1
|
Bender AA, Holiski CK, Embree M, Hennkens HM, Klaehn JR, Lundgreen E, Roberts AG, Zalupski PR, Mastren T. Pursuing theranostics: a multimodal architecture approach. SENSORS & DIAGNOSTICS 2025; 4:35-43. [PMID: 39493501 PMCID: PMC11528688 DOI: 10.1039/d4sd00221k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
Theranostics is a field of nuclear medicine which uses the same targeting vector and chelating system for both a diagnostic and therapeutic radionuclide, allowing for uniformity in imaging and treatment. This growing field requires the development of more flexible chelate systems that permit novel targeting strategies. Toward this end, a multimodal architecture has been realized, making use of a phosphazene-based core and click chemistry to achieve a flexible and customizable scaffold. The six arm phosphazene-based core can scaffold six DTPA chelating motifs or a mixed set of 3 : 3 DTPA : DFO chelates resulting in two multimodal compounds, pDbDt and pDbDtDf, respectively. Terbium complexes displayed strong luminescence, supporting that the structures act as an organic antenna for luminescence. Metal displacement titration studies confirmed the desired structures as well as the capability for heterometallic labeling of the structures. These structures were found to have high thermal and biological stability in vitro. Radiolabeling of each compound resulted in high molar activity labeling of each compound: 169 MBq nmol-1: [161Tb]Tb-pDbDt, 170 MBq nmol-1: [89Zr]Zr-pDbDtDf, and the mixed radiolabeling illustrated chelation of both radionuclides in a 1 : 1 ratio. This multimodal architecture is promising as a heterometallic structure for coupling of both a diagnostic and a therapeutic radionuclide with a highly customizable core structure.
Collapse
Affiliation(s)
- Aidan A Bender
- Nuclear Engineering Program, University of Utah 110 Central Campus Dr. Suite 2000B Salt Lake City UT 84112 USA
| | - Connor K Holiski
- Nuclear Engineering Program, University of Utah 110 Central Campus Dr. Suite 2000B Salt Lake City UT 84112 USA
| | - Mary Embree
- University of Missouri Research Reactor Columbia MO 65211 USA
| | - Heather M Hennkens
- University of Missouri Research Reactor Columbia MO 65211 USA
- Department of Chemistry, University of Missouri Columbia MO 65211 USA
| | - John R Klaehn
- Biological and Chemical Process Sciences, Idaho National Laboratory Idaho Falls ID 83415 USA
| | - Ellie Lundgreen
- Nuclear Engineering Program, University of Utah 110 Central Campus Dr. Suite 2000B Salt Lake City UT 84112 USA
| | - Andrew G Roberts
- Department of Chemistry, University of Utah Salt Lake City UT 84112 USA
| | - Peter R Zalupski
- Aqueous Separations and Radiochemistry, Idaho National Laboratory Idaho Falls ID 83415 USA
| | - Tara Mastren
- Nuclear Engineering Program, University of Utah 110 Central Campus Dr. Suite 2000B Salt Lake City UT 84112 USA
| |
Collapse
|
2
|
Hao JL, Li XY, Liu YT, Lang JX, Liu DJ, Zhang CD. Antibody-drug conjugates in gastric cancer: from molecular landscape to clinical strategies. Gastric Cancer 2024; 27:887-906. [PMID: 38963593 DOI: 10.1007/s10120-024-01529-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a crucial component of targeted therapies in gastric cancer, potentially altering traditional treatment paradigms. Many ADCs have entered rigorous clinical trials based on biological theories and preclinical experiments. Modality trials have also been conducted in combination with monoclonal antibody therapies, chemotherapies, immunotherapies, and other treatments to enhance the efficacy of drug coordination effects. However, ADCs exhibit limitations in treating gastric cancer, including resistance triggered by their structure or other factors. Ongoing intensive researches and preclinical experiments are yielding improvements, while enhancements in drug development processes and concomitant diagnostics during the therapeutic period actively boost ADC efficacy. The optimal treatment strategy for gastric cancer patients is continually evolving. This review summarizes the clinical progress of ADCs in treating gastric cancer, analyzes the mechanisms of ADC combination therapies, discusses resistance patterns, and offers a promising outlook for future applications in ADC drug development and companion diagnostics.
Collapse
Affiliation(s)
- Jia-Lin Hao
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Yu-Tong Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Ji-Xuan Lang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Di-Jie Liu
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Chun-Dong Zhang
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| |
Collapse
|
3
|
Abstract
Evidence implicating Eph receptor tyrosine kinases and their ephrin ligands (that together make up the 'Eph system') in cancer development and progression has been accumulating since the discovery of the first Eph receptor approximately 35 years ago. Advances in the past decade and a half have considerably increased the understanding of Eph receptor-ephrin signalling mechanisms in cancer and have uncovered intriguing new roles in cancer progression and drug resistance. This Review focuses mainly on these more recent developments. I provide an update on the different mechanisms of Eph receptor-ephrin-mediated cell-cell communication and cell autonomous signalling, as well as on the interplay of the Eph system with other signalling systems. I further discuss recent advances in elucidating how the Eph system controls tumour expansion, invasiveness and metastasis, supports cancer stem cells, and drives therapy resistance. In addition to functioning within cancer cells, the Eph system also mediates the reciprocal communication between cancer cells and cells of the tumour microenvironment. The involvement of the Eph system in tumour angiogenesis is well established, but recent findings also demonstrate roles in immune cells, cancer-associated fibroblasts and the extracellular matrix. Lastly, I discuss strategies under evaluation for therapeutic targeting of Eph receptors-ephrins in cancer and conclude with an outlook on promising future research directions.
Collapse
Affiliation(s)
- Elena B Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
4
|
Keinänen O, Sarrett SM, Delaney S, Rodriguez C, Dayts EJ, Capone E, Sauniere F, Ippoliti R, Sala G, Iacobelli S, Zeglis BM. Visualizing Galectin-3 Binding Protein Expression with ImmunoPET. Mol Pharm 2023; 20:3241-3248. [PMID: 37191353 PMCID: PMC10245371 DOI: 10.1021/acs.molpharmaceut.3c00241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023]
Abstract
Galectin-3 binding protein (Gal-3BP) is a glycoprotein that is overexpressed and secreted by several cancers and has been implicated as a marker of both tumor progression and poor prognosis in melanoma, non-small cell lung cancer, head and neck squamous cell carcinoma, and breast cancer. The expression of Gal-3BP by a variety of neoplasms makes it an enticing target for both diagnostics and therapeutics, including immuno-positron emission tomography (immunoPET) probes and antibody-drug conjugates (ADCs). Herein, we report the development, in vitro characterization, and in vivo evaluation of a pair of Gal-3BP-targeting radioimmunoconjugates for 89Zr-immunoPET. A humanized anti-Gal-3BP antibody, 1959, and its corresponding ADC, 1959-sss/DM4 (DM4 = ravtansine), were modified with desferrioxamine (DFO) to yield DFO-1959 and DFO-1959-sss/DM4 immunoconjugates bearing 1-2 DFO/monoclonal antibody. Both DFO-modified immunoconjugates retained their affinity for Gal-3BP in enzyme-linked immunosorbent assay experiments. The chelator-bearing antibodies were radiolabeled with zirconium-89 (t1/2 ≈ 3.3 d) to produce radioimmunoconjugates ─ [89Zr]Zr-DFO-1959 and [89Zr]Zr-DFO-1959-sss/DM4 ─ with high specific activity (>444 MBq/mg, >12 mCi/mg) and stability (>80% intact after 168 h in human serum at 37 °C). In mice bearing subcutaneous Gal-3BP-secreting A375-MA1 xenografts, [89Zr]Zr-DFO-1959 clearly delineated tumor tissue, reaching a maximum tumoral activity concentration (54.8 ± 15.8%ID/g) and tumor-to-background contrast (tumor-to-blood = 8.0 ± 4.6) at 120 h post-injection. The administration of [89Zr]Zr-DFO-1959 to mice bearing subcutaneous Gal-3BP-expressing melanoma patient-derived xenografts produced similarly promising results. [89Zr]Zr-DFO-1959 and [89Zr]Zr-DFO-1959-sss/DM4 exhibited nearly identical pharmacokinetic profiles in the mice bearing A375-MA1 tumors, though the latter produced higher uptake in the spleen and kidneys. Both [89Zr]Zr-DFO-1959 and [89Zr]Zr-DFO-1959-sss/DM4 effectively visualized Gal-3BP-secreting tumors in murine models of melanoma. These results suggest that both probes could play a role in the clinical imaging of Gal-3BP-expressing malignancies, particularly as companion theranostics for the identification of patients likely to respond to Gal-3BP-targeted therapeutics such as 1959-sss/DM4.
Collapse
Affiliation(s)
- Outi Keinänen
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United
States
- Department
of Chemistry, University of Helsinki, Helsinki 00014, Finland
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10065, New York, United
States
| | - Samantha M. Sarrett
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United
States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10065, New York, United
States
- Ph.D.
Program in Biochemistry, Graduate Center
of the City University of New York, New York 10016, New
York, United States
| | - Samantha Delaney
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United
States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10065, New York, United
States
- Ph.D.
Program in Biochemistry, Graduate Center
of the City University of New York, New York 10016, New
York, United States
| | - Cindy Rodriguez
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United
States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10065, New York, United
States
- Ph.D.
Program in Chemistry, Graduate Center of
the City University of New York, New York 10016, New
York, United States
| | - Eric J. Dayts
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United
States
| | - Emily Capone
- Department
of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, Chieti 66100, Italy
- Mediapharma
srl, Chieti 66013, Italy
- Center
for Advanced Studies and Technology, Chieti 66100, Italy
| | | | - Rodolfo Ippoliti
- Department
of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila 67100, Italy
| | - Gianluca Sala
- Department
of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, Chieti 66100, Italy
- Mediapharma
srl, Chieti 66013, Italy
- Center
for Advanced Studies and Technology, Chieti 66100, Italy
| | | | - Brian M. Zeglis
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United
States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10065, New York, United
States
- Ph.D.
Program in Biochemistry, Graduate Center
of the City University of New York, New York 10016, New
York, United States
- Ph.D.
Program in Chemistry, Graduate Center of
the City University of New York, New York 10016, New
York, United States
- Department
of Radiology, Weill Cornell Medical College, New York 10021, New York, United States
| |
Collapse
|
5
|
Modi A, Pandey P, Uniyal A, Chouhan D, Agrawal S, Allani M, Singh AK, Kumar S, Tiwari V. Disentangling the enigmatic role of ephrin signaling in chronic pain: Moving towards future anti-pain therapeutics. Life Sci 2023:121796. [PMID: 37230378 DOI: 10.1016/j.lfs.2023.121796] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 05/08/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Chronic pain is a common and debilitating condition with a huge social and economic burden worldwide. Currently, available drugs in clinics are not adequately effective and possess a variety of severe side effects leading to treatment withdrawal and poor quality of life. The ongoing search for new therapeutics with minimal side effects for chronic pain management remains a high research priority. Erythropoietin-producing human hepatocellular carcinoma cell receptor (Eph) is a tyrosine kinase receptor that is involved in neurodegenerative disorders, including pain. The Eph receptor interacts with several molecular switches, such as N methyl d-aspartate receptor (NMDAR), mitogen-activated protein kinase (MAPK), calpain 1, caspase 3, protein kinase a (PKA), and protein kinase Cy (PKCy), which in turn regulates pathophysiology of chronic pain. Here we highlight the emerging evidence of the Ephs/ephrin system as a possible near-future therapeutic target for the treatment of chronic pain and discuss the various mechanism of its involvement. We critically analyse the present status of Eph receptor system and conclude that extrapolating the pharmacological and genetic approaches using a strong therapeutic development framework could serve as next-generation analgesics for the management of chronic pain.
Collapse
Affiliation(s)
- Ajay Modi
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Priyanka Pandey
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Ankit Uniyal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Somesh Agrawal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Meghana Allani
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Anurag Kumar Singh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Sonu Kumar
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India.
| |
Collapse
|
6
|
Furukawa T, Kimura H, Sasaki M, Yamada T, Iwasawa T, Yagi Y, Kato K, Yasui H. Novel [ 111 In]In-BnDTPA-EphA2-230-1 Antibody for Single-Photon Emission Computed Tomography Imaging Tracer Targeting of EphA2. ACS OMEGA 2023; 8:7030-7035. [PMID: 36844571 PMCID: PMC9948553 DOI: 10.1021/acsomega.2c07849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
Erythropoietin-producing hepatocellular receptor A2 (EphA2) is overexpressed in cancer cells and causes abnormal cell proliferation. Therefore, it has attracted attention as a target for diagnostic agents. In this study, the EphA2-230-1 monoclonal antibody (EphA2-230-1) was labeled with [111In]In and evaluated as an imaging tracer for single-photon emission computed tomography (SPECT) of EphA2. EphA2-230-1 was conjugated with 2-(4-isothiocyanatobenzyl)-diethylenetriaminepentaacetic acid (p-SCN-BnDTPA) and then labeled with [111In]In. [111In]In-BnDTPA-EphA2-230-1 was evaluated in cell-binding, biodistribution, and SPECT/computed tomography (CT) studies. The cellular uptake ratio of [111In]In-BnDTPA-EphA2-230-1 was 14.0 ± 2.1%/mg protein at 4 h in the cell-binding study. In the biodistribution study, a high uptake of [111In]In-BnDTPA-EphA2-230-1 was observed in tumor tissue (14.6 ± 3.2% injected dose/g at 72 h). The superior accumulation of [111In]In-BnDTPA-EphA2-230-1 in tumors was also confirmed using SPECT/CT. Therefore, [111In]In-BnDTPA-EphA2-230-1 has potential as a SPECT imaging tracer for EphA2.
Collapse
Affiliation(s)
- Takenori Furukawa
- Department
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Science, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hiroyuki Kimura
- Department
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Science, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Minon Sasaki
- Department
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Science, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Takumu Yamada
- Department
of Biomedical Engineering, Faculty of Science and Engineering, Toyo University, 2100 Nakanodai, Kujirai, Kawagoe, Saitama 350-0815, Japan
| | - Takumi Iwasawa
- Department
of Biomedical Engineering, Faculty of Science and Engineering, Toyo University, 2100 Nakanodai, Kujirai, Kawagoe, Saitama 350-0815, Japan
| | - Yusuke Yagi
- Department
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Science, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
- Department
of Radiological Technology, Faculty of Medicinal Science, Kyoto College of Medical Science, 1-3 Imakita, Oyama-higashi, Sonobe,
Nantan, Kyoto 622-0022, Japan
| | - Kazunori Kato
- Department
of Biomedical Engineering, Faculty of Science and Engineering, Toyo University, 2100 Nakanodai, Kujirai, Kawagoe, Saitama 350-0815, Japan
| | - Hiroyuki Yasui
- Department
of Analytical and Bioinorganic Chemistry, Division of Analytical and
Physical Science, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| |
Collapse
|
7
|
Chen Y, Han J, Zhao Y, Zhao X, Zhao M, Zhang J, Wang J. 18F-labeled FGFR1 peptide: a new PET probe for subtype FGFR1 receptor imaging. Front Oncol 2023; 13:1047080. [PMID: 37182162 PMCID: PMC10174317 DOI: 10.3389/fonc.2023.1047080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 04/14/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction The fibroblast growth factor receptor (FGFR) family is highly expressed in a variety of tumor types and represents a new target for cancer therapy. Different FGFR subtype aberrations have been found to exhibit highly variable sensitivity and efficacy to FGFR inhibitors. Methods The present study is the first to suggest an imaging method for assessing FGFR1 expression. The FGFR1-targeting peptide NOTA-PEG2-KAEWKSLGEEAWHSK was synthesized by manual solid-phase peptide synthesis and high-pressure liquid chromatography (HPLC) purification and then labeled with fluorine-18 using NOTA as a chelator. In vitro and in vivo experiments were conducted to evaluate the stability, affinity and specificity of the probe. Tumor targeting efficacy and biodistribution were evaluated by micro-PET/CT imaging in RT-112, A549, SNU-16 and Calu-3 xenografts. Results The radiochemical purity of [18F]F-FGFR1 was 98.66% ± 0.30% (n = 3) with excellent stability. The cellular uptake rate of [18F]F-FGFR1 in the RT-112 cell line (FGFR1 overexpression) was higher than that in the other cell lines and could be blocked by the presence of excess unlabeled FGFR1 peptide. Micro-PET/CT imaging revealed a significant concentration of [18F]F-FGFR1 in RT-112 xenografts with no or very low uptake in nontargeted organs and tissues, which demonstrated that [18F]F-FGFR1 was selectively taken up by FGFR1-positive tumors. Conclusion [18F]F-FGFR1 showed high stability, affinity, specificity and good imaging capacity for FGFR1-overexpressing tumors in vivo, which provides new application potential in the visualization of FGFR1 expression in solid tumors.
Collapse
Affiliation(s)
- Yang Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Zhao
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, China
- *Correspondence: Xinming Zhao,
| | - Mengmeng Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
8
|
Advanced molecular imaging for the characterisation of complex medicines. Drug Discov Today 2022; 27:1716-1723. [DOI: 10.1016/j.drudis.2022.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/18/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023]
|
9
|
Yang Y, Nian S, Li L, Wen X, Liu Q, Zhang B, Lan Y, Yuan Q, Ye Y. Fully human recombinant antibodies against EphA2 from a multi-tumor patient immune library suitable for tumor-targeted therapy. Bioengineered 2021; 12:10379-10400. [PMID: 34709992 PMCID: PMC8810047 DOI: 10.1080/21655979.2021.1996807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Enhanced EphA2 expression is observed in a variety of epithelial-derived malignancies and is an important target for anti-tumor therapy. Currently, Therapeutic monoclonal antibodies against immune checkpoints have shown good efficacy for tumor treatment. In this study, we constructed an immune single-chain fragment variable (scFv) library using peripheral blood mononuclear cells (PBMCs) from 200 patients with a variety of malignant tumors. High affinity scFvs against EphA2 can be easily screened from the immune library using phage display technology. Anti-EphA2 scFvs can be modified into any form of recombinant antibody, including scFv-Fc and full-length IgG1 antibodies, and the recombinant antibody affinity was improved following modification. Among the modified anti-EphA2 antibodies the affinity of 77-IgG1 was significantly increased, reaching a pmol affinity level (10−12). We further demonstrated the binding activity of recombinant antibodies to the EphA2 protein, tumor cells, and tumor tissues using macromolecular interaction techniques, flow cytometry and immunohistochemistry. Most importantly, both the constructed scFvs-Fc, as well as the IgG1 antibodies against EphA2 were able to inhibit the growth of tumor cells to some extent. These results suggest that the immune libraries from patients with malignant tumors are more likely to screen for antibodies with high affinity and therapeutic effect. The constructed fully human scFv immune library has broad application prospects for specific antibody screening. The screened scFv-Fc and IgG1 antibodies against EphA2 can be used for the further study of tumor immunotherapy.
Collapse
Affiliation(s)
- Yaqi Yang
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China
| | - Siji Nian
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China
| | - Lin Li
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China
| | - Xue Wen
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China.,Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Sichuan 646000, P.R. China
| | - Qin Liu
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China
| | - Bo Zhang
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China
| | - Yu Lan
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China
| | - Qing Yuan
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China
| | - Yingchun Ye
- Public Center of Experimental Technology, The school of Basic medical science, Southwest medical university, Luzhou, Sichuan Province, 646000, China
| |
Collapse
|
10
|
Pyo A, You SH, Sik Kim H, Young Kim J, Min JJ, Kim DY, Hong Y. Production of 64Cu-labeled monobody for imaging of human EphA2-expressing tumors. Bioorg Med Chem Lett 2020; 30:127262. [PMID: 32527560 DOI: 10.1016/j.bmcl.2020.127262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/20/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
We previously reported on the monobody E1, which specifically targets the tumor marker hEphA2. In this study, we labeled NOTA-conjugated E1 with 64Cu (64Cu-NOTA-E1) and evaluated biologic characteristics. The uptake of 64Cu-NOTA-E1 in PC3 cells (a human prostate cancer cell line) with high expression of hEphA2 increased in a time-dependent manner. In PC3 xenograft mice, 64Cu-NOTA-E1 injected via the tail vein allowed visualization of tumors on positron emission tomography after 1 h and the highest uptake measured at 24 h post-injection. By contrast, the radioactivity of other tissues either did not increase or decreased over 24 h. This indicates that 64Cu-NOTA-E1 has high tumor uptake and retention, with rapid clearance, and low background values in other tissues. Therefore, 64Cu-NOTA-E1 should be suitable as a novel PET imaging agent for hEphA2-expressing tumors.
Collapse
Affiliation(s)
- Ayoung Pyo
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Sung-Hwan You
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Hyeon Sik Kim
- Medical Photonics Research Center, Korea Photonics Technology Institute, Gwangju, Republic of Korea
| | - Jung Young Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Dong-Yeon Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea.
| | - Yeongjin Hong
- Department of Microbiology, Chonnam National University Medical School, Hwasun, Republic of Korea.
| |
Collapse
|
11
|
Harnessing the Power of Eph/ephrin Biosemiotics for Theranostic Applications. Pharmaceuticals (Basel) 2020; 13:ph13060112. [PMID: 32492868 PMCID: PMC7345574 DOI: 10.3390/ph13060112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
Comprehensive basic biological knowledge of the Eph/ephrin system in the physiologic setting is needed to facilitate an understanding of its role and the effects of pathological processes on its activity, thereby paving the way for development of prospective therapeutic targets. To this end, this review briefly addresses what is currently known and being investigated in order to highlight the gaps and possible avenues for further investigation to capitalize on their diverse potential.
Collapse
|
12
|
Janes PW, Vail ME, Gan HK, Scott AM. Antibody Targeting of Eph Receptors in Cancer. Pharmaceuticals (Basel) 2020; 13:ph13050088. [PMID: 32397088 PMCID: PMC7281212 DOI: 10.3390/ph13050088] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022] Open
Abstract
The Eph subfamily of receptor tyrosine kinases mediate cell-cell communication controlling cell and tissue patterning during development. While generally less active in adult tissues, they often re-emerge in cancers, particularly on undifferentiated or progenitor cells in tumors and the tumor microenvironment, associated with tumor initiation, angiogenesis and metastasis. Eph receptors are thus attractive therapeutic targets, and monoclonal antibodies have been commonly developed and tested for anti-cancer activity in preclinical models, and in some cases in the clinic. This review summarizes 20 years of research on various antibody-based approaches to target Eph receptors in tumors and the tumor microenvironment, including their mode of action, tumor specificity, and efficacy in pre-clinical and clinical testing.
Collapse
|
13
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
14
|
Critical Issues in the Development of Immunotoxins for Anticancer Therapy. J Pharm Sci 2019; 109:104-115. [PMID: 31669121 DOI: 10.1016/j.xphs.2019.10.037] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
Abstract
Immunotoxins (ITs) are attractive anticancer modalities aimed at cancer-specific delivery of highly potent cytotoxic protein toxins. An IT consists of a targeting domain (an antibody, cytokine, or another cell-binding protein) chemically conjugated or recombinantly fused to a highly cytotoxic payload (a bacterial and plant toxin or human cytotoxic protein). The mode of action of ITs is killing designated cancer cells through the effector function of toxins in the cytosol after cellular internalization via the targeted cell-specific receptor-mediated endocytosis. Although numerous ITs of diverse structures have been tested in the past decades, only 3 ITs-denileukin diftitox, tagraxofusp, and moxetumomab pasudotox-have been clinically approved for treating hematological cancers. No ITs against solid tumors have been approved for clinical use. In this review, we discuss critical research and development issues associated with ITs that limit their clinical success as well as strategies to overcome these obstacles. The issues include off-target and on-target toxicities, immunogenicity, human cytotoxic proteins, antigen target selection, cytosolic delivery efficacy, solid-tumor targeting, and developability. To realize the therapeutic promise of ITs, novel strategies for safe and effective cytosolic delivery into designated tumors, including solid tumors, are urgently needed.
Collapse
|
15
|
Abstract
Targeted therapies hold great promise for cancer treatment and may exhibit even greater efficacy when combined with patient selection tools. The clinical impact of identifying likely responders includes reducing the number of unnecessary and ineffective therapies as well as more accurately determining drug effects. Positron emission tomography (PET) imaging using zirconium-89 radiolabeled monoclonal antibodies (mAbs), also referred to as zirconium-89 (89Zr)-immuno-PET, provides a potential biomarker to measure target expression and verify optimal delivery of targeted agents to tumors. Antibody-drug conjugates (ADCs) combine the high affinity and specificity of mAbs with the potency of cytotoxic drugs to target tumor-expressing antigen and destroy cancer cells. Thus, 89Zr-immuno-PET of whole-body biodistribution, pharmacokinetics, and tumor targeting of antibodies and ADCs to predict toxicity and efficacy could help guide individualized treatment. Here, we review how 89Zr-immuno-PET is being used as a companion diagnostic with the development of ADCs. Furthermore, we discuss how 89Zr-immuno-PET may be utilized in future clinical trials as an adjunct tool with novel ADCs to select cancer patients who have the greatest potential to benefit from treatment and improve ADC dosing regimens.
Collapse
Affiliation(s)
- Kendra S Carmon
- 1 Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ali Azhdarinia
- 1 Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
16
|
Nejadmoghaddam MR, Minai-Tehrani A, Ghahremanzadeh R, Mahmoudi M, Dinarvand R, Zarnani AH. Antibody-Drug Conjugates: Possibilities and Challenges. Avicenna J Med Biotechnol 2019; 11:3-23. [PMID: 30800238 PMCID: PMC6359697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 12/31/2017] [Indexed: 11/22/2022] Open
Abstract
The design of Antibody Drug Conjugates (ADCs) as efficient targeting agents for tumor cell is still in its infancy for clinical applications. This approach incorporates the antibody specificity and cell killing activity of chemically conjugated cytotoxic agents. Antibody in ADC structure acts as a targeting agent and a nanoscale carrier to deliver a therapeutic dose of cytotoxic cargo into desired tumor cells. Early ADCs encountered major obstacles including, low blood residency time, low penetration capacity to tumor microenvironment, low payload potency, immunogenicity, unusual off-target toxicity, drug resistance, and the lack of stable linkage in blood circulation. Although extensive studies have been conducted to overcome these issues, the ADCs based therapies are still far from having high-efficient clinical outcomes. This review outlines the key characteristics of ADCs including tumor marker, antibody, cytotoxic payload, and linkage strategy with a focus on technical improvement and some future trends in the pipeline.
Collapse
Affiliation(s)
- Mohammad-Reza Nejadmoghaddam
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Arash Minai-Tehrani
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Ramin Ghahremanzadeh
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Morteza Mahmoudi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Immunology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
| |
Collapse
|
17
|
Muns JA, Montserrat V, Houthoff HJ, Codée-van der Schilden K, Zwaagstra O, Sijbrandi NJ, Merkul E, van Dongen GAMS. In Vivo Characterization of Platinum(II)-Based Linker Technology for the Development of Antibody-Drug Conjugates: Taking Advantage of Dual Labeling with 195mPt and 89Zr. J Nucl Med 2018; 59:1146-1151. [PMID: 29496986 DOI: 10.2967/jnumed.117.206672] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/03/2018] [Indexed: 12/16/2022] Open
Abstract
Linker instability and impaired tumor targeting can affect the tolerability and efficacy of antibody-drug conjugates (ADCs). To improve these ADC characteristics, we recently described the use of a metal-organic linker, [ethylenediamineplatinum(II)]2+, herein called Lx Initial therapy studies in xenograft-bearing mice revealed that trastuzumab-Lx-auristatin F (AF) outperformed its maleimide benchmark trastuzumab-mal-AF and the Food and Drug Administration-approved ado-trastuzumab emtansine, both containing conventional linkers. In this study, we aimed to characterize Lx-based ADCs for in vivo stability and tumor targeting using 195mPt and 89Zr. Methods: The γ-emitter 195mPt was used to produce the radiolabeled Lx [195mPt]Lx89Zr-Desferrioxamine (89Zr-DFO) was conjugated to trastuzumab either via [195mPt]Lx (to histidine residues) or conventionally (to lysine residues) in order to monitor the biodistribution of antibody, payload, and linker separately. Linker stability was determined by evaluating the following ADCs for biodistribution in NCI-N87 xenograft-bearing nude mice 72 h after injection: trastuzumab-[195mPt]Lx-DFO-89Zr, trastuzumab-[195mPt]Lx-AF, and 89Zr-DFO-(Lys)trastuzumab (control), all having drug-to-antibody ratios (DARs) of 2.2-2.5. To assess the influence of DAR on biodistribution, 89Zr-DFO-(Lys)trastuzumab-Lx-AF with an AF-to-antibody ratio of 0, 2.6, or 5.2 was evaluated 96 h after injection. Results: Similar biodistributions were observed for trastuzumab-[195mPt]Lx-DFO-89Zr, trastuzumab-[195mPt]Lx-AF, and 89Zr-DFO-(Lys)trastuzumab irrespective of the isotope used for biodistribution assessment. The fact that Lx follows the antibody biodistribution indicates that the payload-Lx bond is stable in vivo. Uptake of the 3 conjugates, as percentage injected dose (%ID) per gram of tissue, was about 30 %ID/g in tumor tissue but less than 10 %ID/g in most healthy tissues. Trastuzumab-[195mPt]Lx-AF (DAR 2.2) showed a tendency toward faster blood clearance and an elevated liver uptake, which increased significantly to 28.1 ± 4.2 %ID/g at a higher DAR of 5.2, as revealed from the biodistribution and PET imaging studies. Conclusion: As shown by 195mPt/89Zr labeling, ADCs containing the Lx linker are stable in vivo. In the case of trastuzumab-Lx-AF (DARs 2.2 and 2.6), an unimpaired biodistribution was demonstrated.
Collapse
Affiliation(s)
| | | | | | | | - Oene Zwaagstra
- Nuclear Research and Consultancy Group (NRG), Petten, The Netherlands; and
| | | | | | - Guus A M S van Dongen
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|