1
|
Liu Q, Xu X, Yang Z, Zhang J, Li J, Qiao Y, Hu S, Liu X, Guo W, Song S. Granzyme B PET/CT Imaging Evaluates Early Response to Immunotherapy in Gastric Cancer. J Nucl Med 2024; 65:1695-1701. [PMID: 39389628 DOI: 10.2967/jnumed.124.267529] [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: 01/29/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
In several malignancies, only a limited number of patients respond to immune checkpoint inhibitors. Predicting and monitoring responses to these inhibitors represent an unmet clinical need. Here, we developed a PET/CT probe targeting granzyme B, [68Ga]Ga-NOTA-Gly-Gly-Gly-Ile-Glu-Pro-Asp-CHO (GSI), and aimed to investigate whether it can be used to monitor the effects of immune checkpoint inhibitors early in the course of therapy. Methods: Seventy-two patients with gastric cancer (stages III-IV) were recruited for [68Ga]Ga-NOTA-GSI PET/CT imaging after 2 or 3 cycles of the immunotherapy, and 40 patients were included in the final analysis. The SUVmax of primary tumors (SUVmax-t), SUVmax of metastatic lymph nodes (SUVmax-LN), and SUVmax of normal tissues (liver and blood pool) were measured, and their target-to-liver background ratio (TLR) and target-to-blood background ratio (TBR) were denoted for primary tumors as TLRtumor and TBRtumor and for metastatic lymph nodes as TLRLN and TBRLN, respectively. The treatment responses were assessed within 1 wk after full-course treatment according to RECIST version 1.1. Wilcoxon rank-sum tests were used to compare the PET/CT parameters between responders and nonresponders. Receiver operating characteristic curve analysis was used to assess the diagnostic efficacy of [68Ga]Ga-NOTA-GSI PET/CT parameters in identifying responders. Two-tailed P value of less than 0.05 was considered statistically significant. Results: We found that SUVmax-t, TLRtumor, TBRtumor, SUVmax-LN, and TBRLN were higher in responders than in nonresponders (2.49 ± 0.58 vs. 1.55 ± 0.48, P = 0.000; 2.24 ± 0.48 vs. 1.74 ± 0.67, P = 0.007; 1.38 ± 0.43 vs. 0.90 ± 0.23, P = 0.000; 2.24 ± 0.99 vs. 1.42 ± 0.55, P = 0.003; and 1.28 ± 0.68 vs. 0.83 ± 0.32, P = 0.012, respectively). According to receiver operating characteristic curve analysis, the area under the curve for SUVmax-t, TBRtumor, TLRtumor, SUVmax-LN, TLRLN, and TBRLN was 0.886, 0.866, 0.746, 0.772, 0.648, and 0.731, respectively. The threshold of SUVmax-t was 2.05, and its sensitivity and specificity were 81.0% and 84.2%, respectively. In addition, multivariate logistic regression indicated that TBRtumor was an independent predictor of treatment response (P = 0.03). Conclusion: Our results indicated that [68Ga]Ga-NOTA-GSI PET/CT is a promising tool for predicting early response to combined immunotherapy in gastric cancer patients.
Collapse
Affiliation(s)
- Qiufang Liu
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Xiaoping Xu
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Ziyi Yang
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Jianping Zhang
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Jindian Li
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Ying Qiao
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Silong Hu
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Xiaosheng Liu
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Weijian Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China;
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Shaoli Song
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China;
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| |
Collapse
|
2
|
Roohani B, Mendez AS, Dangarwala M, Katz S, Marquez-Nostra B. Nuclear Imaging of Bispecific Antibodies on the Rise. J Nucl Med 2024; 65:1512-1517. [PMID: 39266295 PMCID: PMC11448611 DOI: 10.2967/jnumed.123.267215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 08/08/2024] [Indexed: 09/14/2024] Open
Abstract
Bispecific antibodies (bsAbs) are engineered to target 2 different epitopes simultaneously. About 75% of the 16 clinically approved bsAbs have entered the clinic internationally since 2022. Hence, research on biomedical imaging of various radiolabeled bsAb scaffolds may serve to improve patient selection for bsAb therapy. Here, we provide a comprehensive overview of recent advances in radiolabeled bsAbs for imaging via PET or SPECT. We compare direct targeting and pretargeting approaches in preclinical and clinical studies in oncologic research. Furthermore, we show preclinical applications of imaging bsAbs in neurodegenerative diseases. Finally, we offer perspectives on the future directions of imaging bsAbs based on their challenges and opportunities.
Collapse
Affiliation(s)
- Borna Roohani
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut; and
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Aldred Shane Mendez
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut; and
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mann Dangarwala
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Samantha Katz
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut; and
| | | |
Collapse
|
3
|
Zhang R, Su C, Jia Y, Xing M, Jin S, Zong H. Molecular mechanisms of HER2-targeted therapy and strategies to overcome the drug resistance in colorectal cancer. Biomed Pharmacother 2024; 179:117363. [PMID: 39236476 DOI: 10.1016/j.biopha.2024.117363] [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: 06/11/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
HER2 amplification is one of the mechanisms that induce drug resistance to anti-EGFR therapy in colorectal cancer. In recent years, data from several randomized clinical trials show that anti-HER2 therapies improved the prognosis of patients with HER2-positive colorectal cancer. These results indicate that HER2 is a promising therapeutic target in advanced colorectal cancer. Despite the anti-HER2 therapies including monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates improving the outcomes, less than 30 % of the patients achieve objective response and eventually have drug resistance. It is necessary to explore the primary and secondary mechanisms for the resistance to anti-HER2 therapies, which will pave the way to overcome the drug resistance. Several studies have reported the potential mechanisms for the resistance to anti-HER2 therapies. In this review, we present a comprehensive overview of the recent advances in clinical research, mechanisms of treatment resistance, and strategies for reversing resistance in HER2-positive colorectal cancer patients.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Chang Su
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Yongliang Jia
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Menglu Xing
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Shuiling Jin
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Hong Zong
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
4
|
van Winkel CAJ, Pierik FR, Brouwers AH, de Groot DJA, de Vries EGE, Lub-de Hooge MN. Molecular imaging supports the development of multispecific cancer antibodies. Nat Rev Clin Oncol 2024:10.1038/s41571-024-00946-3. [PMID: 39327536 DOI: 10.1038/s41571-024-00946-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
Multispecific antibodies are engineered antibody derivatives that can bind to two or more distinct epitopes or antigens. Unlike mixtures of monospecific antibodies, the binding properties of multispecific antibodies enable two specific molecules to be physically linked, a characteristic with important applications in cancer therapy. The field of multispecific antibodies is highly dynamic and expanding rapidly; to date, 15 multispecific antibodies have been approved for clinical use, of which 11 were approved for oncological indications, and more than 100 new antibodies are currently in clinical development. Nevertheless, substantial challenges limit the applications of multispecific antibodies in cancer therapy, particularly inefficient targeting of solid tumours and substantial adverse effects. Both PET and single photon emission CT imaging can reveal the biodistribution and complex pharmacology of radiolabelled multispecific antibodies. This Review summarizes the insights obtained from preclinical and clinical molecular imaging studies of multispecific antibodies, focusing on their structural properties, such as molecular weight, shape, target specificity, affinity and avidity. The opportunities associated with use of molecular imaging studies to support the clinical development of multispecific antibody therapies are also highlighted.
Collapse
Affiliation(s)
- Claudia A J van Winkel
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Frank R Pierik
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Derk Jan A de Groot
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
5
|
Lv G, Hu X, Zhang N, Zhu J, Gao X, Xi H, Peng Y, Xie Q, Qiu L, Lin J. Synthesis and immunotherapy efficacy of a PD-L1 small-molecule inhibitor combined with its 131I-iodide labelled isostructural compound. Bioorg Chem 2024; 153:107810. [PMID: 39276489 DOI: 10.1016/j.bioorg.2024.107810] [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: 07/02/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
Although antibody-based immune checkpoint blockades have been successfully used in antitumor immunotherapy, the low response rate is currently the main problem. In this work, a small-molecule programmed cell death-ligand (PD-L1) inhibitor, LG-12, was developed and radiolabeled with 131I to obtain the chemically and biologically identical radiopharmaceutical [131I]LG-12, which aimed to improve the antitumor effect by combination of LG-12 and [131I]LG-12. LG-12 showed high inhibitory activity to PD-1/PD-L1 interaction. The results of cell uptake and biodistribution studies indicated that [131I]LG-12 could specifically bind to PD-L1 in B16-F10 tumors. It could induce immunogenic cell death and the release of high mobility group box 1 and calreticulin. The combination of [131I]LG-12 and LG-12 could significantly inhibit tumor growth and resulted in enhanced antitumor immune response. This PD-L1 small-molecule inhibitor based combination strategy has great potential for tumor treatment.
Collapse
Affiliation(s)
- Gaochao Lv
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xin Hu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Nan Zhang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Junyi Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoqing Gao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Hongjie Xi
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Ying Peng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Quan Xie
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Ling Qiu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
6
|
Qin S, Yang Y, Zhang J, Yin Y, Liu W, Zhang H, Fan X, Yang M, Yu F. Effective Treatment of SSTR2-Positive Small Cell Lung Cancer Using 211At-Containing Targeted α-Particle Therapy Agent Which Promotes Endogenous Antitumor Immune Response. Mol Pharm 2023; 20:5543-5553. [PMID: 37788300 PMCID: PMC10630944 DOI: 10.1021/acs.molpharmaceut.3c00427] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/05/2023]
Abstract
Small cell lung cancer (SCLC) is a neuroendocrine tumor with a high degree of malignancy. Due to limited treatment options, patients with SCLC have a poor prognosis. We have found, however, that intravenously administered octreotide (Oct) armed with astatine-211 ([211At]SAB-Oct) is effective against a somatostatin receptor 2 (SSTR2)-positive SCLC tumor in SCLC tumor-bearing BALB/c nude mice. In biodistribution analysis, [211At]SAB-Oct achieved the highest concentration in the SCLC tumors up to 3 h after injection as time proceeded. A single intravenous injection of [211At]SAB-Oct (370 kBq) was sufficient to suppress SSTR2-positive SCLC tumor growth in treated mice by inducing DNA double-strand breaks. Additionally, a multitreatment course (370 kBq followed by twice doses of 370 kBq for a total of 1110 kBq) inhibited the growth of the tumor compared to the untreated control group without significant off-target toxicity. Surprisingly, we found that [211At]SAB-Oct could up-regulate the expressions of calreticulin and major histocompatibility complex I (MHC-I) on the tumor cell membrane surface, suggesting that α-particle internal irradiation may activate an endogenous antitumor immune response through the regulation of immune cells in the tumor microenvironment, which could synergically enhance the efficacy of immunotherapy. We conclude that [211At]SAB-Oct is a potential new therapeutic option for SSTR2-positive SCLC.
Collapse
Affiliation(s)
- Shanshan Qin
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Yuanyou Yang
- Key
Laboratory of Radiation Physics and Technology, Ministry of Education,
Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, People’s
Republic of China
| | - Jiajia Zhang
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Yuzhen Yin
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Weihao Liu
- Key
Laboratory of Radiation Physics and Technology, Ministry of Education,
Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, People’s
Republic of China
| | - Han Zhang
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Xin Fan
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Mengdie Yang
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| | - Fei Yu
- Department
of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, People’s Republic of China
- Institute
of Nuclear Medicine, Tongji University School
of Medicine, No. 301
Yan-chang-zhong Road, Shanghai 200072, People’s Republic
of China
| |
Collapse
|
7
|
Hurley K, Cao M, Huang H, Wang Y. Targeted Alpha Therapy (TAT) with Single-Domain Antibodies (Nanobodies). Cancers (Basel) 2023; 15:3493. [PMID: 37444603 DOI: 10.3390/cancers15133493] [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: 05/30/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
The persistent threat of cancer necessitates the development of improved and more efficient therapeutic strategies that limit damage to healthy tissues. Targeted alpha therapy (TαT), a novel form of radioimmuno-therapy (RIT), utilizes a targeting vehicle, commonly antibodies, to deliver high-energy, but short-range, alpha-emitting particles specifically to cancer cells, thereby reducing toxicity to surrounding normal tissues. Although full-length antibodies are often employed as targeting vehicles for TαT, their high molecular weight and the presence of an Fc-region lead to a long blood half-life, increased bone marrow toxicity, and accumulation in other tissues such as the kidney, liver, and spleen. The discovery of single-domain antibodies (sdAbs), or nanobodies, naturally occurring in camelids and sharks, has introduced a novel antigen-specific vehicle for molecular imaging and TαT. Given that nanobodies are the smallest naturally occurring antigen-binding fragments, they exhibit shorter relative blood half-lives, enhanced tumor uptake, and equivalent or superior binding affinity and specificity. Nanobody technology could provide a viable solution for the off-target toxicity observed with full-length antibody-based TαT. Notably, the pharmacokinetic properties of nanobodies align better with the decay characteristics of many short-lived α-emitting radionuclides. This review aims to encapsulate recent advancements in the use of nanobodies as a vehicle for TαT.
Collapse
Affiliation(s)
- Kate Hurley
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Meiyun Cao
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Haiming Huang
- Research Center, Forlong Biotechnology Inc., Suzhou 215004, China
| | - Yi Wang
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
8
|
Wang X, Liu W, Li K, Chen K, He S, Zhang J, Gu B, Xu X, Song S. PET imaging of PARP expression using 68Ga-labelled inhibitors. Eur J Nucl Med Mol Imaging 2023; 50:2606-2620. [PMID: 37145164 PMCID: PMC10317875 DOI: 10.1007/s00259-023-06249-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
PURPOSE Imaging the PARP expression using 18F probes has been approved in clinical trials. Nevertheless, hepatobiliary clearance of both 18F probes hindered their application in monitoring abdominal lesions. Our novel 68Ga-labelled probes aim for fewer abdominal signals while ensuring PARP targeting by optimizing the pharmacokinetic properties of radioactive probes. METHODS Three radioactive probes targeted PARP were designed, synthesized, and evaluated based on the PARP inhibitor Olaparib. These 68Ga-labelled radiotracers were assessed in vitro and in vivo. RESULTS Precursors that did not lose binding affinity for PARP were designed, synthesized, and then labelled with 68Ga in high radiochemical purity (> 97%). The 68Ga-labelled radiotracers were stable. Due to the increased expression of PARP-1 in SK-OV-3 cells, the uptake of the three radiotracers by SK-OV-3 cells was significantly greater than that by A549 cells. PET/CT imaging of the SK-OV-3 models indicated that the tumor uptake of 68Ga-DOTA-Olaparib (0.5 h: 2.83 ± 0.55%ID/g; 1 h: 2.37 ± 0.64%ID/g) was significantly higher than that of the other 68Ga-labelled radiotracers. There was a significant difference in the T/M (tumor-to-muscle) ratios between the unblocked and blocked groups as calculated from the PET/CT images (4.07 ± 1.01 vs. 1.79 ± 0.45, P = 0.0238 < 0.05). Tumor autoradiography revealed high accumulation in tumor tissues, further confirming the above data. PARP-1 expression in the tumor was confirmed by immunochemistry. CONCLUSION As the first 68Ga-labelled PARP inhibitor, 68Ga-DOTA-Olaparib displayed high stability and quick PARP imaging in a tumor model. This compound is thus a promising imaging agent that can be used in a personalized PARP inhibitor treatment regimen.
Collapse
Affiliation(s)
- Xiangwei Wang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032 China
| | - Wei Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032 China
| | - Ke Li
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032 China
| | - Kaiwen Chen
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, China
| | - Simin He
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032 China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032 China
| | - Bingxin Gu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032 China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032 China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032 China
| |
Collapse
|
9
|
Cheng Z, Jin Y, Li J, Shi G, Yu L, Shao B, Tian J, Du Y, Yuan Z. Fibronectin-targeting and metalloproteinase-activatable smart imaging probe for fluorescence imaging and image-guided surgery of breast cancer. J Nanobiotechnology 2023; 21:112. [PMID: 36978072 PMCID: PMC10053476 DOI: 10.1186/s12951-023-01868-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Residual lesions in the tumor bed have been a challenge for conventional white-light breast-conserving surgery. Meanwhile, lung micro-metastasis also requires improved detection methods. Intraoperative accurate identification and elimination of microscopic cancer can improve surgery prognosis. In this study, a smart fibronectin-targeting and metalloproteinase-activatable imaging probe CREKA-GK8-QC is developed. CREKA-GK8-QC possesses an average diameter of 21.7 ± 2.5 nm, excellent MMP-9 protein responsiveness and no obvious cytotoxicity. In vivo experiments demonstrate that NIR-I fluorescence imaging of CREKA-GK8-QC precisely detects orthotopic breast cancer and micro-metastatic lesions (nearly 1 mm) of lungs with excellent imaging contrast ratio and spatial resolution. More notably, fluorescence image-guided surgery facilitates complete resection and avoids residual lesions in the tumor bed, improving survival outcomes. We envision that our newly developed imaging probe shows superior capacity for specific and sensitive targeted imaging, as well as providing guidance for accurate surgical resection of breast cancer.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yushen Jin
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing, 100013, China
| | - Jiaqian Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Guangyuan Shi
- University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Leyi Yu
- Haidian Section of Peking University Third Hospital, Beijing, 100080, China
| | - Bing Shao
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing, 100013, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine Science and Engineering, Beihang University, Beijing, 100191, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100080, China.
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China.
| |
Collapse
|
10
|
Positron emission tomography molecular imaging to monitor anti-tumor systemic response for immune checkpoint inhibitor therapy. Eur J Nucl Med Mol Imaging 2023; 50:1671-1688. [PMID: 36622406 PMCID: PMC10119238 DOI: 10.1007/s00259-022-06084-1] [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: 09/23/2022] [Accepted: 12/08/2022] [Indexed: 01/10/2023]
Abstract
Immune checkpoint inhibitors (ICIs) achieve a milestone in cancer treatment. Despite the great success of ICI, ICI therapy still faces a big challenge due to heterogeneity of tumor, and therapeutic response is complicated by possible immune-related adverse events (irAEs). Therefore, it is critical to assess the systemic immune response elicited by ICI therapy to guide subsequent treatment regimens. Positron emission tomography (PET) molecular imaging is an optimal approach in cancer diagnosis, treatment effect evaluation, follow-up, and prognosis prediction. PET imaging can monitor metabolic changes of immunocytes and specifically identify immuno-biomarkers to reflect systemic immune responses. Here, we briefly review the application of PET molecular imaging to date of systemic immune responses following ICI therapy and the associated rationale.
Collapse
|