1
|
Su RL, Cao XW, Zhao J, Wang FJ. A high hydrophobic moment arginine-rich peptide screened by a machine learning algorithm enhanced ADC antitumor activity. J Pept Sci 2024; 30:e3628. [PMID: 38950972 DOI: 10.1002/psc.3628] [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: 03/10/2024] [Revised: 04/15/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024]
Abstract
Cell-penetrating peptides (CPPs) with better biomolecule delivery properties will expand their clinical applications. Using the MLCPP2.0 machine algorithm, we screened multiple candidate sequences with potential cellular uptake ability from the nuclear localization signal/nuclear export signal database and verified them through cell-penetrating fluorescent tracing experiments. A peptide (NCR) derived from the Rev protein of the caprine arthritis-encephalitis virus exhibited efficient cell-penetrating activity, delivering over four times more EGFP than the classical CPP TAT, allowing it to accumulate in lysosomes. Structural and property analysis revealed that a high hydrophobic moment and an appropriate hydrophobic region contribute to the high delivery activity of NCR. Trastuzumab emtansine (T-DM1), a HER2-targeted antibody-drug conjugate, could improve its anti-tumor activity by enhancing targeted delivery efficiency and increasing lysosomal drug delivery. This study designed a new NCR vector to non-covalently bind T-DM1 by fusing domain Z, which can specifically bind to the Fc region of immunoglobulin G and effectively deliver T-DM1 to lysosomes. MTT results showed that the domain Z-NCR vector significantly enhanced the cytotoxicity of T-DM1 against HER2-positive tumor cells while maintaining drug specificity. Our results make a useful attempt to explore the potential application of CPP as a lysosome-targeted delivery tool.
Collapse
Affiliation(s)
- Ruo-Long Su
- Department of Applied Biology, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Xue-Wei Cao
- Department of Applied Biology, East China University of Science and Technology, Shanghai, People's Republic of China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Jian Zhao
- Department of Applied Biology, East China University of Science and Technology, Shanghai, People's Republic of China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Fu-Jun Wang
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, Shanghai, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., Zhejiang, People's Republic of China
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Zhang J, Rinne SS, Yin W, Leitao CD, Björklund E, Abouzayed A, Ståhl S, Löfblom J, Orlova A, Gräslund T, Vorobyeva A. Affibody-Drug Conjugates Targeting the Human Epidermal Growth Factor Receptor-3 Demonstrate Therapeutic Efficacy in Mice Bearing Low Expressing Xenografts. ACS Pharmacol Transl Sci 2024; 7:3228-3240. [PMID: 39416966 PMCID: PMC11475273 DOI: 10.1021/acsptsci.4c00402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024]
Abstract
The outcome of clinical trials evaluating drugs targeting the human epidermal growth factor receptor 3 (HER3) has been poor, with primary concerns related to lack of efficacy. HER3 is considered a difficult target since its overexpression on tumors is relatively low and there is normal expression in many different organs. However, a significant number of patients across different cancer indications have overexpression of HER3 and the development of novel modalities targeting HER3 is therefore warranted. Here, we have investigated the properties of affibody-based drug conjugates targeting HER3. The HER3-targeting affibody molecule ZHER3 was fused in a mono- and bivalent format to an engineered albumin-binding domain (ABD) for in vivo half-life extension and was coupled to the cytotoxic drug DM1 via a non-cleavable maleimidocaproyl (mc) linker. In vivo, a moderate uptake was observed for [99mTc]Tc-labeled ZHER3-ABD-ZHER3-mcDM1 in HER3 expressing BxPC3 tumors (3.5 ± 0.3%IA/g) at 24 h after injection, and clearance was predominately renal-mediated. Treatment of mice with BxPC3 human pancreatic cancer xenografts showed that a combination of ZHER3-ABD-ZHER3-mcDM1 and its cytostatic analog ZHER3-ABD-ZHER3 was efficacious and superior to treatment with only ZHER3-ABD-ZHER3, providing tumor growth inhibition and longer median survival (90 d) in comparison to monotherapy (68 d) and vehicle control (49 d). ZHER3-ABD-ZHER3-mcDM1 was found to be a potent drug conjugate for the treatment of HER3-expressing tumors in mice.
Collapse
Affiliation(s)
- Jie Zhang
- Department
of Protein Science, KTH Royal Institute
of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden
| | - Sara S. Rinne
- Department
of Medicinal Chemistry, Uppsala University, Dag Hammarskjöldsv 14C, 751 83 Uppsala, Sweden
| | - Wen Yin
- Department
of Protein Science, KTH Royal Institute
of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden
| | - Charles Dahlsson Leitao
- Department
of Protein Science, KTH Royal Institute
of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden
| | - Elvira Björklund
- Department
of Medicinal Chemistry, Uppsala University, Dag Hammarskjöldsv 14C, 751 83 Uppsala, Sweden
| | - Ayman Abouzayed
- Department
of Medicinal Chemistry, Uppsala University, Dag Hammarskjöldsv 14C, 751 83 Uppsala, Sweden
| | - Stefan Ståhl
- Department
of Protein Science, KTH Royal Institute
of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden
| | - John Löfblom
- Department
of Protein Science, KTH Royal Institute
of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden
| | - Anna Orlova
- Department
of Medicinal Chemistry, Uppsala University, Dag Hammarskjöldsv 14C, 751 83 Uppsala, Sweden
- Science
for Life Laboratory, Dag Hammarskjöldsv 14C, 751
83 Uppsala, Sweden
| | - Torbjörn Gräslund
- Department
of Protein Science, KTH Royal Institute
of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden
| | - Anzhelika Vorobyeva
- Department
of Immunology, Genetics and Pathology, Uppsala
University, Dag Hammarskjölds
Väg 20, 751 85 Uppsala, Sweden
| |
Collapse
|
3
|
He Y, Tian R, Xu D, Wu Y, Rina S, Chen T, Guan Y, Xie T, Ying T, Xie F, Han J. Preclinical evaluation and pilot clinical study of [ 68Ga]Ga-NOTA-H006 for non-invasive PET imaging of 5T4 oncofetal antigen. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06941-1. [PMID: 39377811 DOI: 10.1007/s00259-024-06941-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Trophoblast glycoprotein, the so-called 5T4, is an oncofetal antigen expressed in many different cancers. However, no 5T4-specific radioligand is employed in the clinic for non-invasive diagnosis. Thus, the aim of the current study was to develop a PET radiotracer for imaging 5T4 expression in preclinical and clinical stages. METHODS A VHH library was constructed by camel immunization. The specificity of the VHHs toward 5T4 antigen was screened through phage display biopanning and periplasmic extract enzyme-linked immunosorbent assay. 1,4,7-Triazacyclononane-1,4,7-triacetate acid (NOTA) derivative was conjugated to the selected VHH. After radiolabeling, microPET/CT and ex vivo biodistribution were conducted using BxPC-3 and MDA-MB-468 tumor-bearing mice. Cold VHH was co-injected with the tracer to challenge its binding in vivo. For the pilot clinical study, PET/CT images were acquired at 1 h after injection of tracer in patients with pathologically confirmed primary and metastatic tumors. RESULTS A library with a capacity of 1.2 × 1012 colony-forming units was constructed after successful camel immunization. Nb1-40 with a median effect concentration of 0.43 nM was selected. After humanization, the resulting H006 maintained a high affinity towards 5T4. [68Ga]Ga-NOTA-H006 with the molar activities of 6.48-54.2 GBq/µmol was prepared with high radiochemical purity (> 98%). Using [68Ga]Ga-NOTA-H006, microPET/CT revealed a clear visualization of 5T4 expression in BxPC-3 tumor-bearing mice. Ex vivo biodistribution showed that the highest tumor-to-blood ratio (∼ 3-fold) and tumor-to-muscle ratio (∼ 5-fold) were achieved at 60 min post-injection. Co-injection of the cold H006 at a dose of 1.5 mg/kg significantly reduced the tumor uptake (p < 0.0001). In the pilot clinical study, [68Ga]Ga-NOTA-H006 demonstrated its capacity to map 5T4-positive lesions in humans and yielded a mean effective dose of 3.4 × 10- 2 mSv/MBq. CONCLUSIONS [68Ga]Ga-NOTA-H006, which can visualize 5T4 expression in vivo, has been successfully developed. This opens up opportunities for non-invasively studying 5T4 expression through nuclear medicine. Further clinical investigations are warranted to explore its clinical value in disease progression and companion diagnosis.
Collapse
Affiliation(s)
- Yingfang He
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Sa Rina
- Huahe Pharmaceutical Co., Ltd, Shanghai, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Tianwu Xie
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China.
| | - Junbin Han
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China.
| |
Collapse
|
4
|
Westerlund K, Oroujeni M, Gestin M, Clinton J, Hani Rosly A, Tano H, Vorobyeva A, Orlova A, Eriksson Karlström A, Tolmachev V. Shorter Peptide Nucleic Acid Probes Improve Affibody-Mediated Peptide Nucleic Acid-Based Pretargeting. ACS Pharmacol Transl Sci 2024; 7:1595-1611. [PMID: 38751640 PMCID: PMC11091976 DOI: 10.1021/acsptsci.4c00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024]
Abstract
Affibody-mediated PNA-based pretargeting shows promise for HER2-expressing tumor radiotherapy. In our recent study, a 15-mer ZHER2:342-HP15 affibody-PNA conjugate, in combination with a shorter 9-mer [177Lu]Lu-HP16 effector probe, emerged as the most effective pretargeting strategy. It offered a superior tumor-to-kidney uptake ratio and more efficient tumor targeting compared to longer radiolabeled effector probes containing 12 or 15 complementary PNA bases. To enhance the production efficiency of our pretargeting system, we here introduce even shorter 6-, 7-, and 8-mer secondary probes, designated as HP19, HP21, and HP20, respectively. We also explore the replacement of the original 15-mer Z-HP15 primary probe with shorter 12-mer Z-HP12 and 9-mer Z-HP9 alternatives. This extended panel of shorter PNA-based probes was synthesized using automated microwave-assisted methods and biophysically screened in vitro to identify shorter probe combinations with the most effective binding properties. In a mouse xenograft model, we evaluated the biodistribution of these probes, comparing them to the Z-HP15:[177Lu]Lu-HP16 combination. Tumor-to-kidney ratios at 4 and 144 h postinjection of the secondary probe showed no significant differences among the Z-HP9:[177Lu]Lu-HP16, Z-HP9:[177Lu]Lu-HP20, and the Z-HP15:[177Lu]Lu-HP16 pairs. Importantly, tumor uptake significantly exceeded, by several hundred-fold, that of most normal tissues, with kidney uptake being the critical organ for radiation therapy. This suggests that using a shorter 9-mer primary probe, Z-HP9, in combination with 9-mer HP16 or 8-mer HP20 secondary probes effectively targets tumors while minimizing the dose-limiting kidney uptake of radionuclide. In conclusion, the Z-HP9:HP16 and Z-HP9:HP20 probe combinations offer good prospects for both cost-effective production and efficient in vivo pretargeting of HER2-expressing tumors.
Collapse
Affiliation(s)
- Kristina Westerlund
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Maryam Oroujeni
- Department
of Immunology, Genetics and
Pathology, Uppsala University, Uppsala 751 23, Sweden
- Affibody
AB, Solna 171
65, Sweden
| | - Maxime Gestin
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Jacob Clinton
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Alia Hani Rosly
- Department
of Immunology, Genetics and
Pathology, Uppsala University, Uppsala 751 23, Sweden
| | - Hanna Tano
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Anzhelika Vorobyeva
- Department
of Immunology, Genetics and
Pathology, Uppsala University, Uppsala 751 23, Sweden
| | - Anna Orlova
- Department
of Medicinal Chemistry, Uppsala University, Uppsala 751 23, Sweden
| | - Amelie Eriksson Karlström
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, Stockholm 106 91, Sweden
| | - Vladimir Tolmachev
- Department
of Immunology, Genetics and
Pathology, Uppsala University, Uppsala 751 23, Sweden
| |
Collapse
|
5
|
Larkina M, Varvashenya R, Yuldasheva F, Plotnikov E, Bezverkhniaia E, Tretyakova M, Zelchan R, Schulga A, Konovalova E, Vorobyeva A, Belousov M, Orlova A, Tolmachev V, Deyev S. Comparative Preclinical Evaluation of HYNIC-Modified Designed Ankyrin Repeat Proteins G3 for the 99mTc-Based Imaging of HER2-Expressing Malignant Tumors. Mol Pharm 2024; 21:1919-1932. [PMID: 38557163 DOI: 10.1021/acs.molpharmaceut.3c01173] [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] [Indexed: 04/04/2024]
Abstract
HER2 status determination is a necessary step for the proper choice of therapy and selection of patients for the targeted treatment of cancer. Targeted radiotracers such as radiolabeled DARPins provide a noninvasive and effective way for the molecular imaging of HER2 expression. This study aimed to evaluate tumor-targeting properties of three 99mTc-labeled DARPin G3 variants containing Gly-Gly-Gly-Cys (G3C), (Gly-Gly-Gly-Ser)3-Cys ((G3S)3C), or Glu-Glu-Glu-Cys (E3C) amino acid linkers at the C-terminus and conjugated to the HYNIC chelating agent, as well as to compare them with the clinically evaluated DARPin G3 labeled with 99mTc(CO)3 using the (HE)3-tag at the N-terminus. The labeling of DARPin G3-HYNIC variants provided radiochemical yields in the range of 50-80%. Labeled variants bound specifically to human HER2-expressing cancer cell lines with affinities in the range of 0.5-3 nM. There was no substantial influence of the linker and HYNIC chelator on the binding of 99mTc-labeled DARPin G3 variants to HER2 in vitro; however, [99mTc]Tc-G3-(G3S)3C-HYNIC had the highest affinity. Comparative biodistribution of [99mTc]Tc-G3-G3C-HYNIC, [99mTc]Tc-G3-(G3S)3C-HYNIC, [99mTc]Tc-G3-E3C-HYNIC, and [99mTc]Tc-(HE)3-G3 in healthy CD1 mice showed that there was a strong influence of the linkers on uptake in normal tissues. [99mTc]Tc-G3-E3C-HYNIC had an increased retention of activity in the liver and the majority of other organs compared to the other conjugates. The tumor uptake of [99mTc]Tc-G3-(G3S)3C-HYNIC and [99mTc]Tc-(HE)3-G3 in Nu/j mice bearing SKOV-3 xenografts was similar. The specificity of tumor targeting in vivo was demonstrated for both tracers. [99mTc]Tc-G3-(G3S)3C-HYNIC provided comparable, although slightly lower tumor-to-lung, tumor-to spleen and tumor-to-liver ratios than [99mTc]Tc-(HE)3-G3. Radiolabeling of DARPin G3-HYNIC conjugates with 99mTc provided the advantage of a single-step radiolabeling procedure; however, the studied HYNIC conjugates did not improve imaging contrast compared to the 99mTc-tricarbonyl-labeled DARPin G3. At this stage, [99mTc]Tc-(HE)3-G3 remains the most promising candidate for the clinical imaging of HER2-overexpressing cancers.
Collapse
Affiliation(s)
- Maria Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Ruslan Varvashenya
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Feruza Yuldasheva
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Evgenii Plotnikov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Ekaterina Bezverkhniaia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Maria Tretyakova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Roman Zelchan
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Nuclear Medicine, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Elena Konovalova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Anzhelika Vorobyeva
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Mikhail Belousov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Sergey Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
6
|
Larkina M, Plotnikov E, Bezverkhniaia E, Shabanova Y, Tretyakova M, Yuldasheva F, Zelchan R, Schulga A, Konovalova E, Vorobyeva A, Garousi J, Gräslund T, Belousov M, Tolmachev V, Deyev S. Comparative Preclinical Evaluation of Peptide-Based Chelators for the Labeling of DARPin G3 with 99mTc for Radionuclide Imaging of HER2 Expression in Cancer. Int J Mol Sci 2022; 23:13443. [PMID: 36362226 PMCID: PMC9653920 DOI: 10.3390/ijms232113443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2023] Open
Abstract
Non-invasive radionuclide imaging of human epidermal growth factor receptor type 2 (HER2) expression in breast, gastroesophageal, and ovarian cancers may stratify patients for treatment using HER2-targeted therapeutics. Designed ankyrin repeat proteins (DARPins) are a promising type of targeting probe for radionuclide imaging. In clinical studies, the DARPin [99mTc]Tc-(HE)3-G3 labeled using a peptide-based chelator His-Glu-His-Glu-His-Glu ((HE)3), provided clear imaging of HER2 expressing breast cancer 2-4 h after injection. The goal of this study was to evaluate if the use of cysteine-containing peptide-based chelators Glu-Glu-Glu-Cys (E3C), Gly-Gly-Gly-Cys (G3C), and Gly-Gly-Gly-Ser-Cys connected via a (Gly-Gly-Gly-Ser)3-linker (designated as G3-(G3S)3C) would further improve the contrast of imaging using 99mTc-labeled derivatives of G3. The labeling of the new variants of G3 provided a radiochemical yield of over 95%. Labeled G3 variants bound specifically to human HER2-expressing cancer cell lines with affinities in the range of 1.9-5 nM. Biodistribution of [99mTc]Tc-G3-G3C, [99mTc]Tc-G3-(G3S)3C, and [99mTc]Tc-G3-E3C in mice was compared with the biodistribution of [99mTc]Tc-(HE)3-G3. It was found that the novel variants provide specific accumulation in HER2-expressing human xenografts and enable discrimination between tumors with high and low HER2 expression. However, [99mTc]Tc-(HE)3-G3 provided better contrast between tumors and the most frequent metastatic sites of HER2-expressing cancers and is therefore more suitable for clinical applications.
Collapse
Affiliation(s)
- Mariia Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Evgenii Plotnikov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Ekaterina Bezverkhniaia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Yulia Shabanova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Maria Tretyakova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Feruza Yuldasheva
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Roman Zelchan
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Nuclear Medicine, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Elena Konovalova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 11417 Stockholm, Sweden
| | - Torbjörn Gräslund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 11417 Stockholm, Sweden
| | - Mikhail Belousov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Sergey Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
7
|
Liu Y, Yu S, Xu T, Bodenko V, Orlova A, Oroujeni M, Rinne SS, Tolmachev V, Vorobyeva A, Gräslund T. Preclinical Evaluation of a New Format of 68Ga- and 111In-Labeled Affibody Molecule Z IGF-1R:4551 for the Visualization of IGF-1R Expression in Malignant Tumors Using PET and SPECT. Pharmaceutics 2022; 14:pharmaceutics14071475. [PMID: 35890370 PMCID: PMC9320461 DOI: 10.3390/pharmaceutics14071475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
The Insulin-like growth factor-1 receptor (IGF-1R) is a molecular target for several monoclonal antibodies undergoing clinical evaluation as anticancer therapeutics. The non-invasive detection of IGF-1R expression in tumors might enable stratification of patients for specific treatment and improve the outcome of both clinical trials and routine treatment. The affibody molecule ZIGF-1R:4551 binds specifically to IGF-1R with subnanomolar affinity. The goal of this study was to evaluate the 68Ga and 111In-labeled affibody construct NODAGA-(HE)3-ZIGF-1R:4551 for the imaging of IGF-1R expression, using PET and SPECT. The labeling was efficient and provided stable coupling of both radionuclides. The two imaging probes, [68Ga]Ga-NODAGA-(HE)3-ZIGF-1R:4551 and [111In]In-NODAGA-(HE)3-ZIGF-1R:4551, demonstrated specific binding to IGF-1R-expressing human cancer cell lines in vitro and to IGF-1R-expressing xenografts in mice. Preclinical PET and SPECT/CT imaging demonstrated visualization of IGF-1R-expressing xenografts already one hour after injection. The tumor-to-blood ratios at 3 h after injection were 7.8 ± 0.2 and 8.0 ± 0.6 for [68Ga]Ga-NODAGA-(HE)3-ZIGF-1R:4551 and [111In]In-NODAGA-(HE)3-ZIGF-1R:4551, respectively. In conclusion, a molecular design of the ZIGF-1R:4551 affibody molecule, including placement of a (HE)3-tag on the N-terminus and site-specific coupling of a NODAGA chelator on the C-terminus, provides a tracer with improved imaging properties for visualization of IGF-1R in malignant tumors, using PET and SPECT.
Collapse
Affiliation(s)
- Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Shengze Yu
- Department of Protein Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden;
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Vitalina Bodenko
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden;
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
- Affibody AB, 17165 Solna, Sweden
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden;
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
- Correspondence: (V.T.); (T.G.); Tel.: +46-704-250782 (V.T.); +46-8790-9627 (T.G.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden;
- Correspondence: (V.T.); (T.G.); Tel.: +46-704-250782 (V.T.); +46-8790-9627 (T.G.)
| |
Collapse
|
8
|
Hu X, Li D, Fu Y, Zheng J, Feng Z, Cai J, Wang P. Advances in the Application of Radionuclide-Labeled HER2 Affibody for the Diagnosis and Treatment of Ovarian Cancer. Front Oncol 2022; 12:917439. [PMID: 35785201 PMCID: PMC9240272 DOI: 10.3389/fonc.2022.917439] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/20/2022] [Indexed: 12/19/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a highly expressed tumor marker in epithelial ovarian cancer, and its overexpression is considered to be a potential factor of poor prognosis. Therefore, monitoring the expression of HER2 receptor in tumor tissue provides favorable conditions for accurate localization, diagnosis, targeted therapy, and prognosis evaluation of cancer foci. Affibody has the advantages of high affinity, small molecular weight, and stable biochemical properties. The molecular probes of radionuclide-labeled HER2 affibody have recently shown broad application prospects in the diagnosis and treatment of ovarian cancer; the aim is to introduce radionuclides into the cancer foci, display systemic lesions, and kill tumor cells through the radioactivity of the radionuclides. This process seamlessly integrates the diagnosis and treatment of ovarian cancer. Current research and development of new molecular probes of radionuclide-labeled HER2 affibody should focus on overcoming the deficiencies of non-specific uptake in the kidney, bone marrow, liver, and gastrointestinal tract, and on reducing the background of the image to improve image quality. By modifying the amino acid sequence; changing the hydrophilicity, surface charge, and lipid solubility of the affibody molecule; and using different radionuclides, chelating agents, and labeling conditions to optimize the labeling method of molecular probes, the specific uptake of molecular probes at tumor sites will be improved, while reducing radioactive retention in non-target organs and obtaining the best target/non-target value. These measures will enable the clinical use of radionuclide-labeled HER2 affibody molecular probes as soon as possible, providing a new clinical path for tumor-specific diagnosis, targeted therapy, and efficacy evaluation. The purpose of this review is to describe the application of radionuclide-labeled HER2 affibody in the imaging and treatment of ovarian cancer, including its potential clinical value and dilemmas.
Collapse
Affiliation(s)
- Xianwen Hu
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dandan Li
- Department of Obstetrics, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, China
| | - Yujie Fu
- Research and Development Department, Jiangsu Yuanben Biotechnology Co., Ltd., Zunyi, China
| | - Jiashen Zheng
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zelong Feng
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiong Cai
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Jiong Cai, ; Pan Wang,
| | - Pan Wang
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Jiong Cai, ; Pan Wang,
| |
Collapse
|
9
|
Experimental Therapy of HER2-Expressing Xenografts Using the Second-Generation HER2-Targeting Affibody Molecule 188Re-ZHER2:41071. Pharmaceutics 2022; 14:pharmaceutics14051092. [PMID: 35631678 PMCID: PMC9146794 DOI: 10.3390/pharmaceutics14051092] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023] Open
Abstract
HER2-targeted radionuclide therapy might be helpful for the treatment of breast, gastric, and ovarian cancers which have developed resistance to antibody and antibody-drug conjugate-based therapies despite preserved high HER2-expression. Affibody molecules are small targeting proteins based on a non-immunoglobulin scaffold. The goal of this study was to test in an animal model a hypothesis that the second-generation HER2-targeting Affibody molecule 188Re-ZHER2:41071 might be useful for treatment of HER2-expressing malignant tumors. ZHER2:41071 was efficiently labeled with a beta-emitting radionuclide rhenium-188 (188Re). 188Re-ZHER2:41071 demonstrated preserved specificity and high affinity (KD = 5 ± 3 pM) of binding to HER2-expressing cells. In vivo studies demonstrated rapid washout of 188Re from kidneys. The uptake in HER2-expressing SKOV-3 xenografts was HER2-specific and significantly exceeded the renal uptake 4 h after injection and later. The median survival of mice, which were treated by three injections of 16 MBq 188Re-ZHER2:41071 was 68 days, which was significantly longer (<0.0001 in the log-rank Mantel-Cox test) than survival of mice in the control groups treated with vehicle (29 days) or unlabeled ZHER2:41071 (27.5 days). In conclusion, the experimental radionuclide therapy using 188Re-ZHER2:41071 enabled enhancement of survival of mice with human tumors without toxicity to the kidneys, which is the critical organ.
Collapse
|
10
|
Xu T, Liu Y, Schulga A, Konovalova E, Deyev S, Tolmachev V, Vorobyeva A. Epithelial cell adhesion molecule‑targeting designed ankyrin repeat protein‑toxin fusion Ec1‑LoPE exhibits potent cytotoxic action in prostate cancer cells. Oncol Rep 2022; 47:94. [PMID: 35315504 PMCID: PMC8968790 DOI: 10.3892/or.2022.8305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Targeted anticancer therapeutics offer the advantage of reducing cytotoxic side effects to normal cells by directing the cytotoxic payload selectively to cancer cells. Designed ankyrin repeat proteins (DARPins) are promising non-immunoglobulin-based scaffold proteins for payload delivery to cancer-associated molecular targets. Epithelial cell adhesion molecule (EpCAM) is overexpressed in 40–60% of prostate cancers (PCs) and is associated with metastasis, increased risk of PC recurrence and resistance to treatment. Here, we investigated the use of DARPin Ec1 for targeted delivery of Pseudomonas exotoxin A variant (LoPE) with low immunogenicity and low non-specific toxicity to EpCAM-expressing prostate cancer cells. Ec1-LoPE fusion protein was radiolabeled with tricarbonyl technetium-99m and its binding specificity, binding kinetics, cellular processing, internalization and cytotoxicity were evaluated in PC-3 and DU145 cell lines. Ec1-LoPE showed EpCAM-specific binding to EpCAM-expressing prostate cancer cells. Rapid internalization mediated potent cytotoxic effect with picomolar IC50 values in both studied cell lines. Taken together, these data support further evaluation of Ec1-LoPE in a therapeutic setting in a prostate cancer model in vivo.
Collapse
Affiliation(s)
- Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin‑Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Sergey Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| |
Collapse
|
11
|
Xu T, Zhang J, Oroujeni M, Tretyakova MS, Bodenko V, Belousov MV, Orlova A, Tolmachev V, Vorobyeva A, Gräslund T. Effect of Inter-Domain Linker Composition on Biodistribution of ABD-Fused Affibody-Drug Conjugates Targeting HER2. Pharmaceutics 2022; 14:pharmaceutics14030522. [PMID: 35335898 PMCID: PMC8949183 DOI: 10.3390/pharmaceutics14030522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/04/2022] Open
Abstract
Targeted drug conjugates based on Affibody molecules fused to an albumin-binding domain (ABD) for half-life extension have demonstrated potent anti-tumor activity in preclinical therapeutic studies. Furthermore, optimization of their molecular design might increase the cytotoxic effect on tumors and minimize systemic toxicity. This study aimed to investigate the influence of length and composition of a linker between the human epidermal growth factor receptor 2 (HER2)-targeted affibody molecule (ZHER2:2891) and the ABD domain on functionality and biodistribution of affibody-drug conjugates containing a microtubulin inhibitor mertansin (mcDM1) (AffiDCs). Two conjugates, having a trimeric (S3G)3 linker or a trimeric (G3S)3 linker were produced, radiolabeled with 99mTc(CO)3, and compared side-by-side in vitro and in vivo with the original ZHER2:2891-G4S-ABD-mcDM1 conjugate having a monomeric G4S linker. Both conjugates with longer linkers had a decreased affinity to HER2 and mouse and human serum albumin in vitro, however, no differences in blood retention were observed in NMRI mice up to 24 h post injection. The use of both (S3G)3 and (G3S)3 linkers reduced liver uptake of AffiDCs by approximately 1.2-fold compared with the use of a G4S linker. This finding provides important insights into the molecular design for the development of targeted drug conjugates with reduced hepatic uptake.
Collapse
Affiliation(s)
- Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.O.); (V.T.)
| | - Jie Zhang
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden; (J.Z.); (T.G.)
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.O.); (V.T.)
- Department of Science and Development, Affibody AB, 171 65 Solna, Sweden
| | - Maria S. Tretyakova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (M.S.T.); (V.B.); (A.O.)
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Vitalina Bodenko
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (M.S.T.); (V.B.); (A.O.)
| | - Mikhail V. Belousov
- Department of Pharmaceutical Analysis, Siberian State Medical University, Ministry of Health of the Russian Federation, 634050 Tomsk, Russia;
- Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (M.S.T.); (V.B.); (A.O.)
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.O.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (M.S.T.); (V.B.); (A.O.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.O.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (M.S.T.); (V.B.); (A.O.)
- Correspondence: ; Tel.: +46-70-838-74-87
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden; (J.Z.); (T.G.)
| |
Collapse
|
12
|
Yamatsugu K, Katoh H, Yamashita T, Takahashi K, Aki S, Tatsumi T, Kaneko Y, Kawamura T, Miura M, Ishii M, Ohkubo K, Osawa T, Kodama T, Ishikawa S, Kanai M, Sugiyama A. Antibody mimetic drug conjugate manufactured by high-yield Escherichia coli expression and non-covalent binding system. Protein Expr Purif 2022; 192:106043. [PMID: 34973460 DOI: 10.1016/j.pep.2021.106043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022]
Abstract
Antibody-drug conjugates (ADCs) are a major therapeutic tool for the treatment of advanced cancer. Malignant cells in advanced cancer often display multiple genetic mutations and become resistant to monotherapy. Therefore, a therapeutic regimen that simultaneously targets multiple molecules with multiple payloads is desirable. However, the development of ADCs is hampered by issues in biopharmaceutical manufacturing and the complexity of the conjugation process of low-molecular-weight payloads to biologicals. Here, we report antibody mimetic-drug conjugates (AMDCs) developed by exploiting the non-covalent binding property of payloads based on high-affinity binding of mutated streptavidin and modified iminobiotin. Miniprotein antibodies were fused to a low immunogenic streptavidin variant, which was then expressed in Escherichia coli inclusion bodies, solubilized, and refolded into functional tetramers. The AMDC developed against human epidermal growth factor receptor 2 (HER2) effectively killed cultured cancer cells using bis-iminobiotin conjugated to photo-activating silicon phthalocyanine. The HER2-targeting AMDC was also effective in vivo against a mouse KPL-4 xenograft model. This AMDC platform provides rapid, stable, and high-yield therapeutics against multiple targets.
Collapse
Affiliation(s)
- Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takefumi Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Kazuki Takahashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Sho Aki
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Toshifumi Tatsumi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yudai Kaneko
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Takeshi Kawamura
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan; Isotope Science Center, The University of Tokyo, 2-11-16 Yayoi, Bunyo-ku, Tokyo, 113-0032, Japan
| | - Mai Miura
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Masazumi Ishii
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Kei Ohkubo
- Institute for Advanced Co-Creation Studies, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Tsuyoshi Osawa
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Tatsuhiko Kodama
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Akira Sugiyama
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan; Isotope Science Center, The University of Tokyo, 2-11-16 Yayoi, Bunyo-ku, Tokyo, 113-0032, Japan.
| |
Collapse
|
13
|
HER3 PET Imaging: 68Ga-Labeled Affibody Molecules Provide Superior HER3 Contrast to 89Zr-Labeled Antibody and Antibody-Fragment-Based Tracers. Cancers (Basel) 2021; 13:cancers13194791. [PMID: 34638277 PMCID: PMC8508546 DOI: 10.3390/cancers13194791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary HER3 is a known driver for oncogenesis and therapy resistance in solid cancers. PET imaging could be a useful tool to non-invasively detect and monitor HER3 expression and aid in the selection of patients for HER3-targeted therapy. PET tracers based on therapeutic antibodies have thus far shown limited success in reliably imaging HER3-expressing tumors in clinical trials. Smaller-sized tracers specifically designed for imaging might be needed for higher contrast imaging and sufficient sensitivity. Our group has previously studied the use of radiolabeled affibody molecules for imaging of HER3 expression. In the present study, we compared four different types of potential PET tracers for imaging of HER3 expression in a preclinical model. We demonstrated that the affibody-based tracer, [68Ga]Ga-ZHER3, could provide overall superior imaging contrast to antibody- and antibody-fragment-based tracers shortly after injection. Our results indicate that HER3-targeting affibody molecules are promising agents for PET imaging of HER3 expression. Abstract HER3 (human epidermal growth factor receptor type 3) is a challenging target for diagnostic radionuclide molecular imaging due to the relatively modest overexpression in tumors and substantial expression in healthy organs. In this study, we compared four HER3-targeting PET tracers based on different types of targeting molecules in a preclinical model: the 89Zr-labeled therapeutic antibody seribantumab, a seribantumab-derived F(ab)2-fragment labeled with 89Zr and 68Ga, and the 68Ga-labeled affibody molecule [68Ga]Ga-ZHER3. The novel conjugates were radiolabeled and characterized in vitro using HER3-expressing BxPC-3 and DU145 human cancer cells. Biodistribution was studied using Balb/c nu/nu mice bearing BxPC-3 xenografts. HER3-negative RAMOS xenografts were used to demonstrate binding specificity in vivo. Autoradiography was conducted on the excised tumors. nanoPET/CT imaging was performed. New conjugates specifically bound to HER3 in vitro and in vivo. [68Ga]Ga-DFO-seribantumab-F(ab’)2 was considered unsuitable for imaging due to the low stability and high uptake in normal organs. The highest tumor-to-non-tumor contrast with [89Zr]Zr-DFO-seribantumab and [89Zr]Zr-DFO-seribantumab-F(ab’)2 was achieved at 96 h and 48 h pi, respectively. Despite lower tumor uptake, [68Ga]Ga-ZHER3 provided the best imaging contrast due to the fastest clearance from blood and normal organs. The results of our study suggest that affibody-based tracers are more suitable for PET imaging of HER3 expression than antibody- and antibody-fragment-based tracers.
Collapse
|
14
|
Xu T, Vorobyeva A, Schulga A, Konovalova E, Vorontsova O, Ding H, Gräslund T, Tashireva LA, Orlova A, Tolmachev V, Deyev SM. Imaging-Guided Therapy Simultaneously Targeting HER2 and EpCAM with Trastuzumab and EpCAM-Directed Toxin Provides Additive Effect in Ovarian Cancer Model. Cancers (Basel) 2021; 13:3939. [PMID: 34439094 PMCID: PMC8393281 DOI: 10.3390/cancers13163939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/23/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022] Open
Abstract
Efficient treatment of disseminated ovarian cancer (OC) is challenging due to its heterogeneity and chemoresistance. Overexpression of human epidermal growth factor receptor 2 (HER2) and epithelial cell adhesion molecule (EpCAM) in approx. 30% and 70% of ovarian cancers, respectively, allows for co-targeted treatment. The clinical efficacy of the monoclonal antibody trastuzumab in patients with HER2-positive breast, gastric and gastroesophageal cancers makes it readily available as the HER2-targeting component. As the EpCAM-targeting component, we investigated the designed ankyrin repeat protein (DARPin) Ec1 fused to a truncated variant of Pseudomonas exotoxin A with reduced immunogenicity and low general toxicity (LoPE). Ec1-LoPE was radiolabeled, evaluated in ovarian cancer cells in vitro and its biodistribution and tumor-targeting properties were studied in vivo. The therapeutic efficacy of Ec1-LoPE alone and in combination with trastuzumab was studied in mice bearing EpCAM- and HER2-expressing SKOV3 xenografts. SPECT/CT imaging enabled visualization of EpCAM and HER2 expression in the tumors. Co-treatment using Ec1-LoPE and trastuzumab was more effective at reducing tumor growth and prolonged the median survival of mice compared with mice in the control and monotherapy groups. Repeated administration of Ec1-LoPE was well tolerated without signs of hepatic or kidney toxicity. Co-treatment with trastuzumab and Ec1-LoPE might be a potential therapeutic strategy for HER2- and EpCAM-positive OC.
Collapse
Affiliation(s)
- Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (O.V.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (O.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Olga Vorontsova
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (O.V.)
| | - Haozhong Ding
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Liubov A. Tashireva
- Cancer Research Institute, Tomsk National Research Medical Center Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (O.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
| | - Sergey M. Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (A.S.); (A.O.); (S.M.D.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
- Bio-Nanophotonic Lab, Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University ‘MEPhI’, 115409 Moscow, Russia
- Center of Biomedical Engineering, Sechenov University, 119991 Moscow, Russia
| |
Collapse
|
15
|
Deyev SM, Xu T, Liu Y, Schulga A, Konovalova E, Garousi J, Rinne SS, Larkina M, Ding H, Gräslund T, Orlova A, Tolmachev V, Vorobyeva A. Influence of the Position and Composition of Radiometals and Radioiodine Labels on Imaging of Epcam Expression in Prostate Cancer Model Using the DARPin Ec1. Cancers (Basel) 2021; 13:cancers13143589. [PMID: 34298801 PMCID: PMC8304184 DOI: 10.3390/cancers13143589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Metastasis-targeting therapy might improve outcomes in oligometastatic prostate cancer. Epithelial cell adhesion molecule (EpCAM) is overexpressed in 40–60% of prostate cancer cases and might be used as a target for specific delivery of toxins and drugs. Radionuclide molecular imaging could enable non-invasive detection of EpCAM and stratification of patients for targeted therapy. Designed ankyrin repeat proteins (DARPins) are scaffold proteins, which can be selected for specific binding to different targets. The DARPin Ec1 binds strongly to EpCAM. To determine an optimal design of Ec1-based probes, we labeled Ec1 at two different positions with four different nuclides (68Ga, 111In, 57Co and 125I) and investigated the impact on Ec1 biodistribution. We found that the C-terminus is the best position for labeling and that 111In and 125I provide the best imaging contrast. This study might be helpful for scientists developing imaging probes based on scaffold proteins. Abstract The epithelial cell adhesion molecule (EpCAM) is intensively overexpressed in 40–60% of prostate cancer (PCa) cases and can be used as a target for the delivery of drugs and toxins. The designed ankyrin repeat protein (DARPin) Ec1 has a high affinity to EpCAM (68 pM) and a small size (18 kDa). Radiolabeled Ec1 might be used as a companion diagnostic for the selection of PCa patients for therapy. The study aimed to investigate the influence of radiolabel position (N- or C-terminal) and composition on the targeting and imaging properties of Ec1. Two variants, having an N- or C-terminal cysteine, were produced, site-specifically conjugated to a DOTA chelator and labeled with cobalt-57, gallium-68 or indium-111. Site-specific radioiodination was performed using ((4-hydroxyphenyl)-ethyl)maleimide (HPEM). Biodistribution of eight radiolabeled Ec1-probes was measured in nude mice bearing PCa DU145 xenografts. In all cases, positioning of a label at the C-terminus provided the best tumor-to-organ ratios. The non-residualizing [125I]I-HPEM label provided the highest tumor-to-muscle and tumor-to-bone ratios and is more suitable for EpCAM imaging in early-stage PCa. Among the radiometals, indium-111 provided the highest tumor-to-blood, tumor-to-lung and tumor-to-liver ratios and could be used at late-stage PCa. In conclusion, label position and composition are important for the DARPin Ec1.
Collapse
Affiliation(s)
- Sergey M. Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
- Bio-Nanophotonic Lab., Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University “MEPhI”, 115409 Moscow, Russia
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
| | - Maria Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Department of Pharmaceutical Analysis, Siberian State Medical University (SSMU), 2, Moscow Trakt, 634050 Tomsk, Russia
| | - Haozhong Ding
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Torbjörn Gräslund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
- Correspondence:
| | - Anzhelika Vorobyeva
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
| |
Collapse
|
16
|
Rinne SS, Orlova A, Tolmachev V. PET and SPECT Imaging of the EGFR Family (RTK Class I) in Oncology. Int J Mol Sci 2021; 22:ijms22073663. [PMID: 33915894 PMCID: PMC8036874 DOI: 10.3390/ijms22073663] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The human epidermal growth factor receptor family (EGFR-family, other designations: HER family, RTK Class I) is strongly linked to oncogenic transformation. Its members are frequently overexpressed in cancer and have become attractive targets for cancer therapy. To ensure effective patient care, potential responders to HER-targeted therapy need to be identified. Radionuclide molecular imaging can be a key asset for the detection of overexpression of EGFR-family members. It meets the need for repeatable whole-body assessment of the molecular disease profile, solving problems of heterogeneity and expression alterations over time. Tracer development is a multifactorial process. The optimal tracer design depends on the application and the particular challenges of the molecular target (target expression in tumors, endogenous expression in healthy tissue, accessibility). We have herein summarized the recent preclinical and clinical data on agents for Positron Emission Tomography (PET) and Single Photon Emission Tomography (SPECT) imaging of EGFR-family receptors in oncology. Antibody-based tracers are still extensively investigated. However, their dominance starts to be challenged by a number of tracers based on different classes of targeting proteins. Among these, engineered scaffold proteins (ESP) and single domain antibodies (sdAb) show highly encouraging results in clinical studies marking a noticeable trend towards the use of smaller sized agents for HER imaging.
Collapse
Affiliation(s)
- Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden; (S.S.R.); (A.O.)
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden; (S.S.R.); (A.O.)
- Science for Life Laboratory, Uppsala University, 752 37 Uppsala, Sweden
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
- Correspondence: ; Tel.: +46-704-250-782
| |
Collapse
|
17
|
Affibody-Derived Drug Conjugates Targeting HER2: Effect of Drug Load on Cytotoxicity and Biodistribution. Pharmaceutics 2021; 13:pharmaceutics13030430. [PMID: 33806887 PMCID: PMC8005000 DOI: 10.3390/pharmaceutics13030430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Affibody molecules hold great promise as carriers of cytotoxic drugs for cancer therapy due to their typically high affinity, easy production, and inherent control of the drug molecules’ loading and spatial arrangement. Here, the impact of increasing the drug load from one to three on the properties of an affibody drug conjugate targeting the human epidermal growth factor receptor 2 (HER2) was investigated. The affibody carrier was recombinantly expressed as a fusion to an albumin-binding domain (ABD) for plasma half-life extension. One or three cysteine amino acids were placed at the C-terminus to which cytotoxic mcDM1 molecules were conjugated. The resulting drug conjugates, ZHER2–ABD–mcDM1 and ZHER2–ABD–mcDM13, were characterized in vitro, and their biodistribution in mice carrying HER2-overexpressing SKOV3 xenografts was determined. Increasing the drug load from one to three led to a decrease in affinity for HER2, but a significantly more potent cytotoxic effect on SKOV3 cells with high HER2 expression. The difference in cytotoxic effect on other cell lines with high HER2 expression was not significant. In vivo, an increase in drug load led to a 1.45-fold higher amount of cytotoxic mcDM1 delivered to the tumors. The increase in drug load also led to more rapid hepatic clearance, warranting further optimization of the molecular design.
Collapse
|
18
|
Oroujeni M, Rinne SS, Vorobyeva A, Loftenius A, Feldwisch J, Jonasson P, Chernov V, Orlova A, Frejd FY, Tolmachev V. Preclinical Evaluation of 99mTc-ZHER2:41071, a Second-Generation Affibody-Based HER2-Visualizing Imaging Probe with a Low Renal Uptake. Int J Mol Sci 2021; 22:ijms22052770. [PMID: 33803361 PMCID: PMC7967187 DOI: 10.3390/ijms22052770] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/28/2021] [Accepted: 03/01/2021] [Indexed: 12/20/2022] Open
Abstract
Radionuclide imaging of HER2 expression in tumours may enable stratification of patients with breast, ovarian, and gastroesophageal cancers for HER2-targeting therapies. A first-generation HER2-binding affibody molecule [99mTc]Tc-ZHER2:V2 demonstrated favorable imaging properties in preclinical studies. Thereafter, the affibody scaffold has been extensively modified, which increased its melting point, improved storage stability, and increased hydrophilicity of the surface. In this study, a second-generation affibody molecule (designated ZHER2:41071) with a new improved scaffold has been prepared and characterized. HER2-binding, biodistribution, and tumour-targeting properties of [99mTc]Tc-labelled ZHER2:41071 were investigated. These properties were compared with properties of the first-generation affibody molecules, [99mTc]Tc-ZHER2:V2 and [99mTc]Tc-ZHER2:2395. [99mTc]Tc-ZHER2:41071 bound specifically to HER2 expressing cells with an affinity of 58 ± 2 pM. The renal uptake for [99mTc]Tc-ZHER2:41071 and [99mTc]Tc-ZHER2:V2 was 25–30 fold lower when compared with [99mTc]Tc-ZHER2:2395. The uptake in tumour and kidney for [99mTc]Tc-ZHER2:41071 and [99mTc]Tc-ZHER2:V2 in SKOV-3 xenografts was similar. In conclusion, an extensive re-engineering of the scaffold did not compromise imaging properties of the affibody molecule labelled with 99mTc using a GGGC chelator. The new probe, [99mTc]Tc-ZHER2:41071 provided the best tumour-to-blood ratio compared to HER2-imaging probes for single photon emission computed tomography (SPECT) described in the literature so far. [99mTc]Tc-ZHER2:41071 is a promising candidate for further clinical translation studies.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/chemistry
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/pharmacology
- Cell Line, Tumor
- Female
- Humans
- Kidney/diagnostic imaging
- Kidney/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms, Experimental/diagnostic imaging
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Radiopharmaceuticals/chemical synthesis
- Radiopharmaceuticals/chemistry
- Radiopharmaceuticals/pharmacokinetics
- Radiopharmaceuticals/pharmacology
- Receptor, ErbB-2/metabolism
- Technetium/chemistry
- Technetium/pharmacokinetics
- Technetium/pharmacology
- Tissue Distribution
- Tomography, Emission-Computed, Single-Photon
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (F.Y.F.); (V.T.)
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden;
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (F.Y.F.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | | | | | - Per Jonasson
- Affibody AB, 171 65 Solna, Sweden; (A.L.); (J.F.); (P.J.)
| | - Vladimir Chernov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Nuclear Medicine Department, Cancer Research Institute, Tomsk National Research Medical Center Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden;
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Correspondence: ; Tel.: +46-073-9922846
| | - Fredrik Y. Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (F.Y.F.); (V.T.)
- Affibody AB, 171 65 Solna, Sweden; (A.L.); (J.F.); (P.J.)
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (F.Y.F.); (V.T.)
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden;
| |
Collapse
|
19
|
Oroujeni M, Xu T, Gagnon K, Rinne SS, Weis J, Garousi J, Andersson KG, Löfblom J, Orlova A, Tolmachev V. The Use of a Non-Conventional Long-Lived Gallium Radioisotope 66Ga Improves Imaging Contrast of EGFR Expression in Malignant Tumours Using DFO-ZEGFR:2377 Affibody Molecule. Pharmaceutics 2021; 13:pharmaceutics13020292. [PMID: 33672373 PMCID: PMC7926986 DOI: 10.3390/pharmaceutics13020292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/13/2021] [Accepted: 02/19/2021] [Indexed: 12/18/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is overexpressed in many malignancies. EGFR-targeted therapy extends survival of patients with disseminated cancers. Radionuclide molecular imaging of EGFR expression would make EGFR-directed treatment more personalized and therefore more efficient. A previous study demonstrated that affibody molecule [68Ga]Ga-DFO-ZEGFR:2377 permits specific positron-emission tomography (PET) imaging of EGFR expression in xenografts at 3 h after injection. We anticipated that imaging at 24 h after injection would provide higher contrast, but this is prevented by the short half-life of 68Ga (67.6 min). Here, we therefore tested the hypothesis that the use of the non-conventional long-lived positron emitter 66Ga (T1/2 = 9.49 h, β+ = 56.5%) would permit imaging with higher contrast. 66Ga was produced by the 66Zn(p,n)66Ga nuclear reaction and DFO-ZEGFR:2377 was efficiently labelled with 66Ga with preserved binding specificity in vitro and in vivo. At 24 h after injection, [66Ga]Ga-DFO-ZEGFR:2377 provided 3.9-fold higher tumor-to-blood ratio and 2.3-fold higher tumor-to-liver ratio than [68Ga]Ga-DFO-ZEGFR:2377 at 3 h after injection. At the same time point, [66Ga]Ga-DFO-ZEGFR:2377 provided 1.8-fold higher tumor-to-blood ratio, 3-fold higher tumor-to-liver ratio, 1.9-fold higher tumor-to-muscle ratio and 2.3-fold higher tumor-to-bone ratio than [89Zr]Zr-DFO-ZEGFR:2377. Biodistribution data were confirmed by whole body PET combined with magnetic resonance imaging (PET/MRI). The use of the positron emitter 66Ga for labelling of DFO-ZEGFR:2377 permits PET imaging of EGFR expression at 24 h after injection and improves imaging contrast.
Collapse
Affiliation(s)
- Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (M.O.); (T.X.); (J.G.)
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (M.O.); (T.X.); (J.G.)
| | - Katherine Gagnon
- GE Healthcare, GEMS PET Systems, 75015 Uppsala, Sweden;
- Department of Medicinal Chemistry, Uppsala University, 75183 Uppsala, Sweden; (S.S.R.); (A.O.)
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 75183 Uppsala, Sweden; (S.S.R.); (A.O.)
| | - Jan Weis
- Department of Medical Physics, Uppsala University Hospital, 75185 Uppsala, Sweden;
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (M.O.); (T.X.); (J.G.)
| | - Ken G. Andersson
- Department of Protein Science, KTH Royal Institute of Technology, 10691 Stockholm, Sweden; (K.G.A.); (J.L.)
| | - John Löfblom
- Department of Protein Science, KTH Royal Institute of Technology, 10691 Stockholm, Sweden; (K.G.A.); (J.L.)
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 75183 Uppsala, Sweden; (S.S.R.); (A.O.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (M.O.); (T.X.); (J.G.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Correspondence:
| |
Collapse
|
20
|
Tano H, Oroujeni M, Vorobyeva A, Westerlund K, Liu Y, Xu T, Vasconcelos D, Orlova A, Karlström AE, Tolmachev V. Comparative Evaluation of Novel 177Lu-Labeled PNA Probes for Affibody-Mediated PNA-Based Pretargeting. Cancers (Basel) 2021; 13:cancers13030500. [PMID: 33525578 PMCID: PMC7865858 DOI: 10.3390/cancers13030500] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Affibody molecules are small, engineered affinity proteins based on a nonimmunoglobulin scaffold. Affibody-based radionuclide imaging probes have demonstrated excellent tumor targeting. However, the renal clearance of affibody molecules is accompanied by high reabsorption and retention of activity in the kidney, which prevents their use for radionuclide therapy. We have previously shown the feasibility of overcoming the high renal uptake using a pretargeting approach for affibody-mediated therapy based on peptide nucleic acid (PNA) hybridization. In this study, we test the hypothesis that shortening the PNA pretargeting probes would further increase the difference between the accumulation of radiometals in tumor xenografts and in kidneys. A series of novel PNA probes has been designed and evaluated in vitro and in vivo. We have found that a variant containing 9 nucleobases enables a two-fold increase of the tumor-to-kidney dose ratio compared with a variant containing 15 nucleobases. This creates preconditions for more efficient therapy of cancer. Abstract Affibody-mediated PNA-based pretargeting is a promising approach to radionuclide therapy of HER2-expressing tumors. In this study, we test the hypothesis that shortening the PNA pretargeting probes would increase the tumor-to-kidney dose ratio. The primary probe ZHER2:342-SR-HP15 and the complementary secondary probes HP16, HP17, and HP18, containing 9, 12, and 15 nucleobases, respectively, and carrying a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator were designed, synthesized, characterized in vitro, and labeled with 177Lu. In vitro pretargeting was studied in HER2-expressing SKOV3 and BT474 cell lines. The biodistribution of these novel probes was evaluated in immunodeficient mice bearing SKOV3 xenografts and compared to the previously studied [177Lu]Lu-HP2. Characterization confirmed the formation of high-affinity duplexes between HP15 and the secondary probes, with the affinity correlating with the length of the complementary PNA sequences. All the PNA-based probes were bound specifically to HER2-expressing cells in vitro. In vivo studies demonstrated HER2-specific uptake of all 177Lu-labeled probes in xenografts in a pretargeting setting. The ratio of cumulated radioactivity in the tumor to the radioactivity in kidneys was dependent on the secondary probe’s size and decreased with an increased number of nucleobases. The shortest PNA probe, [177Lu]Lu-HP16, showed the highest tumor-to-kidney ratio. [177Lu]Lu-HP16 is the most promising secondary probe for affibody-mediated tumor pretargeting.
Collapse
Affiliation(s)
- Hanna Tano
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (H.T.); (K.W.); (D.V.)
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Kristina Westerlund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (H.T.); (K.W.); (D.V.)
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
| | - Daniel Vasconcelos
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (H.T.); (K.W.); (D.V.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Department of Medicinal Chemistry, Dag Hammarskjölds väg 14C, Uppsala University, 751 23 Uppsala, Sweden
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (H.T.); (K.W.); (D.V.)
- Correspondence:
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (A.V.); (Y.L.); (T.X.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
| |
Collapse
|
21
|
Tolmachev V, Orlova A, Sörensen J. The emerging role of radionuclide molecular imaging of HER2 expression in breast cancer. Semin Cancer Biol 2021; 72:185-197. [PMID: 33465471 DOI: 10.1016/j.semcancer.2020.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
Targeting of human epidermal growth factor type 2 (HER2) using monoclonal antibodies, antibody-drug conjugates and tyrosine kinase inhibitors extends survival of patients with HER2-expressing metastatic breast cancer. High expression of HER2 is a predictive biomarker for such specific treatment. Accurate determination of HER2 expression level is necessary for stratification of patients to targeted therapy. Non-invasive in vivo radionuclide molecular imaging of HER2 has a potential of repetitive measurements, addressing issues of heterogeneous expression and conversion of HER2 status during disease progression or in response to therapy. Imaging probes based of several classes of targeting proteins are currently in preclinical and early clinical development. Both preclinical and clinical data suggest that the most promising are imaging agents based on small proteins, such as single domain antibodies or engineered scaffold proteins. These agents permit a very specific high-contrast imaging at the day of injection.
Collapse
Affiliation(s)
- Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden; Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia.
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia; Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden; Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jens Sörensen
- Division of Radiology and Nuclear Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Xu T, Ding H, Vorobyeva A, Oroujeni M, Orlova A, Tolmachev V, Gräslund T. Drug Conjugates Based on a Monovalent Affibody Targeting Vector Can Efficiently Eradicate HER2 Positive Human Tumors in an Experimental Mouse Model. Cancers (Basel) 2020; 13:cancers13010085. [PMID: 33396753 PMCID: PMC7794879 DOI: 10.3390/cancers13010085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Drug conjugates, consisting of a tumor targeting part coupled to a highly toxic molecule, are promising for treatment of many different types of cancer. However, for many patients it is not curative, and investigation of alternative or complimentary types of drug conjugates is motivated. Here, we have devised and studied a novel cancer cell-directed drug conjugate ZHER2:2891-ABD-E3-mcDM1. We found that it could induce efficient shrinkage and, in some cases, complete regression of human tumors implanted in mice, and thus holds promise to become a therapeutic agent for clinical use in the future. Abstract The human epidermal growth factor receptor 2 (HER2) is frequently overexpressed in a variety of cancers and therapies targeting HER2 are routinely used in the clinic. Recently, small engineered scaffold proteins, such as affibody molecules, have shown promise as carriers of cytotoxic drugs, and these drug conjugates may become complements or alternatives to the current HER2-targeting therapies. Here, we investigated if a monovalent HER2-binding affibody molecule, ZHER2:2891, fused with a plasma half-life extending albumin binding domain (ABD), may be used as carrier of the cytotoxic maytansine derivate mcDM1. We found that the resulting drug conjugate, ZHER2:2891-ABD-E3-mcDM1, had strong affinity for its cognate molecular targets: HER2 and serum albumin. ZHER2:2891-ABD-E3-mcDM1 displayed potent cytotoxic activity towards cells with high HER2 expression, with IC50 values ranging from 0.6 to 33 nM. In vivo, an unspecific increase in uptake in the liver, imparted by the hydrophobic mcDM1, was counteracted by incorporation of hydrophilic and negatively charged glutamate residues near the site of mcDM1 conjugation. A dose-escalation experiment showed that increasing doses up to 15.1 mg/kg gave a proportional increase in uptake in xenografted HER2-overexpressing SKOV3 tumors, after which the tumors became saturated. Experimental therapy with four once-weekly injection of 10.3 or 15.1 mg/kg led to efficient regression of tumors in all animals and complete regression in some. Weight loss was detected for some animals in the group receiving the highest dose, suggesting that it was close to the maximum tolerated dose. In conclusion, the monovalent HER2-targeting affibody drug conjugate presented herein have potent anti-tumor activity in vivo.
Collapse
Affiliation(s)
- Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (A.V.); (M.O.); (V.T.)
| | - Haozhong Ding
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden;
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (A.V.); (M.O.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (A.V.); (M.O.); (V.T.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (A.V.); (M.O.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, 114 17 Stockholm, Sweden;
- Correspondence: ; Tel.: +46-(0)8-790-96-27
| |
Collapse
|
23
|
Vorobyeva A, Bezverkhniaia E, Konovalova E, Schulga A, Garousi J, Vorontsova O, Abouzayed A, Orlova A, Deyev S, Tolmachev V. Radionuclide Molecular Imaging of EpCAM Expression in Triple-Negative Breast Cancer Using the Scaffold Protein DARPin Ec1. Molecules 2020; 25:molecules25204719. [PMID: 33066684 PMCID: PMC7587533 DOI: 10.3390/molecules25204719] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/04/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Efficient treatment of disseminated triple-negative breast cancer (TNBC) remains an unmet clinical need. The epithelial cell adhesion molecule (EpCAM) is often overexpressed on the surface of TNBC cells, which makes EpCAM a potential therapeutic target. Radionuclide molecular imaging of EpCAM expression might permit selection of patients for EpCAM-targeting therapies. In this study, we evaluated a scaffold protein, designed ankyrin repeat protein (DARPin) Ec1, for imaging of EpCAM in TNBC. DARPin Ec1 was labeled with a non-residualizing [125I]I-para-iodobenzoate (PIB) label and a residualizing [99mTc]Tc(CO)3 label. Both imaging probes retained high binding specificity and affinity to EpCAM-expressing MDA-MB-468 TNBC cells after labeling. Internalization studies showed that Ec1 was retained on the surface of MDA-MB-468 cells to a high degree up to 24 h. Biodistribution in Balb/c nu/nu mice bearing MDA-MB-468 xenografts demonstrated specific uptake of both [125I]I-PIB-Ec1 and [99mTc]Tc(CO)3-Ec1 in TNBC tumors. [125I]I-PIB-Ec1 had appreciably lower uptake in normal organs compared with [99mTc]Tc(CO)3-Ec1, which resulted in significantly (p < 0.05) higher tumor-to-organ ratios. The biodistribution data were confirmed by micro-Single-Photon Emission Computed Tomography/Computed Tomography (microSPECT/CT) imaging. In conclusion, an indirectly radioiodinated Ec1 is the preferable probe for imaging of EpCAM in TNBC.
Collapse
Affiliation(s)
- Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (J.G.); (O.V.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (E.B.); (A.S.); (A.O.); (S.D.)
- Correspondence:
| | - Ekaterina Bezverkhniaia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (E.B.); (A.S.); (A.O.); (S.D.)
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (E.B.); (A.S.); (A.O.); (S.D.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (J.G.); (O.V.); (V.T.)
| | - Olga Vorontsova
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (J.G.); (O.V.); (V.T.)
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (E.B.); (A.S.); (A.O.); (S.D.)
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Sergey Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (E.B.); (A.S.); (A.O.); (S.D.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
- Bio-Nanophotonic Lab, Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University ‘MEPhI’, 115409 Moscow, Russia
- Center of Biomedical Engineering, Sechenov University, 119991 Moscow, Russia
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (J.G.); (O.V.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634 050 Tomsk, Russia; (E.B.); (A.S.); (A.O.); (S.D.)
| |
Collapse
|
24
|
Vorobyeva A, Konovalova E, Xu T, Schulga A, Altai M, Garousi J, Rinne SS, Orlova A, Tolmachev V, Deyev S. Feasibility of Imaging EpCAM Expression in Ovarian Cancer Using Radiolabeled DARPin Ec1. Int J Mol Sci 2020; 21:ijms21093310. [PMID: 32392820 PMCID: PMC7246691 DOI: 10.3390/ijms21093310] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is overexpressed in 55%–75% of ovarian carcinomas (OC). EpCAM might be used as a target for a treatment of disseminated OC. Designed ankyrin repeats protein (DARPin) Ec1 is a small (18 kDa) protein, which binds to EpCAM with subnanomolar affinity. We tested a hypothesis that Ec1 labeled with a non-residualizing label might serve as a companion imaging diagnostic for stratification of patients for EpCAM-targeting therapy. Ec1 was labeled with 125I using N-succinimidyl-para-iodobenzoate. Binding affinity, specificity, and cellular processing of [125I]I-PIB-Ec1 were evaluated using SKOV-3 and OVCAR-3 ovarian carcinoma cell lines. Biodistribution and tumor-targeting properties of [125I]I-PIB-Ec1 were studied in Balb/c nu/nu mice bearing SKOV-3 and OVCAR-3 xenografts. EpCAM-negative Ramos lymphoma xenografts served as specificity control. Binding of [125I]I-PIB-Ec1 to ovarian carcinoma cell lines was highly specific and had affinity in picomolar range. Slow internalization of [125I]I-PIB-Ec1 by OC cells confirmed utility of non-residualizing label for in vivo imaging. [125I]I-PIB-Ec1 provided 6 h after injection tumor-to-blood ratios of 30 ± 11 and 48 ± 12 for OVCAR-3 and SKOV-3 xenografts, respectively, and high contrast to other organs. Tumor targeting was highly specific. Saturation of tumor uptake at a high dose of Ec1 in SKOV-3 model provided a rationale for dose selection in further studies using therapeutic conjugates of Ec1 for targeted therapy. In conclusion, [125I]I-PIB-Ec1 is a promising agent for visualizing EpCAM expression in OC.
Collapse
Affiliation(s)
- Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.A.); (J.G.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.); (A.O.)
- Correspondence: ; Tel.: +46-18-471-3868
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (E.K.); (S.D.)
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.A.); (J.G.); (V.T.)
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.); (A.O.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (E.K.); (S.D.)
| | - Mohamed Altai
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.A.); (J.G.); (V.T.)
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.A.); (J.G.); (V.T.)
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.); (A.O.)
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (M.A.); (J.G.); (V.T.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.); (A.O.)
| | - Sergey Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.); (A.O.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (E.K.); (S.D.)
- Bio-Nanophotonic Lab, Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University ‘MEPhI’, 115409 Moscow, Russia
- Center of Biomedical Engineering, Sechenov University, 119991 Moscow, Russia
| |
Collapse
|
25
|
Rinne SS, Dahlsson Leitao C, Saleh-Nihad Z, Mitran B, Tolmachev V, Ståhl S, Löfblom J, Orlova A. Benefit of Later-Time-Point PET Imaging of HER3 Expression Using Optimized Radiocobalt-Labeled Affibody Molecules. Int J Mol Sci 2020; 21:ijms21061972. [PMID: 32183096 PMCID: PMC7139902 DOI: 10.3390/ijms21061972] [Citation(s) in RCA: 8] [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: 02/05/2020] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
HER3-binding affibody molecules are a promising format for visualization of HER3 expression. Cobalt-55, a positron-emitting isotope, with a half-life of 17.5 h, allows for next-day imaging. We investigated the influence of the charge of the radiocobalt–chelator complex on the biodistribution of anti-HER3 affibody molecule (HE)3-ZHER3 and compared the best radiocobalt-labeled variant with a recently optimized gallium-labeled variant. Affibody conjugates (HE)3-ZHER3-X (X = NOTA, NODAGA, DOTA, DOTAGA) were labeled with [57Co]Co (surrogate for 55Co). Affinity measurements, binding specificity and cellular processing were studied in two HER3-expressing cancer cell lines. Biodistribution was studied 3 and 24 h post-injection (pi) in mice with HER3-expressing BxPC-3 xenografts and compared to [68Ga]Ga-(HE)3-ZHER3-NODAGA. Micro-single-photon emission tomography/computed tomography (microSPECT/CT) and micro-positron emission tomography/computed tomography (microPET/CT) imaging was performed 3 and 24 h pi. Stably labeled conjugates bound to HER3 with subnanomolar affinity. [57Co]Co-(HE)3-ZHER3-DOTA had the best tumor retention and a significantly lower concentration in blood than other conjugates, leading to superior tumor-to-blood and tumor-to-liver ratios 24 h pi. Compared to [68Ga]Ga-(HE)3-ZHER3-NODAGA 3 h pi, [57Co]Co-(HE)3-ZHER3-DOTA provided superior imaging contrast in liver 24 h pi. Concluding, the composition and charge of the [57Co]Co–chelator complex influenced the uptake in tumors and normal tissue. [57Co]Co-(HE)3-ZHER3-DOTA provided the best imaging properties among the cobalt-labeled conjugates. Delayed imaging of HER3 expression with [57Co]Co-(HE)3-ZHER3-DOTA improved imaging contrast compared to early-time-point imaging with [68Ga]Ga-(HE)3-ZHER3-NODAGA.
Collapse
Affiliation(s)
- Sara S Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | - Charles Dahlsson Leitao
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Zahra Saleh-Nihad
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Science for Life Laboratory, Uppsala University, 752 37 Uppsala, Sweden
| |
Collapse
|
26
|
Affibody Molecules as Targeting Vectors for PET Imaging. Cancers (Basel) 2020; 12:cancers12030651. [PMID: 32168760 PMCID: PMC7139392 DOI: 10.3390/cancers12030651] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
Affibody molecules are small (58 amino acids) engineered scaffold proteins that can be selected to bind to a large variety of proteins with a high affinity. Their small size and high affinity make them attractive as targeting vectors for molecular imaging. High-affinity affibody binders have been selected for several cancer-associated molecular targets. Preclinical studies have shown that radiolabeled affibody molecules can provide highly specific and sensitive imaging on the day of injection; however, for a few targets, imaging on the next day further increased the imaging sensitivity. A phase I/II clinical trial showed that 68Ga-labeled affibody molecules permit an accurate and specific measurement of HER2 expression in breast cancer metastases. This paper provides an overview of the factors influencing the biodistribution and targeting properties of affibody molecules and the chemistry of their labeling using positron emitters.
Collapse
|
27
|
Baranowska-Kortylewicz J, Sharp JG, McGuire TR, Joshi S, Coulter DW. Alpha-Particle Therapy for Multifocal Osteosarcoma: A Hypothesis. Cancer Biother Radiopharm 2020; 35:418-424. [PMID: 32073902 DOI: 10.1089/cbr.2019.3112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Osteosarcoma (OST) is the most common bone tumor in children and adolescents with a second peak of incidence in elderly adults usually diagnosed as secondary tumors in Paget's disease or irradiated bone. Subjects with metastatic disease or whose disease relapses after the initial therapy have a poor prognosis. Moreover, multifocal OST contains tumor-initiating cells that are resistant to chemotherapy. The use of aggressive therapies in an attempt to eradicate these cells can have long-term negative consequences in these vulnerable patient populations. 227Th-labeled molecular probes based on ligands to OST-associated receptors such as IGF-1R (insulin-like growth factor receptor 1), HER2 (human epidermal growth factor receptor 2), and PSMA (prostate-specific membrane antigen) are expected to detect and treat osseous and nonosseous sites of multifocal OST. Published reports indicate that 227Th has limited myelotoxicity, can be stably chelated to its carriers and, as it decays at targeted sites, 227Th produces 223Ra that is subsequently incorporated into the areas of increased osteoblastic activity, that is, osseous metastatic lesions. Linear energy transfer of α particles emitted by 227Th and its daughter 223Ra is within the range of the optimum relative biological effectiveness. The radiotoxicity of α particles is virtually independent of the phase in the cell cycle, oxygenation, and the dose rate. For these reasons, even resistant OST cells remain susceptible to killing by high-energy α particles, which can also kill adjacent quiescent OST cells or cells with low expression of targeted receptors. Systemic side effects are minimized by the limited range of these intense radiations. Quantitative single-photon emission computed tomography of 227Th and 223Ra is feasible. Additionally, the availability of radionuclide pairs, for example, 89Zr for positron emission tomography and 227Th for therapy, establish a strong basis for the theranostic use of 227Th in the individualized treatment of multifocal OST.
Collapse
Affiliation(s)
- Janina Baranowska-Kortylewicz
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - John G Sharp
- Department of Genetics Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Shantharam Joshi
- Department of Genetics Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Don W Coulter
- Division of Hematology/Oncology, Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
28
|
Rinne SS, Xu T, Dahlsson Leitao C, Ståhl S, Löfblom J, Orlova A, Tolmachev V, Vorobyeva A. Influence of Residualizing Properties of the Radiolabel on Radionuclide Molecular Imaging of HER3 Using Affibody Molecules. Int J Mol Sci 2020; 21:ijms21041312. [PMID: 32075258 PMCID: PMC7072899 DOI: 10.3390/ijms21041312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023] Open
Abstract
Human epidermal growth factor receptor type 3 (HER3) is an emerging therapeutic target in several malignancies. To select potential responders to HER3-targeted therapy, radionuclide molecular imaging of HER3 expression using affibody molecules could be performed. Due to physiological expression of HER3 in normal organs, high imaging contrast remains challenging. Due to slow internalization of affibody molecules by cancer cells, we hypothesized that labeling (HE)3-ZHER3:08698-DOTAGA affibody molecule with non-residualizing [125I]-N-succinimidyl-4-iodobenzoate (PIB) label would improve the tumor-to-normal organs ratios compared to previously reported residualizing radiometal labels. The [125I]I-PIB-(HE)3-ZHER3:08698-DOTAGA was compared side-by-side with [111In]In-(HE)3-ZHER3:08698-DOTAGA. Both conjugates demonstrated specific high-affinity binding to HER3-expressing BxPC-3 and DU145 cancer cells. Biodistribution in mice bearing BxPC-3 xenografts at 4 and 24 h pi showed faster clearance of the [125I]I-PIB label compared to the indium-111 label from most tissues, except blood. This resulted in higher tumor-to-organ ratios in HER3-expressing organs for [125I]I-PIB-(HE)3-ZHER3:08698-DOTAGA at 4 h, providing the tumor-to-liver ratio of 2.4 ± 0.3. The tumor uptake of both conjugates was specific, however, it was lower for the [125I]I-PIB label. In conclusion, the use of non-residualizing [125I]I-PIB label for HER3-targeting affibody molecule provided higher tumor-to-liver ratio than the indium-111 label, however, further improvement in tumor uptake and retention is needed.
Collapse
Affiliation(s)
- Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden; (S.S.R.); (A.O.)
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (V.T.)
| | - Charles Dahlsson Leitao
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (C.D.L.); (S.S.); (J.L.)
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (C.D.L.); (S.S.); (J.L.)
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (C.D.L.); (S.S.); (J.L.)
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden; (S.S.R.); (A.O.)
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
- Centrum for Oncotheranostics, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (V.T.)
- Centrum for Oncotheranostics, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (V.T.)
- Centrum for Oncotheranostics, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
- Correspondence: ; Tel.: +46-18-471-3868
| |
Collapse
|
29
|
Deyev SM, Vorobyeva A, Schulga A, Abouzayed A, Günther T, Garousi J, Konovalova E, Ding H, Gräslund T, Orlova A, Tolmachev V. Effect of a radiolabel biochemical nature on tumor-targeting properties of EpCAM-binding engineered scaffold protein DARPin Ec1. Int J Biol Macromol 2019; 145:216-225. [PMID: 31863835 DOI: 10.1016/j.ijbiomac.2019.12.147] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/27/2022]
Abstract
Radionuclide-based imaging of molecular therapeutic targets might facilitate stratifying patients for specific biotherapeutics. New type of imaging probes, based on designed ankyrin repeat proteins (DARPins), have demonstrated excellent contrast of imaging of human epidermal growth factor type 2 (HER2) expression in preclinical models. We hypothesized that labeling approaches, which result in lipophilic radiometabolites (non-residualizing labels), would provide the best imaging contrast for DARPins that internalize slowly after binding to cancer cells. The hypothesis was tested using DARPin Ec1 that binds to epithelial cell adhesion molecule (EpCAM). EpCAM is a promising therapeutic target. Ec1 was labeled with 125I using two methods to obtain the non-residualizing labels, while residualizing labels were obtained by labeling it with 99mTc. All labeled Ec1 variants preserved target specificity and picomolar binding affinity to EpCAM-expressing pancreatic adenocarcinoma BxPC-3 cells. In murine models, all the variants provided similar tumor uptake. However, 125I-PIB-H6-Ec1 had noticeably lower retention in normal tissues, which provided appreciably higher tumor-to-organ ratios. Furthermore, 125I-PIB-H6-Ec1 demonstrated the highest imaging contrast in preclinical models than any other EpCAM-imaging agent tested so far. In conclusion, DARPin Ec1 in combination with a non-residualizing label is a promising probe for imaging EpCAM expression a few hours after injection.
Collapse
Affiliation(s)
- Sergey M Deyev
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Research School of Chemistry and Applied Biomedical Sciences, Research Tomsk Polytechnic University, Tomsk, Russia; Center of Biomedical Engineering, Sechenov University, Moscow, Russia
| | - Anzhelika Vorobyeva
- Research School of Chemistry and Applied Biomedical Sciences, Research Tomsk Polytechnic University, Tomsk, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Alexey Schulga
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Research School of Chemistry and Applied Biomedical Sciences, Research Tomsk Polytechnic University, Tomsk, Russia
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Tyran Günther
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Haozhong Ding
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anna Orlova
- Research School of Chemistry and Applied Biomedical Sciences, Research Tomsk Polytechnic University, Tomsk, Russia; Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden; Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Research School of Chemistry and Applied Biomedical Sciences, Research Tomsk Polytechnic University, Tomsk, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
30
|
Garousi J, Huizing FJ, Vorobyeva A, Mitran B, Andersson KG, Leitao CD, Frejd FY, Löfblom J, Bussink J, Orlova A, Heskamp S, Tolmachev V. Comparative evaluation of affibody- and antibody fragments-based CAIX imaging probes in mice bearing renal cell carcinoma xenografts. Sci Rep 2019; 9:14907. [PMID: 31624303 PMCID: PMC6797765 DOI: 10.1038/s41598-019-51445-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Carbonic anhydrase IX (CAIX) is a cancer-associated molecular target for several classes of therapeutics. CAIX is overexpressed in a large fraction of renal cell carcinomas (RCC). Radionuclide molecular imaging of CAIX-expression might offer a non-invasive methodology for stratification of patients with disseminated RCC for CAIX-targeting therapeutics. Radiolabeled monoclonal antibodies and their fragments are actively investigated for imaging of CAIX expression. Promising alternatives are small non-immunoglobulin scaffold proteins, such as affibody molecules. A CAIX-targeting affibody ZCAIX:2 was re-designed with the aim to decrease off-target interactions and increase imaging contrast. The new tracer, DOTA-HE3-ZCAIX:2, was labeled with 111In and characterized in vitro. Tumor-targeting properties of [111In]In-DOTA-HE3-ZCAIX:2 were compared head-to-head with properties of the parental variant, [99mTc]Tc(CO)3-HE3-ZCAIX:2, and the most promising antibody fragment-based tracer, [111In]In-DTPA-G250(Fab’)2, in the same batch of nude mice bearing CAIX-expressing RCC xenografts. Compared to the 99mTc-labeled parental variant, [111In]In-DOTA-HE3-ZCAIX:2 provides significantly higher tumor-to-lung, tumor-to-bone and tumor-to-liver ratios, which is essential for imaging of CAIX expression in the major metastatic sites of RCC. [111In]In-DOTA-HE3-ZCAIX:2 offers significantly higher tumor-to-organ ratios compared with [111In]In-G250(Fab’)2. In conclusion, [111In]In-DOTA-HE3-ZCAIX:2 can be considered as a highly promising tracer for imaging of CAIX expression in RCC metastases based on our results and literature data.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fokko J Huizing
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ken G Andersson
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Charles Dahlsson Leitao
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Sandra Heskamp
- Department of Radiology and Nuclear medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
31
|
Oroujeni M, Abouzayed A, Lundmark F, Mitran B, Orlova A, Tolmachev V, Rosenström U. Evaluation of Tumor-Targeting Properties of an Antagonistic Bombesin Analogue RM26 Conjugated with a Non-Residualizing Radioiodine Label Comparison with a Radiometal-Labelled Counterpart. Pharmaceutics 2019; 11:pharmaceutics11080380. [PMID: 31382362 PMCID: PMC6724035 DOI: 10.3390/pharmaceutics11080380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 12/24/2022] Open
Abstract
Radiolabelled antagonistic bombesin analogues are successfully used for targeting of gastrin-releasing peptide receptors (GRPR) that are overexpressed in prostate cancer. Internalization of antagonistic bombesin analogues is slow. We hypothesized that the use of a non-residualizing radioiodine label might not affect the tumour uptake but would reduce the retention in normal organs, where radiopharmaceutical would be internalized. To test this hypothesis, tyrosine was conjugated via diethylene glycol linker to N-terminus of an antagonistic bombesin analogue RM26 to form Tyr-PEG2-RM26. [111In]In-DOTA-PEG2-RM26 was used as a control with a residualizing label. Tyr-PEG2-RM26 was labelled with 125I with 95% radiochemical purity and retained binding specificity to GRPR. The IC50 values for Tyr-PEG2-RM26 and DOTA-PEG2-RM26 were 1.7 ± 0.3 nM and 3.3 ± 0.5 nM, respectively. The cellular processing of [125I]I-Tyr-PEG2-RM26 by PC-3 cells showed unusually fast internalization. Biodistribution showed that uptake in pancreas and tumour was GRPR-specific for both radioconjugates. Blood clearance of [125I]I-Tyr-PEG2-RM26 was appreciably slower and activity accumulation in all organs was significantly higher than for [111In]In-DOTA-PEG2-RM26. Tumor uptake of [111In]In-DOTA-PEG2-RM26 was significantly higher than for [125I]I-Tyr-PEG2-RM26, resulting in higher tumour-to-organ ratio for [111In]In-DOTA-PEG2-RM26 at studied time points. Incorporation of amino acids with hydrophilic side-chains next to tyrosine might overcome the problems associated with the use of tyrosine as a prosthetic group for radioiodination.
Collapse
Affiliation(s)
- Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Fanny Lundmark
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, SE-750 03 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden.
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
32
|
Indirect Radioiodination of DARPin G3 Using N-succinimidyl- Para-Iodobenzoate Improves the Contrast of HER2 Molecular Imaging. Int J Mol Sci 2019; 20:ijms20123047. [PMID: 31234471 PMCID: PMC6627094 DOI: 10.3390/ijms20123047] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 02/08/2023] Open
Abstract
Radionuclide molecular imaging of human epidermal growth factor receptor 2 (HER2) in breast and gastroesophageal cancer might be used to stratify patients for HER2-targeted therapy as well as monitor treatment response and disease progression. Designed ankyrin repeat proteins (DARPins) are small engineered scaffold proteins with favorable properties for molecular imaging. Herein we compared two methods for labeling the anti-HER2 DARPin (HE)3-G3, direct and indirect radioiodination. We hypothesized that the use of N-succinimidyl-para-iodobenzoate (SPIB) for radioiodination would facilitate the clearance of radiometabolites and improve the contrast of imaging. Both radiolabeled (HE)3-G3 variants preserved their binding specificity and high affinity to HER2-expressing cells. The specificity of tumor targeting in vivo was also demonstrated. A biodistribution comparison of [125I]I-(HE)3-G3 and [125I]I-PIB-(HE)3-G3, in mice bearing HER2 expressing SKOV3 xenografts, showed rapid clearance of [125I]I-PIB-(HE)3-G3 from normal organs and tissues and low accumulation of activity in organs with NaI-symporter expression. Both radiolabeled (HE)3-G3 variants had equal tumor uptake. Consequently, the indirect label provided higher tumor-to-blood and tumor-to-organ ratios compared with the direct label. Comparative Single Photon Emission Computed Tomography (SPECT)/CT imaging of HER2 expression in SKOV3 xenografts, using both radiolabeled DARPins, demonstrated the superior imaging contrast of the indirect label. Indirect radioiodination of (HE)3-G3 using SPIB could be further applied for SPECT and PET imaging with iodine-123 and iodine-124.
Collapse
|
33
|
Site-specific conjugation of recognition tags to trastuzumab for peptide nucleic acid-mediated radionuclide HER2 pretargeting. Biomaterials 2019; 203:73-85. [PMID: 30877838 DOI: 10.1016/j.biomaterials.2019.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 01/11/2023]
Abstract
Pretargeting is a promising strategy to reach high imaging contrast in a shorter time than by targeting with directly radiolabeled monoclonal antibodies (mAbs). One of problems in pretargeting is a site-specific, reproducible and uniform conjugation of recognition tags to mAbs. To solve this issue we propose a photoconjugation to covalently couple a recognition tag to a mAb via a photoactivatable Z domain. The Z-domain, a 58-amino acid protein derived from the IgG-binding B-domain of Staphylococcus aureus protein A, has a well-characterized binding site in the Fc portion of IgG. We tested the feasibility of this approach using pretargeting based on hybridization between peptide nucleic acids (PNAs). We have used photoconjugation to couple trastuzumab with the PNA-based hybridization probe, HP1. A complementary [57Co]Co-labeled PNA hybridization probe ([57Co]Co-HP2) was used as the secondary targeting probe. In vitro studies demonstrated that trastuzumab-ZHP1 bound specifically to human epidermal growth factor receptor 2 (HER2)-expressing cells with nanomolar affinity. The binding of the secondary [57Co]Co-HP2 probe to trastuzumab-PNA-pretreated cells was in the picomolar affinity range. A two-fold increase in SKOV-3 tumor targeting was achieved when [57Co]Co-HP2 (0.7 nmol) was injected 48 h after injection of trastuzumab-ZHP1 (0.5 nmol) compared with trastuzumab-ZHP1 alone (0.8 ± 0.2 vs. 0.33 ± 0.06 %ID/g). Tumor accumulation of [57Co]Co-HP2 was significantly reduced by pre-saturation with trastuzumab or when no trastuzumab-ZHP1 was preinjected. A tumor-to-blood uptake ratio of 1.5 ± 0.3 was achieved resulting in a clear visualization of HER2-expressing xenografts as confirmed by SPECT imaging. In conclusion, the feasibility of stable site-specific coupling of a PNA-based recognition tag to trastuzumab and successful pretargeting has been demonstrated. This approach can hopefully be used for a broad range of mAbs and recognition tags.
Collapse
|
34
|
Deyev S, Vorobyeva A, Schulga A, Proshkina G, Güler R, Löfblom J, Mitran B, Garousi J, Altai M, Buijs J, Chernov V, Orlova A, Tolmachev V. Comparative Evaluation of Two DARPin Variants: Effect of Affinity, Size, and Label on Tumor Targeting Properties. Mol Pharm 2019; 16:995-1008. [PMID: 30608701 DOI: 10.1021/acs.molpharmaceut.8b00922] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Designed ankyrin repeat proteins (DARPins) are small engineered scaffold proteins that can be selected for binding to desirable molecular targets. High affinity and small size of DARPins render them promising probes for radionuclide molecular imaging. However, detailed knowledge on many factors influencing their imaging properties is still lacking. We have evaluated two human epidermal growth factor 2 (HER2)-specific DARPins with different size and binding properties. DARPins 9_29-H6 and G3-H6 were radiolabeled with iodine-125 and tricarbonyl technetium-99m and evaluated in vitro. A side-by-side comparison of biodistribution and tumor targeting was performed. HER2-specific tumor accumulation of G3-H6 was demonstrated. A combination of smaller size and higher affinity resulted in a higher tumor uptake of G3-H6 in comparison to 9_29-H6. Technetium-99m labeled G3-H6 demonstrated a better biodistribution profile than 9_29-H6, with several-fold lower uptake in liver. Radioiodinated G3-H6 showed the best tumor-to-organ ratios. The combined effect of affinity, molecular weight, scaffold composition, and nonresidualizing properties of iodine label provided radioiodinated G3-H6 with high clinical potential for imaging of HER2.
Collapse
Affiliation(s)
- Sergey Deyev
- Molecular Immunology Laboratory , Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russia.,National Research Tomsk Polytechnic University, Tomsk , Russia.,Bio-Nanophotonic Lab., Institute of Engineering Physics for Biomedicine (PhysBio) , National Research Nuclear University "MEPhI" , Moscow , Russia
| | | | - Alexey Schulga
- Molecular Immunology Laboratory , Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russia
| | - Galina Proshkina
- Molecular Immunology Laboratory , Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russia
| | - Rezan Güler
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health , KTH Royal Institute of Technology , Stockholm , Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health , KTH Royal Institute of Technology , Stockholm , Sweden
| | | | | | | | | | - Vladimir Chernov
- Nuclear Medicine Department, Cancer Research Institute , Tomsk National Research Medical Center Russian Academy of Sciences , Tomsk , Russia
| | | | | |
Collapse
|
35
|
Garousi J, Lindbo S, Borin J, von Witting E, Vorobyeva A, Oroujeni M, Mitran B, Orlova A, Buijs J, Tolmachev V, Hober S. Comparative evaluation of dimeric and monomeric forms of ADAPT scaffold protein for targeting of HER2-expressing tumours. Eur J Pharm Biopharm 2018; 134:37-48. [PMID: 30408518 DOI: 10.1016/j.ejpb.2018.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/19/2018] [Accepted: 11/04/2018] [Indexed: 12/23/2022]
Abstract
ADAPTs are small engineered non-immunoglobulin scaffold proteins, which have demonstrated very promising features as vectors for radionuclide tumour targeting. Radionuclide imaging of human epidermal growth factor 2 (HER2) expression in vivo might be used for stratification of patients for HER2-targeting therapies. ADAPT6, which specifically binds to HER2, has earlier been shown to have very promising features for in vivo targeting of HER2 expressing tumours. In this study we tested the hypothesis that dimerization of ADAPT6 would increase the apparent affinity to HER2 and accordingly improve tumour targeting. To find an optimal molecular design of dimers, a series of ADAPT dimers with different linkers, -SSSG- (DiADAPT6L1), -(SSSG)2- (DiADAPT6L2), and -(SSSG)3- (DiADAPT6L3) was evaluated. Dimers in combination with optimal linker lengths demonstrated increased apparent affinity to HER2. The best variants, DiADAPT6L2 and DiADAPT6L3 were site-specifically labelled with 111In and 125I, and compared with a monomeric ADAPT6 in mice bearing HER2-expressing tumours. Despite higher affinity, both dimers had lower tumour uptake and lower tumour-to-organ ratios compared to the monomer. We conclude that improved affinity of a dimeric form of ADAPT does not compensate the disadvantage of increased size. Therefore, increase of affinity should be obtained by affinity maturation and not by dimerization.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Sarah Lindbo
- Department of Protein Technology, KTH - Royal Institute of Technology, SE-10691 Stockholm, Sweden
| | - Jesper Borin
- Department of Protein Technology, KTH - Royal Institute of Technology, SE-10691 Stockholm, Sweden
| | - Emma von Witting
- Department of Protein Technology, KTH - Royal Institute of Technology, SE-10691 Stockholm, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jos Buijs
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden.
| | - Sophia Hober
- Department of Protein Technology, KTH - Royal Institute of Technology, SE-10691 Stockholm, Sweden
| |
Collapse
|
36
|
Influence of Molecular Design on the Targeting Properties of ABD-Fused Mono- and Bi-Valent Anti-HER3 Affibody Therapeutic Constructs. Cells 2018; 7:cells7100164. [PMID: 30314301 PMCID: PMC6210767 DOI: 10.3390/cells7100164] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/23/2018] [Accepted: 10/08/2018] [Indexed: 01/26/2023] Open
Abstract
Overexpression of human epidermal growth factor receptor type 3 (HER3) is associated with tumour cell resistance to HER-targeted therapies. Monoclonal antibodies (mAbs) targeting HER3 are currently being investigated for treatment of various types of cancers. Cumulative evidence suggests that affibody molecules may be appropriate alternatives to mAbs. We previously reported a fusion construct (3A3) containing two HER3-targeting affibody molecules flanking an engineered albumin-binding domain (ABD035) included for the extension of half-life in circulation. The 3A3 fusion protein (19.7 kDa) was shown to delay tumour growth in mice bearing HER3-expressing xenografts and was equipotent to the mAb seribantumab. Here, we have designed and explored a series of novel formats of anti-HER3 affibody molecules fused to the ABD in different orientations. All constructs inhibited heregulin-induced phosphorylation in HER3-expressing BxPC-3 and DU-145 cell lines. Biodistribution studies demonstrated extended the half-life of all ABD-fused constructs, although at different levels. The capacity of our ABD-fused proteins to accumulate in HER3-expressing tumours was demonstrated in nude mice bearing BxPC-3 xenografts. Formats where the ABD was located on the C-terminus of affibody binding domains (3A, 33A, and 3A3) provided the best tumour targeting properties in vivo. Further development of these promising candidates for treatment of HER3-overexpressing tumours is therefore justified.
Collapse
|
37
|
Preclinical Evaluation of [ 68Ga]Ga-DFO-ZEGFR:2377: A Promising Affibody-Based Probe for Noninvasive PET Imaging of EGFR Expression in Tumors. Cells 2018; 7:cells7090141. [PMID: 30231504 PMCID: PMC6162391 DOI: 10.3390/cells7090141] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/13/2018] [Accepted: 09/15/2018] [Indexed: 01/10/2023] Open
Abstract
Radionuclide imaging of epidermal growth factor receptor (EGFR) expression in tumors may stratify patients for EGFR-targeting therapies and predict response or resistance to certain treatments. Affibody molecules, which are nonimmunoglobulin scaffold proteins, have a high potential as probes for molecular imaging. In this study, maleimido derivative of desferrioxamine B (DFO) chelator was site-specifically coupled to the C-terminal cysteine of the anti-EGFR affibody molecule ZEGFR:2377, and the DFO-ZEGFR:2377 conjugate was labeled with the generator-produced positron-emitting radionuclide 68Ga. Stability, specificity of binding to EGFR-expressing cells, and processing of [68Ga]Ga-DFO-ZEGFR:2377 by cancer cells after binding were evaluated in vitro. In vivo studies were performed in nude mice bearing human EGFR-expressing A431 epidermoid cancer xenografts. The biodistribution of [68Ga]Ga-DFO-ZEGFR:2377 was directly compared with the biodistribution of [89Zr]Zr-DFO-ZEGFR:2377. DFO-ZEGFR:2377 was efficiently (isolated yield of 73 ± 3%) and stably labeled with 68Ga. Binding of [68Ga]Ga-DFO-ZEGFR:2377 to EGFR-expressing cells in vitro was receptor-specific and proportional to the EGFR expression level. In vivo saturation experiment demonstrated EGFR-specific accumulation of [68Ga]Ga-DFO-ZEGFR:2377 in A431 xenografts. Compared to [89Zr]Zr-DFO-ZEGFR:2377, [68Ga]Ga-DFO-ZEGFR:2377 demonstrated significantly (p < 0.05) higher uptake in tumors and lower uptake in spleen and bones. This resulted in significantly higher tumor-to-organ ratios for [68Ga]Ga-DFO-ZEGFR:2377. In conclusion, [68Ga]Ga-DFO-ZEGFR:2377 is a promising probe for imaging of EGFR expression.
Collapse
|
38
|
Affibody-derived drug conjugates: Potent cytotoxic molecules for treatment of HER2 over-expressing tumors. J Control Release 2018; 288:84-95. [PMID: 30172673 DOI: 10.1016/j.jconrel.2018.08.040] [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] [Received: 04/17/2018] [Revised: 08/07/2018] [Accepted: 08/29/2018] [Indexed: 01/18/2023]
Abstract
Patients with HER2-positive tumors often suffer resistance to therapy, warranting development of novel treatment modalities. Affibody molecules are small affinity proteins which can be engineered to bind to desired targets. They have in recent years been found to allow precise targeting of cancer specific molecular signatures such as the HER2 receptor. In this study, we have investigated the potential of an affibody molecule targeting HER2, ZHER2:2891, conjugated with the cytotoxic maytansine derivate MC-DM1, for targeted cancer therapy. ZHER2:2891 was expressed as a monomer (ZHER2:2891), dimer ((ZHER2:2891)2) and dimer with an albumin binding domain (ABD) for half-life extension ((ZHER2:2891)2-ABD). All proteins had a unique C-terminal cysteine that could be used for efficient and site-specific conjugation with MC-DM1. The resulting affibody drug conjugates were potent cytotoxic molecules for human cells over-expressing HER2, with sub-nanomolar IC50-values similar to trastuzumab emtansine, and did not affect cells with low HER2 expression. A biodistribution study of a radiolabeled version of (ZHER2:2891)2-ABD-MC-DM1, showed that it was taken up by the tumor. The major site of off-target uptake was the kidneys and to some extent the liver. (ZHER2:2891)2-ABD-MC-DM1 was found to have a half-life in circulation of 14 h. The compound was tolerated well by mice at 8.5 mg/kg and was shown to extend survival of mice bearing HER2 over-expressing tumors. The findings in this study show that affibody molecules are a promising class of engineered affinity proteins to specifically deliver small molecular drugs to cancer cells and that such conjugates are potential candidates for clinical evaluation on HER2-overexpressing cancers.
Collapse
|
39
|
Mitran B, Güler R, Roche FP, Lindström E, Selvaraju RK, Fleetwood F, Rinne SS, Claesson-Welsh L, Tolmachev V, Ståhl S, Orlova A, Löfblom J. Radionuclide imaging of VEGFR2 in glioma vasculature using biparatopic affibody conjugate: proof-of-principle in a murine model. Theranostics 2018; 8:4462-4476. [PMID: 30214632 PMCID: PMC6134937 DOI: 10.7150/thno.24395] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 04/21/2018] [Indexed: 01/09/2023] Open
Abstract
Vascular endothelial growth factor receptor-2 (VEGFR2) is a key mediator of angiogenesis and therefore a promising therapeutic target in malignancies including glioblastoma multiforme (GBM). Molecular imaging of VEGFR2 expression may enable patient stratification for antiangiogenic therapy. The goal of the current study was to evaluate the capacity of the novel anti-VEGFR2 biparatopic affibody conjugate (ZVEGFR2-Bp2) for in vivo visualization of VEGFR2 expression in GBM. Methods: ZVEGFR2-Bp2 coupled to a NODAGA chelator was generated and radiolabeled with indium-111. The VEGFR2-expressing murine endothelial cell line MS1 was used to evaluate in vitro binding specificity and affinity, cellular processing and targeting specificity in mice. Further tumor targeting was studied in vivo in GL261 glioblastoma orthotopic tumors. Experimental imaging was performed. Results: [111In]In-NODAGA-ZVEGFR2-Bp2 bound specifically to VEGFR2 (KD=33±18 pM). VEGFR2-mediated accumulation was observed in liver, spleen and lungs. The tumor-to-organ ratios 2 h post injection for mice bearing MS1 tumors were approximately 11 for blood, 15 for muscles and 78 for brain. Intracranial GL261 glioblastoma was visualized using SPECT/CT. The activity uptake in tumors was significantly higher than in normal brain tissue. The tumor-to-cerebellum ratios after injection of 4 µg [111In]In-NODAGA-ZVEGFR2-Bp2 were significantly higher than the ratios observed for the 40 µg injected dose and for the non-VEGFR2 binding size-matched conjugate, demonstrating target specificity. Microautoradiography of cryosectioned CNS tissue was in good agreement with the SPECT/CT images. Conclusion: The anti-VEGFR2 affibody conjugate [111In]In-NODAGA-ZVEGFR2-Bp2 specifically targeted VEGFR2 in vivo and visualized its expression in a murine GBM orthotopic model. Tumor-to-blood ratios for [111In]In-NODAGA-ZVEGFR2-Bp2 were higher compared to other VEGFR2 imaging probes. [111In]In-NODAGA-ZVEGFR2-Bp2 appears to be a promising probe for in vivo noninvasive visualization of tumor angiogenesis in glioblastoma.
Collapse
|
40
|
Vorobyeva A, Westerlund K, Mitran B, Altai M, Rinne S, Sörensen J, Orlova A, Tolmachev V, Karlström AE. Development of an optimal imaging strategy for selection of patients for affibody-based PNA-mediated radionuclide therapy. Sci Rep 2018; 8:9643. [PMID: 29942011 PMCID: PMC6018533 DOI: 10.1038/s41598-018-27886-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/08/2018] [Indexed: 01/03/2023] Open
Abstract
Affibody molecules are engineered scaffold proteins, which demonstrated excellent binding to selected tumor-associated molecular abnormalities in vivo and highly sensitive and specific radionuclide imaging of Her2-expressing tumors in clinics. Recently, we have shown that peptide nucleic acid (PNA)-mediated affibody-based pretargeted radionuclide therapy using beta-emitting radionuclide 177Lu extended significantly survival of mice bearing human Her2-expressing tumor xenografts. In this study, we evaluated two approaches to use positron emission tomography (PET) for stratification of patients for affibody-based pretargeting therapy. The primary targeting probe ZHER2:342-SR-HP1 and the secondary probe HP2 (both conjugated with DOTA chelator) were labeled with the positron-emitting radionuclide 68Ga. Biodistribution of both probes was measured in BALB/C nu/nu mice bearing either SKOV-3 xenografts with high Her2 expression or DU-145 xenografts with low Her2 expression. 68Ga-HP2 was evaluated in the pretargeting setting. Tumor uptake of both probes was compared with the uptake of pretargeted 177Lu-HP2. The uptake of both 68Ga-ZHER2:342-SR-HP1 and 68Ga-HP2 depended on Her2-expression level providing clear discrimination of between tumors with high and low Her2 expression. Tumor uptake of 68Ga-HP2 correlated better with the uptake of 177Lu-HP2 than the uptake of 68Ga-ZHER2:342-SR-HP1. The use of 68Ga-HP2 as a theranostics counterpart would be preferable approach for clinical translation.
Collapse
Affiliation(s)
- Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina Westerlund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Mohamed Altai
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sara Rinne
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jens Sörensen
- Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Medical Imaging Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
41
|
Comparative Evaluation of Radioiodine and Technetium-Labeled DARPin 9_29 for Radionuclide Molecular Imaging of HER2 Expression in Malignant Tumors. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:6930425. [PMID: 29977173 PMCID: PMC6011117 DOI: 10.1155/2018/6930425] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/14/2018] [Accepted: 04/22/2018] [Indexed: 02/08/2023]
Abstract
High expression of human epidermal growth factor receptor 2 (HER2) in breast and gastroesophageal carcinomas is a predictive biomarker for treatment using HER2-targeted therapeutics (antibodies trastuzumab and pertuzumab, antibody-drug conjugate trastuzumab DM1, and tyrosine kinase inhibitor lapatinib). Radionuclide molecular imaging of HER2 expression might permit stratification of patients for HER2-targeting therapies. In this study, we evaluated a new HER2-imaging probe based on the designed ankyrin repeat protein (DARPin) 9_29. DARPin 9_29 was labeled with iodine-125 by direct radioiodination and with [99mTc]Tc(CO)3 using the C-terminal hexahistidine tag. DARPin 9_29 preserved high specificity and affinity of binding to HER2-expressing cells after labeling. Uptake of [125I]I-DARPin 9_29 and [99mTc]Tc(CO)3-DARPin 9_29 in HER2-positive SKOV-3 xenografts in mice at 6 h after injection was 3.4 ± 0.7 %ID/g and 2.9 ± 0.7 %ID/g, respectively. This was significantly (p < 0.00005) higher than the uptake of the same probes in HER2-negative Ramos lymphoma xenografts, 0.22 ± 0.09 %ID/g and 0.30 ± 0.05 %ID/g, respectively. Retention of [125I]I-DARPin 9_29 in the lung, liver, spleen, and kidneys was appreciably lower compared with [99mTc]Tc(CO)3-DARPin 9_29, which resulted in significantly (p < 0.05) higher tumor-to-organ ratios. The biodistribution data were confirmed by SPECT/CT imaging. In conclusion, radioiodine is a preferable label for DARPin 9_29.
Collapse
|
42
|
Tolmachev V, Grönroos TJ, Yim CB, Garousi J, Yue Y, Grimm S, Rajander J, Perols A, Haaparanta-Solin M, Solin O, Ferdani R, Orlova A, Anderson CJ, Karlström AE. Molecular design of radiocopper-labelled Affibody molecules. Sci Rep 2018; 8:6542. [PMID: 29695813 PMCID: PMC5916907 DOI: 10.1038/s41598-018-24785-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/10/2018] [Indexed: 12/18/2022] Open
Abstract
The use of long-lived positron emitters 64Cu or 61Cu for labelling of Affibody molecules may improve breast cancer patients’ stratification for HER-targeted therapy. Previous animal studies have shown that the use of triaza chelators for 64Cu labelling of synthetic Affibody molecules is suboptimal. In this study, we tested a hypothesis that the use of cross-bridged chelator, CB-TE2A, in combination with Gly-Glu-Glu-Glu spacer for labelling of Affibody molecules with radiocopper would improve imaging contrast. CB-TE2A was coupled to the N-terminus of synthetic Affibody molecules extended either with a glycine (designation CB-TE2A-G-ZHER2:342) or Gly-Glu-Glu-Glu spacer (CB-TE2A-GEEE-ZHER2:342). Biodistribution and targeting properties of 64Cu-CB-TE2A-G-ZHER2:342 and 64Cu-CB-TE2A-GEEE-ZHER2:342 were compared in tumor-bearing mice with the properties of 64Cu-NODAGA-ZHER2:S1, which had the best targeting properties in the previous study. 64Cu-CB-TE2A-GEEE-ZHER2:342 provided appreciably lower uptake in normal tissues and higher tumor-to-organ ratios than 64Cu-CB-TE2A-G-ZHER2:342 and 64Cu-NODAGA-ZHER2:S1. The most pronounced was a several-fold difference in the hepatic uptake. At the optimal time point, 6 h after injection, the tumor uptake of 64Cu-CB-TE2A-GEEE-ZHER2:342 was 16 ± 6%ID/g and tumor-to-blood ratio was 181 ± 52. In conclusion, a combination of the cross-bridged CB-TE2A chelator and Gly-Glu-Glu-Glu spacer is preferable for radiocopper labelling of Affibody molecules and, possibly, other scaffold proteins having high renal re-absorption.
Collapse
Affiliation(s)
- Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Tove J Grönroos
- Turku PET Centre, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | - Cheng-Bin Yim
- Turku PET Centre, University of Turku, Turku, Finland.,Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ying Yue
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Sebastian Grimm
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Johan Rajander
- Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Anna Perols
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Merja Haaparanta-Solin
- Turku PET Centre, University of Turku, Turku, Finland.,Department of Chemistry, University of Turku, Turku, Finland
| | - Olof Solin
- Turku PET Centre, University of Turku, Turku, Finland.,Turku PET Centre, Åbo Akademi University, Turku, Finland.,Department of Chemistry, University of Turku, Turku, Finland
| | | | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Carolyn J Anderson
- Departments of Medicine, Radiology, Bioengineering and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15203, USA
| | | |
Collapse
|
43
|
Garousi J, Lindbo S, Mitran B, Buijs J, Vorobyeva A, Orlova A, Tolmachev V, Hober S. Comparative evaluation of tumor targeting using the anti-HER2 ADAPT scaffold protein labeled at the C-terminus with indium-111 or technetium-99m. Sci Rep 2017; 7:14780. [PMID: 29116215 PMCID: PMC5676751 DOI: 10.1038/s41598-017-15366-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/20/2017] [Indexed: 12/29/2022] Open
Abstract
ABD-Derived Affinity Proteins (ADAPTs) is a novel class of engineered scaffold proteins derived from an albumin-binding domain of protein G. The use of ADAPT6 derivatives as targeting moiety have provided excellent preclinical radionuclide imaging of human epidermal growth factor 2 (HER2) tumor xenografts. Previous studies have demonstrated that selection of nuclide and chelator for its conjugation has an appreciable effect on imaging properties of scaffold proteins. In this study we performed a comparative evaluation of the anti-HER2 ADAPT having an aspartate-glutamate-alanine-valine-aspartate-alanine-asparagine-serine (DEAVDANS) N-terminal sequence and labeled at C-terminus with 99mTc using a cysteine-containing peptide based chelator, glycine-serine-serine-cysteine (GSSC), and a similar variant labeled with 111In using a maleimido derivative of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator. Both 99mTc-DEAVDANS-ADAPT6-GSSC and 111In-DEAVDANS-ADAPT6-GSSC-DOTA accumulated specifically in HER2-expressing SKOV3 xenografts. The tumor uptake of both variants did not differ significantly and average values were in the range of 19–21%ID/g. However, there was an appreciable variation in uptake of conjugates in normal tissues that resulted in a notable difference in the tumor-to-organ ratios. The 111In-DOTA label provided 2–6 fold higher tumor-to-organ ratios than 99mTc-GSSC and is therefore the preferable label for ADAPTs.
Collapse
Affiliation(s)
- Javad Garousi
- Institute for Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sarah Lindbo
- School of Biotechnology, Division of Protein Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jos Buijs
- Institute for Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anzhelika Vorobyeva
- Institute for Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Institute for Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Sophia Hober
- School of Biotechnology, Division of Protein Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
44
|
Rosestedt M, Andersson KG, Mitran B, Rinne SS, Tolmachev V, Löfblom J, Orlova A, Ståhl S. Evaluation of a radiocobalt-labelled affibody molecule for imaging of human epidermal growth factor receptor 3 expression. Int J Oncol 2017; 51:1765-1774. [PMID: 29039474 DOI: 10.3892/ijo.2017.4152] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/25/2017] [Indexed: 11/06/2022] Open
Abstract
The human epidermal growth factor receptor 3 (HER3) is involved in the development of cancer resistance towards tyrosine kinase-targeted therapies. Several HER3‑targeting therapeutics are currently under clinical evaluation. Non-invasive imaging of HER3 expression could improve patient management. Affibody molecules are small engineered scaffold proteins demonstrating superior properties as targeting probes for molecular imaging compared with monoclonal antibodies. Feasibility of in vivo HER3 imaging using affibody molecules has been previously demonstrated. Preclinical studies have shown that the contrast when imaging using anti-HER3 affibody molecules can be improved over time. We aim to develop an agent for PET imaging of HER3 expression using the long-lived positron-emitting radionuclide cobalt-55 (55Co) (T1/2=17.5 h). A long-lived cobalt isotope 57Co was used as a surrogate for 55Co in this study. The anti-HER3 affibody molecule HEHEHE-ZHER3-NOTA was labelled with radiocobalt with high yield, purity and stability. Biodistribution of 57Co-HEHEHE-ZHER3-NOTA was measured in mice bearing DU145 (prostate carcinoma) and LS174T (colorectal carcinoma) xenografts at 3 and 24 h post injection (p.i.). Tumour-to-blood ratios significantly increased between 3 and 24 h p.i. (p<0.05). At 24 h p.i., tumour-to-blood ratios were 6 for DU145 and 8 for LS174T xenografts, respectively. HER3‑expressing xenografts were clearly visualized in a preclinical imaging setting already 3 h p.i., and contrast further improved at 24 h p.i. In conclusion, the radiocobalt-labelled anti-HER3 affibody molecule, HEHEHE-ZHER3-NOTA, is a promising tracer for imaging of HER3 expression in tumours.
Collapse
Affiliation(s)
- Maria Rosestedt
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | - Ken G Andersson
- Division of Protein Technology, KTH - Royal Institute of Technology, SE-106 91 Stockholm, Sweden
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | - Sara S Rinne
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 83 Uppsala, Sweden
| | - John Löfblom
- Division of Protein Technology, KTH - Royal Institute of Technology, SE-106 91 Stockholm, Sweden
| | - Anna Orlova
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | - Stefan Ståhl
- Division of Protein Technology, KTH - Royal Institute of Technology, SE-106 91 Stockholm, Sweden
| |
Collapse
|
45
|
Lindbo S, Garousi J, Mitran B, Altai M, Buijs J, Orlova A, Hober S, Tolmachev V. Radionuclide Tumor Targeting Using ADAPT Scaffold Proteins: Aspects of Label Positioning and Residualizing Properties of the Label. J Nucl Med 2017; 59:93-99. [PMID: 28864631 DOI: 10.2967/jnumed.117.197202] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/10/2017] [Indexed: 12/13/2022] Open
Abstract
Visualization of cancer-associated alterations of molecular phenotype using radionuclide imaging is a noninvasive approach to stratifying patients for targeted therapies. The engineered albumin-binding domain-derived affinity protein (ADAPT) is a promising tracer for radionuclide molecular imaging because of its small size (6.5 kDa), which satisfies the precondition for efficient tumor penetration and rapid clearance. Previous studies demonstrated that the human epidermal growth factor receptor type 2 (HER2)-targeting ADAPT6 labeled with radiometals at the N terminus is able to image HER2 expression in xenografts a few hours after injection. The aim of this study was to evaluate whether the use of a nonresidualizing label or placement of the labels at the C terminus would further improve the targeting properties of ADAPT6. Methods: Two constructs, Cys2-ADAPT6 and Cys59-ADAPT6, having the (HE)3DANS sequence at the N terminus were produced and site-specifically labeled using 111In-DOTA or 125I-iodo-((4-hydroxyphenyl)ethyl) maleimide (HPEM). The conjugates were compared in vitro and in vivo. HER2-targeting properties and biodistribution were evaluated in BALB/C nu/nu mice bearing ovarian carcinoma cell (SKOV-3) xenografts. Results: Specific HER2 binding and high affinity were preserved after labeling. Both Cys2-ADAPT6 and Cys59-ADAPT6 were internalized slowly by HER2-expressing cancer cells. Depending on the label position, uptake at 4 h after injection varied from 10% to 22% of the injected dose per gram of tumor tissue. Regardless of terminus position, the 125I-HPEM label provided more than 140-fold lower renal uptake than the 111In-DOTA label at 4 after injection. The tumor-to-organ ratios were, in contrast, higher for both of the 111In-DOTA-labeled ADAPT variants in other organs. Tumor-to-blood ratios for 111In-labeled Cys2-ADAPT6 and Cys59-ADAPT6 did not differ significantly (250-280), but 111In-DOTA-Cys59-ADAPT6 provided significantly higher tumor-to-lung, tumor-to-liver, tumor-to-spleen, and tumor-to-muscle ratios. Radioiodinated variants had similar tumor-to-organ ratios, but 125I-HPEM-Cys59-ADAPT6 had significantly higher tumor uptake and a higher tumor-to-kidney ratio. Conclusion: Residualizing properties of the label strongly influence the targeting properties of ADAPT6. The position of the radiolabel influences targeting as well, although to a lesser extent. Placement of a label at the C terminus yields the best biodistribution features for both radiometal and radiohalogen labels. Low renal retention of the radioiodine label creates a precondition for radionuclide therapy using 131I-labeled HPEM-Cys59-ADAPT6.
Collapse
Affiliation(s)
- Sarah Lindbo
- School of Biotechnology, Division of Protein Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Javad Garousi
- Institute for Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden; and
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Mohamed Altai
- Institute for Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden; and
| | - Jos Buijs
- Institute for Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden; and
| | - Anna Orlova
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Sophia Hober
- School of Biotechnology, Division of Protein Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Vladimir Tolmachev
- Institute for Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden; and
| |
Collapse
|
46
|
Garousi J, Andersson KG, Dam JH, Olsen BB, Mitran B, Orlova A, Buijs J, Ståhl S, Löfblom J, Thisgaard H, Tolmachev V. The use of radiocobalt as a label improves imaging of EGFR using DOTA-conjugated Affibody molecule. Sci Rep 2017; 7:5961. [PMID: 28729680 PMCID: PMC5519605 DOI: 10.1038/s41598-017-05700-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/01/2017] [Indexed: 02/04/2023] Open
Abstract
Several anti-cancer therapies target the epidermal growth factor receptor (EGFR). Radionuclide imaging of EGFR expression in tumours may aid in selection of optimal cancer therapy. The 111In-labelled DOTA-conjugated ZEGFR:2377 Affibody molecule was successfully used for imaging of EGFR-expressing xenografts in mice. An optimal combination of radionuclide, chelator and targeting protein may further improve the contrast of radionuclide imaging. The aim of this study was to evaluate the targeting properties of radiocobalt-labelled DOTA-ZEGFR:2377. DOTA-ZEGFR:2377 was labelled with 57Co (T1/2 = 271.8 d), 55Co (T1/2 = 17.5 h), and, for comparison, with the positron-emitting radionuclide 68Ga (T1/2 = 67.6 min) with preserved specificity of binding to EGFR-expressing A431 cells. The long-lived cobalt radioisotope 57Co was used in animal studies. Both 57Co-DOTA-ZEGFR:2377 and 68Ga-DOTA-ZEGFR:2377 demonstrated EGFR-specific accumulation in A431 xenografts and EGFR-expressing tissues in mice. Tumour-to-organ ratios for the radiocobalt-labelled DOTA-ZEGFR:2377 were significantly higher than for the gallium-labelled counterpart already at 3 h after injection. Importantly, 57Co-DOTA-ZEGFR:2377 demonstrated a tumour-to-liver ratio of 3, which is 7-fold higher than the tumour-to-liver ratio for 68Ga-DOTA-ZEGFR:2377. The results of this study suggest that the positron-emitting cobalt isotope 55Co would be an optimal label for DOTA-ZEGFR:2377 and further development should concentrate on this radionuclide as a label.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ken G Andersson
- Department of Protein Technology, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Johan H Dam
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Birgitte B Olsen
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jos Buijs
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stefan Ståhl
- Department of Protein Technology, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Technology, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
47
|
Altai M, Westerlund K, Velletta J, Mitran B, Honarvar H, Karlström AE. Evaluation of affibody molecule-based PNA-mediated radionuclide pretargeting: Development of an optimized conjugation protocol and 177Lu labeling. Nucl Med Biol 2017; 54:1-9. [PMID: 28810153 DOI: 10.1016/j.nucmedbio.2017.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/30/2017] [Accepted: 07/04/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION We have previously developed a pretargeting approach for affibody-mediated cancer therapy based on PNA-PNA hybridization. In this article we have further developed this approach by optimizing the production of the primary agent, ZHER2:342-SR-HP1, and labeling the secondary agent, HP2, with the therapeutic radionuclide 177Lu. We also studied the biodistribution profile of 177Lu-HP2 in mice, and evaluated pretargeting with 177Lu-HP2 in vitro and in vivo. METHODS The biodistribution profile of 177Lu-HP2 was evaluated in NMRI mice and compared to the previously studied 111In-HP2. Pretargeting using 177Lu-HP2 was studied in vitro using the HER2-expressing cell lines BT-474 and SKOV-3, and in vivo in mice bearing SKOV-3 xenografts. RESULTS AND CONCLUSION Using an optimized production protocol for ZHER2:342-SR-HP1 the ligation time was reduced from 15h to 30min, and the yield increased from 45% to 70%. 177Lu-labeled HP2 binds specifically in vitro to BT474 and SKOV-3 cells pre-treated with ZHER2:342-SR-HP1. 177Lu-HP2 was shown to have a more rapid blood clearance compared to 111In-HP2 in NMRI mice, and the measured radioactivity in blood was 0.22±0.1 and 0.68±0.07%ID/g for 177Lu- and 111In-HP2, respectively, at 1h p.i. In contrast, no significant difference in kidney uptake was observed (4.47±1.17 and 3.94±0.58%ID/g for 177Lu- and 111In-HP2, respectively, at 1h p.i.). Co-injection with either Gelofusine or lysine significantly reduced the kidney uptake for 177Lu-HP2 (1.0±0.1 and 1.6±0.2, respectively, vs. 2.97±0.87%ID/g in controls at 4h p.i.). 177Lu-HP2 accumulated in SKOV-3 xenografts in BALB/C nu/nu mice when administered after injection of ZHER2:342-SR-HP1. Without pre-injection of ZHER2:342-SR-HP1, the uptake of 177Lu-HP2 was about 90-fold lower in tumor (0.23±0.08 vs. 20.7±3.5%ID/g). The tumor-to-kidney radioactivity accumulation ratio was almost 5-fold higher in the group of mice pre-injected with ZHER2:342-SR-HP1. In conclusion, 177Lu-HP2 was shown to be a promising secondary agent for affibody-mediated tumor pretargeting in vivo.
Collapse
Affiliation(s)
- Mohamed Altai
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina Westerlund
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Justin Velletta
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Hadis Honarvar
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Amelie Eriksson Karlström
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
48
|
99mTc-radiolabeled GE11-modified peptide for ovarian tumor targeting. ACTA ACUST UNITED AC 2017; 25:13. [PMID: 28464952 PMCID: PMC5414288 DOI: 10.1186/s40199-017-0179-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 04/25/2017] [Indexed: 01/01/2023]
Abstract
Background Ovarian cancer is a serious threat for women health and the early diagnosis of this cancer might improves the survival rate of patients. The use of the targeted radiopharmaceuticals could be a non-invasive and logical method for tumor imaging. The aim of this study was to radiolabel GE11 peptide as a new specific probe for imaging of ovarian tumor. Methods HYNIC-SSS-GE11 peptide was labeled with 99mTc using tricine as a coligand. The 99mTc-tricine-HYNIC-SSS-GE11 peptide was evaluated for specific cellular binding in three cell lines with different levels of EGFR expression. Tumor targeting was assessed in SKOV3 tumor bearing mice. Results By using tricine as a coligand, labeling yield was more than 98% and the stability of the radiolabelled peptide in human serum up to 4 h was 96%. The in vitro cell uptake test showed that this radiolabeled peptide had a good affinity to SKOV3 cells with dissociation constant of 73 nM. The in vivo results showed a tumor/muscle ratio of 3.2 at 4 h following injection of 99mTc-tricine-HYNIC-SSS-GE11 peptide. Conclusions Results of this study showed that 99mTc-tricine-HYNIC-SSS-GE11 peptide could be a promising tool for diagnosis and staging of ovarian cancer. Graphical Abstract 99mTc-tricine-HYNIC-SSS-GE11, a novl targeted agent for ovarian tumor imaging![]()
Collapse
|
49
|
Comparative Evaluation of Anti-HER2 Affibody Molecules Labeled with 64Cu Using NOTA and NODAGA. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:8565802. [PMID: 29097939 PMCID: PMC5612711 DOI: 10.1155/2017/8565802] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/08/2017] [Indexed: 01/04/2023]
Abstract
Imaging using affibody molecules enables discrimination between breast cancer metastases with high and low expression of HER2, making appropriate therapy selection possible. This study aimed to evaluate if the longer half-life of 64Cu (T1/2 = 12.7 h) would make 64Cu a superior nuclide compared to 68Ga for PET imaging of HER2 expression using affibody molecules. The synthetic ZHER2:S1 affibody molecule was conjugated with the chelators NOTA or NODAGA and labeled with 64Cu. The tumor-targeting properties of 64Cu-NOTA-ZHER2:S1 and 64Cu-NODAGA-ZHER2:S1 were evaluated and compared with the targeting properties of 68Ga-NODAGA-ZHER2:S1 in mice. Both 64Cu-NOTA-ZHER2:S1 and 64Cu-NODAGA-ZHER2:S1 demonstrated specific targeting of HER2-expressing xenografts. At 2 h after injection of 64Cu-NOTA-ZHER2:S1, 64Cu-NODAGA-ZHER2:S1, and 68Ga-NODAGA-ZHER2:S1, tumor uptakes did not differ significantly. Renal uptake of 64Cu-labeled conjugates was dramatically reduced at 6 and 24 h after injection. Notably, radioactivity uptake concomitantly increased in blood, lung, liver, spleen, and intestines, which resulted in decreased tumor-to-organ ratios compared to 2 h postinjection. Organ uptake was lower for 64Cu-NODAGA-ZHER2:S1. The most probable explanation for this biodistribution pattern was the release and redistribution of renal radiometabolites. In conclusion, monoamide derivatives of NOTA and NODAGA may be suboptimal chelators for radiocopper labeling of anti-HER2 affibody molecules and, possibly, other scaffold proteins with high renal uptake.
Collapse
|
50
|
In vivo evaluation of a novel format of a bivalent HER3-targeting and albumin-binding therapeutic affibody construct. Sci Rep 2017; 7:43118. [PMID: 28230065 PMCID: PMC5322329 DOI: 10.1038/srep43118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/19/2017] [Indexed: 01/21/2023] Open
Abstract
Overexpression of human epidermal growth factor receptor 3 (HER3) is involved in resistance to several therapies for malignant tumours. Currently, several anti-HER3 monoclonal antibodies are under clinical development. We introduce an alternative approach to HER3-targeted therapy based on engineered scaffold proteins, i.e. affibody molecules. We designed a small construct (22.5 kDa, denoted 3A3), consisting of two high-affinity anti-HER3 affibody molecules flanking an albumin-binding domain ABD, which was introduced for prolonged residence in circulation. In vitro, 3A3 efficiently inhibited growth of HER3-expressing BxPC-3 cells. Biodistribution in mice was measured using 3A3 that was site-specifically labelled with 111In via a DOTA chelator. The residence time of 111In-DOTA-3A3 in blood was extended when compared with the monomeric affibody molecule. 111In-DOTA-3A3 accumulated specifically in HER3-expressing BxPC-3 xenografts in mice. However, 111In-DOTA-3A3 cleared more rapidly from blood than a size-matched control construct 111In-DOTA-TAT, most likely due to sequestering of 3A3 by mErbB3, the murine counterpart of HER3. Repeated dosing and increase of injected protein dose decreased uptake of 111In-DOTA-3A3 in mErbB3-expressing tissues. Encouragingly, growth of BxPC-3 xenografts in mice was delayed in an experimental (pilot-scale) therapy study using 3A3. We conclude that the 3A3 affibody format seems promising for treatment of HER3-overexpressing tumours.
Collapse
|