selective CD46 epitope, and developed a microtubule inhibitor-based antibody drug conjugate that is in a multi-center phase I trial for mCRPC (NCT03575819). Here we report the development of a novel CD46-targeted alpha therapy based on YS5. We conjugated 212Pb, an in vivo generator of alpha-emitting 212Bi and 212Po, to YS5 through the chelator TCMC to create the radioimmunoconjugate, 212Pb-TCMC-YS5. We characterized 212Pb-TCMC-YS5 in vitro and established a safe dose in vivo. We next studied therapeutic efficacy of a single dose of 212Pb-TCMC-YS5 using three prostate cancer small animal models: a subcutaneous mCRPC cell line-derived xenograft (CDX) model (subcu-CDX), an orthotopically grafted mCRPC CDX model (ortho-CDX), and a prostate cancer patient-derived xenograft model (PDX). In all three models, a single dose of 0.74 MBq (20 µCi) 212Pb-TCMC-YS5 was well tolerated and caused potent and sustained inhibition of established tumors, with significant increases of survival in treated animals. A lower dose (0.37 MBq or 10 µCi 212Pb-TCMC-YS5) was also studied on the PDX model, which also showed a significant effect on tumor growth inhibition and prolongation of animal survival. These results demonstrate that 212Pb-TCMC-YS5 has an excellent therapeutic window in preclinical models including PDXs, opening a direct path for clinical translation of this novel CD46-targeted alpha radioimmunotherapy for mCRPC treatment.
Collapse
Affiliation(s)
- Jun Li
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Guangdong, 518036, China
| | - Tao Huang
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
| | - Jun Hua
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Chongqing Cancer Hospital, Chongqing University, Chongqing, China
| | - Qiong Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Ping Chen
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Guangdong, 518036, China
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Allan Li
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Zhongqiu Xie
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
| | - Sui Shen
- Department of Radiation Oncology, University of Alabama, Birmingham, AL, 35233, USA
| | - Weibin Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA
| | - Daniel Gioeli
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22903, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
| | - Robert Dreicer
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Medicine, University of Virginia, Charlottesville, VA, 22903, USA
| | - Hui Li
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA.
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA.
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA.
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
5
|
Chou J, Egusa EA, Wang S, Badura ML, Lee F, Bidkar AP, Zhu J, Shenoy T, Trepka K, Robinson TM, Steri V, Huang J, Wang Y, Small EJ, Chan E, Stohr BA, Ashworth A, Delafontaine B, Rottey S, Cooke KS, Hashemi Sadraei N, Yu B, Salvati M, Bailis JM, Feng FY, Flavell RR, Aggarwal R. Immunotherapeutic Targeting and PET Imaging of DLL3 in Small-Cell Neuroendocrine Prostate Cancer. Cancer Res 2023; 83:301-315. [PMID: 36351060 DOI: 10.1158/0008-5472.can-22-1433] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/06/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022]
Abstract
Effective treatments for de novo and treatment-emergent small-cell/neuroendocrine (t-SCNC) prostate cancer represent an unmet need for this disease. Using metastatic biopsies from patients with advanced cancer, we demonstrate that delta-like ligand 3 (DLL3) is expressed in de novo and t-SCNC and is associated with reduced survival. We develop a PET agent, [89Zr]-DFO-DLL3-scFv, that detects DLL3 levels in mouse SCNC models. In multiple patient-derived xenograft models, AMG 757 (tarlatamab), a half-life-extended bispecific T-cell engager (BiTE) immunotherapy that redirects CD3-positive T cells to kill DLL3-expressing cells, exhibited potent and durable antitumor activity. Late relapsing tumors after AMG 757 treatment exhibited lower DLL3 levels, suggesting antigen loss as a resistance mechanism, particularly in tumors with heterogeneous DLL3 expression. These findings have been translated into an ongoing clinical trial of AMG 757 in de novo and t-SCNC, with a confirmed objective partial response in a patient with histologically confirmed SCNC. Overall, these results identify DLL3 as a therapeutic target in SCNC and demonstrate that DLL3-targeted BiTE immunotherapy has significant antitumor activity in this aggressive prostate cancer subtype. SIGNIFICANCE The preclinical and clinical evaluation of DLL3-directed immunotherapy, AMG 757, and development of a PET radiotracer for noninvasive DLL3 detection demonstrate the potential of targeting DLL3 in SCNC prostate cancer.
Collapse
Affiliation(s)
- Jonathan Chou
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Emily A Egusa
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Radiation Oncology and Urology, University of California, San Francisco, California
| | - Sinan Wang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Michelle L Badura
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Radiation Oncology and Urology, University of California, San Francisco, California.,Department of Biology, Santa Clara University, Santa Clara, California
| | - Fei Lee
- Oncology Research, Amgen Research, Amgen, South San Francisco, California
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Jun Zhu
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Radiation Oncology and Urology, University of California, San Francisco, California
| | - Tanushree Shenoy
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Kai Trepka
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Radiation Oncology and Urology, University of California, San Francisco, California.,Medical Scientist Training Program, University of California, San Francisco, California
| | - Troy M Robinson
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Radiation Oncology and Urology, University of California, San Francisco, California
| | - Veronica Steri
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Jiaoti Huang
- Department of Pathology, Duke University, Durham, North Carolina
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer, Vancouver, British Columbia.,Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eric J Small
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Emily Chan
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Pathology, University of California, San Francisco, California
| | - Bradley A Stohr
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Pathology, University of California, San Francisco, California
| | - Alan Ashworth
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | | | | | - Keegan S Cooke
- Oncology Research, Amgen Research, Amgen, Thousand Oaks, California
| | | | - Brian Yu
- Global Development, Amgen, Thousand Oaks, California
| | - Mark Salvati
- Global Development, Amgen, Thousand Oaks, California
| | - Julie M Bailis
- Oncology Research, Amgen Research, Amgen, South San Francisco, California
| | - Felix Y Feng
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Radiation Oncology and Urology, University of California, San Francisco, California
| | - Robert R Flavell
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Rahul Aggarwal
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| |
Collapse
|
6
|
Wang S, Li J, Hua J, Su Y, Beckford-Vera DR, Zhao W, Jayaraman M, Huynh TL, Zhao N, Wang YH, Huang Y, Qin F, Shen S, Gioeli D, Dreicer R, Sriram R, Egusa EA, Chou J, Feng FY, Aggarwal R, Evans MJ, Seo Y, Liu B, Flavell RR, He J. Molecular Imaging of Prostate Cancer Targeting CD46 Using ImmunoPET. Clin Cancer Res 2020; 27:1305-1315. [PMID: 33293372 DOI: 10.1158/1078-0432.ccr-20-3310] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/19/2020] [Accepted: 12/03/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We recently identified CD46 as a novel therapeutic target in prostate cancer. In this study, we developed a CD46-targeted PET radiopharmaceutical, [89Zr]DFO-YS5, and evaluated its performance for immunoPET imaging in murine prostate cancer models. EXPERIMENTAL DESIGN [89Zr]DFO-YS5 was prepared and its in vitro binding affinity for CD46 was measured. ImmunoPET imaging was conducted in male athymic nu/nu mice bearing DU145 [AR-, CD46+, prostate-specific membrane antigen-negative (PSMA-)] or 22Rv1 (AR+, CD46+, PSMA+) tumors, and in NOD/SCID gamma mice bearing patient-derived adenocarcinoma xenograft, LTL-331, and neuroendocrine prostate cancers, LTL-331R and LTL-545. RESULTS [89Zr]DFO-YS5 binds specifically to the CD46-positive human prostate cancer DU145 and 22Rv1 xenografts. In biodistribution studies, the tumor uptake of [89Zr]DFO-YS5 was 13.3 ± 3.9 and 11.2 ± 2.5 %ID/g, respectively, in DU145 and 22Rv1 xenografts, 4 days postinjection. Notably, [89Zr]DFO-YS5 demonstrated specific uptake in the PSMA- and AR-negative DU145 model. [89Zr]DFO-YS5 also showed uptake in the patient-derived LTL-331 and -331R models, with particularly high uptake in the LTL-545 neuroendocrine prostate cancer tumors (18.8 ± 5.3, 12.5 ± 1.8, and 32 ± 5.3 %ID/g in LTL-331, LTL-331R, and LTL-545, respectively, at 4 days postinjection). CONCLUSIONS [89Zr]DFO-YS5 is an excellent PET imaging agent across a panel of prostate cancer models, including in both adenocarcinoma and neuroendocrine prostate cancer, both cell line- and patient-derived xenografts, and both PSMA-positive and -negative tumors. It demonstrates potential for clinical translation as an imaging agent, theranostic platform, and companion biomarker in prostate cancer.
Collapse
Affiliation(s)
- Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Jun Li
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia.,Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Jun Hua
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia.,Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Denis R Beckford-Vera
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Walter Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Mayuri Jayaraman
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Tony L Huynh
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Ning Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Yung-Hua Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Yangjie Huang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Fujun Qin
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Sui Shen
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel Gioeli
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia.,University of Virginia Cancer Center, Charlottesville, Virginia
| | - Robert Dreicer
- University of Virginia Cancer Center, Charlottesville, Virginia.,Departments of Medicine and Urology, University of Virginia, Charlottesville, Virginia
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Emily A Egusa
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Jonathan Chou
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Felix Y Feng
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Rahul Aggarwal
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Division of Hematology and Oncology, University of California, San Francisco, San Francisco, California
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, San Francisco, California. .,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California. .,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia. .,University of Virginia Cancer Center, Charlottesville, Virginia
| |
Collapse
|
7
|
He J, Feng J, Su Y, Seo Y, Liu B. Quantitative 99mTc Labeling Kit for HYNIC-Conjugated Single Chain Antibody Fragments Targeting Malignant Mesothelioma. Bioconjug Chem 2020; 31:1750-1755. [PMID: 32628464 DOI: 10.1021/acs.bioconjchem.0c00319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Single chain antibody fragment (scFv) is a promising agent for imaging and targeted therapy. The objective of the study is to evaluate a kit formulation for 99mTc labeling of scFv for tumor imaging. The scFv was engineered to contain a cysteine tag to accommodate the specific conjugation of HYNIC and subsequent 99mTc labeling. The labeling conditions were formulated to allow instantaneous one-pot quantitative labeling. The reproducibility of labeling was evaluated at various time points during kit storage at -20 °C. In vitro cell binding experiments and HPLC analysis were performed to assess binding affinity and radiolabel stability, respectively. In vivo tumor targeting study was performed in xenograft models with biodistribution studied at 1, 3, and 24 h post-injection. The optimized kit with 5 μg SnF2, pH 5.5, and 50 μg GH along with as low as 15 μg of HYNIC-cys-scFv provided high labeling yield (>95%), high specific activity (1.8 × 107 Ci/Mol), and robust reproducibility with shelf life up to 90 days when stored at -20 °C. The in vitro cell binding study showed the labeled scFv maintained the binding capability with an apparent KD of ∼27 nM. The animal study using tumor-bearing mice showed high tumor uptake at 16.9%ID/g 24 h post-injection along with rapid blood clearance (0.18%ID/g) and kidney excretion (44%ID/g), resulting in very high contrast (tumor/muscle >200:1). A kit formulation for 99mTc labeling of scFvs targeting mesothelioma was developed based on specific HYNIC conjugation and GH (Glucoheptonate) as a coligand, producing not only high specific activity, but also improved tumor uptake. This convenient one-pot labeling method has the potential for translation into clinical use and is applicable to other scFvs as well.
Collapse
Affiliation(s)
- Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia 22908, United States.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Jinjin Feng
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, California 94143, United States
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143, United States
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, California 94143, United States.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143, United States
| |
Collapse
|
8
|
Su Y, Liu Y, Behrens CR, Bidlingmaier S, Lee NK, Aggarwal R, Sherbenou DW, Burlingame AL, Hann BC, Simko JP, Premasekharan G, Paris PL, Shuman MA, Seo Y, Small EJ, Liu B. Targeting CD46 for both adenocarcinoma and neuroendocrine prostate cancer. JCI Insight 2018; 3:121497. [PMID: 30185663 DOI: 10.1172/jci.insight.121497] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022] Open
Abstract
Although initially responsive to androgen signaling inhibitors (ASIs), metastatic castration-resistant prostate cancer (mCRPC) inevitably develops and is incurable. In addition to adenocarcinoma (adeno), neuroendocrine prostate cancer (NEPC) emerges to confer ASI resistance. We have previously combined laser capture microdissection and phage antibody display library selection on human cancer specimens and identified novel internalizing antibodies binding to tumor cells residing in their tissue microenvironment. We identified the target antigen for one of these antibodies as CD46, a multifunctional protein that is best known for negatively regulating the innate immune system. CD46 is overexpressed in primary tumor tissue and CRPC (localized and metastatic; adeno and NEPC), but expressed at low levels on normal tissues except for placental trophoblasts and prostate epithelium. Abiraterone- and enzalutamide-treated mCRPC cells upregulate cell surface CD46 expression. Genomic analysis showed that the CD46 gene is gained in 45% abiraterone-resistant mCRPC patients. We conjugated a tubulin inhibitor to our macropinocytosing anti-CD46 antibody and showed that the resulting antibody-drug conjugate (ADC) potently and selectively kills both adeno and NEPC cell lines in vitro (sub-nM EC50) but not normal cells. CD46 ADC regressed and eliminated an mCRPC cell line xenograft in vivo in both subcutaneous and intrafemoral models. Exploratory toxicology studies of the CD46 ADC in non-human primates demonstrated an acceptable safety profile. Thus, CD46 is an excellent target for antibody-based therapy development, which has potential to be applicable to both adenocarcinoma and neuroendocrine types of mCRPC that are resistant to current treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Rahul Aggarwal
- Department of Medicine.,Helen Diller Family Comprehensive Cancer Center
| | | | | | | | - Jeffry P Simko
- Helen Diller Family Comprehensive Cancer Center.,Department of Pathology
| | | | - Pamela L Paris
- Helen Diller Family Comprehensive Cancer Center.,Department of Urology, and
| | | | - Youngho Seo
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | - Eric J Small
- Department of Medicine.,Helen Diller Family Comprehensive Cancer Center.,Department of Urology, and
| | - Bin Liu
- Department of Anesthesia.,Helen Diller Family Comprehensive Cancer Center
| |
Collapse
|
9
|
Nanomedicine for cancer diagnosis and therapy: advancement, success and structure-activity relationship. Ther Deliv 2018; 8:1003-1018. [PMID: 29061101 DOI: 10.4155/tde-2017-0062] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Multifunctional nanoparticles (NPs), composed of organic and inorganic materials, have been explored as promising drug-delivery vehicles for cancer diagnosis and therapy. The success of nanosystems has been attributed to its smaller size, biocompatibility, selective tumor accumulation and reduced toxicity. The relationship among numbers of molecules in payload, NP diameter and encapsulation efficacy have crucial role in clinical translation. Advancement of bioengineering, and systematic fine-tuning of functional components to NPs have diversified their optical and theranostic properties. In this review, we summarize wide varieties of NPs, such as ultrasmall polymer-lipid hybrid NPs, dendrimers, liposomes, quantum dots, carbon nanotubes, gold NPs and iron oxide NPs. We also discuss their tumor targetability, tissue penetration, pharmacokinetics, and therapeutic and diagnostic properties. [Formula: see text].
Collapse
|
10
|
Sau S, Tatiparti K, Alsaab HO, Kashaw SK, Iyer AK. A tumor multicomponent targeting chemoimmune drug delivery system for reprograming the tumor microenvironment and personalized cancer therapy. Drug Discov Today 2018; 23:1344-1356. [PMID: 29551455 DOI: 10.1016/j.drudis.2018.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 02/11/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
Nanoparticle library engineered with tunable size, shape, and geometry will provide a better idea of targeting multicomponent of tumor microenvironment consisting of epithelial cells, tumor hypoxia, tumor immune cells and angiogenic blood vessels.
Collapse
Affiliation(s)
- Samaresh Sau
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Katyayani Tatiparti
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Hashem O Alsaab
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Sushil K Kashaw
- Department of Pharmaceutical Sciences, Dr Harisingh Gour Central University, Sagar, MP 470003, India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Molecular Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
11
|
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: 5.1] [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
|
12
|
Ha KD, Bidlingmaier SM, Su Y, Lee NK, Liu B. Identification of Novel Macropinocytosing Human Antibodies by Phage Display and High-Content Analysis. Methods Enzymol 2017; 585:91-110. [PMID: 28109445 PMCID: PMC8671048 DOI: 10.1016/bs.mie.2016.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Internalizing antibodies have great potential for the development of targeted therapeutics. Antibodies that internalize via the macropinocytosis pathway are particularly promising since macropinocytosis is capable of mediating rapid, bulk uptake and is selectively upregulated in many cancers. We hereby describe a method for identifying antibodies that internalize via macropinocytosis by screening phage-displayed single-chain antibody selection outputs with an automated fluorescent microscopy-based high-content analysis platform. Furthermore, this method can be similarly applied to other endocytic pathways if other fluorescent, pathway-specific, soluble markers are available.
Collapse
Affiliation(s)
| | | | | | | | - Bin Liu
- Corresponding author Department of Anesthesia, University of California at San Francisco, 1001 Potrero Ave., Box 1305, San Francisco, CA 94110-1305,
| |
Collapse
|
13
|
Sherbenou DW, Aftab BT, Su Y, Behrens CR, Wiita A, Logan AC, Acosta-Alvear D, Hann BC, Walter P, Shuman MA, Wu X, Atkinson JP, Wolf JL, Martin TG, Liu B. Antibody-drug conjugate targeting CD46 eliminates multiple myeloma cells. J Clin Invest 2016; 126:4640-4653. [PMID: 27841764 DOI: 10.1172/jci85856] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 10/06/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple myeloma is incurable by standard approaches because of inevitable relapse and development of treatment resistance in all patients. In our prior work, we identified a panel of macropinocytosing human monoclonal antibodies against CD46, a negative regulator of the innate immune system, and constructed antibody-drug conjugates (ADCs). In this report, we show that an anti-CD46 ADC (CD46-ADC) potently inhibited proliferation in myeloma cell lines with little effect on normal cells. CD46-ADC also potently eliminated myeloma growth in orthometastatic xenograft models. In primary myeloma cells derived from bone marrow aspirates, CD46-ADC induced apoptosis and cell death, but did not affect the viability of nontumor mononuclear cells. It is of clinical interest that the CD46 gene resides on chromosome 1q, which undergoes genomic amplification in the majority of relapsed myeloma patients. We found that the cell surface expression level of CD46 was markedly higher in patient myeloma cells with 1q gain than in those with normal 1q copy number. Thus, genomic amplification of CD46 may serve as a surrogate for target amplification that could allow patient stratification for tailored CD46-targeted therapy. Overall, these findings indicate that CD46 is a promising target for antibody-based treatment of multiple myeloma, especially in patients with gain of chromosome 1q.
Collapse
|
14
|
Iizuka A, Kondou R, Nonomura C, Ashizawa T, Ohshima K, Kusuhara M, Isaka M, Ohde Y, Yamaguchi K, Akiyama Y. Unstable B7-H4 cell surface expression and T-cell redirection as a means of cancer therapy. Oncol Rep 2016; 36:2625-2632. [PMID: 27632942 DOI: 10.3892/or.2016.5084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/07/2016] [Indexed: 11/06/2022] Open
Abstract
Tumor immune regulation has been demonstrated in clinical studies using antibodies targeted to the B7/CD28 family. B7 homolog 4 (B7-H4) negatively regulates immune responses and is overexpressed in many types of human cancer, indicating that B7-H4 may be a potential target of cancer therapy. B7-H4 expression is affected by the microenvironment, and its presence has been reported in cancer tissues and immune cells. We found an upregulation of B7-H4 expression using comprehensive whole exome sequencing and gene expression profiling (project HOPE) launched by the Shizuoka Cancer Center based on tumor tissue samples from 1,058 cancer patients. We were successful in producing monoclonal antibodies for B7-H4 and demonstrated B7-H4 dimerization and rapid cell surface disappearance by antibody cross-linking in breast cancer cells, even under typical conditions. These observations may explain why antibody-dependent cellular cytotoxicity (ADCC) did not function in vivo on the B7-H4-expressing tumor cells. Unstable cell surface antigens are not suitable as targets for ADCC, and we therefore performed an indirect ADCC-redirecting T-cell cytotoxicity assay to study B7-H4 using polyclonal anti-mouse IgG antibody-mediated linking. Our results showed the possibility of targeting the B7-H4 molecule as a means of treating cancer.
Collapse
Affiliation(s)
- Akira Iizuka
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Ryota Kondou
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Chizu Nonomura
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Tadashi Ashizawa
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Keiichi Ohshima
- Division of Medical Genetics, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Masatoshi Kusuhara
- Division of Regional Resources, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Mitsuhiro Isaka
- Division of Thoracic Surgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Yasuhisa Ohde
- Division of Thoracic Surgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Ken Yamaguchi
- Office of the President, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Yasuto Akiyama
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| |
Collapse
|
15
|
Li C, Feng H, Xia X, Wang L, Gao B, Zhang Y, Lan X. (99m) Tc-labeled tetramer and pentamer of single-domain antibody for targeting epidermal growth factor receptor in xenografted tumors. J Labelled Comp Radiopharm 2016; 59:305-12. [PMID: 27123559 DOI: 10.1002/jlcr.3399] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 03/04/2016] [Accepted: 03/17/2016] [Indexed: 01/01/2023]
Abstract
The single-domain antibody EG2 can be fused with right-handed coiled-coil (RHCC) and human cartilage oligomeric matrix protein (COMP), to form the multivalent antibodies EG2-RHCC and EG2-COMP. We labeled these two antibodies with (99m) Tc and assessed their targeting efficiency for epidermal growth factor receptor (EGFR). Cell binding, uptake, efflux, and blocking studies were performed with EGFR high- and/or low-expressing cells with (99m) Tc-labeled EG2-RHCC or EG2-COMP. Single photon-emission computed tomography (SPECT) imaging and biodistribution studies were further carried out. Both (99m) Tc-EG2-RHCC and (99m) Tc-EG2-COMP can specially bind to EGFR in vitro. SPECT imaging showed that A431, which expresses high levels of EGFR, was clearly visible 6 h after (99m) Tc-EG2-COMP injection; however, it was not detectable after administration of (99m) Tc-EG2-RHCC. Uptake of both antibodies by the non-EGFR-secreting OCM-1 tumors was low. EG2-COMP shows promise in identifying EGFR over-expression in tumors; however, EG2-RHCC may not be suitable for targeting EGFR in vivo.
Collapse
Affiliation(s)
- Chongjiao Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China.,Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongyan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lifei Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Bin Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Science and Technology of China, Hefei, China.,China-Japan Joint Laboratory of Molecular Immunology and Microbiology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
16
|
Li C, Zhang Y, Wang L, Feng H, Xia X, Ma J, Yuan H, Gao B, Lan X. A novel multivalent (99m)Tc-labeled EG2-C4bpα antibody for targeting the epidermal growth factor receptor in tumor xenografts. Nucl Med Biol 2015; 42:547-54. [PMID: 25779037 DOI: 10.1016/j.nucmedbio.2015.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/09/2015] [Accepted: 01/26/2015] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The C4b binding protein (C4bp) α/β-chain C-terminal effectively induces polymerization during protein synthesis. Using this fragment and the single-domain antibody EG2, which targets the epidermal growth factor receptor (EGFR), we generated the novel multimeric antibody EG2-C4bpα. We radiolabeled EG2-C4bpα with (99m)Tc and evaluated its targeting efficiency and pharmacokinetics in tumor xenografts. METHODS EGFR expression and EGFR-EG2-C4bpα binding was evaluated in A431 and OCM-1 cells by Western blotting and flow cytometry, respectively. EG2-C4bpα was radiolabeled with [(99m)Tc(CO)3(OH2)3](+) using a tricarbonyl vial followed by purification on a PD-10 column. In vitro studies with (99m)Tc-EG2-C4bpα were performed in A431 and/or OCM-1 cells. Single photon emission computed tomography (SPECT) imaging and biodistribution studies were carried out in (99m)Tc-EG2-C4bpα-injected mice bearing A431- and OCM-1-derived tumors. EGFR immunofluorescent staining in A431 and OCM-1 tumors was performed. RESULTS A431 cells showed higher EGFR expression levels than OCM-1 cells, and flow cytometry confirmed EG2-C4bpα bound more A431 cells than OCM-1 cells. (99m)Tc-EG2-C4bpα was successfully prepared with radiochemical yields of 30.3-50.4%. The binding affinity of (99m)Tc-EG2-C4bpα to A431 cells was approximately 20 nM. (99m)Tc-EG2-C4bpα specifically bound A431 cells and this binding was blocked by 41% in the presence of 50 nM excess unlabeled EG2-C4bpα. In vivo radioactivity uptake in A431 tumors was detected 2h after (99m)Tc-EG2-C4bpα administration and sustained up to 18h. The highest ratio of A431 tumor-to-muscle and tumor-to-blood was 3.69 ± 0.48 at 10h and 0.77 ± 0.14 at 20 h, respectively. Excess unlabeled EG2-C4bpα blocked radioactivity uptake in A431 tumors by 55% at 10h. (99m)Tc-EG2-C4bpα was barely detectable in OCM-1 tumors, and biodistribution analysis confirmed that radioactivity uptake was significantly lower than in A431 tumors. CONCLUSIONS (99m)Tc-EG2-C4bpα specifically and efficiently targets EGFR over-expressing tumors suggesting that EG2-C4bpα may be a promising antibody alternative for future diagnostic application and potential radioimmunotherapy. However, the high activity in the blood and liver, and the relative low ratio of tumor-to-blood should be noticed and improved.
Collapse
Affiliation(s)
- Chongjiao Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, PR China; Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, PR China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, PR China
| | - Lifei Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China; College of Life Sciences, University of Science and Technology of China, Hefei 230026, PR China
| | - Hongyan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, PR China
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, PR China
| | - Juan Ma
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China; College of Life Sciences, University of Science and Technology of China, Hefei 230026, PR China
| | - Hui Yuan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, PR China
| | - Bin Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China; College of Life Sciences, University of Science and Technology of China, Hefei 230026, PR China; China-Japan Joint Laboratory of Molecular Immunology and Microbiology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China.
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, PR China.
| |
Collapse
|
17
|
Williams RM, Hajiran CJ, Nayeem S, Sooter LJ. Identification of an antibody fragment specific for androgen-dependent prostate cancer cells. BMC Biotechnol 2014; 14:81. [PMID: 25186190 PMCID: PMC4158339 DOI: 10.1186/1472-6750-14-81] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 08/29/2014] [Indexed: 12/13/2022] Open
Abstract
Background Prostate cancer is the most-diagnosed non-skin cancer among males in the US, and the second leading cause of cancer-related death. Current methods of treatment and diagnosis are not specific for the disease. This work identified an antibody fragment that binds selectively to a molecule on the surface of androgen-dependent prostate cancer cells but not benign prostatic cells. Results Antibody fragment identification was achieved using a library screening and enrichment strategy. A library of 109 yeast-displayed human non-immune antibody fragments was enriched for those that bind to androgen-dependent prostate cancer cells, but not to benign prostatic cells or purified prostate-specific membrane antigen (PSMA). Seven rounds of panning and fluorescence-activated cell sorting (FACS) screening yielded one antibody fragment identified from the enriched library. This molecule, termed HiR7.8, has a low-nanomolar equilibrium dissociation constant (Kd) and high specificity for androgen-dependent prostate cancer cells. Conclusions Antibody fragment screening from a yeast-displayed library has yielded one molecule with high affinity and specificity. With further pre-clinical development, it is hoped that the antibody fragment identified using this screening strategy will be useful in the specific detection of prostate cancer and in targeted delivery of therapeutic agents for increased efficacy and reduced side effects.
Collapse
Affiliation(s)
| | | | | | - Letha J Sooter
- Department of Basic Pharmaceutical Sciences, West Virginia University, 1 Medical Center Drive, PO Box 9530, Morgantown, WV 26506, USA.
| |
Collapse
|
18
|
Biological evaluation of 131I- and CF750-labeled Dmab(scFv)-Fc antibodies for xenograft imaging of CD25-positive tumors. BIOMED RESEARCH INTERNATIONAL 2014; 2014:459676. [PMID: 24864244 PMCID: PMC4017786 DOI: 10.1155/2014/459676] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/17/2014] [Accepted: 03/17/2014] [Indexed: 02/05/2023]
Abstract
A Dmab(scFv)-Fc antibody containing the single chain variable fragment of a humanized daclizumab antibody and the Fc fragment of a human IgG1 antibody was produced via recombinant expression in Pichia pastoris. The Dmab(scFv)-Fc antibody forms a dimer in solution, and it specifically binds CD25-positive tumor cells and tumor tissues. For tumor imaging, the Dmab(scFv)-Fc antibody was labeled with the 131I isotope and CF750 fluorescent dye, respectively. After intravenous injection of mice bearing CD25-positive tumor xenografts, tumor uptake of the (131)I-Dmab(scFv)-Fc antibody was visible at 1 h, and clear images were obtained at 5 h using SPECT/CT. After systemic administration of the CF750-Dmab(scFv)-Fc antibody, tumor uptake was present as early as 1 h, and tumor xenografts could be kinetically imaged within 9 h after injection. These results indicate that the Dmab(scFv)-Fc antibody rapidly and specifically targets CD25-positive tumor cells, suggesting the potential of this antibody as an imaging agent for the diagnosis of lymphomatous-type ATLL.
Collapse
|
19
|
Badar A, Williams J, de Rosales RTM, Tavaré R, Kampmeier F, Blower PJ, Mullen GED. Optimising the radiolabelling properties of technetium tricarbonyl and His-tagged proteins. EJNMMI Res 2014; 4:14. [PMID: 24606843 PMCID: PMC4015829 DOI: 10.1186/2191-219x-4-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 02/21/2014] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND To date, the majority of protein-based radiopharmaceuticals have been radiolabelled using non-site-specific conjugation methods, with little or no control to ensure retained protein function post-labelling. The incorporation of a hexahistidine sequence (His-tag) in a recombinant protein can be used to site-specifically radiolabel with 99mTc-tricarbonyl ([99mTc(CO)3]+). This chemistry has been made accessible via a technetium tricarbonyl kit; however, reports of radiolabelling efficiencies and specific activities have varied greatly from one protein to another. Here, we aim to optimise the technetium tricarbonyl radiolabelling method to produce consistently >95% radiolabelling efficiencies with high specific activities suitable for in vivo imaging. METHODS Four different recombinant His-tagged proteins (recombinant complement receptor 2 (rCR2) and three single chain antibodies, α-CD33 scFv, α-VCAM-1 scFv and α-PSMA scFv), were used to study the effect of kit volume, ionic strength, pH and temperature on radiolabelling of four proteins. RESULTS We used 260 and 350 μL [99mTc(CO)3]+ kits enabling us to radiolabel at higher [99mTc(CO)3]+ and protein concentrations in a smaller volume and thus increase the rate at which maximum labelling efficiency and specific activity were reached. We also demonstrated that increasing the ionic strength of the reaction medium by increasing [Na+] from 0.25 to 0.63 M significantly increases the rate at which all four proteins reach a >95% labelling efficiency by at least fourfold, as compared to the conventional IsoLink® kit (Covidien, Petten, The Netherlands) and 0.25 M [Na+]. CONCLUSION We have found optimised kit and protein radiolabelling conditions suitable for the reproducible, fast, efficient radiolabelling of proteins without the need for post-labelling purification.
Collapse
Affiliation(s)
- Adam Badar
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London WC1E 6BT, UK
| | - Jennifer Williams
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Rafael TM de Rosales
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Richard Tavaré
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California–Los Angeles, Los Angeles, CA 90095-1735, USA
| | - Florian Kampmeier
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Philip J Blower
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Gregory ED Mullen
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| |
Collapse
|
20
|
LeBeau AM, Sevillano N, King ML, Duriseti S, Murphy ST, Craik CS, Murphy LL, VanBrocklin HF. Imaging the urokinase plasminongen activator receptor in preclinical breast cancer models of acquired drug resistance. Am J Cancer Res 2014; 4:267-79. [PMID: 24505235 PMCID: PMC3915090 DOI: 10.7150/thno.7323] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 12/15/2013] [Indexed: 11/30/2022] Open
Abstract
Subtype-targeted therapies can have a dramatic impact on improving the quality and quantity of life for women suffering from breast cancer. Despite an initial therapeutic response, cancer recurrence and acquired drug-resistance are commonplace. Non-invasive imaging probes that identify drug-resistant lesions are urgently needed to aid in the development of novel drugs and the effective utilization of established therapies for breast cancer. The protease receptor urokinase plasminogen activator receptor (uPAR) is a target that can be exploited for non-invasive imaging. The expression of uPAR has been associated with phenotypically aggressive breast cancer and acquired drug-resistance. Acquired drug-resistance was modeled in cell lines from two different breast cancer subtypes, the uPAR negative luminal A subtype and the uPAR positive triple negative subtype cell line MDA-MB-231. MCF-7 cells, cultured to be resistant to tamoxifen (MCF-7 TamR), were found to significantly over-express uPAR compared to the parental cell line. uPAR expression was maintained when resistance was modeled in triple-negative breast cancer by generating doxorubicin and paclitaxel resistant MDA-MB-231 cells (MDA-MB-231 DoxR and MDA-MB-231 TaxR). Using the antagonistic uPAR antibody 2G10, uPAR was imaged in vivo by near-infrared (NIR) optical imaging and 111In-single photon emission computed tomography (SPECT). Tumor uptake of the 111In-SPECT probe was high in the three drug-resistant xenografts (> 46 %ID/g) and minimal in uPAR negative xenografts at 72 hours post-injection. This preclinical study demonstrates that uPAR can be targeted for imaging breast cancer models of acquired resistance leading to potential clinical applications.
Collapse
|
21
|
Kogelberg H, Miranda E, Burnet J, Ellison D, Tolner B, Foster J, Picón C, Thomas GJ, Meyer T, Marshall JF, Mather SJ, Chester K. Generation and characterization of a diabody targeting the αvβ6 integrin. PLoS One 2013; 8:e73260. [PMID: 24023846 PMCID: PMC3762766 DOI: 10.1371/journal.pone.0073260] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 07/19/2013] [Indexed: 01/01/2023] Open
Abstract
The αvβ6 integrin is up-regulated in cancer and wound healing but it is not generally expressed in healthy adult tissue. There is increasing evidence that it has a role in cancer progression and will be a useful target for antibody-directed cancer therapies. We report a novel recombinant diabody antibody fragment that targets specifically αvβ6 and blocks its function. The diabody was engineered with a C-terminal hexahistidine tag (His tag), expressed in Pichia pastoris and purified by IMAC. Surface plasmon resonance (SPR) analysis of the purified diabody showed affinity in the nanomolar range. Pre-treatment of αvβ6-expressing cells with the diabody resulted in a reduction of cell migration and adhesion to LAP, demonstrating biological function-blocking activity. After radio-labeling, using the His-tag for site-specific attachment of (99m)Tc, the diabody retained affinity and targeted specifically to αvβ6-expressing tumors in mice bearing isogenic αvβ6 +/- xenografts. Furthermore, the diabody was specifically internalized into αvβ6-expressing cells, indicating warhead targeting potential. Our results indicate that the new αvβ6 diabody has a range of potential applications in imaging, function blocking or targeted delivery/internalization of therapeutic agents.
Collapse
Affiliation(s)
- Heide Kogelberg
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Enrique Miranda
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Jerome Burnet
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - David Ellison
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Berend Tolner
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Julie Foster
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Carmen Picón
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Gareth J. Thomas
- Cancer Sciences Unit, University of Southampton, Southampton, United Kingdom
| | - Tim Meyer
- UCL Cancer Institute, University College London, London, United Kingdom
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Stephen J. Mather
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Kerry Chester
- UCL Cancer Institute, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
22
|
Abstract
Functional radionuclide imaging modalities, now commonly combined with anatomical imaging modalities computed tomography (CT) or magnetic resonance imaging (single photon emission computed tomography [SPECT]/CT, positron emission tomography [PET]/CT, and PET/magnetic resonance imaging), are promising tools for the management of prostate cancer, particularly for therapeutic implications. Sensitive detection capability of prostate cancer using these imaging modalities is one issue; however, the treatment of prostate cancer using the information that can be obtained from functional radionuclide imaging techniques is another challenging area. There are not many SPECT or PET radiotracers that can cover the full spectrum of the management of prostate cancer from initial detection to staging, prognosis predictor, and all the way to treatment response assessment. However, when used appropriately, the information from functional radionuclide imaging improves, and sometimes significantly changes, the whole course of the cancer management. The limitations of using SPECT and PET radiotracers with regard to therapeutic implications are not so much different from their limitations solely for the task of detecting prostate cancer; however, the specific imaging target and how this target is reliably imaged by SPECT and PET can potentially make significant impact in the treatment of prostate cancer. Finally, although the localized prostate cancer is considered manageable, there is still significant need for improvement in noninvasive imaging of metastatic prostate cancer, in treatment guidance, and in response assessment from functional imaging, including radionuclide-based techniques. In this review article, we present the rationale of using functional radionuclide imaging and the therapeutic implications for each of radionuclide imaging agent that have been studied in human subjects.
Collapse
Affiliation(s)
- Carina Mari Aparici
- Department of Radiology and Biomedical Imaging, University of California, and Nuclear Medicine Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94107, USA
| | | |
Collapse
|
23
|
Jiang H, Kasten BB, Liu H, Qi S, Liu Y, Tian M, Barnes CL, Zhang H, Cheng Z, Benny PD. Novel, cysteine-modified chelation strategy for the incorporation of [M(I)(CO)(3)](+) (M = Re, (99m)Tc) in an α-MSH peptide. Bioconjug Chem 2012; 23:2300-12. [PMID: 23110503 DOI: 10.1021/bc300509k] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Engineering peptide-based targeting agents with residues for site-specific and stable complexation of radionuclides is a highly desirable strategy for producing diagnostic and therapeutic agents for cancer and other diseases. In this report, a model N-S-N(Py) ligand (3) and a cysteine-derived α-melanocyte stimulating hormone (α-MSH) peptide (6) were used as novel demonstrations of a widely applicable chelation strategy for incorporation of the [M(I)(CO)(3)](+) (M = Re, (99m)Tc) core into peptide-based molecules for radiopharmaceutical applications. The structural details of the core ligand-metal complexes as model systems were demonstrated by full chemical characterization of fac-[Re(I)(CO)(3)(N,S,N(Py)-3)](+) (4) and comparative high-performance liquid chromatography (HPLC) analysis between 4 and [(99m)Tc(I)(CO)(3)(N,S,N(Py)-3)](+) (4a). The α-MSH analogue bearing the N-S-N(Py) chelate on a modified cysteine residue (6) was generated and complexed with [M(I)(CO)(3)](+) to confirm the chelation strategy's utility when applied in a peptide-based targeting agent. Characterization of the Re(I)(CO)(3)-6 peptide conjugate (7) confirmed the efficient incorporation of the metal center, and the (99m)Tc(I)(CO)(3)-6 analogue (7a) was explored as a potential single photon emission computed tomography (SPECT) compound for imaging the melanocortin 1 receptor (MC1R) in melanoma. Peptide 7a showed excellent radiolabeling yields and in vitro stability during amino acid challenge and serum stability assays. In vitro B16F10 melanoma cell uptake of 7a reached a modest value of 2.3 ± 0.08% of applied activity at 2 h at 37 °C, while this uptake was significantly reduced by coincubation with a nonlabeled α-MSH analogue, NAPamide (3.2 μM) (P < 0.05). In vivo SPECT/X-ray computed tomography (SPECT/CT) imaging and biodistribution of 7a were evaluated in a B16F10 melanoma xenografted mouse model. SPECT/CT imaging clearly visualized the tumor at 1 h post injection (p.i.) with high tumor-to-background contrast. Blocking studies with coinjected NAPamide (10 mg per kg of mouse body weight) confirmed the in vivo specificity of 7a for MC1R-positive tumors. Biodistribution results with 7a yielded a moderate tumor uptake of 1.20 ± 0.09 percentage of the injected radioactive dose per gram of tissue (% ID/g) at 1 h p.i. Relatively high uptake of 7a was also seen in the kidneys and liver at 1 h p.i. (6.55 ± 0.36% ID/g and 4.44 ± 0.17% ID/g, respectively), although reduced kidney uptake was seen at 4 h p.i. (3.20 ± 0.48% ID/g). These results demonstrate the utility of the novel [M(I)(CO)(3)](+) chelation strategy when applied in a targeting peptide.
Collapse
Affiliation(s)
- Han Jiang
- Department of Nuclear Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Center of Excellence in Medical Molecular Imaging of Zhejiang State, Hangzhou, 310009, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lütje S, Boerman OC, van Rij CM, Sedelaar M, Helfrich W, Oyen WJG, Mulders PFA. Prospects in radionuclide imaging of prostate cancer. Prostate 2012; 72:1262-72. [PMID: 22127918 DOI: 10.1002/pros.22462] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 10/31/2011] [Indexed: 11/07/2022]
Abstract
Prostate cancer is the most common malignancy in men in the Western world and represents a major health problem with substantial morbidity and mortality. Sensitivity and specificity of digital rectal examination (DRE) and evaluation of prostate specific antigen (PSA) are excellent methods for diagnosis of prostate cancer, but have limited value for staging. Imaging of prostate cancer has become increasingly important to improve staging and management of prostate cancer patients. Conventional imaging modalities, such as transrectal ultrasound and computed tomography, show limited accuracy for a reliable assessment of prostate cancer. Diagnostic value of magnetic resonance imaging has improved by dynamic contrast enhancement (DCI-MRI) and diffusion-weighted magnetic resonance imaging (DWI). Recently, substantial progress has been made in the development of functional and molecular imaging modalities, such as positron emission tomography using radiolabeled metabolic tracers, receptor-binding ligands, amino acids, peptides, or antibodies. Here, we review the value of these novel radionuclide imaging techniques in the assessment of prostate cancer.
Collapse
Affiliation(s)
- Susanne Lütje
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
25
|
Iyer AK, Lan X, Zhu X, Su Y, Feng J, Zhang X, Gao D, Seo Y, Vanbrocklin HF, Broaddus VC, Liu B, He J. Novel human single chain antibody fragments that are rapidly internalizing effectively target epithelioid and sarcomatoid mesotheliomas. Cancer Res 2011; 71:2428-32. [PMID: 21447742 DOI: 10.1158/0008-5472.can-10-3484] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human antibodies targeting all subtypes of mesothelioma could be useful to image and treat this deadly disease. Here we report tumor targeting of a novel internalizing human single chain antibody fragment (scFv) labeled with (⁹⁹m)Tc ((⁹⁹m)Tc-M40) in murine models of mesothelioma of both epithelioid (M28) and sarcomatoid (VAMT-1) origins. (⁹⁹m)Tc-M40 was taken up rapidly and specifically by both subtype tumor cells in vitro, with 68% to 92% internalized within 1 hour. The specificity of binding was evidenced by blocking (up to 95%) with 10-fold excess of unlabeled M40. In animal studies, tumors of both subtypes were clearly visualized by SPECT/CT as early as 1 hour postinjection of (⁹⁹m)Tc-M40. Tumor uptake measured as percent of injected dose per gram tissue (%ID/g) at 3 hours was 4.38 and 5.84 for M28 and VAMT-1 tumors, respectively, significantly greater than all organs or tissues studied (liver, 2.62%ID/g; other organs or tissues <1.7%ID/g), except the kidneys (130.7%ID/g), giving tumor-to-blood ratios of 5:1 and 7:1 and tumor-to-muscle ratios of 45:1 and 60:1, for M28 and VAMT-1, respectively. The target-mediated uptake was confirmed by a nearly 70% reduction in tumor activity following administration of 10-fold excess of unlabeled scFv. Taken together, these results indicate that M40 can rapidly and specifically target epithelioid and sarcomatoid tumor cells, demonstrating the potential of this agent as a versatile targeting ligand for imaging and therapy of all subtypes of mesothelioma.
Collapse
Affiliation(s)
- Arun K Iyer
- Center for Molecular and Functional Imaging, Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhu X, Bidlingmaier S, Hashizume R, James CD, Berger MS, Liu B. Identification of internalizing human single-chain antibodies targeting brain tumor sphere cells. Mol Cancer Ther 2010; 9:2131-41. [PMID: 20587664 DOI: 10.1158/1535-7163.mct-09-1059] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive form of primary brain tumor for which there is no curative treatment to date. Resistance to conventional therapies and tumor recurrence pose major challenges to treatment and management of this disease, and therefore new therapeutic strategies need to be developed. Previous studies by other investigators have shown that a subpopulation of GBM cells can grow as neurosphere-like cells when cultured in restrictive medium and exhibits enhanced tumor-initiating ability and resistance to therapy. We report here the identification of internalizing human single-chain antibodies (scFv) targeting GBM tumor sphere cells. We selected a large naive phage antibody display library on the glycosylation-dependent CD133 epitope-positive subpopulation of GBM cells grown as tumor spheres and identified internalizing scFvs that target tumor sphere cells broadly, as well as scFvs that target the CD133-positive subpopulation. These scFvs were found to be efficiently internalized by GBM tumor sphere cells. One scFv GC4 inhibited self-renewal of GBM tumor sphere cells in vitro. We have further developed a full-length human IgG1 based on this scFv, and found that it potently inhibits proliferation of GBM tumor sphere cells and GBM cells grown in regular nonselective medium. Taken together, these results show that internalizing human scFvs targeting brain tumor sphere cells can be readily identified from a phage antibody display library, which could be useful for further development of novel therapies that target subpopulations of GBM cells to combat recurrence and resistance to treatment.
Collapse
Affiliation(s)
- Xiaodong Zhu
- Department of Anesthesia, University of California at San Francisco, San Francisco, California 94110, USA
| | | | | | | | | | | |
Collapse
|