1
|
Zhang L, Zhang H. Recent advances of affibody molecules in biomedical applications. Bioorg Med Chem 2024; 113:117923. [PMID: 39278106 DOI: 10.1016/j.bmc.2024.117923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Affibody molecules are 58-amino-acid peptides with a molecular weight of about 6.5 kDa, derived from the Z domain of Staphylococcal Protein A. Since they have been used as substitutes for antibodies in biomedicine, several therapeutic affibody molecules have been developed for clinical use. Additionally, affibody molecules have been designed for a range of different applications. This review focuses on the progress made in the last five years in the field of affibody molecules and their potential uses in medical imaging, especially in oncology and cancer treatment. It covers areas such as molecular imaging, targeted delivery of toxic drugs, and their use in combination with nanoparticles. We also highlight some current biomedical applications where affibody molecules are commonly used as a "guide." Due to their many advantages, affibody molecules offer significant potential for applications in both biochemical and medical fields.
Collapse
Affiliation(s)
- Liuyanlin Zhang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, MOE Key Laboratory of Molecular Biophysics, Wuhan 430074, China
| | - Houjin Zhang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, MOE Key Laboratory of Molecular Biophysics, Wuhan 430074, 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
|
Nagy Á, Abouzayed A, Kanellopoulos P, Landmark F, Bezverkhniaia E, Tolmachev V, Orlova A, Eriksson Karlström A. Evaluation of ABD-Linked RM26 Conjugates for GRPR-Targeted Drug Delivery. ACS OMEGA 2024; 9:36122-36133. [PMID: 39220525 PMCID: PMC11359615 DOI: 10.1021/acsomega.4c00489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/10/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Targeting the gastrin-releasing peptide receptor (GRPR) with the bombesin analogue RM26, a 9 aa peptide, has been a promising strategy for cancer theranostics, with recent success in radionuclide imaging of prostate cancer. However, therapeutic application of the short peptide RM26 would require a longer half-life to prevent fast clearance from the circulation. Conjugation to an albumin-binding domain (ABD) is a viable strategy to extend the in vivo half-life of peptides and proteins. We previously reported an ABD-fused RM26 peptide targeting GRPR (ABD-RM26 Gen 1) that showed prolonged and stable tumor uptake over 144 h; however, the observed high kidney uptake indicated that the conjugate's binding to albumin was reduced and that this could be an obstacle for its use as a delivery system for targeted therapy, especially for radiotherapy. Here, we have designed, produced, and preclinically evaluated a series of novel ABD-RM26 conjugates with the aim of improving the conjugate's binding to albumin and decreasing the kidney uptake. We developed three second-generation constructs with varying formats, differing in the relative positions of the targeting moieties and the radionuclide chelator. The produced conjugates were radiolabeled with indium-111 and evaluated in vitro and in vivo. All constructs displayed improved biophysical characteristics, biodistribution, and lower kidney uptake compared to previously reported first-generation molecules. The ABD-RM26 Gen 2A conjugate showed the best biodistribution profile with a nearly 6-fold reduction in kidney uptake. However, the ABD-RM26 Gen 2A conjugate's binding to GRPR was compromised. This conjugate's assembly of albumin- and GRPR-binding moieties might be used for further development of drug conjugates for targeted therapy/radiotherapy of GRPR-expressing cancers.
Collapse
Affiliation(s)
- Ábel Nagy
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| | - Ayman Abouzayed
- Department
of Medicinal Chemistry, Uppsala University, 752 37 Uppsala, Sweden
| | | | - Fredrika Landmark
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| | - Ekaterina Bezverkhniaia
- Department
of Medicinal Chemistry, Uppsala University, 752 37 Uppsala, Sweden
- Research
Centrum for Oncotheranostics, Research School of Chemistry and Applied
Biomedical Sciences, Tomsk Polytechnic University, 634009 Tomsk, Russia
| | - Vladimir Tolmachev
- Department
of Immunology, Genetics and Pathology, Uppsala
University, 752 37 Uppsala, Sweden
| | - Anna Orlova
- Department
of Medicinal Chemistry, Uppsala University, 752 37 Uppsala, Sweden
- Science for
Life Laboratory, Uppsala University, 752 37 Uppsala, Sweden
| | - Amelie Eriksson Karlström
- Department
of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology
and Health, KTH Royal Institute of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| |
Collapse
|
4
|
Zhang J, Bodenko V, Larkina M, Bezverkhniaia E, Xu T, Liao Y, Abouzayed A, Plotnikov E, Tretyakova M, Yuldasheva F, Belousov MV, Orlova A, Tolmachev V, Gräslund T, Vorobyeva A. Half-life extension via ABD-fusion leads to higher tumor uptake of an affibody-drug conjugate compared to PAS- and XTENylation. J Control Release 2024; 370:468-478. [PMID: 38697314 DOI: 10.1016/j.jconrel.2024.04.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
A critical parameter during the development of protein therapeutics is to endow them with suitable pharmacokinetic and pharmacodynamic properties. Small protein drugs are quickly eliminated by kidney filtration, and in vivo half-life extension is therefore often desired. Here, different half-life extension technologies were studied where PAS polypeptides (PAS300, PAS600), XTEN polypeptides (XTEN288, XTEN576), and an albumin binding domain (ABD) were compared for half-life extension of an anti-human epidermal growth factor receptor 2 (HER2) affibody-drug conjugate. The results showed that extension with the PAS or XTEN polypeptides or the addition of the ABD lowered the affinity for HER2 to some extent but did not negatively affect the cytotoxic potential. The half-lives in mice ranged from 7.3 h for the construct including PAS300 to 11.6 h for the construct including PAS600. The highest absolute tumor uptake was found for the construct including the ABD, which was 60 to 160% higher than the PASylated or XTENylated constructs, even though it did not have the longest half-life (9.0 h). A comparison of the tumor-to-normal-organ ratios showed the best overall performance of the ABD-fused construct. In conclusion, PASylation, XTENylation, and the addition of an ABD are viable strategies for half-life extension of affibody-drug conjugates, with the best performance observed for the construct including the ABD.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, Stockholm 114 17, Sweden
| | - Vitalina Bodenko
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Scientific and Educational Laboratory of Chemical and Pharmaceutical Research, Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Maria Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Ekaterina Bezverkhniaia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Tianqi Xu
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Yunqi Liao
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Evgenii Plotnikov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Maria Tretyakova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Feruza Yuldasheva
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Mikhail V Belousov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, Stockholm 114 17, Sweden.
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden.
| |
Collapse
|
5
|
Deyev SM, Oroujeni M, Garousi J, Gräslund T, Li R, Rosly AHB, Orlova A, Konovalova E, Schulga A, Vorobyeva A, Tolmachev V. Preclinical Evaluation of HER2-Targeting DARPin G3: Impact of Albumin-Binding Domain (ABD) Fusion. Int J Mol Sci 2024; 25:4246. [PMID: 38673831 PMCID: PMC11050402 DOI: 10.3390/ijms25084246] [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: 03/07/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Designed ankyrin repeat protein (DARPin) G3 is an engineered scaffold protein. This small (14.5 kDa) targeting protein binds with high affinity to human epidermal growth factor receptor 2 (HER2). HER2 is overexpressed in several cancers. The use of the DARPin G3 for radionuclide therapy is complicated by its high renal reabsorption after clearance via the glomeruli. We tested the hypothesis that a fusion of the DARPin G3 with an albumin-binding domain (ABD) would prevent rapid renal excretion and high renal reabsorption resulting in better tumour targeting. Two fusion proteins were produced, one with the ABD at the C-terminus (G3-ABD) and another at the N-terminus (ABD-G3). Both variants were labelled with 177Lu. The binding properties of the novel constructs were evaluated in vitro and their biodistribution was compared in mice with implanted human HER2-expressing tumours. Fusion with the ABD increased the retention time of both constructs in blood compared with the non-ABD-fused control. The effect of fusion with the ABD depended strongly on the order of the domains in the constructs, resulting in appreciably better targeting properties of [177Lu]Lu-G3-ABD. Our data suggest that the order of domains is critical for the design of targeting constructs based on scaffold proteins.
Collapse
Affiliation(s)
- Sergey M. Deyev
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.M.D.); (E.K.); (A.S.)
- 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; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
- Affibody AB, 171 65 Solna, Sweden
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
- Department of Protein Science, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden; (T.G.); (R.L.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden; (T.G.); (R.L.)
| | - Ruonan Li
- Department of Protein Science, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden; (T.G.); (R.L.)
| | - Alia Hani Binti Rosly
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden;
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.M.D.); (E.K.); (A.S.)
| | - Alexey Schulga
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.M.D.); (E.K.); (A.S.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
| |
Collapse
|
6
|
Li Y, Zhang J, Cai Z, Gao X, Zhang L, Lu Z, Wang X, Yu P, Li J, Fang F. Disitamab Vedotin (RC48) for HER2-positive advanced breast cancer: a case report and literature review. Front Oncol 2023; 13:1286392. [PMID: 38074635 PMCID: PMC10704599 DOI: 10.3389/fonc.2023.1286392] [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: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND/AIM Human epidermal growth factor receptor 2 (HER2)-positive breast cancer is associated with a higher risk of metastasis and poorer overall survival (OS) due to HER2 gene overexpression/amplification. Although anti-HER2 targeted therapy has shown survival benefits in HER2-positive advanced breast cancer (ABC) patients, long-term treatment often leads to drug resistance, complicating further treatment options. RC48, an antibody-drug conjugate (ADC), combines the benefits of antibody targeting with the cytotoxic effects of a small molecule drug. CASE REPORT We present a case involving a female patient with HER2-positive ABC who developed drug resistance and disease progression following multi-line anti-HER2 targeted therapy. In this instance, RC48 exhibited anti-tumor activity in an ABC patient resistant to HER2-targeted therapy. After eight treatment cycles with 120 mg of RC48, the tumor size decreased and stabilized. CONCLUSION This case report underscores the potential clinical value of RC48 as a promising treatment alternative for patients resistant to HER2 targeted therapies.
Collapse
Affiliation(s)
- Yang Li
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jingjiao Zhang
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhengang Cai
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Xue Gao
- Department of Pathology, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Lina Zhang
- Imaging and Nuclear Medicine Department, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhi Lu
- Nuclear Medicine Department, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Xiaojie Wang
- Department of Radiotherapy, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Peiyao Yu
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jia Li
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Fengqi Fang
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Jia D, Liu H, Zheng S, Yuan D, Sun R, Wang F, Li Y, Li H, Yuan F, Fan Q, Zhao Z. ICG-Dimeric Her2-Specific Affibody Conjugates for Tumor Imaging and Photothermal Therapy for Her2-Positive Tumors. Mol Pharm 2023; 20:427-437. [PMID: 36315025 DOI: 10.1021/acs.molpharmaceut.2c00708] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Human epidermal growth factor receptor 2 (Her2) is abundantly expressed in various solid tumors. The Her2-specific Affibody (ZHer2:2891) has been clinically tested in patients with Her2-positive breast cancer and is regarded as an ideal drug carrier for tumor diagnosis and targeted treatment. Indocyanine green (ICG) can be used as a photosensitizer for photothermal therapy (PTT), in addition to fluorescent dyes for tumor imaging. In this study, a dimeric Her2-specific Affibody (ZHer2) based on ZHer2:2891 was prepared using the E. coli expression system and then coupled to ICG through an N-hydroxysuccinimide (NHS) ester reactive group to construct a novel bifunctional protein drug (named ICG-ZHer2) for tumor diagnosis and PTT. In vitro, ICG-ZHer2-mediated PTT selectively and efficiently killed Her2-positive BT-474 and SKOV-3 tumor cells rather than Her2-negative HeLa tumor cells. In vivo, ICG-ZHer2 specifically accumulated in Her2-positive SKOV-3 tumor grafts rather than Her2-negative HeLa tumor grafts; high-contrast tumor optical images were obtained. However, Her2-negative HeLa tumor grafts were not detected. More importantly, ICG-ZHer2-mediated PTT exhibited a significantly enhanced antitumor effect in mice bearing SKOV-3 tumor grafts owing to the good photothermal properties of ICG-ZHer2. Of note, ICG-ZHer2 did not exhibit acute toxicity in mice during short-term treatment. Overall, our findings indicate that ICG-ZHer2 is a promising bifunctional drug for Her2-positive tumor diagnosis and PTT.
Collapse
Affiliation(s)
- Dianlong Jia
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Huimin Liu
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Shuhui Zheng
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Dandan Yuan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ruohan Sun
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Fei Wang
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yang Li
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Hui Li
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zhenxiong Zhao
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 317700, China
| |
Collapse
|
8
|
Sarvari P, Sarvari P, Ramírez-Díaz I, Mahjoubi F, Rubio K. Advances of Epigenetic Biomarkers and Epigenome Editing for Early Diagnosis in Breast Cancer. Int J Mol Sci 2022; 23:ijms23179521. [PMID: 36076918 PMCID: PMC9455804 DOI: 10.3390/ijms23179521] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/02/2022] Open
Abstract
Epigenetic modifications are known to regulate cell phenotype during cancer progression, including breast cancer. Unlike genetic alterations, changes in the epigenome are reversible, thus potentially reversed by epi-drugs. Breast cancer, the most common cause of cancer death worldwide in women, encompasses multiple histopathological and molecular subtypes. Several lines of evidence demonstrated distortion of the epigenetic landscape in breast cancer. Interestingly, mammary cells isolated from breast cancer patients and cultured ex vivo maintained the tumorigenic phenotype and exhibited aberrant epigenetic modifications. Recent studies indicated that the therapeutic efficiency for breast cancer regimens has increased over time, resulting in reduced mortality. Future medical treatment for breast cancer patients, however, will likely depend upon a better understanding of epigenetic modifications. The present review aims to outline different epigenetic mechanisms including DNA methylation, histone modifications, and ncRNAs with their impact on breast cancer, as well as to discuss studies highlighting the central role of epigenetic mechanisms in breast cancer pathogenesis. We propose new research areas that may facilitate locus-specific epigenome editing as breast cancer therapeutics.
Collapse
Affiliation(s)
- Pourya Sarvari
- Department of Clinical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran P.O. Box 14965/161, Iran
| | - Pouya Sarvari
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico
| | - Ivonne Ramírez-Díaz
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico
- Facultad de Biotecnología, Campus Puebla, Universidad Popular Autónoma del Estado de Puebla (UPAEP), Puebla 72410, Mexico
| | - Frouzandeh Mahjoubi
- Department of Clinical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran P.O. Box 14965/161, Iran
| | - Karla Rubio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico
- Licenciatura en Médico Cirujano, Universidad de la Salud del Estado de Puebla (USEP), Puebla 72000, Mexico
- Correspondence:
| |
Collapse
|
9
|
Garousi J, Xu T, Liu Y, Vorontsova O, Hober S, Orlova A, Tolmachev V, Gräslund T, Vorobyeva A. Experimental HER2-Targeted Therapy Using ADAPT6-ABD-mcDM1 in Mice Bearing SKOV3 Ovarian Cancer Xenografts: Efficacy and Selection of Companion Imaging Counterpart. Pharmaceutics 2022; 14:pharmaceutics14081612. [PMID: 36015242 PMCID: PMC9415843 DOI: 10.3390/pharmaceutics14081612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
Overexpression of the human epidermal growth factor receptor 2 (HER2) in breast and gastric cancer is exploited for targeted therapy using monoclonal antibodies and antibody-drug conjugates. Small engineered scaffold proteins, such as the albumin binding domain (ABD) derived affinity proteins (ADAPTs), are a promising new format of targeting probes for development of drug conjugates with well-defined structure and tunable pharmacokinetics. Radiolabeled ADAPT6 has shown excellent tumor-targeting properties in clinical trials. Recently, we developed a drug conjugate based on the HER2-targeting ADAPT6 fused to an albumin binding domain (ABD) for increased bioavailability and conjugated to DM1 for cytotoxic action, designated as ADAPT6-ABD-mcDM1. In this study, we investigated the therapeutic efficacy of this conjugate in mice bearing HER2-expressing SKOV3 ovarian cancer xenografts. A secondary aim was to evaluate several formats of imaging probes for visualization of HER2 expression in tumors. Administration of ADAPT6-ABD-mcDM1 provided a significant delay of tumor growth and increased the median survival of the mice, in comparison with both a non-targeting homologous construct (ADAPTNeg-ABD-mcDM1) and the vehicle-treated groups, without inducing toxicity to liver or kidneys. Moreover, the evaluation of imaging probes showed that small scaffold proteins, such as 99mTc(CO)3-ADAPT6 or the affibody molecule 99mTc-ZHER2:41071, are well suited as diagnostic companions for potential stratification of patients for ADAPT6-ABD-mcDM1–based therapy.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Protein Science, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (J.G.); (S.H.)
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (O.V.); (A.V.)
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (O.V.); (A.V.)
| | - Olga Vorontsova
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (O.V.); (A.V.)
| | - Sophia Hober
- Department of Protein Science, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (J.G.); (S.H.)
| | - Anna Orlova
- 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.); (Y.L.); (O.V.); (A.V.)
- Correspondence: (V.T.); (T.G.); Tel.: +46-70-425-07-82 (V.T.); +46-(0)8-790-96-27 (T.G.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden; (J.G.); (S.H.)
- Correspondence: (V.T.); (T.G.); Tel.: +46-70-425-07-82 (V.T.); +46-(0)8-790-96-27 (T.G.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (O.V.); (A.V.)
| |
Collapse
|
10
|
Targeting Tumor Cells Overexpressing the Human Epidermal Growth Factor Receptor 3 with Potent Drug Conjugates Based on Affibody Molecules. Biomedicines 2022; 10:biomedicines10061293. [PMID: 35740315 PMCID: PMC9219639 DOI: 10.3390/biomedicines10061293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/22/2022] Open
Abstract
Increasing evidence suggests that therapy targeting the human epidermal growth factor receptor 3 (HER3) could be a viable route for targeted cancer therapy. Here, we studied a novel drug conjugate, ZHER3-ABD-mcDM1, consisting of a HER3-targeting affibody molecule, coupled to the cytotoxic tubulin polymerization inhibitor DM1, and an albumin-binding domain for in vivo half-life extension. ZHER3-ABD-mcDM1 showed a strong affinity to the extracellular domain of HER3 (KD 6 nM), and an even stronger affinity (KD 0.2 nM) to the HER3-overexpressing pancreatic carcinoma cell line, BxPC-3. The drug conjugate showed a potent cytotoxic effect on BxPC-3 cells with an IC50 value of 7 nM. Evaluation of a radiolabeled version, [99mTc]Tc-ZHER3-ABD-mcDM1, showed a relatively high rate of internalization, with a 27% internalized fraction after 8 h. Further in vivo evaluation showed that it could target BxPC-3 (pancreatic carcinoma) and DU145 (prostate carcinoma) xenografts in mice, with an uptake peaking at 6.3 ± 0.4% IA/g at 6 h post-injection for the BxPC-3 xenografts. The general biodistribution showed uptake in the liver, lung, salivary gland, stomach, and small intestine, organs known to express murine ErbB3 naturally. The results from the study show that ZHER3-ABD-mcDM1 is a highly potent and selective drug conjugate with the ability to specifically target HER3 overexpressing cells. Further pre-clinical and clinical development is discussed.
Collapse
|