Skip to main content

REVIEW article

Front. Oncol., 31 May 2022
Sec. Cancer Imaging and Image-directed Interventions
This article is part of the Research Topic Cellular Metabolism and Molecular Imaging in Cancer View all 7 articles

Advances in Clinical Oncology Research on 99mTc-3PRGD2 SPECT Imaging

  • Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China

The integrin alpha(α)v beta(β)3 receptor is ubiquitous in malignant tumors and has a certain level of specificity for tumors. Technetium-99m hydrazinonicotinamide-dimeric cyclic arginyl-glycyl-aspartic acid peptide with three polyethylene glycol spacers (99mTc-3PRGD2) can bind specifically to the integrin αvβ3 receptor with high selectivity and strong affinity. Thus, it can specifically mark tumors and regions with angiogenesis for tumor detection and be used in single-photon emission computed tomography (SPECT) imaging. This modality has good application value for diagnosing and treating tumor lesions, such as those in the lung, breast, esophagus, head, and neck. This review provides an overview of the current clinical research progress of 99mTc-3PRGD2 SPECT imaging for tumor lesions, including for the diagnosis and differential diagnosis of tumors in different body parts, evaluation of related metastases, and evaluation of efficacy. In addition, the future clinical application prospects and possibilities of 99mTc-3PRGD2 SPECT imaging are further discussed.

Introduction

Cancer is currently the leading cause of death worldwide, with a global estimated 19.3 million new cancer cases and almost 10.0 million cancer deaths recorded in 2020 (1). Early detection, diagnosis, and treatment are key measures for reducing mortality attributed to malignant tumors and prolonging survival time. The integrin alpha(α)v beta(β)3 receptor is frequently involved in the occurrence and development of malignant tumors (2, 3); it mediates cell–cell and cell–extracellular matrix adhesion (4, 5) and is related to tumor angiogenesis and metastasis (3, 68). The integrin αvβ3 receptor is highly expressed in activated endothelial cells and proliferating tumor cells; however, it is either not expressed or expressed at very low levels in normal endothelial cells, dormant vascular cells, and other normal cells (3, 9) and has a certain level of specificity. Therefore, the integrin αvβ3 receptor is a valuable target for diagnosing and treating malignant tumors.

Polypeptides containing the arginine-glycine-aspartate (Arg-Gly-Asp [RGD]) sequence can bind specifically to the integrin αvβ3 receptor with high selectivity and strong affinity (10). Hence, these polypeptides can specifically demarcate lesions and their angiogenesis for tumor detection and have promising prospects for tumor diagnosis and treatment. Radiolabeled RGD peptides and their analogs have been intensively studied for their application in the non-invasive imaging of integrin αvβ3 receptor expression (1113).

The technetium-99m hydrazinonicotinamide-dimeric cyclic RGD peptide with three polyethylene glycol spacers (99mTc-3PRGD2) is a 99mTc-labeled molecular probe used in nuclear medicine for single-photon emission computed tomography (SPECT). Its core ligand, hydrazinonicotinamide-3PRGD2, is a new type of RGD dimer that can bind specifically to the integrin αvβ3 receptor with high selectivity and affinity. In addition, 99mTc-3PRGD2 has rapid blood clearance and a high level of safety with no adverse reactions having been observed in animal models and humans to date (14, 15). 99mTc-3PRGD2 SPECT imaging is widely used in clinical research because of its high diagnostic performance and excellent cost-effectiveness, which further highlight its potential for clinical applications. Herein, we review the advances in clinical research on 99mTc-3PRGD2 SPECT imaging for tumor lesions over the past decade.

Lungs

Globally, lung cancer is the leading cause of cancer-related death (1). Conventional radiography and computed tomography (CT) offer limited accuracy for lung cancer diagnosis and are poor evaluation methods for lung cancer-related metastasis. 2-deoxy-2-(18F)fluoro-D-glucose (FDG) positron emission tomography (PET) has become the standard of care in the initial management of patients with lung cancer, especially for those with non-small cell lung cancer (16). However, false-positive FDG PET/CT results in nodal staging have been shown in patients with coexistent inflammatory or infectious diseases (17, 18). In addition, a study (19) reported that 68Ga-NOTA-PRGD2 PET/CT has similar sensitivity and higher specificity than 18F-FDG PET/CT for the detection and differentiation of lung lesions. Moreover, it is superior to 18F-FDG PET/CT in that it can be used to judge metastatic lymph nodes with higher specificity. Nevertheless, PET is a relatively expensive technology, and PET scanners are not widely available (20). These limitations can interfere with clinical use. 99mTc-3PRGD2 SPECT may be a more advantageous imaging modality.

Diagnosis and Differential Diagnosis

Zhu et al. (21) examined the effectiveness of 99mTc-3PRGD2 SPECT imaging for the assessment of lung cancer and found that with a low 99mTc-3PRGD2 background in the lungs and mediastinum, most lung malignancies were prominent on 1-h images with tumor-to-background ratios significantly higher than those in benign lesions. Furthermore, most lymph-node and bone metastases could also be detected. Thus, their findings suggested that 99mTc-3PRGD2 SPECT imaging is a sensitive tool for detecting lung cancer (sensitivity, 93–97%). Wang et al. (22) also reported that 99mTc-3PRGD2 SPECT/CT has certain value for the diagnosis of benign and malignant lung lesions. In the diagnosis of solitary pulmonary nodules, Ma et al. (23) demonstrated the feasibility of using 99mTc-3PRGD2 SPECT imaging for the non-invasive differential diagnosis of solitary pulmonary nodules. These results showed that the sensitivity of 99mTc-3PRGD2 SPECT imaging for malignant lesions was higher than that of CT, with CT interpretation, SPECT visual analysis, and SPECT semiquantitative analysis showing sensitivity/specificity of 80/67%, 100/67%, and 100/67%, respectively. All solitary pulmonary nodules classified as indeterminate with CT were unequivocally diagnosed with 99mTc-3PRGD2 SPECT imaging. Furthermore, immunohistochemical findings confirmed that integrin αvβ3 was expressed in nodules showing 99mTc-3PRGD2 uptake. Zhang et al. (24) also showed that 99mTc-3PRGD2 SPECT imaging could more accurately detect malignant solitary pulmonary nodules (the sensitivity/specificity of CT interpretation, SPECT visual analysis, and SPECT semiquantitative analysis were 82.4/71.4%, 100/71.4%, and 100/71.4%, respectively). Li et al. (25) showed that chest thin layer CT has an incremental value over 99mTc-3PRGD2 SPECT/CT imaging for the differential diagnosis of benign and malignant pulmonary lesions. In a comparison between the diagnostic performance of 99mTc-3PRGD2 SPECT imaging and 18F-FDG PET/CT imaging for lung tumors, Jin et al. (26) found no significant difference between the two methods (z=0.82; p=0.410). Moreover, their experiment further confirmed that the intratumoral accumulation level of 99mTc-3PRGD2 was positively correlated with integrin αvβ3 expression (r=0.84; p=0.001) and microvessel density (r=0.63; p=0.011).

Evaluation of Metastasis

Miao et al. (27) evaluated the diagnostic performance of 99mTc-3PRGD2 SPECT imaging and conventional 99mTc-MDP bone scanning for bone metastasis in patients with lung cancer. They reported that 99mTc-MDP bone scanning provided better contrast for most lesions, whereas 99mTc-3PRGD2 SPECT imaging appeared to be more effective at excluding pseudo-positive lesions and detecting osteolytic bone metastasis. A study by Shao et al. (28) further confirmed that the targeted binding of 99mTc-3PRGD2 with integrin αvβ3 receptors on tumor cells and osteoclasts offers certain advantages for the early detection of osteolytic bone metastasis in lung cancer (the diagnostic sensitivity of 99mTc-3PRGD2 SPECT imaging and 99mTc-MDP imaging for osteolytic bone metastasis was 84.8% and 25.0%, respectively). Thus, 99mTc-3PRGD2 SPECT imaging can serve as an effective supplement to conventional 99mTc-MDP bone scanning. For the evaluation of lymph-node metastasis in non-small cell lung cancer, Jin et al. (26) demonstrated that 99mTc-3PRGD2 SPECT imaging had a high diagnostic specificity (94.6%), which can compensate for deficiencies in the specificity of 18F-FDG PET/CT, and hence is of significant value for surgical decision-making in lung cancer.

Evaluation of Efficacy and Prognosis

Ma et al. (29) examined the feasibility of using 99mTc-3PRGD2 SPECT imaging to predict the efficacy of chemoradiotherapy plus bevacizumab in the treatment of advanced non-squamous non-small cell lung cancer. Their findings suggested that after two cycles of chemoradiotherapy plus bevacizumab administration, 99mTc-3PRGD2 SPECT imaging could predict the patient’s treatment response and prognosis. A study by Zhang et al. (30) confirmed the feasibility of using 99mTc-3PRGD2 SPECT imaging to evaluate the early response to treatment with epidermal growth factor receptor tyrosine kinase inhibitors among patients with advanced lung adenocarcinoma (sensitivity, 80.0%; specificity, 87.5%), as well as for predicting the prognosis and progression-free survival. Chen et al. (31) confirmed that 99mTc-3PRGD2 SPECT scanning is a promising modality for predicting the tumor response in patients with advanced non-small cell lung cancer early in the course of bevacizumab therapy (sensitivity, 81.8%; specificity, 91.7%; negative predictive value, 84.6%). In addition, Yang et al. (32) explored the use of 99mTc-3PRGD2 SPECT imaging for the evaluation of the clinical effect of recombinant endostatin (Endostar) for the inhibition of tumor angiogenesis. They concluded that since 99mTc-3PRGD2 SPECT imaging compares the lesion T/N value (uptake ratio of the tumor to that of the normal contralateral lung tissue) before and after treatment, as well as the changes in the difference between the T/N values, 99mTc-3PRGD2 SPECT has some clinical value in evaluating the efficacy of recombinant endostatin administration. Moreover, the T/N value was also significantly correlated with patient prognosis (r=0.879, p<0.001).

Breast

Breast cancer is the most common malignancy among women worldwide and a major cause of cancer-related death among women aged 20–59 years (1). Early detection and diagnosis can substantially reduce breast cancer mortality. Conventional mammography is not ideal for identifying malignant tumors and has a high false-negative rate (33, 34). Magnetic resonance imaging and ultrasonography have limited value in differentiating benign abnormalities from malignancies (35). 18F-FDG PET/CT plays a very important role in the diagnosis and treatment of breast cancer (36, 37). Nevertheless, 18F-FDG PET/CT application is significantly restricted owing to high cost and low availability, as PET is a relatively expensive technology and neither PET scanners nor the cyclotrons required to produce isotopes for PET are widely available (20). Therefore, 99mTc-3PRGD2 SPECT imaging could be an excellent alternative.

Diagnosis and Differential Diagnosis

In a study exploring the diagnostic value of 99mTc-3PRGD2 SPECT imaging for palpable and non-palpable breast lesions, Liu et al. (38) found that 99mTc-3PRGD2 SPECT imaging had greater diagnostic value than mammography for the overall evaluation of breast lesions, showing sensitivity, specificity, accuracy, positive predictive value, and negative predictive value of 83/78%, 73/61%, 77/68%, 69/59%, and 85/80%, respectively. This modality may also enable response monitoring of breast cancer therapy through longitudinal imaging. Li et al. (39) conducted a comparative study on the value of 99mTc-3PRGD2 SPECT/CT and conventional ultrasonography in distinguishing benign and malignant breast lesions and proved that 99mTc-3PRGD2 SPECT/CT can distinguish benign and malignant breast lesions from an angiogenesis perspective with anatomical and functional information. Moreover, this modality can further identify benign and malignant lesions to reduce unnecessary punctures when ultrasonography is unavailable. Liu et al. (40) found that the T/NT ratio is related to the TNM staging of breast cancer, lymph-node metastasis, and HER-2. In addition, Ma et al. (41) compared the diagnostic performance of 99mTc-3PRGD2 SPECT imaging and Tc-99m-MIBI SPECT imaging for malignant breast lesions and found that 99mTc-3PRGD2 SPECT imaging did not offer significant advantages for the detection of primary breast cancer over Tc-99m-MIBI SPECT imaging (area under the receiver operating characteristic curve: 0.851 vs. 0.781; p>0.05). Furthermore, based on a comparison between the diagnostic performance of 99mTc-3PRGD2 SPECT imaging and 18F-FDG PET/CT imaging, Chen et al. (42) concluded that 99mTc-3PRGD2 SPECT imaging was useful for the diagnosis and staging of breast cancer, but its sensitivity for detecting small lymph-node metastases appeared to be lower than that of 18F-FDG PET/CT imaging (sensitivity: 78.05% vs. 85.37%; p>0.05). Their study also confirmed that the expression of integrin αvβ3 was correlated with 99mTc-3PRGD2 uptake (r=0.582; p=0.001) and was especially significantly elevated in patients with HER2-positive and stage III–IV disease (p<0.05).

Evaluation of Efficacy

Ji et al. (43) explored the application of 99mTc-3PRGD2 SPECT imaging to evaluate the efficacy of neoadjuvant chemotherapy in patients with stage II and III breast cancer. Their findings indicated that 99mTc-3PRGD2 SPECT imaging could be used to detect changes in tumor pathology and treatment efficacy in this group of patients (sensitivity, specificity, and negative predictive value: 92.9%, 93.3%, and 93.3%, respectively). It was especially advantageous for determining the efficacy of neoadjuvant chemotherapy among patients with HER2-positive breast cancer.

Esophagus

Esophageal cancer is a major public health concern in China (1). Accurate initial staging of esophageal cancer is the key to selecting the best treatment plan for patients, while complete resection of all cancerous lesions is the key to curing this disease. Lymph-node metastasis is usually the first metastasis to occur in esophageal cancer, and the number of metastatic lymph nodes is significantly negatively correlated with the survival rate (44). In current clinical practice, esophageal cancer is commonly diagnosed using CT and endoscopic ultrasonography, which imposes major constraints on the preoperative assessment of lymph-node metastasis (45, 46). Therefore, it is necessary to identify a more accurate approach for the diagnosis of esophageal cancer and preoperative assessment of lymph-node metastasis.

Diagnosis and Differential Diagnosis

Gao et al. (47) evaluated the diagnostic value of 99mTc-3PRGD2 SPECT imaging for esophageal space-occupying lesions and found that 99mTc-3PRGD2 SPECT had clinical potential for the diagnosis of esophageal cancer, with an especially high diagnostic sensitivity for distal lymph-node metastases. Their study also confirmed that 99mTc-3PRGD2 uptake in esophageal cancer was unrelated to the pathological type of the tumor (p>0.05) but was significantly positively correlated with the percentage of integrin αvβ3-positive cells (r=0.976). In addition, Chen et al. (48) and Zheng et al. (49) demonstrated that 99mTc-3PRGD2 SPECT imaging was useful for diagnosing esophageal space-occupying lesions and assessing their angiogenesis.

Preoperative Assessment of Lymph-Node Metastasis

By comparing the diagnostic value of 99mTc-3PRGD2 SPECT imaging with that of CT for lymph-node metastasis in esophageal cancer, Lv et al. (50) showed that 99mTc-3PRGD2 SPECT imaging was more accurate than CT for the preoperative assessment of lymph-node metastasis (sensitivity, specificity, accuracy, positive predictive value, and negative predictive value: 80.95/59.52%, 86.51/73.02%, 85.12/69.64%, 66.67/42.37%, and 93.16/84.40%, respectively) and, therefore, was more helpful for the determination of treatment plans. However, Zheng et al. (49) speculated that 99mTc-3PRGD2 SPECT imaging may be less sensitive than 18F-FDG PET/CT imaging for the detection of metastatic lesions in small lymph nodes.

Head and Neck

Differentiated Thyroid Cancer

Gao et al. (51) assessed the value of 99mTc-3PRGD2 SPECT imaging for the monitoring of differentiated thyroid cancer recurrence. Their results indicated that among patients with differentiated thyroid cancer with high serum thyroglobulin levels and negative radioiodine whole-body scan results, 99mTc-3PRGD2 SPECT showed higher sensitivity (96.6%) and positive predictive value (93.3%) for monitoring recurrence, while the probability of obtaining a positive SPECT finding was related to the thyroglobulin levels (p=0.006). In addition, a study by Zhao et al. (52) revealed that 99mTc-3PRGD2 SPECT imaging could trace radioactive iodine-refractory differentiated thyroid cancer metastases and could be used for the localization and growth evaluation of such lesions. Thus, the study provided a new therapeutic target and new imaging method for monitoring the efficacy of certain anti-angiogenetic therapies.

Choroidal Melanoma

Yan et al. (53) explored the feasibility of employing 99mTc-3PRGD2 SPECT imaging for the diagnosis of choroidal melanoma and monitoring of the tumor response to plaque brachytherapy. Their results indicated that 99mTc-3PRGD2 SPECT imaging is a suitable modality for diagnosing choroidal melanoma and evaluating the accuracy of plaque brachytherapy, showing a significantly lower tumor-to-occipital bone ratio at 3 months post-plaque brachytherapy compared to that at baseline. Furthermore, follow up for a minimum of 20 months after plaque brachytherapy suggested that the co-analysis of 99mTc-3PRGD2 SPECT imaging with the tumor volume may comprise a promising prognostic predictor for patients with choroidal melanoma.

Tumors of the Sellar Region

Hou et al. (54) reported one case of combined 18F-FDG PET/CT and 99mTc-3PRGD2 SPECT/CT imaging for the diagnosis of pituitary metastases where the tumor-to-cerebellum ratio of 99mTc-3PRGD2 SPECT/CT was significantly higher than that of 18F-FDG PET/CT. Thus, they inferred that 99mTc-3PRGD2 SPECT/CT might be useful for differentiating pituitary adenomas from metastases. Further investigations are needed regarding the use of 99mTc-3PRGD2 SPECT imaging for the examination of tumors in the sellar region.

Brain Glioma

Gao et al. (55) conducted a preliminary evaluation of the clinical application of 99mTc-3PRGD2 SPECT imaging for brain gliomas and revealed that the tumor T/N ratio (ratio of abnormal nuclide accumulation to little or no nuclide accumulation) was significantly positively correlated with the percentage of integrin αvβ3-positive cells (R2 = 0.9253; p<0.05). Their results, therefore, confirmed the feasibility of diagnosing brain gliomas using 99mTc-3PRGD2 SPECT imaging.

Other

Jin et al. (56) reported a case of benign metastasizing leiomyoma that showed low uptake on 18F-FDG PET/CT imaging but significant uptake on 99mTc-3PRGD2 SPECT imaging, which they inferred as being attributable to the active angiogenic process in the lesions. Hence, 99mTc-3PRGD2 may have potential diagnostic value for benign metastasizing leiomyoma.

Discussion

Cancer ranks as the leading cause of death and an important barrier to increasing life expectancy worldwide (57). In recent years, PET/CT technology has gradually matured and has been widely used for the diagnosis, staging, and treatment efficacy evaluation of cancer. However, 18F-FDG, the most commonly used PET/CT imaging agent, is not specific for malignant tumors, and non-malignant inflammation and infectious disease can also simulate the high intake of malignant lesions (5861). The tripeptide sequence of RGD can specifically bind to the integrin αvβ3 receptor (10). Accordingly, various radiolabeled RGD-based peptides have been developed for noninvasive imaging of integrin αvβ3 expression (11, 19, 6269). Compared with the tracers for PET, such as 18F-Ga-lacto-RGD (11, 62), 18F-FPPRGD2 (63, 64), and 68Ga-RGD2 (19, 67), 99mTc-3PRGD2 is superior as an easy-labeling, cost-effective SPECT tracer using the generator-produced 99mTc and broadly available SPECT system, rather than relying on an onsite cyclotron and expensive PET system (26). Moreover, the preparation procedure for 99mTc-3PRGD2 is simple, efficient, and reproducible, allowing a kit formulation and easy availability for routine clinical use (15, 70), which can better meet clinical needs.

In the past decade, 99mTc-3PRGD2 SPECT for integrin αvβ3 receptor imaging has made rapid progress in clinical research, and its application for various tumor lesions has been widely demonstrated. 99mTc-3PRGD2 SPECT imaging can be applied to the diagnosis of tumors positive for the integrin αvβ3 receptor, determination of the location and extent of tumors and metastases, monitoring of postoperative residual or recurrent lesions, auxiliary diagnosis of neovascular density, and efficacy and prognostic evaluation of tumor anti-neovascular therapy. With the progress and success of 99mTc-3PRGD2 SPECT imaging for a wide range of clinical applications, it provides a better understanding of the pathophysiological changes and therapeutic responses of lesions, and it can be expected to have a bright prospect for its future use. Considering the good diagnostic performance of 99mTc 3PRGD2 SPECT imaging, it may be possible to non-invasively evaluate some tumoral histopathological subtypes, such as those of non-small cell lung cancer. Concurrently, immunotherapy for malignant tumors is one of the most popular topics at present. It is difficult to predict the response after immunotherapy and evaluate the efficacy of immunotherapy with traditional imaging, but molecular imaging based on the integrin αvβ3 receptor may be an effective means for predicting and evaluating immunotherapy. In addition, non-oncology applications of RGD peptide-based SPECT imaging have been reported in the literature, showing exciting prospects and may become a hot topic for future research (71).

Unfortunately, although rapid progress has been recently made in tumor-related applications of 99mTc-3PRGD2 SPECT imaging, most advances have remained at the clinical trial stage and have numerous shortcomings. The main limitations are the low sensitivity and spatial resolution of the SPECT/CT scanner and the substantial impact of attenuation and scattering, resulting in lower image quality. However, these deficiencies are expected to be resolved to a great extent with the introduction of next-generation SPECT/CT scanners.

In this review, we presented an overview of the current clinical development of 99mTc-3PRGD2 SPECT imaging in oncology and further discussed its potential future clinical applications. With further research and clinical transformation, as well as update and improvement of SPECT imaging equipment and scanning sequences, 99mTc-3PRGD2 SPECT imaging undoubtedly has marked application prospects, and we await further breakthroughs on several grounds, including early diagnosis of tumors, dynamic evaluation of tumor treatment efficacy and prognosis, and individualized guidance for molecular targeted therapy.

Author Contributions

LX drafted the manuscript. JX contributed with discussions and critical revision of the manuscript. Both authors reviewed, commented, and revised the manuscript. Both authors contributed to the article and approved the submitted version.

Funding

This work was supported by the 345 Talent Project from Shengjing Hospital of China Medical University [grant/award number M0441].

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

The authors would like to thank Ming Du and Lu Wang for reading the manuscript and providing insightful suggestions.

References

1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin (2021) 71(3):209–49. doi: 10.3322/caac.21660

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Jin H, Varner J. Integrins. Roles in Cancer Development and as Treatment Targets. Br J Cancer (2004) 90(3):561–5. doi: 10.1038/sj.bjc.6601576

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Niu G, Chen X. Why Integrin as a Primary Target for Imaging and Therapy. Theranostics (2011) 1:30–47. doi: 10.7150/thno/v01p0030

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Serini G, Valdembri D, Bussolino F. Integrins and Angiogenesis: A Sticky Business. Exp Cell Res (2006) 312(5):651–8. doi: 10.1016/j.yexcr.2005.10.020

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Wolfenson H, Iskratsch T, Sheetz MP. Early Events in Cell Spreading as a Model for Quantitative Analysis of Biomechanical Events. Biophys J (2014) 107(11):2508–14. doi: 10.1016/j.bpj.2014.10.041

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Eliceiri BP, Cheresh DA. The Role of Alphav Integrins During Angiogenesis: Insights Into Potential Mechanisms of Action and Clinical Development. J Clin Invest (1999) 103(9):1227–30. doi: 10.1172/JCI6869

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Hood JD, Cheresh DA. Role of Integrins in Cell Invasion and Migration. Nat Rev Cancer (2002) 2(2):91–100. doi: 10.1038/nrc727

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Ruoslahti E. Specialization of Tumour Vasculature. Nat Rev Cancer (2002) 2(2):83–90. doi: 10.1038/nrc724

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Desgrosellier JS, Cheresh DA. Integrins in Cancer: Biological Implications and Therapeutic Opportunities. Nat Rev Cancer (2010) 10(1):9–22. doi: 10.1038/nrc2748

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Haubner R, Wester HJ, Reuning U, Senekowitsch-Schmidtke R, Diefenbach B, Kessler H, et al. Radiolabeled Alpha(V)Beta3 Integrin Antagonists: A New Class of Tracers for Tumor Targeting. J Nucl Med (1999) 40(6):1061–71.

PubMed Abstract | Google Scholar

11. Beer AJ, Haubner R, Sarbia M, Goebel M, Luderschmidt S, Grosu AL, et al. Positron Emission Tomography Using [18F]Galacto-RGD Identifies the Level of Integrin Alpha(V)Beta3 Expression in Man. Clin Cancer Res (2006) 12(13):3942–9. doi: 10.1158/1078-0432.CCR-06-0266

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Zhang X, Xiong Z, Wu Y, Cai W, Tseng JR, Gambhir SS, et al. Quantitative PET Imaging of Tumor Integrin Alphavbeta3 Expression With 18F-FRGD2. J Nucl Med (2006) 47(1):113–21.

PubMed Abstract | Google Scholar

13. Liu Z, Jia B, Shi J, Jin X, Zhao H, Li F, et al. Tumor Uptake of the RGD Dimeric Probe 99mtc-G3-2p4-RGD2 Is Correlated With Integrin Alphavbeta3 Expressed on Both Tumor Cells and Neovasculature. Bioconjug Chem (2010) 21(3):548–55. doi: 10.1021/bc900547d

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Shi J, Wang L, Kim YS, Zhai S, Liu Z, Chen X, et al. Improving Tumor Uptake and Excretion Kinetics of 99mtc-Labeled Cyclic Arginine-Glycine-Aspartic (RGD) Dimers With Triglycine Linkers. J Med Chem (2008) 51(24):7980–90. doi: 10.1021/jm801134k

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Jia B, Liu Z, Zhu Z, Shi J, Jin X, Zhao H, et al. Blood Clearance Kinetics, Biodistribution, and Radiation Dosimetry of a Kit-Formulated Integrin Alphavbeta3-Selective Radiotracer 99mtc-3PRGD2 in Non-Human Primates. Mol Imaging Biol (2011) 13(4):730–6. doi: 10.1007/s11307-010-0385-y

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Chang AJ, Dehdashti F, Bradley JD. The Role of Positron Emission Tomography for Non-Small Cell Lung Cancer. Pract Radiat Oncol (2011) 1(4):282–8. doi: 10.1016/j.prro.2011.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Birim O, Kappetein AP, Stijnen T, Bogers AJ. Meta-Analysis of Positron Emission Tomographic and Computed Tomographic Imaging in Detecting Mediastinal Lymph Node Metastases in Nonsmall Cell Lung Cancer. Ann Thorac Surg (2005) 79(1):375–82. doi: 10.1016/j.athoracsur.2004.06.041

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Bille A, Pelosi E, Skanjeti A, Arena V, Errico L, Borasio P, et al. Preoperative Intrathoracic Lymph Node Staging in Patients With Non-Small-Cell Lung Cancer: Accuracy of Integrated Positron Emission Tomography and Computed Tomography. Eur J Cardiothorac Surg (2009) 36(3):440–5. doi: 10.1016/j.ejcts.2009.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Zheng K, Liang N, Zhang J, Lang L, Zhang W, Li S, et al. 68Ga-NOTA-PRGD2 PET/CT for Integrin Imaging in Patients With Lung Cancer. J Nucl Med (2015) 56(12):1823–7. doi: 10.2967/jnumed.115.160648

PubMed Abstract | CrossRef Full Text | Google Scholar

20. MacManus MP, Hicks RJ, Ball DL, Ciavarella F, Binns D, Hogg A, et al. Imaging With F-18 FDG PET Is Superior to Tl-201 SPECT in the Staging of non-Small Cell Lung Cancer for Radical Radiation Therapy. Australas Radiol (2001) 45(4):483–90. doi: 10.1046/j.1440-1673.2001.00960.x

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Zhu Z, Miao W, Li Q, Dai H, Ma Q, Wang F, et al. 99mtc-3PRGD2 for Integrin Receptor Imaging of Lung Cancer: A Multicenter Study. J Nucl Med (2012) 53(5):716–22. doi: 10.2967/jnumed.111.098988

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Wang S, Liu X, Li M, Luo S, Liu L, Zhang R. Diagnostic Value of 99Tcm-3PRGD2 SPECT/CT in Benign and Malignant Lung Lesions. Chin J Med Imaging Technol (2017) 33(10):1512–6. doi: 10.13929/j.1003-3289.201701074

CrossRef Full Text | Google Scholar

23. Ma Q, Ji B, Jia B, Gao S, Ji T, Wang X, et al. Differential Diagnosis of Solitary Pulmonary Nodules Using 99mtc-3P4-RGD2 Scintigraphy. Eur J Nucl Med Mol Imaging (2011) 38(12):2145–52. doi: 10.1007/s00259-011-1901-2

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Zhang H, Gao S, Chen B, Cheng G. Comparison of the Accuracy of 99mtc-3P4-RGD2 SPECT and CT in Diagnosing Solitary Pulmonary Nodules. Oncol Lett (2016) 12(4):2517–23. doi: 10.3892/ol.2016.5030

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Li Y, He C, He W, Li J, Yang J. Incremental Value of Chest Thin Layer CT Over 99Tcm-3PRGD2 SPECT/CT Imaging in the Diagnosis of Solitary Pulmonary Space Occupying. Chin J Nucl Med Mol Imaging (2020) 40(11):647–51. doi: 10.3760/cma.j.cn321828-20191129-00280

CrossRef Full Text | Google Scholar

26. Jin X, Liang N, Wang M, Meng Y, Jia B, Shi X, et al. Integrin Imaging With 99mtc-3PRGD2 SPECT/CT Shows High Specificity in the Diagnosis of Lymph Node Metastasis From Non-Small Cell Lung Cancer. Radiology (2016) 281(3):958–66. doi: 10.1148/radiol.2016150813

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Miao W, Zheng S, Dai H, Wang F, Jin X, Zhu Z, et al. Comparison of 99mtc-3PRGD2 Integrin Receptor Imaging With 99mtc-MDP Bone Scan in Diagnosis of Bone Metastasis in Patients With Lung Cancer: A Multicenter Study. PloS One (2014) 9(10):e111221. doi: 10.1371/journal.pone.0111221

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Shao G, Cui C, Luo R, Meng Q, Yang R, Liang K, et al. Imaging of Bone Metastasis From Lung Cancer With 99Tcm-3P4-RGD2. Acta Universitatis Med Nanjing (Natural Sci) (2015) 35(11):1659–63. doi: 10.7655/NYDXBNS20151135

CrossRef Full Text | Google Scholar

29. Ma Q, Min K, Wang T, Chen B, Wen Q, Wang F, et al. 99mtc-3PRGD2 SPECT/CT Predicts the Outcome of Advanced Nonsquamous non-Small Cell Lung Cancer Receiving Chemoradiotherapy Plus Bevacizumab. Ann Nucl Med (2015) 29(6):519–27. doi: 10.1007/s12149-015-0975-5

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Zhang Z, Zhao X, Ding C, Wang J, Zhang J, Wang F. 99mtc-3PRGD2 SPECT/CT Imaging for Monitoring Early Response of EGFR-TKIs Therapy in Patients With Advanced-Stage Lung Adenocarcinoma. Cancer Biother Radiopharm (2016) 31(7):238–45. doi: 10.1089/cbr.2016.2052

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Chen B, Zhao G, Ma Q, Ji B, Ji T, Xin H, et al. 99mtc-3P-RGD2 SPECT to Monitor Early Response to Bevacizumab Therapy in Patients With Advanced non-Small Cell Lung Cancer. Int J Clin Exp Pathol (2015) 8(12):16064–72.

PubMed Abstract | Google Scholar

32. Yang Z, Ye L, Fang S, Gu W. Clinical Value of 99mtc-3PRGD2 Molecular Image in Evaluating Effect of Recombinant Endostatin. Cancer Res Prev Treat (2018) 45(05):306–10. doi: 10.3971/j.issn.1000-8578.2018.17.0596

CrossRef Full Text | Google Scholar

33. Murphy IG, Dillon MF, Doherty AO, McDermott EW, Kelly G, O'Higgins N, et al. Analysis of Patients With False Negative Mammography and Symptomatic Breast Carcinoma. J Surg Oncol (2007) 96(6):457–63. doi: 10.1002/jso.20801

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Rosenberg RD, Hunt WC, Williamson MR, Gilliland FD, Wiest PW, Kelsey CA, et al. Effects of Age, Breast Density, Ethnicity, and Estrogen Replacement Therapy on Screening Mammographic Sensitivity and Cancer Stage at Diagnosis: Review of 183,134 Screening Mammograms in Albuquerque, New Mexico. Radiology (1998) 209(2):511–8. doi: 10.1148/radiology.209.2.9807581

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Kolb TM, Lichy J, Newhouse JH. Comparison of the Performance of Screening Mammography, Physical Examination, and Breast US and Evaluation of Factors That Influence Them: An Analysis of 27,825 Patient Evaluations. Radiology (2002) 225(1):165–75. doi: 10.1148/radiol.2251011667

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Berry JD, Cook GJ. Positron Emission Tomography in Oncology. Br Med Bull (2006) 79-80:171–86. doi: 10.1093/bmb/ldl013

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Kumar R, Halanaik D, Malhotra A. Clinical Applications of Positron Emission Tomography-Computed Tomography in Oncology. Indian J Cancer (2010) 47(2):100–19. doi: 10.4103/0019-509X.62997

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Liu L, Song Y, Gao S, Ji T, Zhang H, Ji B, et al. 99mtc-3PRGD2 Scintimammography in Palpable and Nonpalpable Breast Lesions. Mol Imaging (2014) 13(5). doi: 10.2310/7290.2014.00010

CrossRef Full Text | Google Scholar

39. Li W, Liu H, Qin L, Cui Y, Niu J, Wu Z, et al. The Evaluation of 99Tcm-3PRGD2 Integrin Receptor Imaging in the Differential Diagnosis in Benign and Malignant Breast Lesions Comparision With Ultrasound. Int J Radiat Med Nucl Med (2018) 42(03):242–7. doi: 10.3760/cma.j.issn.1673-4114.2018.03.009

CrossRef Full Text | Google Scholar

40. Liu H, Li W, Qin L, Cui Y, Niu J, Wu Z, et al. Relationship Between the Semi-Quantitative Index of 99Tcm-3PRGD2 SPECT/CT Imaging and Clinical Pathological Features of Breast Cancer. Chin J Nucl Med Mol Imaging (2018) 38(12):786–9. doi: 10.3760/cma.j.issn.2095-2848.2018.12.003

CrossRef Full Text | Google Scholar

41. Ma Q, Chen B, Gao S, Ji T, Wen Q, Song Y, et al. 99mtc-3P4-RGD2 Scintimammography in the Assessment of Breast Lesions: Comparative Study With 99mtc-MIBI. PloS One (2014) 9(9):e108349. doi: 10.1371/journal.pone.0108349

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Chen Z, Fu F, Li F, Zhu Z, Yang Y, Chen X, et al. Comparison of [99mtc]3PRGD2 Imaging and [18F]FDG PET/CT in Breast Cancer and Expression of Integrin Alphavbeta3 in Breast Cancer Vascular Endothelial Cells. Mol Imaging Biol (2018) 20(5):846–56. doi: 10.1007/s11307-018-1178-y

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Ji B, Chen B, Wang T, Song Y, Chen M, Ji T, et al. 99mtc-3PRGD2 SPECT to Monitor Early Response to Neoadjuvant Chemotherapy in Stage II and III Breast Cancer. Eur J Nucl Med Mol Imaging (2015) 42(9):1362–70. doi: 10.1007/s00259-015-3062-1

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Mariette C, Piessen G, Briez N, Triboulet JP. The Number of Metastatic Lymph Nodes and the Ratio Between Metastatic and Examined Lymph Nodes are Independent Prognostic Factors in Esophageal Cancer Regardless of Neoadjuvant Chemoradiation or Lymphadenectomy Extent. Ann Surg (2008) 247(2):365–71. doi: 10.1097/SLA.0b013e31815aaadf

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Goei R, Lamers RJS, Engelshove HA, Oei KT. Computed Tomographic Staging of Esophageal Carcinoma: A Study on Interobserver Variation and Correlation With Pathological Findings. Eur J Radiol (1992) 15(1):40–4. doi: 10.1016/0720-048x(92)90201-j

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Zhang X, Watson DI, Lally C, Bessell JR. Endoscopic Ultrasound for Preoperative Staging of Esophageal Carcinoma. Surg Endosc (2005) 19(12):1618–21. doi: 10.1007/s00464-005-0250-2

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Gao S, Ma Q, Wen Q, Jia B, Liu Z, Chen Z, et al. 99mtc-3P4 -RGD2 Radiotracers for SPECT/CT of Esophageal Tumor. Nucl Sci Techniques (2013) 24(04):40–5. doi: 10.13538/j.1001-8042/nst.2013.04.006

CrossRef Full Text | Google Scholar

48. Chen Z, Guo Y, Zhang Y, Li D, Gao S, Ma Q, et al. The Value of 99mtc-3PRGD2-SPECT /CT in the Diagnosis of Space Occupying Disease of Esophagus. Modern Oncol (2014) 22(09):2156–60. doi: 10.3969/j.issn.1672-4992.2014.09.47

CrossRef Full Text | Google Scholar

49. Zheng S, Chen Z, Huang C, Chen Y, Miao W. [99mtc]3PRGD2 for Integrin Receptor Imaging of Esophageal Cancer: A Comparative Study With [18F]FDG PET/Ct. Ann Nucl Med (2019) 33(2):135–43. doi: 10.1007/s12149-018-1315-3

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Lv N, Gao S, Bai L, Ji B, Xue J, Ge X, et al. Advantages of 99mtc-3PRGD2 SPECT Over CT in the Preoperative Assessment of Lymph Node Metastasis in Patients With Esophageal Cancer. Ann Nucl Med (2019) 33(1):39–46. doi: 10.1007/s12149-018-1300-x

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Gao R, Zhang GJ, Wang YB, Liu Y, Wang F, Jia X, et al. Clinical Value of 99mtc-3PRGD2 SPECT/CT in Differentiated Thyroid Carcinoma With Negative 131i Whole-Body Scan and Elevated Thyroglobulin Level. Sci Rep (2018) 8(1):473. doi: 10.1038/s41598-017-19036-9

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Zhao D, Jin X, Li F, Liang J, Lin Y. Integrin Alphavbeta3 Imaging of Radioactive Iodine-Refractory Thyroid Cancer Using 99mtc-3PRGD2. J Nucl Med (2012) 53(12):1872–7. doi: 10.2967/jnumed.112.107821

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Yan B, Fu T, Liu Y, Wei W, Dai H, Fang W, et al. 99mtc-3PRGD2 Single-Photon Emission Computed Tomography/Computed Tomography for the Diagnosis of Choroidal Melanoma: A Preliminary STROBE-Compliant Observational Study. Med (Baltimore) (2018) 97(40):e12441. doi: 10.1097/MD.0000000000012441

CrossRef Full Text | Google Scholar

54. Hou Y, Zhu Z, Jin X, Wang R, Xing B. Combined 18f-FDG PET/CT and 99mtc 3PRGD2 SPECT/CT Imaging in a Case of Pituitary Metastases. Clin Nucl Med (2013) 38(7):550–2. doi: 10.1097/RLU.0b013e318292aa2f

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Gao S, Ma Q, Cui Q, Liu L, Zhou X, Jia B, et al. A Pilot Study on 99mtc-3PRGD2 Scintigraphy in Diagnosis of Brain Glioma. Nucl Sci Techniques (2013) 24(02):36–40. doi: 10.13538/j.1001-8042/nst.2013.02.009

CrossRef Full Text | Google Scholar

56. Jin X, Meng Y, Zhu Z, Jing H, Li F. Elevated 99mtc 3PRGD2 Activity in Benign Metastasizing Leiomyoma. Clin Nucl Med (2013) 38(2):117–9. doi: 10.1097/RLU.0b013e318279f14d

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Bray F, Laversanne M, Weiderpass E, Soerjomataram I. The Ever-Increasing Importance of Cancer as a Leading Cause of Premature Death Worldwide. Cancer (2021) 127(16):3029–30. doi: 10.1002/cncr.33587

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Yu JQ, Kumar R, Xiu Y, Alavi A, Zhuang H. Diffuse FDG Uptake in the Lungs in Aspiration Pneumonia on Positron Emission Tomographic Imaging. Clin Nucl Med (2004) 29(9):567–8. doi: 10.1097/01.rlu.0000134986.58984.5f

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Sim YT, Goh YG, Dempsey MF, Han S, Poon FW. PET-CT Evaluation of Solitary Pulmonary Nodules: Correlation With Maximum Standardized Uptake Value and Pathology. Lung (2013) 191(6):625–32. doi: 10.1007/s00408-013-9500-6

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Cheng CY, Huang WS, Shen DH, Fan YM, Hsu HH, Cherng SC, et al. FDG PET/CT Demonstrated Rapid Progression of Mediastinal Lymphadenopathy in Sarcoidosis. Clin Nucl Med (2007) 32(2):117–21. doi: 10.1097/01.rlu.0000252217.14064.0f

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Shie P, Farukhi I, Hughes RS, Oz OK. Round Pneumonia Mimicking Pulmonary Malignancy on F-18 FDG PET/Ct. Clin Nucl Med (2007) 32(1):55–6. doi: 10.1097/01.rlu.0000249628.58514.fd

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Beer AJ, Haubner R, Goebel M, Luderschmidt S, Spilker ME, Wester HJ, et al. Biodistribution and Pharmacokinetics of the Alphavbeta3-Selective Tracer 18F-Galacto-RGD in Cancer Patients. J Nucl Med (2005) 46(8):1333–41.

PubMed Abstract | Google Scholar

63. Iagaru A, Mosci C, Shen B, Chin FT, Mittra E, Telli ML, et al. 18f-FPPRGD2 PET/CT: Pilot Phase Evaluation of Breast Cancer Patients. Radiology (2014) 273(2):549–59. doi: 10.1148/radiol.14140028

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Mittra ES, Goris ML, Iagaru AH, Kardan A, Burton L, Berganos R, et al. Pilot Pharmacokinetic and Dosimetric Studies of 18F-FPPRGD2: A PET Radiopharmaceutical Agent for Imaging αvβ3 Integrin Levels. Radiology (2011) 260(1):182–91. doi: 10.1148/radiol.11101139

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Yoon HJ, Kang KW, Chun IK, Cho N, Im SA, Jeong S, et al. Correlation of Breast Cancer Subtypes, Based on Estrogen Receptor, Progesterone Receptor, and HER2, With Functional Imaging Parameters From 68Ga-RGD PET/CT and 18F-FDG PET/Ct. Eur J Nucl Med Mol Imaging (2014) 41(8):1534–43. doi: 10.1007/s00259-014-2744-4

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Doss M, Kolb HC, Zhang JJ, Belanger MJ, Stubbs JB, Stabin MG, et al. Biodistribution and Radiation Dosimetry of the Integrin Marker 18F-RGD-K5 Determined From Whole-Body PET/CT in Monkeys and Humans. J Nucl Med (2012) 53(5):787–95. doi: 10.2967/jnumed.111.088955

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Kang F, Wang Z, Li G, Wang S, Liu D, Zhang M, et al. Inter-Heterogeneity and Intra-Heterogeneity of Alphavbeta3 in Non-Small Cell Lung Cancer and Small Cell Lung Cancer Patients as Revealed by 68Ga-RGD2 PET Imaging. Eur J Nucl Med Mol Imaging (2017) 44(9):1520–8. doi: 10.1007/s00259-017-3696-2

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Bach-Gansmo T, Danielsson R, Saracco A, Wilczek B, Bogsrud TV, Fangberget A, et al. Integrin Receptor Imaging of Breast Cancer: A Proof-of-Concept Study to Evaluate 99mtc-NC100692. J Nucl Med (2006) 47(9):1434–9.

PubMed Abstract | Google Scholar

69. McParland BJ, Miller MP, Spinks TJ, Kenny LM, Osman S, Khela MK, et al. The Biodistribution and Radiation Dosimetry of the Arg-Gly-Asp Peptide 18F-AH111585 in Healthy Volunteers. J Nucl Med (2008) 49(10):1664–7. doi: 10.2967/jnumed.108.052126

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Wang L, Shi J, Kim YS, Zhai S, Jia B, Zhao H, et al. Improving Tumor-Targeting Capability and Pharmacokinetics of 99mtc-Labeled Cyclic RGD Dimers With PEG4 Linkers. Mol Pharm (2009) 6(1):231–45. doi: 10.1021/mp800150r

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Ebenhan T, Kleynhans J, Zeevaart JR, Jeong JM, Sathekge M. Non-Oncological Applications of RGD-Based Single-Photon Emission Tomography and Positron Emission Tomography Agents. Eur J Nucl Med Mol Imaging (2021) 48(5):1414–33. doi: 10.1007/s00259-020-04975-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: 99mTc-3PRGD2, integrin αvβ3 receptor, oncology, single-photon emission computed tomography (SPECT), molecular imaging

Citation: Xiao L and Xin J (2022) Advances in Clinical Oncology Research on 99mTc-3PRGD2 SPECT Imaging. Front. Oncol. 12:898764. doi: 10.3389/fonc.2022.898764

Received: 17 March 2022; Accepted: 05 May 2022;
Published: 31 May 2022.

Edited by:

Bradley T Scroggins, National Cancer Institute (NIH), United States

Reviewed by:

Lei Kang, First Hospital, Peking University, China
Kun Zheng, Peking Union Medical College Hospital (CAMS), China

Copyright © 2022 Xiao and Xin. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jun Xin, xinj@sj-hospital.org

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.