Next Article in Journal
Genetic Polymorphisms and Genetic Risk Scores Contribute to the Risk of Coronary Artery Disease (CAD) in a North Indian Population
Previous Article in Journal
Exploring the Utility of Long Non-Coding RNAs for Assessing the Health Consequences of Vaping
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

EGFR- and Integrin αVβ3-Targeting Peptides as Potential Radiometal-Labeled Radiopharmaceuticals for Cancer Theranostics

by
Cibele Rodrigues Toledo
1,
Ahmed A. Tantawy
1,2,
Leonardo Lima Fuscaldi
3,
Luciana Malavolta
3 and
Carolina de Aguiar Ferreira
1,4,5,6,*
1
The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
2
Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
3
Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo 01221-020, Brazil
4
Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
5
Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
6
Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(15), 8553; https://doi.org/10.3390/ijms25158553
Submission received: 26 June 2024 / Revised: 29 July 2024 / Accepted: 30 July 2024 / Published: 5 August 2024
(This article belongs to the Special Issue Targeted Radioligand Therapy and Immunotherapy for Cancer Treatment)

Abstract

:
The burgeoning field of cancer theranostics has witnessed advancements through the development of targeted molecular agents, particularly peptides. These agents exploit the overexpression or mutations of specific receptors, such as the Epidermal Growth Factor receptor (EGFR) and αVβ3 integrin, which are pivotal in tumor growth, angiogenesis, and metastasis. Despite the extensive research into and promising outcomes associated with antibody-based therapies, peptides offer a compelling alternative due to their smaller size, ease of modification, and rapid bioavailability, factors which potentially enhance tumor penetration and reduce systemic toxicity. However, the application of peptides in clinical settings has challenges. Their lower binding affinity and rapid clearance from the bloodstream compared to antibodies often limit their therapeutic efficacy and diagnostic accuracy. This overview sets the stage for a comprehensive review of the current research landscape as it relates to EGFR- and integrin αVβ3-targeting peptides. We aim to delve into their synthesis, radiolabeling techniques, and preclinical and clinical evaluations, highlighting their potential and limitations in cancer theranostics. This review not only synthesizes the extant literature to outline the advancements in peptide-based agents targeting EGFR and integrin αVβ3 but also identifies critical gaps that could inform future research directions. By addressing these gaps, we contribute to the broader discourse on enhancing the diagnostic precision and therapeutic outcomes of cancer treatments.

1. Introduction

The area of cancer theranostics has seen remarkable progress, with the development of several targeting agents, particularly peptides. These molecules exploit the upregulation of specific receptors, such as the Epidermal Growth Factor receptor (EGFR) and integrin αVβ3. These receptors recognize specific amino acid sequences like tyrosine kinase domain-targeting peptides and RGD (Arg-Gly-Asp) derivatives, respectively, making them prime targets for therapeutic and diagnostic applications [1,2].
Peptides are particularly notable in this context due to their low molecular weight, generally consisting of up to 50 amino acid residues. This attribute allows for enhanced penetration into tumor tissues and swift clearance from the bloodstream and non-targeted tissues, which improves imaging accuracy and reduces treatment-related toxicity. Additionally, peptides are characterized by low antigenicity, and they can undergo chemical modifications during the radiolabeling process without losing activity, further enhancing their suitability for theranostic applications [3,4]. Over the past decades, the identification and utilization of peptides that target EGFR and integrin αVβ3 have significantly increased.
In this comprehensive review, we provide a thorough summary of the current literature concerning the utilization of radiolabeled EGFR- and integrin αVβ3-targeting peptides in cancer imaging and peptide receptor radionuclide therapy (PRRT). We discuss the various EGFR- and integrin αVβ3-targeting peptides developed to date, emphasizing their efficacy as targeting ligands and outlining their advantages and disadvantages when compared to alternative targeting agents. Our discussion extends to their applications in both preclinical and clinical studies, whether labeled with common imaging radionuclides for positron emission tomography (PET) and single-photon emission computed tomography (SPECT) or with therapeutic radionuclides for cancer radionuclide therapy. Lastly, we assess the challenges associated with translating these radiolabeled peptides to clinical use and contemplate the prospects for future research in this field.

2. EGFR and Integrin αVβ3 as Cancer Targets

The EGFR, also called ERBB-1 (Erythroblastic Leukemia Viral Oncogene Homologue-1) or HER-1 (Human Epidermal Growth Factor Receptor-1), is a 170 kDa single-chain transmembrane glycoprotein belonging to the ERBB family of receptor tyrosine kinases. EGFR presents three domains: (a) a hydrophobic transmembrane domain, which is associated with dimerization interaction among receptors; (b) an intracellular tyrosine kinase domain responsible for substrate phosphorylation; and (c) an extracellular EGFR-binding domain (sEGFR), which binds ligands which stimulate EGFR [5,6]. The sEGFR consists of four domains (I, II, III and IV), and exhibits two conformations: an open conformation when bound to a ligand and a closed, auto-inhibited conformation [1]. Ligand binding induces conformational changes, exposing a dimerization arm that facilitates the formation of homodimers and heterodimers with other ERBB family members. This dimerization activates the kinase domain, which consists of two lobes, with a cleft between them containing four critical structural sites: (1) the catalytic region, (2) the hinge site, (3) the activation loop, and (4) the kinase specificity pocket [7,8,9,10].
The aberrant activation of those EGFR signaling pathways, due to factors like cell-surface overexpression, autocrine activation, and mutations in the EGFR gene, is observed in various cancers, including lung, head and neck, cervical, colorectal, and brain cancers [11]. Among the various EGFR mutations, the most prevalent is EGFRvIII, an oncogene formed by a large extracellular deletion [12].
While EGFR itself is a critical target in cancer therapy due to its overexpression and mutations in various tumors, it is essential to recognize that the downstream signaling pathways activated by EGFR are the primary drivers of cancer progression. EGFR activation triggers two key signaling cascades in cancer: the PI3K/AKT/mTOR and MAPK pathways. The PI3K/AKT/mTOR pathway regulates cell growth, proliferation, and survival [13,14], while the MAPK pathway controls gene expression and cell cycle progression [11,15]. Both pathways are frequently dysregulated in cancers, contributing to uncontrolled growth and therapy resistance. NF-κβ [16] and GTPase path signaling are also activated [17]. The result of these intracellular pathways is the increased transcription of genes responsible for cell growth, survival, and/or migration [18]. Significant crosstalk between these pathways can lead to compensatory mechanisms, making single-pathway inhibition often ineffective. Understanding these interactions is crucial for developing combination therapies that target multiple pathways simultaneously to overcome resistance and improve cancer treatment outcomes.
On the other hand, integrins are a family of transmembrane cell surface receptors that mediate cell–matrix and cell–cell interactions [19]. These heterodimeric glycoproteins consist of noncovalently connected α and β subunits. In mammals, 18 α and 8 β subunits combine to form at least 24 distinct integrin heterodimers [20,21]. Each integrin subunit has a large extracellular domain (750–1000 residues), a single transmembrane domain, and a short cytoplasmatic domain [22,23]. Integrins bind to specific endogenous ligands, including cell surface counter-receptors, soluble ligands, and extracellular matrix (ECM) proteins [24]. Eight integrin heterodimers can recognize RGD-containing peptides [25]. Ligand binding triggers interactions between the cytoplasmatic domain, intracellular proteins, and cytoskeletal filaments, initiating signaling pathways through the Src kinases, which are activated by FAK phosphorylation [26]. This signaling controls key cellular functions, including adhesion, migration, proliferation, differentiation, and survival, that are critical for tissue homeostasis, development, and repair [27]. Integrin dysregulation is involved in the pathogenesis of several diseases characterized by altered angiogenesis, inflammation, or infection [28].
Integrin αVβ3, also known as the vitronectin receptor, is of particular interest in cancer research. While αVβ3 is expressed at low levels or is undetectable in most healthy adult epithelia, it is overexpressed in various tumor cells (breast, lung, glioblastoma, prostate, and melanoma), the tumor-associated vasculature, and invasive tumor fronts [23]. Interactions between αVβ3 and its ECM ligands activate intracellular signaling pathways like PI3K/Akt and ERK/MAPK, promoting cancer progression. The engagement of αVβ3 with the ECM also facilitates crosstalk with growth factor receptors and proteases to further enhance tumor cell survival and motility [29]. Notably, αVβ3 expression is associated with the acquisition of a cancer stem cell phenotype, conferring tumors with enhanced initiation and therapy resistance capabilities [30].
Therefore, targeting EGFR and/or integrin αVβ3 with specific inhibitors or antibodies/small molecules can disrupt these signaling pathways, inhibiting tumor growth and spread.

3. EGFR- and Integrin αvβ3-Targeting Peptides

3.1. EGFR-Targeting Peptides

Several EGFR-targeting peptides have been identified and developed as potential diagnostic and therapeutic agents. EGFR peptides can be obtained from experimental synthesis, phage display libraries, computer-aided design, and natural sources. The most widely explored peptide is GE11 [31,32,33,34,35,36,37,38]. GE11 is a dodecapeptide that is identified through phage display screening. It exhibits high affinity and selectivity for EGFR. It binds to EGFR with a dissociation constant (Kd) of 22 nM, which is lower than that of the natural ligand EGF (Kd = 2 nM) but significantly higher than seen for non-specific binding to proteins like bovine serum albumin. This lower affinity compared to EGF is attributed to GE11’s smaller size and its binding to only one EGFR region, in contrast to EGF’s interaction with multiple domains [18]. De Paiva et al. [39] identified the binding site between GE11 and sEGFR using molecular dynamics and molecular docking simulation. According to these researchers, GE11 acts as an inhibitor. The optimal conformation of GE11 and sEGFR occurs at domains II and IV, which may block the exposure of the dimerization arm and prevent dimer formation [1,40]. Importantly, while GE11 binds to EGFR with high specificity, it does not exhibit mitogenic activity. This characteristic makes it an attractive option for targeted drug delivery without stimulating tumor growth. Recent studies have shown that GE11 can enhance nanoparticle endocytosis through an alternative EGFR-dependent, actin-driven pathway [18]. This mechanism allows for the maintenance of EGFR levels on the cell surface after GE11 binding, potentially enabling prolonged receptivity to GE11-conjugated therapeutics. Other peptides like EBP (CMYIEALDKYAC) and D4 (LARLLT) have also shown promise in EGFR targeting. EBP was experimentally synthesized and demonstrated high affinity for EGFR, while D4 was developed through computer-aided design approaches [1]. Recent research by Tripathi et al. (2024) [16] demonstrated the anticancer potential of short peptides, derived from the conserved regions of the MIEN1 protein. Their study highlighted that a six-amino-acid peptide, LA3IK, effectively inhibited EGF-mediated NF-kB nuclear translocation in breast cancer cells. This finding underscores the therapeutic promise of targeting the MIEN1 signaling pathway to impede cancer progression.
In addition to targeting wild-type EGFR, there has been significant progress in developing peptides that are specific to EGFR mutations, such as EGFRvIII, which is commonly associated with aggressive cancers like glioblastoma. EGFRvIII, characterized by the deletion of exons 2–7, leads to a constitutively active receptor that drives tumorigenesis. Recent studies have utilized phage display technology to identify cyclic peptides that selectively bind to EGFRvIII. For instance, novel cyclic peptides P6 and P9 have shown high specificity for EGFRvIII-expressing cells, enhancing targeted drug delivery and cytotoxicity in non-small-cell lung cancer (NSCLC) and glioblastoma models [41]. These advancements underscore the potential of peptide-based therapies to effectively target specific EGFR mutations, offering new avenues for precision oncology.
These peptides, along with others listed in Table 1, represent a diverse array of EGFR-targeting strategies, each with unique binding properties and potential applications in cancer diagnostics and therapeutics.

3.2. Integrin αvβ3-Targeting Peptides

Peptides containing the Arg-Gly-Asp (RGD) motif are the most studied αVβ3 ligands [19]. Table 2 summarizes some of the integrin αVβ3-targeting peptides explored in cancer research.

4. Radiolabeled Peptides as Valuable Tools for Imaging and the Treatment of Cancer

The process of radiolabeling peptides involves attaching radioactive isotopes to peptides, which can then be detected using imaging techniques to provide real-time, non-invasive insights into the molecular environment of tumors. This capability not only aids in the accurate diagnosis and staging of cancer but also facilitates the monitoring of treatment responses and the detection of metastases. Furthermore, when these radiolabeled peptides are designed to carry therapeutic radionuclides, they serve a dual function by also providing targeted radionuclide therapy, delivering cytotoxic radiation directly to tumor cells and thereby reducing the tumor burden while sparing normal tissues [60].

4.1. Strategies for Radiolabeling Peptides

Peptide radiolabeling employs two primary methods: direct and indirect labeling (Figure 1).

4.1.1. Direct Labeling

The radioisotope is covalently attached to the peptide. This is commonly performed with radioiodines like iodine-125 (125I, t1/2 = 59.4 days; Eγ = 35 keV) and iodine-131 (131I, t1/2 = 8 days; 90% β = 606 keV) via the electrophilic radioiodination of the tyrosine side chain aromatic ring. Oxidizing agents such as Chloramine T or Iodo-Gen® are used to convert iodide into an electrophilic iodate that is substituted onto the tyrosine’s aromatic ring at room temperature [61]. This procedure offers the advantage of not modifying the amino acid sequence. Examples of direct iodination include 125I-labeling of GE11 [31] and 131I labeling of GRGDYV [62].
Direct 99mTc- (t1/2 = 6 h; Eγ = 140 keV) labeling is also performed for peptides with disulfide bonds or via the formation of a [99mTc(CO)3]+ complex that binds the histidine side chain imidazole ring. This two-step approach first makes the [99mTc(H2O)3(CO)3]+ core, which then labels the histidine to form a stable complex, as demonstrated for GRGDHV [62]. The study of Baishya and coworkers evaluated two [99mTc(CO)3]+-labeled tetrapeptides and one [99mTc(CO)3]+-labeled hexapeptide by changing the amino acid sequence of the RGD motif for potential use as tumor-targeting radiopharmaceuticals [63]. Comparative in vivo studies of [99mTc(CO)3]+-labeled PEGylated and non-PEGylated cRGDfK demonstrated that the addition of a PEG7 unit increased the melanoma tumor uptake and slowed the clearance from other organs, decreasing target-to-background ratios [64]. However, 99mTc peptide labeling more often uses indirect methods.

4.1.2. Indirect Labeling

The bifunctional chelator or prosthetic group is attached to the peptide, which then complexes with the radiometal. Typically, the chelator is coupled to the peptide first to simplify the radiosynthesis. Linkers can also be added between the chelator and peptide [65].
For integrin αVβ3 imaging, 18F (t1/2 = 109.7 min; 97% β+; Eβ+max = 635 keV) has been used most, followed by 68Ga and 64Cu. 18F-Galacto-RGD was the first RGD PET tracer in humans [66,67], and it was made by conjugating a sugar amino acid to c(RGDfK) and labeling with 18F-fluoropropionate. This radiolabeling process is time-consuming, which presents a hurdle for large-scale clinical studies. Similar challenges are observed when labeling other tracers like 18F-FPPRGD and 18F-RGD [25,66,67,68,69,70]. Other 18F-RGD tracers, such as 18F-FPPRGD and 18F-Alfatide I/II, provide a more efficient production process with easier and faster radiosynthesis (40 min and 20 min for kit radiolabeling) and higher yields (42%, radiochemical purity > 95%) [69].
As an alternative to 18F in peptide labeling, 68Ga presents advantageous physical characteristics. 68Ga (t1/2 = 68 min; 89% β+; Eβ+max = 1920 keV) can be eluted from an in-house 68Ge/68Ga generator and its half-life aligns well with the pharmacokinetics of several peptides. Many chelators, such as DOTA, NODAGA, NOTA, 1,4,7-triazacyclononane-1,4,7-tris[(2-carboxyethyl)methylenephosphinic acid (TRAP), and tris-hydroxypyridinone (THP), form stable complexes with 68Ga, allowing for labeling in short reaction times, often at room temperature [25,69,71,72,73]. A study conducted by Lang et al. (2011) demonstrated that, for the same chelator–peptide complex (NOTA-PRGD2), the labeling yield required to form a 68Ga complex was higher than that needed to form a 18FAl complex [67,74].
64Cu (t1/2 = 12.7 h; 19% β+, Eβ+max = 656 keV; 38% β, Eβmax = 573 keV; 43% EC) also stands out as an attractive radionuclide for PET imaging because of its favorable decay characteristics, coupled with the ability to produce it at high specific activity levels on small biomedical cyclotrons [75].
Concerning EGFR-targeting peptides, most radiolabeling approaches were performed using 99mTc [33,34,76,77,78].
Over 80% of the radiopharmaceuticals utilized for SPECT imaging rely on 99mTc-labeled compounds due to their favorable nuclear properties and widespread availability through 99Mo/99mTc generators at a low cost. With a half-life of 6 h, 99mTc allows radiopharmacists ample time for radiosynthesis, while still permitting physicians to obtain clinically pertinent images. Among the chelating agents employed for the labeling of 99mTc compounds, HYNIC (6-hydrazinonicotinic acid) is the most extensively utilized [33,76,78].
Table 3, Table 4, Table 5 and Table 6 offer an overview of the different radiolabeling strategies used for EGFR- and integrin αVβ3-targeting peptides.

5. Preclinical Studies of Radiolabeled EGFR and Integrin αvβ3-Targeting Peptides

5.1. EGFR-Targeting Peptides

The GE11 peptide has been labeled with various radionuclides, including 18F [32,79], 111In [79], 64Cu [36,81,132], 99mTc [33,34,37], and 68Ga [82,133,134,135]. However, its EGFR-targeting efficacy is controversial. Some studies reported good binding affinity and tumor uptake in cell lines and murine models [31,32,33,34,37]. Figure 2 illustrates dynamic PET/CT images of [18F]F-FP-Lys-GE11 from U-87 MG tumor-bearing mice, time–activity curves, and tumor/organ rates.
Other studies cast doubt on GE11’s EGFR affinity [36,82]. Striese et al. [36,82] and Judmann et al. [36,82] attributed this lack of targeting efficacy to GE11’s high hydrophobicity, which may cause peptide aggregation and limit its interactions with EGFR. They also proposed that the cell uptake reported in other GE11 studies may be facilitated by highly hydrophilic linkers or constructs with multiple peptide copies, suggesting that developing small-molecule GE11-based radioligands may not be a promising approach to obtaining alternatives to GE11. Few attempts have been made in terms of other peptide scaffolds [50,76,77,78,83]. Table 7 summarizes all the collected preclinical data.

Use of EGFR-Targeting Peptides for Therapeutic Purposes

No studies have yet been conducted that utilize radiotherapeutic isotopes for the treatment of conditions via EGFR-targeting peptides. This gap in the research might be attributed to the controversial efficacy of EGFR targeting itself. This inconsistency in the efficacy of EGFR targeting could potentially prevent the development of therapeutic applications involving radiotherapeutic isotopes, as the foundational premise of specific and effective EGFR targeting remains under debate [36,82].

5.2. Integrin αVβ3-Targeting Peptides

The development of radiotracers for imaging purposes employs various strategies [136]. The multimerization of the cRGD scaffold is an approach that leverages the polyvalence effect to increase the binding affinity to integrin αVβ3. This method suggests that radiotracers derived from multimeric peptides exhibit higher tumor uptake and better tumor/background ratios than their monomeric counterparts [104,137,138]. However, this can also lead to increased non-specific uptake in non-target tissues [52]. Guo et al. [139] showed that the 18F-labeled RGD dimer ([18F]F-FP-PRGD2) had a greater binding affinity than the monomer ([18F]F-FP-RGD) in mice bearing MDA-MB-435 tumor xenografts (difference in %ID/g uptake: RGD2/RGD~1.5, p = 0.0045). Chen et al. [68] also demonstrated the higher tumor uptake of [18F]F-FPRGD2 than [18F]F-FPRGD at all time points in a glioblastoma xenograft mouse model. The dimeric tracer showed predominantly renal excretion, while the monomeric tracer was excreted primarily through the biliary route, resulting in higher tumor/background ratios [69].
Pegylation has been shown to extend the radiotracer’s circulation time and modulate its clearance, without affecting rapid elimination from the liver and kidneys [25,67]. Glycosylation, involving the conjugation of a sugar amino acid to the peptide structure, also enhances hydrophilicity and blood circulation time. One notable example of a glycosylated radiotracer is [18F]F-Galacto-RGD, which demonstrates significant tumor uptake, fast blood clearance, and predominantly renal elimination [25,66,140].
In preclinical studies, RGD radiotracers have been evaluated in cancer models of breast cancer, lung cancer, melanoma, and especially glioblastoma. The U-87 MG cell line is the most reported among xenograft models [141]. Figure 3 illustrates the preclinical study conducted by Oxboel et al. [87], who used radiolabeled E{c(RGDFk)2 to image U-87 MG and H727 tumors.
Relevant preclinical studies performed with radiotracers targeting integrin αVβ3 are shown in Table 8 and Table 9.

Use of Integrin αVβ3-Targeting Peptides for Therapeutic Purposes

The radioisotopes that are mainly in use are the β emitters, like Lutetium-177 (177Lu) and Yttrium-90 (90Y) (Table 10) [125,126,127,128,129,130,131]. 177Lu emits both β particles and γ rays, which allows, respectively, radionuclide therapy and SPECT imaging to confirm the distribution and localization of the therapeutic agent. 177Lu has a half-life of 6.7 d and a tissue penetration range of about 2 mm, making it suitable for treating small- to medium-sized tumors. Conversely, 90Y is a pure β emitter with a higher energy and longer range (approximately 11 mm) than 177Lu. These characteristics make it effective in treating larger tumors. However, its shorter half-life of 2.7 d requires more precise timing in clinical applications [129,144]. The efficacy of 177Lu- and 90Y-labeled integrin αVβ3-targeting peptides in animal models has shown promising results. However, some challenges need to be addressed to enhance the efficacy and safety of these therapies: the heterogeneity of tumor expression, the development of resistance, and radiation toxicity (this can affect surrounding healthy tissues) [145]. Further research is needed to explore the use of other therapeutic radioisotopes or hybrid peptides that might offer better therapeutic profiles.

6. Clinical Studies

Table 11 summarizes some clinical studies performed using radiolabeled RGD-based peptides for cancer imaging and therapy. These trials show promising efficacy in terms of tumor detection, staging, and monitoring treatment responses, highlighting their potential in enhancing the precision of cancer diagnostics and therapy.

7. Dual-Targeting Peptides

Achieving optimal single-target tumor imaging and therapy is challenging due to receptor heterogeneity, low binding affinities, and suboptimal in vivo pharmacokinetics. These limitations hinder the generation of high-quality diagnostic images and the effective application of monomeric radiopeptides [82]. The solution is the development of heterodimeric peptides (HPs) that link two distinct specific peptide ligands [160]. To date, only a few HPs for bispecific EGFR and integrin αVβ3 targeting have been described [132,133,135,161], highlighting the novelty and limited availability of such agents.
Yu et al. [133] designed [68Ga]Ga-NOTA-RGD-Cys-6-Ahx-GE11. Chen et al. [161] subsequently investigated the in vitro and in vivo properties of this radiotracer, comparing it with those of monomeric radiopeptides. The superiority of HPs was noticed in terms of binding affinities and tumor uptake in biodistribution and PET/CT imaging studies, as shown in Figure 4. Specifically, in the PET/CT imaging study at 2 h p.i., the tumor uptake values and tumor/muscle ratios were 3.5 ± 0.6%ID/g and 4.4 ± 1.0 for HPs, 2.4 ± 0.3%ID/g and 2.9 ± 0.7 for [68Ga]Ga-NOTA-GE11, and 2.8 ± 0.5%ID/g and 3.1 ± 0.7 for [68Ga]Ga-NOTA-RGD, respectively. However, in all cases, the liver and kidneys presented high activity levels, highlighting the need for further ligand structure modifications to achieve better pharmacokinetics [161,162].
Braun et al. [135] refined the radiotracer developed by Chen et al. [161]. They replaced the cysteine building block with (NH2-propyl)2Gly-OH to achieve a more uniform structure and used NODA as the chelator. PEG spacers were also incorporated. [68Ga]Ga-NODA-(PEG3-GE11-PEG3-c(RGDyK)) and [68Ga]Ga-NODA-(PEG5-GE11-PEG5-c(RGDfK)) were synthesized. In vitro cell (A431) uptake studies demonstrated favorable integrin αVβ3-specific receptor affinities for these two bispecific agents. However, they did not exhibit receptor-specific interactions with the EGFR in the in vitro studies. These in vitro findings were corroborated by PET/CT imaging in tumor-bearing mice, which showed that the observed tumor uptake was only mediated by integrin αVβ3 and not by EGFR binding [135].
In the same year, Li et al. [132] synthesized [64Cu]Cu-NOTA-RGD-GE11. The bispecific agent was compared to its monomeric units. [64Cu]Cu-NOTA-RGD-GE11 demonstrated significantly enhanced tumor uptake (4.6 ± 0.2%ID/g) compared to monomeric agents (1.2 ± 0.2%ID/g for [64Cu]Cu-NOTA-RGD and 0.8 ± 0.1%ID/g for [64Cu]Cu-NOTA-GE11) at 2 h p.i. in mice bearing BxPC3 xenografts, as shown in Figure 5. The tumor uptake of the HPs was effectively inhibited in the presence of both non-radioactive c(RGDyK) and GE11, suggesting that both peptides exhibited receptor-specific interactions with their respective targets. These findings underscore the potential of dual-targeting peptides in improving the specificity and effectiveness of cancer therapeutics and diagnostics, paving the way for future clinical applications.

8. Conclusions and Future Directions

Herein, we undertook a comprehensive review of the development and application of EGFR- and integrin αVβ3-targeting peptides as potential radiometal-labeled radiopharmaceuticals for cancer theranostics. The use of these peptides in both diagnostic and therapeutic contexts offers a dual benefit by enabling precise tumor imaging and targeted therapy, potentially leading to better patient outcomes.
Despite the promising aspects of peptide-based radiopharmaceuticals, several challenges and limitations persist. One major challenge is the inherent lower binding affinity and the rapid clearance from the bloodstream of peptides, which can limit their therapeutic efficacy and diagnostic accuracy. Also, the renal toxicity associated with radiometal-labeled peptides accumulation poses a significant concern for patient safety. The small size of peptides, while beneficial for tumor penetration, also contributes to their rapid degradation and clearance, necessitating frequent or higher dosages.
To overcome these challenges, several emerging strategies and technologies are being explored. One approach is the modification of peptides to enhance their stability and binding affinity. This includes the use of cyclization, PEGylation, and the incorporation of non-natural amino acids, which can improve metabolic stability and reduce renal clearance. Another strategy is the development of multivalent and multimeric peptide systems that can increase functional affinity and selectivity towards target receptors. Additionally, the exploration of alternative targeting moieties, such as small-molecule ligands or scaffold proteins, offers a potential avenue for increasing the therapeutic index of these potential radiopharmaceuticals.
The clinical translation of EGFR- and integrin αVβ3-targeting peptides faces several hurdles that must be addressed in order to realize their full potential. The optimization of peptide structures to enhance receptor binding and stability, coupled with advanced radiolabeling techniques, is crucial for improving the efficacy and safety profiles of these agents. Clinical trials are essential for evaluating the therapeutic benefits, potential side effects, and overall patient outcomes associated with these novel potential radiopharmaceuticals. Furthermore, regulatory approval will be pivotal in determining the feasibility of incorporating these targeted therapies into standard clinical practice.
While EGFR- and integrin αVβ3-targeting peptides hold significant promise for enhancing cancer diagnosis and treatment, extensive research and development are still required to address the existing challenges. With continued advancements in peptide engineering and radiolabeling technologies, these agents can become integral components of precision oncology, offering more effective and personalized treatment options for cancer patients. In addition, while the targeting of EGFR and integrin is a valid therapeutic strategy, it is the downstream signaling pathways, particularly PI3K/AKT/mTOR and MAPK, that drive cancer progression and therapeutic resistance. Future research and therapeutic development should focus on these pathways to achieve more effective cancer treatments. By integrating inhibitors of these pathways with EGFR-targeted therapies, it may be possible to enhance treatment efficacy and overcome resistance mechanisms, leading to better patient outcomes.

Author Contributions

Conceptualization, C.d.A.F., L.M. and L.L.F.; formal analysis, data acquisition and curation, C.R.T. and A.A.T.; writing—original draft preparation, C.R.T. and A.A.T.; writing—review and editing, C.R.T., L.L.F., C.d.A.F. and L.M.; visualization, funding acquisition, supervision and administration, L.L.F., C.d.A.F. and L.M. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by Cancer Theranostics Innovation Center (CancerThera)/Centros de Pesquisa, Inovação e Difusão (CEPID)/Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP), grant number 2021/10265-8.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tang, H.; Pan, Y.; Zhang, Y.; Tang, H. Challenges for the application of EGFR-targeting peptide GE11 in tumor diagnosis and treatment. J. Control. Release 2022, 349, 592–605. [Google Scholar] [CrossRef]
  2. Li, Z.B.; Cai, W.; Cao, Q.; Chen, K.; Wu, Z.; He, L.; Chen, X. 64Cu-labeled tetrameric and octameric RGD peptides for small-animal PET of tumor αvβ3 integrin expression. J. Nucl. Med. 2007, 48, 1162–1171. [Google Scholar] [CrossRef] [PubMed]
  3. Zhou, M.; Zou, X.; Cheng, K.; Zhong, S.; Su, Y.; Wu, T.; Tao, Y.; Cong, L.; Yan, B.; Jiang, Y. The role of cell-penetrating peptides in potential anti-cancer therapy. Clin. Transl. Med. 2022, 12, e822. [Google Scholar] [CrossRef] [PubMed]
  4. Evans, B.J.; King, A.T.; Katsifis, A.; Matesic, L.; Jamie, J.F. Methods to Enhance the Metabolic Stability of Peptide-Based PET Radiopharmaceuticals. Molecules 2020, 25, 2314. [Google Scholar] [CrossRef]
  5. Herbst, R.S. Review of epidermal growth factor receptor biology. Int. J. Radiat. Oncol. Biol. Phys. 2004, 59, 21–26. [Google Scholar] [CrossRef] [PubMed]
  6. Bessman, N.J.; Freed, D.M.; Lemmon, M.A. Putting together structures of epidermal growth factor receptors. Curr. Opin. Struct. Biol. 2014, 29, 95–101. [Google Scholar] [CrossRef] [PubMed]
  7. Guy, P.M.; Platko, J.V.; Cantley, L.C.; Cerione, R.A.; Carraway, K.L., 3rd. Insect cell-expressed p180erbB3 possesses an impaired tyrosine kinase activity. Proc. Natl. Acad. Sci. USA 1994, 91, 8132–8136. [Google Scholar] [CrossRef]
  8. Hubbard, S.R. Crystal structure of the activated insulin receptor tyrosine kinase in complex with peptide substrate and ATP analog. EMBO J. 1997, 16, 5572–5581. [Google Scholar] [CrossRef]
  9. Mohammadi, M.; Schlessinger, J.; Hubbard, S.R. Structure of the FGF receptor tyrosine kinase domain reveals a novel autoinhibitory mechanism. Cell 1996, 86, 577–587. [Google Scholar] [CrossRef]
  10. Kamath, S.; Buolamwini, J.K. Targeting EGFR and HER-2 receptor tyrosine kinases for cancer drug discovery and development. Med. Res. Rev. 2006, 26, 569–594. [Google Scholar] [CrossRef]
  11. Uribe, M.L.; Marrocco, I.; Yarden, Y. EGFR in Cancer: Signaling Mechanisms, Drugs, and Acquired Resistance. Cancers 2021, 13, 2748. [Google Scholar] [CrossRef] [PubMed]
  12. Gan, H.K.; Cvrljevic, A.N.; Johns, T.G. The epidermal growth factor receptor variant III (EGFRvIII): Where wild things are altered. FEBS J. 2013, 280, 5350–5370. [Google Scholar] [CrossRef] [PubMed]
  13. Rascio, F.; Spadaccino, F.; Rocchetti, M.T.; Castellano, G.; Stallone, G.; Netti, G.S.; Ranieri, E. The Pathogenic Role of PI3K/AKT Pathway in Cancer Onset and Drug Resistance: An Updated Review. Cancers 2021, 13, 3949. [Google Scholar] [CrossRef] [PubMed]
  14. Stefani, C.; Miricescu, D.; Stanescu, S., II; Nica, R.I.; Greabu, M.; Totan, A.R.; Jinga, M. Growth Factors, PI3K/AKT/mTOR and MAPK Signaling Pathways in Colorectal Cancer Pathogenesis: Where Are We Now? Int. J. Mol. Sci. 2021, 22, 10260. [Google Scholar] [CrossRef] [PubMed]
  15. Braicu, C.; Buse, M.; Busuioc, C.; Drula, R.; Gulei, D.; Raduly, L.; Rusu, A.; Irimie, A.; Atanasov, A.G.; Slaby, O.; et al. A Comprehensive Review on MAPK: A Promising Therapeutic Target in Cancer. Cancers 2019, 11, 1618. [Google Scholar] [CrossRef] [PubMed]
  16. Tripathi, A.K.; Desai, P.P.; Tyagi, A.; Lampe, J.B.; Srivastava, Y.; Donkor, M.; Jones, H.P.; Dzyuba, S.V.; Crossley, E.; Williams, N.S.; et al. Short peptides based on the conserved regions of MIEN1 protein exhibit anticancer activity by targeting the MIEN1 signaling pathway. J. Biol. Chem. 2024, 300, 105680. [Google Scholar] [CrossRef] [PubMed]
  17. Shostak, K.; Chariot, A. EGFR and NF-κB: Partners in cancer. Trends Mol. Med. 2015, 21, 385–393. [Google Scholar] [CrossRef] [PubMed]
  18. Genta, I.; Chiesa, E.; Colzani, B.; Modena, T.; Conti, B.; Dorati, R. GE11 Peptide as an Active Targeting Agent in Antitumor Therapy: A Minireview. Pharmaceutics 2017, 10, 2. [Google Scholar] [CrossRef] [PubMed]
  19. Ruoslahti, E.; Pierschbacher, M.D. Arg-Gly-Asp: A versatile cell recognition signal. Cell 1986, 44, 517–518. [Google Scholar] [CrossRef]
  20. Cai, W.; Chen, X. Anti-angiogenic cancer therapy based on integrin αvβ3 antagonism. Anti-Cancer Agents Med. Chem. 2006, 6, 407–428. [Google Scholar] [CrossRef]
  21. Hynes, R.O. Integrins: Bidirectional, allosteric signaling machines. Cell 2002, 110, 673–687. [Google Scholar] [CrossRef]
  22. Alghisi, G.C.; Rüegg, C. Vascular integrins in tumor angiogenesis: Mediators and therapeutic targets. Endothelium 2006, 13, 113–135. [Google Scholar] [CrossRef] [PubMed]
  23. Hood, J.D.; Cheresh, D.A. Role of integrins in cell invasion and migration. Nat. Rev. Cancer 2002, 2, 91–100. [Google Scholar] [CrossRef] [PubMed]
  24. Eble, J.A.; Haier, J. Integrins in cancer treatment. Curr. Cancer Drug Targets 2006, 6, 89–105. [Google Scholar] [CrossRef] [PubMed]
  25. Debordeaux, F.; Chansel-Debordeaux, L.; Pinaquy, J.B.; Fernandez, P.; Schulz, J. What about αvβ3 integrins in molecular imaging in oncology? Nucl. Med. Biol. 2018, 62–63, 31–46. [Google Scholar] [CrossRef] [PubMed]
  26. Guo, W.; Giancotti, F.G. Integrin signalling during tumour progression. Nat. Rev. Mol. Cell Biol. 2004, 5, 816–826. [Google Scholar] [CrossRef] [PubMed]
  27. Desgrosellier, J.S.; Cheresh, D.A. Integrins in cancer: Biological implications and therapeutic opportunities. Nat. Rev. Cancer 2010, 10, 9–22. [Google Scholar] [CrossRef] [PubMed]
  28. Mezu-Ndubuisi, O.J.; Maheshwari, A. The role of integrins in inflammation and angiogenesis. Pediatr. Res. 2021, 89, 1619–1626. [Google Scholar] [CrossRef] [PubMed]
  29. Kumar, C. Integrin αvβ3 as a therapeutic target for blocking tumor-induced angiogenesis. Curr. Drug Targets 2003, 4, 123–131. [Google Scholar] [CrossRef]
  30. Seguin, L.; Desgrosellier, J.S.; Weis, S.M.; Cheresh, D.A. Integrins and cancer: Regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol. 2015, 25, 234–240. [Google Scholar] [CrossRef]
  31. Li, Z.; Zhao, R.; Wu, X.; Sun, Y.; Yao, M.; Li, J.; Xu, Y.; Gu, J. Identification and characterization of a novel peptide ligand of epidermal growth factor receptor for targeted delivery of therapeutics. FASEB J. 2005, 19, 1978–1985. [Google Scholar] [CrossRef] [PubMed]
  32. Li, X.; Hu, K.; Liu, W.; Wei, Y.; Sha, R.; Long, Y.; Han, Y.; Sun, P.; Wu, H.; Li, G.; et al. Synthesis and evaluation of [18F]FP-Lys-GE11 as a new radiolabeled peptide probe for epidermal growth factor receptor (EGFR) imaging. Nucl. Med. Biol. 2020, 90–91, 84–92. [Google Scholar] [CrossRef] [PubMed]
  33. Rahmanian, N.; Hosseinimehr, S.J.; Khalaj, A.; Noaparast, Z.; Abedi, S.M.; Sabzevari, O. Tc labeled HYNIC-EDDA/tricine-GE11 peptide as a successful tumor targeting agent. Med. Chem. Res. 2018, 27, 890–902. [Google Scholar] [CrossRef]
  34. Rahmanian, N.; Hosseinimehr, S.J.; Khalaj, A.; Noaparast, Z.; Abedi, S.M.; Sabzevari, O. 99mTc-radiolabeled GE11-modified peptide for ovarian tumor targeting. DARU J. Pharm. Sci. 2017, 25, 13. [Google Scholar] [CrossRef] [PubMed]
  35. Abourbeh, G.; Shir, A.; Mishani, E.; Ogris, M.; Rödl, W.; Wagner, E.; Levitzki, A. PolyIC GE11 polyplex inhibits EGFR-overexpressing tumors. IUBMB Life 2012, 64, 324–330. [Google Scholar] [CrossRef] [PubMed]
  36. Striese, F.; Sihver, W.; Gao, F.; Bergmann, R.; Walther, M.; Pietzsch, J.; Steinbach, J.; Pietzsch, H.J. Exploring pitfalls of 64Cu-labeled EGFR-targeting peptide GE11 as a potential PET tracer. Amino Acids 2018, 50, 1415–1431. [Google Scholar] [CrossRef]
  37. Jiao, H.; Zhao, X.; Han, J.; Zhang, J.; Wang, J. Synthesis of a novel 99mTc labeled GE11 peptide for EGFR SPECT imaging. Int. J. Radiat. Biol. 2020, 96, 1443–1451. [Google Scholar] [CrossRef] [PubMed]
  38. Judmann, B.; Wängler, B.; Schirrmacher, R.; Fricker, G.; Wängler, C. Towards Radiolabeled EGFR-Specific Peptides: Alternatives to GE11. Pharmaceuticals 2023, 16, 273. [Google Scholar] [CrossRef] [PubMed]
  39. De Paiva, I.M.; Vakili, M.R.; Soleimani, A.H.; Tabatabaei Dakhili, S.A.; Munira, S.; Paladino, M.; Martin, G.; Jirik, F.R.; Hall, D.G.; Weinfeld, M.; et al. Biodistribution and Activity of EGFR Targeted Polymeric Micelles Delivering a New Inhibitor of DNA Repair to Orthotopic Colorectal Cancer Xenografts with Metastasis. Mol. Pharm. 2022, 19, 1825–1838. [Google Scholar] [CrossRef]
  40. Decker, S.; Taschauer, A.; Geppl, E.; Pirhofer, V.; Schauer, M.; Pöschl, S.; Kopp, F.; Richter, L.; Ecker, G.F.; Sami, H.; et al. Structure-based peptide ligand design for improved epidermal growth factor receptor targeted gene delivery. Eur. J. Pharm. Biopharm. 2022, 176, 211–221. [Google Scholar] [CrossRef]
  41. Furman, O.; Zaporozhets, A.; Tobi, D.; Bazylevich, A.; Firer, M.A.; Patsenker, L.; Gellerman, G.; Lubin, B.C.R. Novel Cyclic Peptides for Targeting EGFR and EGRvIII Mutation for Drug Delivery. Pharmaceutics 2022, 14, 1505. [Google Scholar] [CrossRef] [PubMed]
  42. Ai, S.; Duan, J.; Liu, X.; Bock, S.; Tian, Y.; Huang, Z. Biological evaluation of a novel doxorubicin-peptide conjugate for targeted delivery to EGF receptor-overexpressing tumor cells. Mol. Pharm. 2011, 8, 375–386. [Google Scholar] [CrossRef] [PubMed]
  43. Stroobant, P.; Rice, A.P.; Gullick, W.J.; Cheng, D.J.; Kerr, I.M.; Waterfield, M.D. Purification and characterization of vaccinia virus growth factor. Cell 1985, 42, 383–393. [Google Scholar] [CrossRef] [PubMed]
  44. Cao, X.W.; Yang, X.Z.; Du, X.; Fu, L.Y.; Zhang, T.Z.; Shan, H.W.; Zhao, J.; Wang, F.J. Structure optimisation to improve the delivery efficiency and cell selectivity of a tumour-targeting cell-penetrating peptide. J. Drug Target. 2018, 26, 777–792. [Google Scholar] [CrossRef] [PubMed]
  45. Hamzeh-Mivehroud, M.; Mahmoudpour, A.; Dastmalchi, S. Identification of new peptide ligands for epidermal growth factor receptor using phage display and computationally modeling their mode of binding. Chem. Biol. Drug Des. 2012, 79, 246–259. [Google Scholar] [CrossRef] [PubMed]
  46. Zhou, J.; Joshi, B.P.; Duan, X.; Pant, A.; Qiu, Z.; Kuick, R.; Owens, S.R.; Wang, T.D. EGFR Overexpressed in Colonic Neoplasia Can be Detected on Wide-Field Endoscopic Imaging. Clin. Transl. Gastroenterol. 2015, 6, e101. [Google Scholar] [CrossRef] [PubMed]
  47. Song, S.; Liu, D.; Peng, J.; Deng, H.; Guo, Y.; Xu, L.X.; Miller, A.D.; Xu, Y. Novel peptide ligand directs liposomes toward EGF-R high-expressing cancer cells in vitro and in vivo. FASEB J. 2009, 23, 1396–1404. [Google Scholar] [CrossRef] [PubMed]
  48. Xiang, Z.; Yang, X.; Xu, J.; Lai, W.; Wang, Z.; Hu, Z.; Tian, J.; Geng, L.; Fang, Q. Tumor detection using magnetosome nanoparticles functionalized with a newly screened EGFR/HER2 targeting peptide. Biomaterials 2017, 115, 53–64. [Google Scholar] [CrossRef] [PubMed]
  49. Sachdeva, S.; Joo, H.; Tsai, J.; Jasti, B.; Li, X. A Rational Approach for Creating Peptides Mimicking Antibody Binding. Sci. Rep. 2019, 9, 997. [Google Scholar] [CrossRef]
  50. Sobral, D.V.; Fuscaldi, L.L.; Durante, A.C.R.; Rangel, M.G.; Oliveira, L.R.; Mendonça, F.F.; Miranda, A.C.C.; Cabeza, J.M.; Montor, W.R.; Cabral, F.R.; et al. Radiochemical and biological properties of peptides designed to interact with EGF receptor: Relevance for glioblastoma. Nucl. Med. Biol. 2020, 88–89, 14–23. [Google Scholar] [CrossRef]
  51. Gu, Y.; Dong, B.; He, X.; Qiu, Z.; Zhang, J.; Zhang, M.; Liu, H.; Pang, X.; Cui, Y. The challenges and opportunities of αvβ3-based therapeutics in cancer: From bench to clinical trials. Pharmacol. Res. 2023, 189, 106694. [Google Scholar] [CrossRef]
  52. Hernandez, R.; Czerwinski, A.; Chakravarty, R.; Graves, S.A.; Yang, Y.; England, C.G.; Nickles, R.J.; Valenzuela, F.; Cai, W. Evaluation of two novel 64Cu-labeled RGD peptide radiotracers for enhanced PET imaging of tumor integrin αvβ3. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 1859–1868. [Google Scholar] [CrossRef]
  53. Zuo, H. iRGD: A Promising Peptide for Cancer Imaging and a Potential Therapeutic Agent for Various Cancers. J. Oncol. 2019, 2019, 9367845. [Google Scholar] [CrossRef]
  54. Wang, Y.; Xiao, W.; Zhang, Y.; Meza, L.; Tseng, H.; Takada, Y.; Ames, J.B.; Lam, K.S. Optimization of RGD-Containing Cyclic Peptides against αvβ3 Integrin. Mol. Cancer Ther. 2016, 15, 232–240. [Google Scholar] [CrossRef]
  55. Indrevoll, B.; Kindberg, G.M.; Solbakken, M.; Bjurgert, E.; Johansen, J.H.; Karlsen, H.; Mendizabal, M.; Cuthbertson, A. NC-100717: A versatile RGD peptide scaffold for angiogenesis imaging. Bioorg. Med. Chem. Lett. 2006, 16, 6190–6193. [Google Scholar] [CrossRef] [PubMed]
  56. Koivunen, E.; Wang, B.; Ruoslahti, E. Phage Libraries Displaying Cyclic Peptides with Different Ring Sizes: Ligand Specificities of the RGD-Directed Integrins. Bio/Technology 1995, 13, 265–270. [Google Scholar] [CrossRef]
  57. Del Gatto, A.; Zaccaro, L.; Grieco, P.; Novellino, E.; Zannetti, A.; Del Vecchio, S.; Iommelli, F.; Salvatore, M.; Pedone, C.; Saviano, M. Novel and Selective αvβ3 Receptor Peptide Antagonist:  Design, Synthesis, and Biological Behavior. J. Med. Chem. 2006, 49, 3416–3420. [Google Scholar] [CrossRef]
  58. Zhang, L.; Shan, X.; Meng, X.; Gu, T.; Guo, L.; An, X.; Jiang, Q.; Ge, H.; Ning, X. Novel Integrin αvβ3-Specific Ligand for the Sensitive Diagnosis of Glioblastoma. Mol. Pharm. 2019, 16, 3977–3984. [Google Scholar] [CrossRef]
  59. Ma, Y.; Ai, G.; Zhang, C.; Zhao, M.; Dong, X.; Han, Z.; Wang, Z.; Zhang, M.; Liu, Y.; Gao, W.; et al. Novel Linear Peptides with High Affinity to αvβ3 Integrin for Precise Tumor Identification. Theranostics 2017, 7, 1511–1523. [Google Scholar] [CrossRef]
  60. Kręcisz, P.; Czarnecka, K.; Królicki, L.; Mikiciuk-Olasik, E.; Szymański, P. Radiolabeled Peptides and Antibodies in Medicine. Bioconjug. Chem. 2021, 32, 25–42. [Google Scholar] [CrossRef]
  61. Durante, A.C.R.; Sobral, D.V.; Miranda, A.C.C.; Almeida, É.L.d.V.; Fuscaldi, L.; de Barboza, M.R.F.F.; Malavolta, L. Comparative Study of Two Oxidizing Agents, Chloramine T and Iodo-Gen®, for the Radiolabeling of β-CIT with Iodine-131: Relevance for Parkinson’s Disease. Pharmaceuticals 2019, 12, 25. [Google Scholar] [CrossRef]
  62. Sobral, D.V.; Fuscaldi, L.L.; Durante, A.C.R.; Mendonca, F.F.; de Oliveira, L.R.; Miranda, A.C.C.; Mejia, J.; Montor, W.R.; de Barboza, M.F.; Malavolta, L. Comparative Evaluation of Radiochemical and Biological Properties of 131I- and [99mTc]Tc(CO)3-Labeled RGD Analogues Planned to Interact with the αvβ3 Integrin Expressed in Glioblastoma. Pharmaceuticals 2022, 15, 116. [Google Scholar] [CrossRef]
  63. Baishya, R.; Nayak, D.K.; Chatterjee, N.; Halder, K.K.; Karmakar, S.; Debnath, M.C. Synthesis, Characterization, and Biological Evaluation of 99mTc(CO)3-Labeled Peptides for Potential Use as Tumor Targeted Radiopharmaceuticals. Chem. Biol. Drug Des. 2014, 83, 58–70. [Google Scholar] [CrossRef]
  64. Vats, K.; Satpati, D.; Sharma, R.; Sarma, H.D.; Banerjee, S. Synthesis and comparative in vivo evaluation of 99mTc(CO)3-labeled PEGylated and non-PEGylated cRGDfK peptide monomers. Chem. Biol. Drug Des. 2017, 89, 371–378. [Google Scholar] [CrossRef]
  65. Brechbiel, M.W. Bifunctional chelates for metal nuclides. Q. J. Nucl. Med. Mol. Imaging 2008, 52, 166–173. [Google Scholar]
  66. Haubner, R.; Weber, W.A.; Beer, A.J.; Vabuliene, E.; Reim, D.; Sarbia, M.; Becker, K.F.; Goebel, M.; Hein, R.; Wester, H.J.; et al. Noninvasive visualization of the activated αvβ3 integrin in cancer patients by positron emission tomography and [18F]Galacto-RGD. PLoS Med. 2005, 2, e70. [Google Scholar] [CrossRef]
  67. Chen, H.; Niu, G.; Wu, H.; Chen, X. Clinical Application of Radiolabeled RGD Peptides for PET Imaging of Integrin αvβ3. Theranostics 2016, 6, 78–92. [Google Scholar] [CrossRef]
  68. Chen, X.; Hou, Y.; Tohme, M.; Park, R.; Khankaldyyan, V.; Gonzales-Gomez, I.; Bading, J.R.; Laug, W.E.; Conti, P.S. Pegylated Arg-Gly-Asp Peptide: 64Cu Labeling and PET Imaging of Brain Tumor αvβ3-Integrin Expression. J. Nucl. Med. 2004, 45, 1776. [Google Scholar]
  69. Liolios, C.; Sachpekidis, C.; Kolocouris, A.; Dimitrakopoulou-Strauss, A.; Bouziotis, P. PET Diagnostic Molecules Utilizing Multimeric Cyclic RGD Peptide Analogs for Imaging Integrin αvβ3 Receptors. Molecules 2021, 26, 1792. [Google Scholar] [CrossRef] [PubMed]
  70. Liu, S.; Liu, Z.; Chen, K.; Yan, Y.; Watzlowik, P.; Wester, H.J.; Chin, F.T.; Chen, X. 18F-labeled galacto and PEGylated RGD dimers for PET imaging of αvβ3 integrin expression. Mol. Imaging Biol. 2010, 12, 530–538. [Google Scholar] [CrossRef] [PubMed]
  71. Cui, J.; Yue, J.B. Current status and advances in arginine-glycine-aspartic acid peptide-based molecular imaging to evaluate the effects of anti-angiogenic therapies. Precis. Radiat. Oncol. 2019, 3, 29–34. [Google Scholar] [CrossRef]
  72. Imberti, C.; Terry, S.Y.; Cullinane, C.; Clarke, F.; Cornish, G.H.; Ramakrishnan, N.K.; Roselt, P.; Cope, A.P.; Hicks, R.J.; Blower, P.J.; et al. Enhancing PET Signal at Target Tissue in Vivo: Dendritic and Multimeric Tris(hydroxypyridinone) Conjugates for Molecular Imaging of αvβ3 Integrin Expression with Gallium-68. Bioconjug. Chem. 2017, 28, 481–495. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, J.H.; Lee, J.S.; Kang, K.W.; Lee, H.Y.; Han, S.W.; Kim, T.Y.; Lee, Y.S.; Jeong, J.M.; Lee, D.S. Whole-body distribution and radiation dosimetry of 68Ga-NOTA-RGD, a positron emission tomography agent for angiogenesis imaging. Cancer Biother. Radiopharm. 2012, 27, 65–71. [Google Scholar] [CrossRef] [PubMed]
  74. Lang, L.; Li, W.; Guo, N.; Ma, Y.; Zhu, L.; Kiesewetter, D.O.; Shen, B.; Niu, G.; Chen, X. Comparison study of [18F]FAl-NOTA-PRGD2, [18F]FPPRGD2, and [68Ga]Ga-NOTA-PRGD2 for PET imaging of U87MG tumors in mice. Bioconjug. Chem. 2011, 22, 2415–2422. [Google Scholar] [CrossRef] [PubMed]
  75. Wei, L.; Ye, Y.; Wadas, T.J.; Lewis, J.S.; Welch, M.J.; Achilefu, S.; Anderson, C.J. 64Cu-labeled CB-TE2A and diamsar-conjugated RGD peptide analogs for targeting angiogenesis: Comparison of their biological activity. Nucl. Med. Biol. 2009, 36, 277–285. [Google Scholar] [CrossRef] [PubMed]
  76. Kazemi, Z.; Zahmatkesh, M.H.; Abedi, S.M.; Hosseinimehr, S.J. Biological Evaluation of 99mTc-HYNIC-EDDA/tricine-(Ser)-D4 Peptide for Tumor Targeting. Curr. Radiopharm. 2017, 10, 123–130. [Google Scholar] [CrossRef] [PubMed]
  77. Zahmatkesh, M.H.; Abedi, S.M.; Hosseinimehr, S.J. 99mTc-HYNIC-D4 Peptide: A New Small Radiolabeled Peptide for Non Small Cell Lung Tumor Targeting. Anticancer Agents Med. Chem. 2017, 17, 734–740. [Google Scholar] [CrossRef] [PubMed]
  78. Haddad Zahmatkesh, M.; Abedi, S.M.; Hosseinimehr, S.J. Preparation and biological evaluation of 99mTc-HYNIC-(Ser)3-D4 peptide for targeting and imaging of non-small-cell lung cancer. Future Oncol. 2017, 13, 893–905. [Google Scholar] [CrossRef] [PubMed]
  79. Dissoki, S.; Hagooly, A.; Elmachily, S.; Mishani, E. Labeling approaches for the GE11 peptide, an epidermal growth factor receptor biomarker. J. Label. Compd. Rad. 2011, 54, 693–701. [Google Scholar] [CrossRef]
  80. Ogawa, K.; Takeda, T.; Yokokawa, M.; Yu, J.; Makino, A.; Kiyono, Y.; Shiba, K.; Kinuya, S.; Odani, A. Comparison of Radioiodine- or Radiobromine-Labeled RGD Peptides between Direct and Indirect Labeling Methods. Chem. Pharm. Bull. 2018, 66, 651–659. [Google Scholar] [CrossRef]
  81. Dejesus, O.T. Synthesis of [64Cu]Cu-NOTA-Bn-GE11 for PET imaging of EGFR-rich tumors. Curr. Radiopharm. 2012, 5, 15–18. [Google Scholar] [CrossRef]
  82. Judmann, B.; Braun, D.; Schirrmacher, R.; Wangler, B.; Fricker, G.; Wangler, C. Toward the Development of GE11-Based Radioligands for Imaging of Epidermal Growth Factor Receptor-Positive Tumors. ACS Omega 2022, 7, 27690–27702. [Google Scholar] [CrossRef]
  83. Kim, M.H.; Kim, S.G.; Kim, D.W. A novel dual-labeled small peptide as a multimodal imaging agent for targeting wild-type EGFR in tumors. PLoS ONE 2022, 17, e0263474. [Google Scholar] [CrossRef]
  84. Wu, Y.; Zhang, X.; Xiong, Z.; Cheng, Z.; Fisher, D.R.; Liu, S.; Gambhir, S.S.; Chen, X. microPET imaging of glioma integrin αvβ3 expression using 64Cu-labeled tetrameric RGD peptide. J. Nucl. Med. 2005, 46, 1707–1718. [Google Scholar]
  85. Shi, J.; Kim, Y.S.; Zhai, S.; Liu, Z.; Chen, X.; Liu, S. Improving tumor uptake and pharmacokinetics of 64Cu-labeled cyclic RGD peptide dimers with Gly3 and PEG4 linkers. Bioconjug. Chem. 2009, 20, 750–759. [Google Scholar] [CrossRef]
  86. Liu, S.; Vorobyova, I.; Park, R.; Conti, P.S. Biodistribution and Radiation Dosimetry of the Integrin Marker 64Cu-BaBaSar-RGD2 Determined from Whole-Body PET/CT in a Non-human Primate. Front. Phys. 2017, 5, 54. [Google Scholar] [CrossRef]
  87. Oxboel, J.; Brandt-Larsen, M.; Schjoeth-Eskesen, C.; Myschetzky, R.; El-Ali, H.H.; Madsen, J.; Kjaer, A. Comparison of two new angiogenesis PET tracers 68Ga-NODAGA-E[c(RGDyK)]2 and 64Cu-NODAGA-E[c(RGDyK)]2; in vivo imaging studies in human xenograft tumors. Nucl. Med. Biol. 2014, 41, 259–267. [Google Scholar] [CrossRef]
  88. Cai, H.; Li, Z.; Huang, C.W.; Park, R.; Conti, P.S. 64Cu labeled AmBaSar-RGD2 for micro-PET imaging of integrin αvβ3 expression. Curr. Radiopharm. 2011, 4, 68–74. [Google Scholar] [CrossRef]
  89. Minamimoto, R.; Karam, A.; Jamali, M.; Barkhodari, A.; Gambhir, S.S.; Dorigo, O.; Iagaru, A. Pilot prospective evaluation of 18F-FPPRGD2 PET/CT in patients with cervical and ovarian cancer. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 1047–1055. [Google Scholar] [CrossRef] [PubMed]
  90. Toriihara, A.; Duan, H.; Thompson, H.M.; Park, S.; Hatami, N.; Baratto, L.; Fan, A.C.; Iagaru, A. 18F-FPPRGD2 PET/CT in patients with metastatic renal cell cancer. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 1518–1523. [Google Scholar] [CrossRef] [PubMed]
  91. Mittra, E.S.; Goris, M.L.; Iagaru, A.H.; Kardan, A.; Burton, L.; Berganos, R.; Chang, E.; Liu, S.; Shen, B.; Chin, F.T.; et al. Pilot pharmacokinetic and dosimetric studies of 18F-FPPRGD2: A PET radiopharmaceutical agent for imaging αvβ3 integrin levels. Radiology 2011, 260, 182–191. [Google Scholar] [CrossRef]
  92. Glaser, M.; Morrison, M.; Solbakken, M.; Arukwe, J.; Karlsen, H.; Wiggen, U.; Champion, S.; Kindberg, G.M.; Cuthbertson, A. Radiosynthesis and Biodistribution of Cyclic RGD Peptides Conjugated with Novel [18F]Fluorinated Aldehyde-Containing Prosthetic Groups. Bioconjug. Chem. 2008, 19, 951–957. [Google Scholar] [CrossRef]
  93. Cai, H.; Conti, P.S. RGD-based PET tracers for imaging receptor integrin αvβ3 expression. J. Label. Comp. Radiopharm. 2013, 56, 264–279. [Google Scholar] [CrossRef]
  94. Doss, M.; Kolb, H.C.; Zhang, J.J.; Bélanger, M.J.; Stubbs, J.B.; Stabin, M.G.; Hostetler, E.D.; Alpaugh, R.K.; von Mehren, M.; Walsh, J.C.; 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, 787–795. [Google Scholar] [CrossRef]
  95. Kolb, H.; Walsh, J.; Walsh, J.; Liang, Q.; Zhao, T.; Gao, D.; Secrest, J.; Gomez, L.; Scott, P. 18F-RGD-K5: A cyclic triazole-bearing RGD peptide for imaging integrin αvβ3 expression in vivo. J. Nucl. Med. 2009, 50, 329. [Google Scholar]
  96. Gaertner, F.C.; Kessler, H.; Wester, H.J.; Schwaiger, M.; Beer, A.J. Radiolabelled RGD peptides for imaging and therapy. Eur. J. Nucl. Med. Mol. Imaging 2012, 39 (Suppl. S1), S126–S138. [Google Scholar] [CrossRef]
  97. Liu, S.; Liu, H.; Jiang, H.; Xu, Y.; Zhang, H.; Cheng, Z. One-step radiosynthesis of 18F-AlF-NOTA-RGD2 for tumor angiogenesis PET imaging. Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 1732–1741. [Google Scholar] [CrossRef]
  98. Guo, J.; Guo, N.; Lang, L.; Kiesewetter, D.O.; Xie, Q.; Li, Q.; Eden, H.S.; Niu, G.; Chen, X. 18F-alfatide II and 18F-FDG dual-tracer dynamic PET for parametric, early prediction of tumor response to therapy. J. Nucl. Med. 2014, 55, 154–160. [Google Scholar] [CrossRef]
  99. Provost, C.; Prignon, A.; Rozenblum-Beddok, L.; Bruyer, Q.; Dumont, S.; Merabtene, F.; Nataf, V.; Bouteiller, C.; Talbot, J.N. Comparison and evaluation of two RGD peptides labelled with 68Ga or 18F for PET imaging of angiogenesis in animal models of human glioblastoma or lung carcinoma. Oncotarget 2018, 9, 19307–19316. [Google Scholar] [CrossRef] [PubMed]
  100. Vatsa, R.; Shukla, J.; Kumar, S.; Chakraboarty, S.; Dash, A.; Singh, G.; Mittal, B.R. Effect of Macro-Cyclic Bifunctional Chelators DOTA and NODAGA on Radiolabeling and In Vivo Biodistribution of Ga-68 Cyclic RGD Dimer. Cancer Biother. Radiopharm. 2019, 34, 427–435. [Google Scholar] [CrossRef] [PubMed]
  101. Chakraborty, S.; Chakravarty, R.; Vatsa, R.; Bhusari, P.; Sarma, H.D.; Shukla, J.; Mittal, B.R.; Dash, A. Toward realization of ‘mix-and-use’ approach in ⁶⁸Ga radiopharmacy: Preparation, evaluation and preliminary clinical utilization of ⁶⁸Ga-labeled NODAGA-coupled RGD peptide derivative. Nucl. Med. Biol. 2016, 43, 116–123. [Google Scholar] [CrossRef]
  102. Shi, J.; Jin, Z.; Liu, X.; Fan, D.; Sun, Y.; Zhao, H.; Zhu, Z.; Liu, Z.; Jia, B.; Wang, F. PET Imaging of Neovascularization with 68Ga-3PRGD2 for Assessing Tumor Early Response to Endostar Antiangiogenic Therapy. Mol. Pharm. 2014, 11, 3915–3922. [Google Scholar] [CrossRef]
  103. Kazmierczak, P.M.; Todica, A.; Gildehaus, F.J.; Hirner-Eppeneder, H.; Brendel, M.; Eschbach, R.S.; Hellmann, M.; Nikolaou, K.; Reiser, M.F.; Wester, H.J.; et al. 68Ga-TRAP-(RGD)3 Hybrid Imaging for the In Vivo Monitoring of αvβ3-Integrin Expression as Biomarker of Anti-Angiogenic Therapy Effects in Experimental Breast Cancer. PLoS ONE 2016, 11, e0168248. [Google Scholar] [CrossRef]
  104. Liu, Z.; Niu, G.; Shi, J.; Liu, S.; Wang, F.; Liu, S.; Chen, X. 68Ga-labeled cyclic RGD dimers with Gly3 and PEG4 linkers: Promising agents for tumor integrin αvβ3 PET imaging. Eur. J. Nucl. Med. Mol. Imaging 2009, 36, 947–957. [Google Scholar] [CrossRef]
  105. Knetsch, P.A.; Zhai, C.; Rangger, C.; Blatzer, M.; Haas, H.; Kaeopookum, P.; Haubner, R.; Decristoforo, C. [68Ga]FSC-(RGD)3 a trimeric RGD peptide for imaging αvβ3 integrin expression based on a novel siderophore derived chelating scaffold-synthesis and evaluation. Nucl. Med. Biol. 2015, 42, 115–122. [Google Scholar] [CrossRef]
  106. Kondo, N.; Wakamori, K.; Hirata, M.; Temma, T. Radioiodinated bicyclic RGD peptide for imaging integrin αvβ3 in cancers. Biochem. Biophys. Res. Commun. 2020, 528, 168–173. [Google Scholar] [CrossRef] [PubMed]
  107. Shi, J.; Wang, L.; Kim, Y.S.; Zhai, S.; Liu, Z.; Chen, X.; Liu, S. Improving tumor uptake and excretion kinetics of 99mTc-labeled cyclic arginine-glycine-aspartic (RGD) dimers with triglycine linkers. J. Med. Chem. 2008, 51, 7980–7990. [Google Scholar] [CrossRef]
  108. Zhou, Y.; Kim, Y.S.; Chakraborty, S.; Shi, J.; Gao, H.; Liu, S. 99mTc-labeled cyclic RGD peptides for noninvasive monitoring of tumor integrin αvβ3 expression. Mol. Imaging 2011, 10, 386–397. [Google Scholar] [CrossRef]
  109. Liang, Y.; Jia, X.; Wang, Y.; Liu, Y.; Yao, X.; Bai, Y.; Han, P.; Chen, S.; Yang, A.; Gao, R. Evaluation of integrin αvβ3-targeted imaging for predicting disease progression in patients with high-risk differentiated thyroid cancer (using 99mTc-3PRGD2). Cancer Imaging 2022, 22, 72. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, L.; Shi, J.; Kim, Y.-S.; Zhai, S.; Jia, B.; Zhao, H.; Liu, Z.; Wang, F.; Chen, X.; Liu, S. Improving Tumor-Targeting Capability and Pharmacokinetics of 99mTc-Labeled Cyclic RGD Dimers with PEG4 Linkers. Mol. Pharm. 2009, 6, 231–245. [Google Scholar] [CrossRef] [PubMed]
  111. Zhao, Z.Q.; Yang, Y.; Fang, W.; Liu, S. Comparison of biological properties of 99mTc-labeled cyclic RGD Peptide trimer and dimer useful as SPECT radiotracers for tumor imaging. Nucl. Med. Biol. 2016, 43, 661–669. [Google Scholar] [CrossRef]
  112. Bolzati, C.; Salvarese, N.; Carpanese, D.; Seraglia, R.; Meléndez-Alafort, L.; Rosato, A.; Capasso, D.; Saviano, M.; Del Gatto, A.; Comegna, D.; et al. [99mTc][Tc(N)PNP43]-Labeled RGD Peptides As New Probes for a Selective Detection of αvβ3 Integrin: Synthesis, Structure-Activity and Pharmacokinetic Studies. J. Med. Chem. 2018, 61, 9596–9610. [Google Scholar] [CrossRef]
  113. Decristoforo, C.; Faintuch-Linkowski, B.; Rey, A.; von Guggenberg, E.; Rupprich, M.; Hernandez-Gonzales, I.; Rodrigo, T.; Haubner, R. [99mTc]HYNIC-RGD for imaging integrin αvβ3 expression. Nucl. Med. Biol. 2006, 33, 945–952. [Google Scholar] [CrossRef]
  114. Echavidre, W.; Durivault, J.; Gotorbe, C.; Blanchard, T.; Pagnuzzi, M.; Vial, V.; Raes, F.; Broisat, A.; Villeneuve, R.; Amblard, R.; et al. Integrin-αvβ3 is a Therapeutically Targetable Fundamental Factor in Medulloblastoma Tumorigenicity and Radioresistance. Cancer Res. Commun. 2023, 3, 2483–2496. [Google Scholar] [CrossRef]
  115. Sancey, L.; Ardisson, V.; Riou, L.M.; Ahmadi, M.; Marti-Batlle, D.; Boturyn, D.; Dumy, P.; Fagret, D.; Ghezzi, C.; Vuillez, J.-P. In vivo imaging of tumour angiogenesis in mice with the αvβ3 integrin-targeted tracer 99mTc-RAFT-RGD. Eur. J. Nucl. Med. Mol. Imaging 2007, 34, 2037–2047. [Google Scholar] [CrossRef]
  116. Liu, S.; Hsieh, W.Y.; Jiang, Y.; Kim, Y.S.; Sreerama, S.G.; Chen, X.; Jia, B.; Wang, F. Evaluation of a 99mTc-labeled cyclic RGD tetramer for noninvasive imaging integrin αvβ3-positive breast cancer. Bioconjug. Chem. 2007, 18, 438–446. [Google Scholar] [CrossRef]
  117. Shi, J.; Wang, L.; Kim, Y.S.; Zhai, S.; Jia, B.; Wang, F.; Liu, S. 99mTcO(MAG2-3G3-dimer): A new integrin αvβ3-targeted SPECT radiotracer with high tumor uptake and favorable pharmacokinetics. Eur. J. Nucl. Med. Mol. Imaging 2009, 36, 1874–1884. [Google Scholar] [CrossRef]
  118. Ji, S.; Czerwinski, A.; Zhou, Y.; Shao, G.; Valenzuela, F.; Sowiński, P.; Chauhan, S.; Pennington, M.; Liu, S. 99mTc-Galacto-RGD2: A novel 99mTc-labeled cyclic RGD peptide dimer useful for tumor imaging. Mol. Pharm. 2013, 10, 3304–3314. [Google Scholar] [CrossRef]
  119. Fu, J.; Xie, Y.; Fu, T.; Qiu, F.; Yu, F.; Qu, W.; Yao, X.; Zhang, A.; Yang, Z.; Shao, G.; et al. [99mTc]Tc-Galacto-RGD2 integrin αvβ3-targeted imaging as a surrogate for molecular phenotyping in lung cancer: Real-world data. EJNMMI Res. 2021, 11, 59. [Google Scholar] [CrossRef] [PubMed]
  120. Terry, S.Y.; Abiraj, K.; Frielink, C.; van Dijk, L.K.; Bussink, J.; Oyen, W.J.; Boerman, O.C. Imaging integrin αvβ3 on blood vessels with 111In-RGD2 in head and neck tumor xenografts. J. Nucl. Med. 2014, 55, 281–286. [Google Scholar] [CrossRef] [PubMed]
  121. Decristoforo, C.; Hernandez Gonzalez, I.; Carlsen, J.; Rupprich, M.; Huisman, M.; Virgolini, I.; Wester, H.J.; Haubner, R. 68Ga- and 111In-labelled DOTA-RGD peptides for imaging of αvβ3 integrin expression. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 1507–1515. [Google Scholar] [CrossRef] [PubMed]
  122. Zheng, Y.; Ji, S.; Tomaselli, E.; Yang, Y.; Liu, S. Comparison of biological properties of 111In-labeled dimeric cyclic RGD peptides. Nucl. Med. Biol. 2015, 42, 137–145. [Google Scholar] [CrossRef] [PubMed]
  123. Shi, J.; Kim, Y.S.; Chakraborty, S.; Zhou, Y.; Wang, F.; Liu, S. Impact of bifunctional chelators on biological properties of 111In-labeled cyclic peptide RGD dimers. Amino Acids 2011, 41, 1059–1070. [Google Scholar] [CrossRef] [PubMed]
  124. Josefsson, A.; Cortez, A.G.; Yu, J.; Majumdar, S.; Bhise, A.; Hobbs, R.F.; Nedrow, J.R. Evaluation of targeting αvβ3 in breast cancers using RGD peptide-based agents. Nucl. Med. Biol. 2024, 128–129, 108880. [Google Scholar] [CrossRef] [PubMed]
  125. Dijkgraaf, I.; Kruijtzer, J.A.; Frielink, C.; Corstens, F.H.; Oyen, W.J.; Liskamp, R.M.; Boerman, O.C. αvβ3 integrin-targeting of intraperitoneally growing tumors with a radiolabeled RGD peptide. Int. J. Cancer 2007, 120, 605–610. [Google Scholar] [CrossRef] [PubMed]
  126. Ju, C.H.; Jeong, J.M.; Lee, Y.S.; Kim, Y.J.; Lee, B.C.; Lee, D.S.; Chung, J.K.; Lee, M.C.; Jeong, S.Y. Development of a ¹⁷⁷Lu-labeled RGD derivative for targeting angiogenesis. Cancer Biother. Radiopharm. 2010, 25, 687–691. [Google Scholar] [CrossRef] [PubMed]
  127. Shi, J.; Fan, D.; Dong, C.; Liu, H.; Jia, B.; Zhao, H.; Jin, X.; Liu, Z.; Li, F.; Wang, F. Anti-tumor effect of integrin targeted 177Lu-3PRGD2 and combined therapy with Endostar. Theranostics 2014, 4, 256–266. [Google Scholar] [CrossRef] [PubMed]
  128. Bozon-Petitprin, A.; Bacot, S.; Gauchez, A.S.; Ahmadi, M.; Bourre, J.C.; Marti-Batlle, D.; Perret, P.; Broisat, A.; Riou, L.M.; Claron, M.; et al. Targeted radionuclide therapy with RAFT-RGD radiolabelled with 90Y or 177Lu in a mouse model of αvβ3-expressing tumours. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 252–263. [Google Scholar] [CrossRef] [PubMed]
  129. Kang, C.S.; Chen, Y.; Lee, H.; Liu, D.; Sun, X.; Kweon, J.; Lewis, M.R.; Chong, H.S. Synthesis and evaluation of a new bifunctional NETA chelate for molecular targeted radiotherapy using 90Y or 177Lu. Nucl. Med. Biol. 2015, 42, 242–249. [Google Scholar] [CrossRef]
  130. Chen, H.; Jacobson, O.; Niu, G.; Weiss, I.D.; Kiesewetter, D.O.; Liu, Y.; Ma, Y.; Wu, H.; Chen, X. Novel “Add-On” Molecule Based on Evans Blue Confers Superior Pharmacokinetics and Transforms Drugs to Theranostic Agents. J. Nucl. Med. 2017, 58, 590–597. [Google Scholar] [CrossRef]
  131. Zhao, L.; Chen, H.; Guo, Z.; Fu, K.; Yao, L.; Fu, L.; Guo, W.; Wen, X.; Jacobson, O.; Zhang, X.; et al. Targeted Radionuclide Therapy in Patient-Derived Xenografts Using 177Lu-EB-RGD. Mol. Cancer Ther. 2020, 19, 2034–2043. [Google Scholar] [CrossRef] [PubMed]
  132. Li, H.; Peng, W.; Zhen, Z.; Zhang, W.; Liao, S.; Wu, X.; Wang, L.; Xuan, A.; Gao, Y.; Xu, J. Integrin αvβ3 and EGFR dual-targeted [64Cu]Cu-NOTA-RGD-GE11 heterodimer for PET imaging in pancreatic cancer mouse model. Nucl. Med. Biol. 2023, 124–125, 108364. [Google Scholar] [CrossRef] [PubMed]
  133. Yu, H.M.; Chen, J.H.; Lin, K.L.; Lin, W.J. Synthesis of 68Ga-labeled NOTA-RGD-GE11 heterodimeric peptide for dual integrin and epidermal growth factor receptor-targeted tumor imaging. J. Label. Comp. Radiopharm. 2015, 58, 299–303. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, H.; Zhao, L.; Fu, K.; Lin, Q.; Wen, X.; Jacobson, O.; Sun, L.; Wu, H.; Zhang, X.; Guo, Z.; et al. Integrin αvβ3-targeted radionuclide therapy combined with immune checkpoint blockade immunotherapy synergistically enhances anti-tumor efficacy. Theranostics 2019, 9, 7948–7960. [Google Scholar] [CrossRef] [PubMed]
  135. Braun, D.; Judmann, B.; Cheng, X.; Wangler, B.; Schirrmacher, R.; Fricker, G.; Wangler, C. Synthesis, Radiolabeling, and In Vitro and In Vivo Characterization of Heterobivalent Peptidic Agents for Bispecific EGFR and Integrin αvβ3 Targeting. ACS Omega 2023, 8, 2793–2807. [Google Scholar] [CrossRef] [PubMed]
  136. Li, L.; Chen, X.; Yu, J.; Yuan, S. Preliminary Clinical Application of RGD-Containing Peptides as PET Radiotracers for Imaging Tumors. Front. Oncol. 2022, 12, 837952. [Google Scholar] [CrossRef] [PubMed]
  137. Shi, J.; Wang, F.; Liu, S. Radiolabeled cyclic RGD peptides as radiotracers for tumor imaging. Biophys. Rep. 2016, 2, 1–20. [Google Scholar] [CrossRef] [PubMed]
  138. Liu, Z.; Liu, S.; Wang, F.; Liu, S.; Chen, X. Noninvasive imaging of tumor integrin expression using 18F-labeled RGD dimer peptide with PEG4 linkers. Eur. J. Nucl. Med. Mol. Imaging 2009, 36, 1296–1307. [Google Scholar] [CrossRef] [PubMed]
  139. Guo, N.; Lang, L.; Li, W.; Kiesewetter, D.O.; Gao, H.; Niu, G.; Xie, Q.; Chen, X. Quantitative Analysis and Comparison Study of [18F]AlF-NOTA-PRGD2, [18F]FPPRGD2 and [68Ga]Ga-NOTA-PRGD2 Using a Reference Tissue Model. PLoS ONE 2012, 7, e37506. [Google Scholar] [CrossRef]
  140. Haubner, R.; Kuhnast, B.; Mang, C.; Weber, W.A.; Kessler, H.; Wester, H.J.; Schwaiger, M. [18F]Galacto-RGD: Synthesis, radiolabeling, metabolic stability, and radiation dose estimates. Bioconjug. Chem. 2004, 15, 61–69. [Google Scholar] [CrossRef]
  141. Eo, J.S.; Jeong, J.M. Angiogenesis Imaging Using 68Ga-RGD PET/CT: Therapeutic Implications. Semin. Nucl. Med. 2016, 46, 419–427. [Google Scholar] [CrossRef]
  142. Jeong, J.M.; Hong, M.K.; Chang, Y.S.; Lee, Y.S.; Kim, Y.J.; Cheon, G.J.; Lee, D.S.; Chung, J.K.; Lee, M.C. Preparation of a promising angiogenesis PET imaging agent: 68Ga-labeled c(RGDyK)-isothiocyanatobenzyl-1,4,7-triazacyclononane-1,4,7-triacetic acid and feasibility studies in mice. J. Nucl. Med. 2008, 49, 830–836. [Google Scholar] [CrossRef] [PubMed]
  143. Notni, J.; Pohle, K.; Wester, H.-J. Be spoilt for choice with radiolabelled RGD peptides: Preclinical evaluation of 68Ga-TRAP(RGD)3. Nucl. Med. Biol. 2013, 40, 33–41. [Google Scholar] [CrossRef]
  144. Echavidre, W.; Picco, V.; Faraggi, M.; Montemagno, C. Integrin-αvβ3 as a Therapeutic Target in Glioblastoma: Back to the Future? Pharmaceutics 2022, 14, 1053. [Google Scholar] [CrossRef]
  145. De Jong, M.; Breeman, W.A.P.; Valkema, R.; Bernard, B.F.; Krenning, E.P. Combination Radionuclide Therapy Using 177Lu- and 90Y-Labeled Somatostatin Analogs. J. Nucl. Med. 2005, 46, 13S–17S. [Google Scholar]
  146. Sivolapenko, G.B.; Skarlos, D.; Pectasides, D.; Stathopoulou, E.; Milonakis, A.; Sirmalis, G.; Stuttle, A.; Courtenay-Luck, N.S.; Konstantinides, K.; Epenetos, A.A. Imaging of metastatic melanoma utilising a technetium-99m labelled RGD-containing synthetic peptide. Eur. J. Nucl. Med. 1998, 25, 1383–1389. [Google Scholar] [CrossRef]
  147. Beer, A.J.; Haubner, R.; Goebel, M.; Luderschmidt, S.; Spilker, M.E.; Wester, H.J.; Weber, W.A.; Schwaiger, M. Biodistribution and pharmacokinetics of the αvβ3-selective tracer 18F-galacto-RGD in cancer patients. J. Nucl. Med. 2005, 46, 1333–1341. [Google Scholar]
  148. Zhao, D.; Jin, X.; Li, F.; Liang, J.; Lin, Y. Integrin αvβ3 imaging of radioactive iodine-refractory thyroid cancer using 99mTc-3PRGD2. J. Nucl. Med. 2012, 53, 1872–1877. [Google Scholar] [CrossRef]
  149. Iagaru, A.; Mosci, C.; Mittra, E.; Zaharchuk, G.; Fischbein, N.; Harsh, G.; Li, G.; Nagpal, S.; Recht, L.; Gambhir, S.S. Glioblastoma Multiforme Recurrence: An Exploratory Study of 18F FPPRGD2 PET/CT. Radiology 2015, 277, 497–506. [Google Scholar] [CrossRef] [PubMed]
  150. Gao, S.; Wu, H.; Li, W.; Zhao, S.; Teng, X.; Lu, H.; Hu, X.; Wang, S.; Yu, J.; Yuan, S. A pilot study imaging integrin αvβ3 with RGD PET/CT in suspected lung cancer patients. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 2029–2037. [Google Scholar] [CrossRef] [PubMed]
  151. Haubner, R.; Finkenstedt, A.; Stegmayr, A.; Rangger, C.; Decristoforo, C.; Zoller, H.; Virgolini, I.J. [68Ga]NODAGA-RGD-Metabolic stability, biodistribution, and dosimetry data from patients with hepatocellular carcinoma and liver cirrhosis. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 2005–2013. [Google Scholar] [CrossRef]
  152. Clausen, M.M.; Carlsen, E.A.; Christensen, C.; Madsen, J.; Brandt-Larsen, M.; Klausen, T.L.; Holm, S.; Loft, A.; Berthelsen, A.K.; Kroman, N.; et al. First-in-Human Study of [68Ga]Ga-NODAGA-E[c(RGDyK)]2 PET for Integrin αvβ3 Imaging in Patients with Breast Cancer and Neuroendocrine Neoplasms: Safety, Dosimetry and Tumor Imaging Ability. Diagnostics 2022, 12, 851. [Google Scholar] [CrossRef]
  153. Carlsen, E.A.; Loft, M.; Loft, A.; Czyzewska, D.; Andreassen, M.; Langer, S.W.; Knigge, U.; Kjaer, A. Prospective Phase II Trial of [68Ga]Ga-NODAGA-E[c(RGDyK)]2 PET/CT Imaging of Integrin αvβ3 for Prognostication in Patients with Neuroendocrine Neoplasms. J. Nucl. Med. 2023, 64, 252–259. [Google Scholar] [CrossRef]
  154. Chen, S.H.; Wang, H.M.; Lin, C.Y.; Chang, J.T.; Hsieh, C.H.; Liao, C.T.; Kang, C.J.; Yang, L.Y.; Yen, T.C. RGD-K5 PET/CT in patients with advanced head and neck cancer treated with concurrent chemoradiotherapy: Results from a pilot study. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 1621–1629. [Google Scholar] [CrossRef]
  155. Li, L.; Ma, L.; Shang, D.; Liu, Z.; Yu, Q.; Wang, S.; Teng, X.; Zhang, Q.; Hu, X.; Zhao, W.; et al. Pretreatment PET/CT imaging of angiogenesis based on 18F-RGD tracer uptake may predict antiangiogenic response. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 940–947. [Google Scholar] [CrossRef]
  156. Zheng, S.; Chen, Y.; Miao, W. 99mTc-3PRGD2 for Integrin Receptor Imaging of Esophageal Cancer, Compared Study with 18F-FDG PET/CT. J. Nucl. Med. 2018, 59, 1409. [Google Scholar]
  157. Lobeek, D.; Rijpkema, M.; Terry, S.Y.A.; Molkenboer-Kuenen, J.D.M.; Joosten, L.; van Genugten, E.A.J.; van Engen-van Grunsven, A.C.H.; Kaanders, J.; Pegge, S.A.H.; Boerman, O.C.; et al. Imaging angiogenesis in patients with head and neck squamous cell carcinomas by [68Ga]Ga-DOTA-E-[c(RGDfK)]2 PET/CT. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 2647–2655. [Google Scholar] [CrossRef]
  158. Jiang, Y.; Liu, Q.; Wang, G.; Sui, H.; Wang, R.; Wang, J.; Zhu, Z. A prospective head-to-head comparison of 68Ga-NOTA-3P-TATE-RGD and 68Ga-DOTATATE in patients with gastroenteropancreatic neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 4218–4227. [Google Scholar] [CrossRef]
  159. Zhao, L.; Wen, X.; Xu, W.; Pang, Y.; Sun, L.; Wu, X.; Xu, P.; Zhang, J.; Guo, Z.; Lin, Q.; et al. Clinical Evaluation of 68Ga-FAPI-RGD for Imaging of Fibroblast Activation Protein and Integrin αvβ3 in Various Cancer Types. J. Nucl. Med. 2023, 64, 1210–1217. [Google Scholar] [CrossRef] [PubMed]
  160. Judmann, B.; Braun, D.; Wangler, B.; Schirrmacher, R.; Fricker, G.; Wangler, C. Current State of Radiolabeled Heterobivalent Peptidic Ligands in Tumor Imaging and Therapy. Pharmaceuticals 2020, 13, 173. [Google Scholar] [CrossRef]
  161. Chen, C.J.; Chan, C.H.; Lin, K.L.; Chen, J.H.; Tseng, C.H.; Wang, P.Y.; Chien, C.Y.; Yu, H.M.; Lin, W.J. 68Ga-labelled NOTA-RGD-GE11 peptide for dual integrin and EGFR-targeted tumour imaging. Nucl. Med. Biol. 2019, 68–69, 22–30. [Google Scholar] [CrossRef]
  162. Ahmadi, M.; Ahmadyousefi, Y.; Salimi, Z.; Mirzaei, R.; Najafi, R.; Amirheidari, B.; Rahbarizadeh, F.; Kheshti, J.; Safari, A.; Soleimani, M. Innovative Diagnostic Peptide-Based Technologies for Cancer Diagnosis: Focus on EGFR-Targeting Peptides. ChemMedChem 2023, 18, e202200506. [Google Scholar] [CrossRef]
Figure 1. Radiolabeling strategies: (A) direct (B) indirect.
Figure 1. Radiolabeling strategies: (A) direct (B) indirect.
Ijms 25 08553 g001
Figure 2. Preclinical study using radiolabeled GE11 for imaging of EGFR-positive tumors (A) Dynamic PET/CT images of [18F]F-FP-Lys-GE11 from U-87 MG tumor-bearing mice at 5, 15, 30, 60, 90, and 120 min p.i. The red arrow indicates tumor. (B) Time–activity curves. (C) Tumor/organ ratios (tumor/muscle, tumor/liver, tumor/kidney). Reproduced with permission from reference [32].
Figure 2. Preclinical study using radiolabeled GE11 for imaging of EGFR-positive tumors (A) Dynamic PET/CT images of [18F]F-FP-Lys-GE11 from U-87 MG tumor-bearing mice at 5, 15, 30, 60, 90, and 120 min p.i. The red arrow indicates tumor. (B) Time–activity curves. (C) Tumor/organ ratios (tumor/muscle, tumor/liver, tumor/kidney). Reproduced with permission from reference [32].
Ijms 25 08553 g002
Figure 3. Preclinical studies using radiolabeled RGD dimer to image integrin αVβ3-positive tumors. (A) Mice bearing U-87 MG or H727 tumors scanned with [68Ga]Ga-NODAGA-RGD2 at 1, 2, and 4 h p.i.; (B) Mice bearing U-87 MG or H727 tumors scanned with [64Cu]Cu-NODAGA-RGD2 at 1, 4, and 18 h p.i. Tumors are indicated by arrows; (C) [68Ga]Ga-NODAGA-RGD2 uptake in U-87 MG or H727 tumors; (D) [64Cu]Cu-NODAGA-RGD2 in U-87 MG or H727 tumors uptake. Reproduced with permission from reference [87]. * p < 0.05 for 1 versus 4 h p.i.; *** p < 0.001 for 1 versus 4 or 18 h p.i.; # p < 0.05 for 4 versus 18 h p.i.; ### p < 0.001 for 4 versus 18 h p.i.
Figure 3. Preclinical studies using radiolabeled RGD dimer to image integrin αVβ3-positive tumors. (A) Mice bearing U-87 MG or H727 tumors scanned with [68Ga]Ga-NODAGA-RGD2 at 1, 2, and 4 h p.i.; (B) Mice bearing U-87 MG or H727 tumors scanned with [64Cu]Cu-NODAGA-RGD2 at 1, 4, and 18 h p.i. Tumors are indicated by arrows; (C) [68Ga]Ga-NODAGA-RGD2 uptake in U-87 MG or H727 tumors; (D) [64Cu]Cu-NODAGA-RGD2 in U-87 MG or H727 tumors uptake. Reproduced with permission from reference [87]. * p < 0.05 for 1 versus 4 h p.i.; *** p < 0.001 for 1 versus 4 or 18 h p.i.; # p < 0.05 for 4 versus 18 h p.i.; ### p < 0.001 for 4 versus 18 h p.i.
Ijms 25 08553 g003
Figure 4. (A) PET/CT imaging of lung tumor-bearing mice (NCI-H292 cells) 2 h p.i. of [68Ga]Ga-NOTA-RGD-Cys-6-Ahx-GE11, [68Ga]Ga-NOTA-RGD, [68Ga]Ga-NOTA-GE11, [68Ga]Ga-NOTA-RGD-Cys-6-Ahx-GE11 + blocking. Arrows indicate tumors. (B) Tumor/muscle (T/M) ratio of [68Ga]Ga-NOTA-RGD-Cys-6-Ahx-GE11, [68Ga]Ga-NOTA-RGD, [68Ga]Ga-NOTA-GE11. Reproduced with permission from reference [161].
Figure 4. (A) PET/CT imaging of lung tumor-bearing mice (NCI-H292 cells) 2 h p.i. of [68Ga]Ga-NOTA-RGD-Cys-6-Ahx-GE11, [68Ga]Ga-NOTA-RGD, [68Ga]Ga-NOTA-GE11, [68Ga]Ga-NOTA-RGD-Cys-6-Ahx-GE11 + blocking. Arrows indicate tumors. (B) Tumor/muscle (T/M) ratio of [68Ga]Ga-NOTA-RGD-Cys-6-Ahx-GE11, [68Ga]Ga-NOTA-RGD, [68Ga]Ga-NOTA-GE11. Reproduced with permission from reference [161].
Ijms 25 08553 g004
Figure 5. (A) PET/CT imaging and (B,C) biodistribution of pancreas adenocarcinoma-bearing mice (BxPC3 cells) 2 h p.i. of [64Cu]Cu-NOTA-GE11, [64Cu]Cu-NOTA-RGD, [64Cu]Cu-NOTA-RGD-GE11 and in the presence of blocking dose of GE11, c(RGDyk), or both unlabeled peptides. The arrows indicate the tumors. Significant differences are indicated by: * p < 0.05, ** p < 0.01 and *** p < 0.001. Reproduced with permission from reference [132].
Figure 5. (A) PET/CT imaging and (B,C) biodistribution of pancreas adenocarcinoma-bearing mice (BxPC3 cells) 2 h p.i. of [64Cu]Cu-NOTA-GE11, [64Cu]Cu-NOTA-RGD, [64Cu]Cu-NOTA-RGD-GE11 and in the presence of blocking dose of GE11, c(RGDyk), or both unlabeled peptides. The arrows indicate the tumors. Significant differences are indicated by: * p < 0.05, ** p < 0.01 and *** p < 0.001. Reproduced with permission from reference [132].
Ijms 25 08553 g005
Table 1. Examples of EGFR-targeting peptides.
Table 1. Examples of EGFR-targeting peptides.
NameSequenceSourceRef.
EBPCMYIEALDKYACExperimentally synthesized.[42]
S3RCSHGYTGIRCQAVVLFrom the third loop structure and linear C-terminal region of vaccinia virus growth factor (VGF).[43,44]
GE11YHWYGYTPQNVIScreened out from a phage display peptide library.[18,31]
P1SYPIPDT[45]
P2HTSDQTN[45]
QRHQRHKPRE[46]
D4LARLLTComputer-aided design approach.[47]
P75KYFPPLALYNPTEYFYFrom one-bead-one-compound (OBOC) library.[48]
Pep 11WSGENGPGFYDYEADesigned using knob-socket model.[49]
---EEEEYFELV DEDEYFELVExperimentally synthesized.[50]
Table 2. Examples of integrin αVβ3-targeting peptides.
Table 2. Examples of integrin αVβ3-targeting peptides.
ExamplesComments
RGD peptides
RGD, c(RGDfK), c(RGDyK)RGD peptides can present linear or cyclic formats (cRGD) with different levels of selectivity and specificity. Linear RGD peptides are more sensitive to chemical degradation [51]. Cyclization aims to decrease susceptibility to enzymatic degradation, enhancing tracer stability [52]. c(RGDyK) and c(RGDfK) have been extensively studied, with c(RGDyK) showing superior affinity for integrin αVβ3 [25].
iRGDiRGD is a 9-amino-acid cRGD derived from phage display screening [53].
LXW7 and LXW64LXW7 (cGRGDdvc-NH2) was derived from the OBOC library and LXW64 (cGRGDd-DNaI1-c-NH2) is its optimized version [54].
RGD-4C and NC-100717RGD-4 C is a double-cysteine-bridged peptide and the development of NC-100717 was based on the PEGylation of RGD-4C [51,55,56].
c(RGDf[NMe]V (cilenglitide) and RGDechiCilenglitide acts as a selective αVβ3 antagonist. Preclinical studies have demonstrated its antiangiogenic and antitumor effects in various cancer models [45]. While cilengitide was well tolerated in phase I/II clinical trials [46,47], it failed to show efficacy in phase III studies [48], likely due to the complexity and plasticity of integrin signaling networks.
RGDechi is a designed αVβ3 antagonist based on cilenglitide structures combined with echistatin C-terminal tails [57].
Non-RGD peptides
ATN-161ATN-161 (Ac-PHSCN-NH2) is a capped pentapeptide synthesized from the fibronectin—PHSRN sequence and it can be used alone or along with radiotherapy and chemotherapy to prevent metastasis and tumor development [49,50,51]. In phase I of a clinical study, ATN-161 was used for aggressive solid tumors and demonstrated good toleration and safety [53].
RWrNK and RWrNMRWrNK and RWrNM are linear peptides that contains an unnatural d-arginine (r). They present great water solubility and the ability to pass through the blood–brain tumor barrier. They have been investigated for glioblastoma diagnosis [58,59].
Table 3. Direct labeling strategies for EGFR- and integrin αVβ3-targeting peptides.
Table 3. Direct labeling strategies for EGFR- and integrin αVβ3-targeting peptides.
EGFR-Targeting
RadionuclidePeptide/SequenceFormulationRCY * (%)RCP ** (%)Ref.
124IGE11 [124I]I-GE11 [31,35]
[124I]I-GE11≥47≥98[79]
131IEEEEYFELV [131I]I-EEEEYFELV>84>90[50]
DEDEYFELV[131I]I-DEDEYFELV>91>90
Integrin αVβ3-Targeting
125Ic(RGDyK)[125I]I-c(RGDyK)≥89>95[80]
77Br[77Br]Br-c(RGDyK)≥73>95
131IGRGDYV[131I]I-GRGDYV>95>94[62]
99mTcGRGDHV[99mTc]Tc(CO)3-GRGDHV>95>94
* RCY: radiochemical yield; ** CP: radiochemical purity.
Table 4. Indirect labeling strategies for EGFR-targeting peptides.
Table 4. Indirect labeling strategies for EGFR-targeting peptides.
EGFR-Targeting
RadionuclideChelator/
Prosthetic Group
Linker/SpacerPeptideFormulationRCY * (%)RCP ** (%)Ref.
64Cup-SCN-Bn-NOTA GE11 [64Cu]Cu-NOTA-GE114690[81]
β-alanina + ethylene glycol-based linker[64Cu]Cu-NOTA-linker-β-Ala-GE11 >99[36]
[64Cu]Cu-NOTA-linker-β-Ala-GE11-NH2 >99
18FN-succinimidyl 4-fluorobenzoate (SFB)-Gly-Gly-Gly-Lys-(GGGK)[18F]F-SFB-GGGK-GE11 95[79]
F-PEG4-propyne[18F]F-F-PEG4-propyne-GGGK-GE11 98
4-nitrophenyl-2-fluoropropionate (NFP) GE11/GE11-Lys [18F]F-NFP-Lys-GE11 7>99[32]
68GaNODAGAPEG (different lengths)GE11 [68Ga]Ga-NODAGA-PEGn-GE11 ≥97[82]
PEG5[68Ga]Ga-NODAGA-PEG5-GE115–74≥98[38]
D4 [68Ga]Ga-NODAGA-PEG5-D4
P1 [68Ga]-NODAGA-PEG5-P1
P2 [68Ga]Ga-NODAGA-PEG5-P2
CPP [68Ga]Ga-NODAGA-PEG5-CPP
EGBP [68Ga]Ga-NODAGA-PEG5-EGPB
QRH [68Ga]Ga-NODAGA-PEG5-QRH
Pep11 [68Ga]Ga-NODAGA-PEG5-Pep11
124IN-succinimidyl 4-iodobenzoate (SIB)GGGKGE11 [124I]I-SIB-GGGK-GE113098[79]
111Inp-SCN-Bn-NOTA[111In]In-NOTA-GGGK-GE11 100
99mTcHYNIC + tricineSeryl-seryl-serine residue (SSS)[99mTc]Tc-tricine-HYNIC-SSS-GE11>9895[34]
D4 [99mTc]Tc-tricine-HYNIC-SSS-D49898[78]
[99mTc]Tc-tricine-HYNIC-D49898[77]
HYNIC + tricine + EDDASSS[99mTc]Tc-tricine/EDDA-HYNIC-SSS-D4 98[76]
GE11 [99mTc]Tc-tricine/EDDA-HYNIC-SSS-GE11>9999[33]
-Gly-Gly-Gly-Cys-(GGGC) [99mTc]Tc-GGGC-GE11>98>90[37]
ECG (tripeptide–several nitrogen atoms and one sulfur atom)Histidine-containing spacer peptide (GHEG)P1 [99mTc]Tc-ECG-SYPIPDT-ECG-TAMRA>95 [83]
* RCY: radiochemical yield; ** RCP: radiochemical purity.
Table 5. Indirect labeling strategies for integrin αVβ3-targeting peptides.
Table 5. Indirect labeling strategies for integrin αVβ3-targeting peptides.
RadionuclideChelator/Prosthetic GroupLinker/SpacerPeptideFormulationRCY * (%)RCP ** (%)Ref.
64CuDOTA E{E[c(RGDfK)]2)2[64Cu]Cu-DOTA-E{E[c(RGDfK)]2)2/[64Cu]Cu-DOTA-RGD27595[84]
CB-TE2A (4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2] hexadecane)) c(RGDyK)[64Cu]Cu-CB-TE2A-c(RGDyK) 95[75]
Hexaazamacrobicyclic sarcophagine (Sar) based chelator (diamSar) c(RGDfD)[64Cu]Cu-diamSar-c(RGDfD) 95
DOTAPEG4E[c(RGDfK)]2[64Cu]Cu-DOTA-PEG4-E[c(RGDfK)]29595[85]
DOTATriglycine (3G)E[c(RGDfK)]2[64Cu]Cu-DOTA-G3-E[c(RGDfK)]29595
Sar-based chelator
(BaBaSar)
E[c(RGDfK)]2[64Cu]Cu-BaBaSar-E[c(RGDfK)]2/[64Cu]Cu-BaBaSar-RGD29599[86]
NODAGA E[c(RGDfK)]2[64Cu]Cu-NODAGA-E[c(RGDyK)]2/[64Cu]Cu-RGD250>89[69,87]
Sar-based chelator
(AmBaSar)
E[c(RGDfK)]2[64Cu]Cu-AmBaSar-RGD2/[64Cu]Cu-AmBaSar-E[c(RGDfK)]2>9098[88]
NOTAPEG4-SAA4c(RGDfK)[64Cu]Cu-NOTA-PEG4-SAA4-c(RGDfK)>90 [52]
NOTAPEG2c(RGDfK)[64Cu]Cu-NOTA-PEG2-c(RGDfK)>90
18FNFPPEG3E{c(RGDyk)2[18F]F-FPPRGD2>7399[70,89,90,91]
NFPPEG2, SAA and 1,2,3-triazolec(RGDfK)[18F]F-Galacto-RGD/[18F]F P-SAA-RGD>2498[66]
p-fluorobenzaldeyde aminooxy-bearing RGD peptide (AH111585)[18F]F-fluciclatide/[18F]F-AH111585>1795[92,93]
pentyne tosylate c(RGDyK)[18F]F-RGD-K53595[94,95,96]
SFB E[c(RGDyK)]2[18F]F-FB-RGD2>2099[68]
SFB c(RGDyK)[18F]F-FB-RGD>3599[68]
Al(NOTA)PEG3E[c(RGDyK)]2[18F]F-AlF-NOTA-PRGD2 [18F]F-AlF-NOTA-PEG3-E[c(RGDyK)]2/[18F]F-Altatide I>4095[97]
Al(NOTA)PEG4E[c(RGDfk)]2[18F]F-NOTA-E[PEG4-c(RGDfk)]2/[18F]F-AlF-NOTA-2P-RGD2/[18F]F-Alfatide II>4095[98]
68GaNODAGA c(RGDfK)[68Ga]Ga-RGD/[68Ga]Ga-NODAGA-c(RGDfK)9698[71,99]
NODAGA E[c(RGDfK)]2[68Ga]Ga-NODAGA-E[c(RGDyK)]2/[68Ga]Ga-RGD250>89[87,100,101]
DOTAPEG4E[c(RGDfK)]2[68Ga]Ga-3PRGD2/[68Ga]Ga (PEG4-E[PEG4-c(RGDfK)]2).9098[71,102]
TRAP PEG4c(RGDfK)3[68Ga]Ga-TRAP-(RGD)3≥9599[71,103]
p-SCN-Bn-NOTA3 PEG4E[c(RGDfK)]2[68Ga]Ga-NOTA-PRGD2/[68Ga]Ga NOTA-PEG4-E[c(RGDfK)]24295[69,104]
p-SCN-Bn-NOTA3GE[c(RGDfK)]2[68Ga]Ga-NOTA-E[G3c(RGDfK)]24395[69,104]
p-SCN-Bn-NOTA c(RGDyK)[68Ga]Ga-NOTA-RGD9899.5[67,73]
Fusarinine-C (FSC) c(RGDfK)3[68Ga]GaFSC-(RGD)39495[105]
THP c(RGDfK)[68Ga]Ga-HP3-(RGD)3>8695[72]
125ISIB c(RGDfK)[124I]I-bcRGD3699[106]
99mTcHYNIC + tricine +TPPTS3GE{c(RGDFk)2[99mTc]Tc-HYNIC-3G-(RGD)2>60>95[107,108]
HYNIC + tricine +TPPTS3 PEG4E{c(RGDFk)2[99mTc]Tc-3PRGD2/[99mTc]Tc-HYNIC-PEG4-E[PEG4-c(RGDfK)]2>95>95[109,110,111]
Cysteine (Cys)PNP = [(CH3)2P(CH2)2N(C2H4OCH3)(CH2)2P(CH3)2]RGDechi[99mTc]Tc(RGDechi-Cys)PNP43>8298[112]
HYNIC + tricine +EDDA c(RGDyK)[99mTc]Tc-HYNIC-RGD>94>95[113]
DOTA RAFT-E[c(RGDfK)]2[99mTc]Tc-RAFT-RGD >90[114,115]
HYNIC + tricine +TPPTS E[c(RGDfK)]2[99mTc]Tc(HYNIC-E E[c(RGDfK) 2]2)(tricine)(TPPTS)]6595[116]
MAG23GE{c(RGDFk)2[99mTc]TcO(MAG2-3G3-E{c(RGDFk)2 95[117]
HYNIC + tricine +TPPTSPEG2, SAA and 1,2,3-triazoleE{c(RGDFk)2[99mTc]Tc-Galacto-RGD2 >95[118,119]
111InDOTA E-[c(RGDfK)]2[111In]In-RGD2/[111In]InDOTA-E-[c(RGDfK)]2 >95[120]
DOTA c(RGDfK)[111In]In-RGD 90[121]
DOTAPEG2, SAA and 1,2,3-triazoleE-[c(RGDfK)]2[111In]In-DOTA-Galacto-RGD2 95[122]
DOTAPEG4E-[c(RGDfK)]2[111In]In-3P-RGD233>95[122]
DTPAPEG4E-[c(RGDfK)]2[111In]In-DTPA-3PRGD237>95[123]
DOTA E-[c(RGDfK)]2[111In]In-DOTA-RGD2>95 [124]
* RCY: radiochemical yield; ** RCP: radiochemical purity.
Table 6. Radiolabeling strategies for αVβ3-targeting peptides aiming therapy.
Table 6. Radiolabeling strategies for αVβ3-targeting peptides aiming therapy.
RadionuclideChelator LinkerPeptideFormulationRadiolabeling Efficiency (%)Ref.
177LuDOTA E-c(RGDfK)[177Lu]Lu-DOTA-E-c(RGDfK)>93[125]
NOTA-SCN c(RGDyK)[177Lu]Lu-NOTA-SCN-c(RGDyK)99[126]
DOTA3 PEG4E[c(RGDfK)]2[177Lu]Lu-DOTA-(PEG4)3-E[c(RGDfK)]2/
[177Lu]Lu-3PRGD2
>99[127]
90Y RAFT(c[-RGDfK-])4[90Y]Y-DOTA-RAFT-(c[-RGDfK-])4/[90Y]Y-RAFT-RGD >90[128]
177Lu[177Lu]Lu-DOTA-RAFT-(c[-RGDfK-])4/
[177Lu]Lu -RAFT-RGD
>97
5p-C-NETA c(RGDyK)[177Lu]Lu-5p-C-NETA-c(RGDyK)99[129]
90Y[90Y]Y-5p-C-NETA-c(RGDyK)99
NOTAEvans blue (EB)c(RGDfK)[90Y]Y-NOTA-EB- c(RGDfK)/[90Y]Y-NMEB-RGD>90[130]
177Lu [177Lu]Lu-NOTA-c(RGDfK)/[177Lu]Lu -RGD>95[131]
EB[177Lu]Lu-NOTA-EB-c(RGDfK)/[177Lu]Lu-EB-RGD>95
225AcDOTA E[c(RGDfK)]2[225Ac]Ac-DOTA- E[c(RGDfK)]2/[225Ac]Ac-DOTA-RGD2>95[124]
Table 7. Preclinical studies performed with radiotracers targeting EGFR.
Table 7. Preclinical studies performed with radiotracers targeting EGFR.
FormulationCell LineIn Vitro
Investigation
Animal ModelIn Vivo InvestigationBiodistribution Tumor Uptake (%ID/g)Imaging Tumor UptakeRef.
[124I]I-GE11SMMC-7721Binding affinity, internalization, MTTHepatocellular carcinomaBiodistribution (at 0.5 and 4 h p.i.), gene delivery and transfection3.2 (4 h p.i.)N/A[31]
[124I]I -GE11A431Binding affinity (no inhibition)Epidermal and glioblastoma Efficiency of treatmentN/AN/A[35]
[64Cu]Cu-NOTA-linker-β-Ala-GE11-NH2 and [64Cu]Cu-NOTA-linker-β-Ala-GE11A431, MDA-MB-435 and FaDuHuman serum stability, cell medium and buffer viability, binding saturation, displacement assays on intact cells and homogenates, immunoblottingHead and neck squamous cell carcinoma (HNSC)Biodistribution (at 5 and 60 min p.i.), PET (at 1 h and 36 h p.i.)<1 (5 min p.i.)Some [64Cu]Cu-NOTA-linker-β-Ala-GE11 accumulation was detected 5 min p.i.[36]
[18F]FP-Lys-GE11A431, U87MGand PC3Receptor binding, partition coefficient, stability, cell uptake and blocking assaysGlioblastoma and prostate cancermetabolism, immunohistochemistry, biodistribution at 2 h p.i., and 2 h dynamic PET/CT imagingN/A3.5 ± 0.4 (U87) and 3.7 ± 0.8 (PC-3)[32]
[99mTc]Tc-tricine-HYNIC-SSS-GE11SKOV3Plasma stability, receptor binding, internalization and, blocking assayOvarian cancerBiodistribution at 1 and, 4 h p.i.~2.3 (1 h p.i.)N/A[34]
[99mTc]Tc-tricine-HYNIC/EDDA-SSS-GE11Receptor binding and, internalizationBiodistribution at 1 and, 4 h p.i., gamma camera imaging at 1 h p.i.3.6 ± 0.7 (1 h p.i.)Good A549 tumor visualization at 1 h p.i.[33]
[99mTc]Tc-GGGC-GE11A549Cellular uptake, retention kinetics, internalization and blocking assaysNon-small-cell lung cancer
(NSCLC)
Biodistribution at 1 and, 4 h p.i., SPECT scans at 1, 2, and 4 h p.i.3.4 ± 0.5 (2 h p.i.)The signal of A549 tumors reached its strongest at 2 h p.i.[37]
[99mTc]Tc-tricine-HYNIC-SSS-D4Stability in solution and human serum, receptor binding, internalization assaysBiodistribution at 1 and, 4 h p.i., gamma camera imaging at 1 h p.i.~7.6 (1 h p.i.)Good A549 tumor visualization at 1 h p.i.[78]
[99mTc]Tc-tricine-HYNIC-D4Stability, receptor bindingBiodistribution at 1 and, 4 h p.i.~8.1 (1 h p.i.)N/A[77]
[99mTc]Tc-tricine/EDDA-HYNIC-SSS-D4Cellular uptake and blocking testBiodistribution at 1 and, 4 h p.i.~2.5 (1 h p.i.)N/A[76]
[99mTc]Tc-SYPIPDT-ECG-TAMRANCI-H460Receptor binding affinity, cellular uptake by microscopyNSCLC Gamma camera imaging and biodistribution at 1, 2, and 3 h p.i., fluorescent imaging and immunohistochemical staining1.9 ± 0.1 (1 h p.i.)2.7 ± 0.6 at 1 h p.i.[83]
[131I]I-EEEEYFELV and [131I}I-DEDEYFELVC6Stability, partition coefficient, serum protein bindingGlioblastoma allograftsBiodistribution at 0.25, 1, and 2 h p.i.), binding, and internalization studies in brain homogenates2.3 ± 0.2 ([131I]I-EEEEYFELV) and 0.6 ± 0.0 ([131I]I-DEDEYFELV) at 1 h p.i.N/A[50]
N/A: not available.
Table 8. Preclinical studies performed with PET tracers targeting integrin αVβ3.
Table 8. Preclinical studies performed with PET tracers targeting integrin αVβ3.
FormulationCell Line In Vitro Investigation Murine Model In Vivo Investigation Biodistribution Tumor Uptake (%ID/g) PET Imaging Tumor UptakeRef.
[18F]F-FPPRGD2U-87 MGReceptor-binding assay (IC50 = 51.8 ± 4.6 nM)GlioblastomaBiodistribution at 1 h p.i., and static microPET scans at 20 min, 1 and 2 h p.i.2.3 ± 0.24.2 ± 0.2 (20 min)[70]
[18F]F-Galacto-RGDReceptor-binding assay (IC50 = 404 ± 38 nM)Static microPET scans at 20 min, 1 and 2 h p.i.N/A2.1 ± 0.2 (20 min)[70]
[18F]F-fluciclatideEA-Hy926Biding affinity (Ki = 2.3 nM) Lewis lung Ccarcinoma (LLC)Biodistribution and PET at 2 h p.i., blocking study1.6 ± 0.11.5 ± 0.3[92]
[18F]F-RGD-K5U-87 MGxGlioblastomaMetabolic stability, biodistribution, PET, blocking study4.2 ± 0.61.4 ± 0.2 (U-87 MG); 1.2 ± 0.4 (A549) (SUVmax)[99]
[18F]F-RGD2Receptor-binding assay IC50 = 2.3 ± 0.7 nMBiodistribution at 0.5, 1 h, 2 h, and 4 h p.i., PET 1 h p.i.4.3 ± 1.0 (2 h p.i.)4.4 ± 0.6 (1 h p.i.)[68]
[18F]F-RGDReceptor-binding assay IC50 = 3.5 ± 0.3 nMBiodistribution at 0.5, 1 h, 2 h, and 4 h p.i., PET 1 h p.i.1.6 ± 0.4 (2 h p.i.) [68]
[18F]F-Alfatide IReceptor-binding assay IC50 = 46 ± 4.4 nMSerum stability, biodistribution at 2 h p.i. and dynamic (2 -35 min) and static PET 1 and 2 h p.i., blocking study2.3 ± 0.9 (2 h p.i.)5.3 ± 1.2% (peak at 3 min p.i); 0.3 ± 0.1 (60 min p.i.)[97]
[18F]F-Alfatide IIU-87 MG and MDA-MB-435 N/AGlioblastoma or breast cancerDual PET imaging (18F-alfatide and 18F-FDG) of mice treated with doxorubicin or paclitaxel, computational modeling (ROI, time–activity curves, dual-tracer input function and tumor time–activity curve separation and kinetics.N/A4.7 ± 1.0%ID/g at 40 min[98]
[68Ga]Ga-RGD U-87 MG and A549N/AGlioblastoma and NSCLC Biodistribution at 80 min p.i. and statistic PET at 1 h p.i.2.9 ± 0.8 (U-87 MG) and 3.9 ± 1.2 (A549)0.9 ± 0.3 (U-87 MG); 0.9 ± 0.3 (A549) (SUVmax)[99]
[68Ga]Ga-RGD2U-87 MG and H727 N/AGlioblastoma and neuroendocrine tumorsPET/CT scans, blocking and biodistribution (in major organs) at 1, 2 and 4 h p.i., dosimetryN/A2.2 ± 0.1 (U-87 MG); 1.5 ± 0.1 (A549) 1 h p.i.[87]
B16-F10In vitro stabilityMelanomaBiodistribution at 10 min, 0.5 and 1 h p.i. and blocking studies at 0.5 h and clinical study4.1 ± 0.9N/A[101]
[68Ga]Ga-3PRGD2U-87 MG and LCCN/ALLC PET (1 h p.i.), immunofluorescence, western blotN/A4.9 ± 1.2[102]
[68Ga]Ga-TRAP-(RGD)3MDA-MB-231N/ABreast cancerImmunohistochemistry analysis, PET/CT at 1 h p.i., autoradiography and immunofluorescence imagingN/A3.0 ± 0.9[103]
[68Ga]Ga-NOTA-PRGD2U-87 MGReceptor-binding assay (IC50 = 88.8 ± 5.4 nM)GlioblastomaBiodistribution at 1 h p.i. and PET imaging at 0.5, 1 and 2 h p.i. 7.0 ± 1.19.0 ± 2.0 (30 min)[104]
[68Ga]Ga-NOTA-E[G3c(RGDfK)]2Receptor-binding assay (IC50 = 61.6 ± 3.3 nM)Biodistribution at 1 h p.i. and PET imaging at 0.5, 1, and 2 h p.i. 8.0 ± 0.910.1 ± 1.8 (0.5 h)[104]
[68Ga]Ga-NOTA-RGDSNUC4Binding assay with human serum (Ki = 1.9 nM)Colon cancer Biodistribution 1 h p.i., blocking study and PET imaging at 1 and 2 h p.i.1.6 ± 0.25.1 ± 1.0[142]
[68Ga]Ga FSC-(RGD)3M21Stability, protein binding, binding affinity, internalization MelanomaImmunohistochemistry and PET/CT at 1 h p.i. N/A3.0 ± 0.9[105]
[68Ga]Ga-HP3-(RGD)3Binding assay (IC50 = 73 ± 22 nM)Biodistribution 1 and 2 h p.i., blocking, and dynamic PET imaging for 1.5 h p.i., stability6.1 ± 0.6 (1 h)qualitative[143]
[64Cu]Cu-RGD2U-87 MG and H727N/AGlioblastoma and neuroendocrine tumors PET/CT scans, blocking and biodistribution in major organs 1, 4, and 18 h p.i., dosimetryN/A2.3 ± 0.2 (U-87 MG); 1.5 ± 0.1 (H727) 1 h p.i.[87]
[64Cu]Cu-DOTA-
E[c(RGDfK)]2
U-87 MGBinding assay (IC50 = 73 ± 22 nM)Glioblastoma Biodistribution 1 h p.i., PET imaging at 15, 30 min, 1, 2, 4, and 18 h p.i., dosimetry9.9 ± 1.1 (0.5 h)8.7 ± 1.5 (1 h p.i.)[84]
[64Cu]Cu-CB-TE2A-c(RGDyK)M21Binding assay (IC50 = 6.0 ± 3.6 nM)Melanoma Biodistribution, blocking and PET imaging at 1, 2, 4 and 24 h pi3.0 ± 0.9 (1 h p.i.)qualitative[75]
[64Cu]Cu-diamSar-c(RGDfD)Binding assay (IC50 = 4.8 ± 0.9 nM)Biodistribution, blocking and PET imaging at 1, 2, 4 and 24 h pi1.5 ± 0.5 (1 h p.i.)qualitative
[64Cu]Cu-DOTA-PEG4-E[PEG4-c(RGDfK)]2U-87 MGBinding assay (IC50 = 74 ± 3 nM)Glioblastoma Biodistribution at 0.5, 1 and 2 h p.i., blocking and PET imaging at 1 h p.i.8.2 ± 2.0 (0.5 h p.i.)qualitative[85]
[64Cu]Cu-DOTA-G3-E[G3-c(RGDfK)]2Binding assay (IC50 = 62 ± 6 nM)Biodistribution at 0.5, 1 and 2 h p.i., blocking and PET imaging at 1 h pi8.5 ± 1.4 (0.5 h p.i.)qualitative
[64Cu]Cu-AmBaSar-RGD2Binding assay (IC50 = 10.0 ± 0.5 nM)PET imaging at 1, 2, 4, and, 20 h p.i., blocking and biodistribution at 20 h1.8 ± 0.4~2.5 (20 h)[88]
[64Cu]Cu-NOTA-PEG4-SAA4-c(RGDfK)Binding assay (IC50 = 444 ± 41 nM)PET imaging at 0.5, and 2 and 4 h p.i, blocking and biodistribution at 4 h p.i2.5 ± 0.2 (0.5 h p.i.)1.1 ± 0.3[52]
[64Cu]Cu-NOTA-(PEG)2-c(RGDfK)Binding assay (IC50 = 288 ± 66 nM)PET imaging at 0.5, and 2 and 4 h p.i, blocking and biodistribution at 4 p.i.~4 (0.5 h p.i.)2.4 ± 0.3
[125I]I-bcRGDU-87 MG and A549Selective (2.1 ± 0.6) and binding assay (1.6% and 0.3% dose/mg for U-87 MG and A549)Biodistribution at 10 min, 0.5, 1 and 2 h p.i. 3.8 ± 0.4 (U-87 MG); 2.1 ± 0.6 (A549) (0.5 h p.i.)N/A[106]
N/A: not available.
Table 9. Preclinical studies performed with SPECT tracers targeting integrin αVβ3.
Table 9. Preclinical studies performed with SPECT tracers targeting integrin αVβ3.
FormulationCell Line In Vitro Investigation Murine Model In Vivo Investigation Biodistribution Tumor Uptake (%ID/g) Ref.
[99mTc]Tc-HYNIC-3G-RGD2U-87 MGPartition coefficient, binding assay U-87 MG (IC50 = 61.1 ± 2.1) Glioblastoma and breast cancer Biodistribution at 0.5, 1 and 2 h p.i., blocking, SPECT/CT and, metabolism9.1 ± 1.8 (MDA-MB-435); 7.7 ± 1.2 (U-87 MG) (2 h p.i.)[107]
[99mTc]Tc-3PRGD2Binding assay (IC50 = 2.4 ± 0.7 nM)Glioblastoma Biodistribution at 0.5, 1 and 2 h p.i., blocking, SPECT/CT (0.5, 1, 2 and 4 h p.i.) and, metabolism9.7 ± 3.2 (2 h p.i.)[110]
[99mTc]Tc (RGDechi-Cys)(PNP)43Flow cytometry, cell uptakeBiodistribution at 0.5 and 2 h p.i. and metabolism~0.4 (2 h p.i.)[112]
[99mTc]Tc-HYNIC-RGDM21Protein binding in fresh human plasma, binding affinity Melanoma Biodistribution at 1 and 4 p.i. and planar γ-camera (10 min) imaging2.1 ± 0.4 (M21); 1.5 ± 0.3 (A549) (4 h p.i.)[113]
[99mTc]Tc-RAFT-RGDB16F0- and TS/A-pcImmunoprecipitation, western blot, blood distributionMelanoma and mammary cancerBiodistribution, immunohistochemistry, autoradiography, planar γ-camera and SPECT imaging2.4 ± 0.5 (B16F0); 2.7 ± 0.8 (TS/A-pc) (1 h p.i.)[115]
[99mTc]Tc-RAFT-RGDDAOY-Luc spheroids or HD-MB03-LucExperiments involving the generation of radioresistant medulloblastoma cell linesIntracranial orthotopic SPECT and bioluminescenceN/A[114]
[99mTc]Tc (HYNIC-E E[c(RGDfK) 2] 2)(tricine)(TPPTS)]MDA-MB-435Solution stability, partition coefficient, binding assay (IC50 = 51 ± 11 nM)Breast cancer Biodistribution at 5 min, 0.5, 1 and 2 h p.i., SPECT at 1, 2 and 4 h p.i. and metabolism7.3 ± 1.3 (2 h p.i.)[116]
[99mTc]TcO(MAG2-3G3-E{c(RGDFk)2U-87 MGBinding assay (IC50 = 3.6 ± 0.6 nM), partition coefficientGlioblastoma Biodistribution at 0.5 and 2 h p.i., planar imaging and metabolic stability8.3 ± 1.5 (2 h p.i.)[117]
[99mTc]Tc-Galacto-RGD2Binding assay (IC50 = 20 ± 2 nM)Biodistribution at 5, 30, 60 and 120 min p.i., blocking, SPECT/CT at 1 h p.i. and metabolism, immunostaining, immunohistochemistry 5.6 ± 1.5 (2 h p.i.)[118]
[111In]In-RGD2/[111In]In-DOTA-E-[c(RGDfK)]2FaDu, SCCNij3, and SCCNij20N/AHNSCCBiodistribution at 1 h p.i., immunohistochemistry, autoradiography and SPECT/CT (3 frames of 20 min)2.2 ± 0.0 (FaDu), 1.9 ± 0.5 (SCCNij3), and 1.2 ± 0.0 (SCCNij20) (1 h p.i.)[120]
[111In]In (DOTA-Galacto-RGD2)U-87 MG and MDA-MB-435Binding assay U-87 MG (27 ± 2 nM)Breast cancerBiodistribution at 1, 4, 24 and 72 h, planar imaging at 1, 4 and 24 h p.i., blocking, immunostaining, immunohistochemistry6.8 ± 1.0 (1 h p.i.)[122]
[111In]In-3P-RGD2Binding assay U-87 MG (29 ± 4 nM)Biodistribution at 1, 4, 24 and 72 h, planar imaging at 1, 4 and 24 h p.i., blocking, immunostaining, immunohistochemistry6.2 ± 1.6 (1 h p.i.)[122]
[111In]In-DOTA-RGD2NT2.5 and MDA-MB-231Serum stabilityBiodistribution at 0.5, 1.5, 3, 6 and 24 h4.8 ± 0.7(NT2.5), 2.2 ± 0.9 (MDA-MB-231) (3 h p.i.)[124]
N/A: not available.
Table 10. Preclinical studies performed with radiotracers targeting integrin αVβ3 aiming therapy.
Table 10. Preclinical studies performed with radiotracers targeting integrin αVβ3 aiming therapy.
FormulationTherapy Dose (MBq)Murine Model In Vivo Investigation Main Findings Ref.
[177Lu]Lu-DOTA-E-c(RGDfK)37OVCAR-3 ovarian cancer Survival studyTreated mice survived 16 weeks more than the untreated group.[125]
[177Lu]Lu-NOTA-SCN-c(RGDyK)0.37CT-26 colon cancer Biodistribution (at 1, 2, 4, 12 and 24 h p.i.)The tumor uptake and the tumor/muscle ratio at 1 h p.i. was 1.7 ± 0.3 and 2.1 ± 0.4%ID/g. The highest uptake was observed in kidneys 7.6 ± 0.7%ID/g.[126]
[177Lu]Lu-3PRGD20.37, 37.74 and 111LCC tumor model Biodistribution (at 1, 4, 24 and 72 h p.i.), gamma imaging (at 4 and 24 h p.i.) and maximum tolerated dose (MTD), immunohistochemistry and hematoxylin-eosin stainingThe tumor uptake at 1 h p.i. was 6.0 ± 0.6%ID/g and remained at 1.2 ± 0.2%ID/g 72 h p.i. Highest uptake was observed in the intestine (5.2 ± 0.5%ID/g) and kidney (4.2 ± 1.1) at 1 h p.i. The MTD was greater than 111 MBq per mouse.[127]
[90Y]Y-RAFT-RGD30–37 (1× or fractionated 2×)U-87 MG glioblastomaBiodistribution at 1, 4, 24 and 48 h p.i. of 0.37 MBq of [90Y]Y-RAFT-RGD, toxicity and, dosimetry The tumor uptake of [90Y]Y-RAFT-RGD 1 h p.i. was quick and high (9.0 ± 4.3% ID/g) and remained at 1.8 ± 0.7% ID/g 48 h p.i. The highest kidney uptake was 13.9 ± 3.5%ID/g at 1 h p.i. The toxicity findings were as follows: reduction in leukocyte and platelet counts and higher serum creatinine levels in the treated groups (compared to control). Radiation dosimetry extrapolation to humans: the whole-body effective dose was estimated at 0.11 mSv/MBq.[128]
[177Lu]Lu-RAFT-RGD30–37SPECT/CT (at 1 and 4 h p.i.), toxicityThe tumor uptake at 1 and 4 h p.i. was 3.3 ± 0.5%ID/g and 3.8 ± 0.9%ID/g, and remained at 1.6 ± 0.0%ID/g 48 h p.i. The tumor/muscle ratio was ~10 at 1 h p.i.The highest activity levels (~6%ID/g) were detected in the kidneys and the bladder. Toxicity findings: reduction in leukocyte and platelet counts in treated groups (compared to control).
[177Lu]Lu-5p-C-NETA-c(RGDyK)2.22Biodistribution at 1, 4 and, 24 h p.iTumor uptake was 1.4 ± 0.6% ID/g 1 h p.i. The kidneys presented the highest uptake 1 h p.i. (3.2 ± 0.5%ID/g)[129]
[90Y]Y-NMEB-RGD7.4, 3.7 and 1.75 MBqAntitumoral radiotherapy efficacy, TUNEL and hematoxilin-eosin stainingThe treated group presented lower tumor volumes. The tumor vasculature was lower in the group receiving a medium dose of 90Y-NMEB-RGD. Also, this group presented more cell apoptosis.[130]
[177Lu]Lu-RGD18.5 and 29.6 Patient-derived xenografts (PDX) from NSCLCSPECT imaging (at 4, 24, 48, 72, and 96 h p.i.) and biodistribution (at 4, 24, 48 and, 72 h p.i.), therapy regimen, CD31 immunohistochemistry[177Lu]Lu-EB-RGD demonstrated higher tumor uptake than [177Lu]Lu-RGD (13.4 ± 1.0 vs. 2.6 ± 1.6%ID/g). The [177Lu]Lu-EB-RGD group showed more cell apoptosis than control and [177Lu]Lu-RGD. A therapy dose of 18.5 MBq [177Lu]Lu-EB-RGD might be strong enough.[131]
[177Lu]Lu-EB-RGD
[225Ac]Ac-DOTA-cRGDfK0.037HER2 (NT2.5 and MDA-MB-231) breast cancer The estimation of maximum tolerated activity (eMTA), biodistribution in normal tissues, α-CameraThe organs receiving the highest mean absorbed dose were kidneys (2.5 Gy) > spleen (1.8 Gy) > liver (1.2 Gy). The eMTA was 150 kBq (kidneys as the limiting tissue). The decay of daughters (213Bi and 221Fr) was also monitored. All the free 221Fr and the majority of the 213Bi decayed at 3 h.[124]
Table 11. Clinical trials using radiolabeled RGD-based peptides.
Table 11. Clinical trials using radiolabeled RGD-based peptides.
Agent Clinical TrialsSUVmaxCancer TypeNo. of Patients Clinical Phase/Study Period Radio-Dose Toxicity Imaging ToolComments Ref.
[99mTc]Tc-RGD-SCRGDSY/[99mTc]Tc-αP2)N/AN/AMetastatic melanoma14
(10 men and 4 women)
Up to 12 months185 to 1222 MBqNo adverse effectSPECT/CTTumor detection.[146]
[18F]F-Galacto-RGDN/ARanged in tumors (1.2 to 9.0)Malignant melanoma, sarcomas, and osseous metastases19
(9 men, 10 women)
Phase I/II133–200 MBqN/APETDemonstrate a highly favorable biodistribution in humans with specific receptor binding, and visualization of αvβ3 expression in tumors with high contrast.[147]
[99mTc]Tc-3 PRGD2N/AN/ADifferentiated thyroid cancer patients with radioactive iodine refractory lesions (RAIR-DTC)10
(2 men and 8 women)
N/A11.1 MBq/kgN/ASPECT/CT[99mTc]Tc-3PRGD2 with SPECT is a promising modality for diagnosing and guiding further treatment of RAIR DTC.[148]
[18F]F-FPPRGD2NCT018066750.8–5.8Glioblastoma multiforme recurrence17
(8 men and 9 women)
Phase I/IIThe [18F]F-FPPRGD2 doses at injection ranged from 3.8 to 9.9 mCi (mean, 8.1 mCi ± 1.7)SafePET/CTFunction as increase uptake of glioblastoma.[149]
[18F]F-alfatideN/A5.4 ± 2.2Lung cancer26N/A(213.3 ± 29.8 MBq)SafePET/CTEffective in the diagnosis of NSCL.[150]
[68Ga]Ga-NODAGA-RGD N/AN/AHepatocellular carcinoma9 menPhase I154–184 MBqWell tolerated and metabolically stable in humansPET/CTUptake in HCC tumors was not enough.[151]
[68Ga]Ga-NODAGA-RGD2N/A>10Locally advanced breast cancer5 women (33 to 68 years old)N/A111–185 MBqN/APET/CTGood uptake in tumor site.[101]
NCT029707864.5–17.7Neuroendocrine neoplasms and breast cancer10 (5 with neoplasm and 5 with breast cancer)Phase I97.3–220 MBqSafe.PET/CTGood image contrast and stable retention in tumor[152]
NCT032712811.4–14.1Neuroendocrine neoplasms113Phase II104–226 MBqNo grade 3–5 adverse eventsPET/CTGreat tumor uptake[153]
[18F]F-RGD-K5NCT01447134For main tumor (5.3–6.0)
For nodal (3.3–4.7)
Head and neck carcinoma (HNC)11Phase II10 mCiN/APET/CTAssessing response to concurrent chemoradiotherapy (CCRT) in patients with advanced HNC.[154]
[18F]F-FPPRGD2NA3.7 ± 1.3Cervical and ovarian tumors6 womenNARanged from 196 to 344 MBqN/APET/CTHave potential for early prediction of response to treatment.[89]
[18F]F-ALF-NOTA-PRGD2NCT033845115.4 ± 2.6Lung cancer, stomach cancer, cervical cancer, gall bladder cancer, breast cancer, nasopharyngeal carcinoma, soft tissue carcinoma, esophageal cancer38
(only 25 met criteria)
Phase IV224.6 ± 38.2 MBqN/APET/CTPredictive for treatment response of antiangiogenic treatment (apatinib).[155]
[99mTc]Tc-3PRGD2NCT02744729Malignant vs. benign lesions
(5.1 ± 2.0 vs. 2.0 ± 0.7)
Esophageal cancer29
(24 men and 5 women)
Early phase I11.1 MBq/kgN/ASPECT/CTValuable for the diagnosis and staging of esophageal cancer.[156]
[68Ga]Ga-RGDNCT04222543Ranged between 4.0 and 12.7Oral squamous cell carcinoma (OSCC).10 Phase II214 ± 9 MBqN/APET/CTProvide insight in angiogenesis as a hallmark of the head and neck squamous cell carcinomas’ tumor microenvironment.[157]
[68Ga]Ga-NOTA-3P-TATE-RGDNCT0281794527.2 ± 13.6Gastroenteropancreatic–neuroendocrine tumors (GEP-NETs)35 Early phase I74–148 MBqN/APET/CTDetection of liver metastases.[158]
[68Ga]Ga-FAPI-RGDNCT05543317Primary tumors: SUVmax 18.0
and lymph node metastases: SUVmax 12.1
Nasopharyngeal carcinoma, small lung cancer, pancreatic cancer; lymph node, brain, lung, liver, bone, and subcutaneous metastasis.22 NA3.0–3.7 MBq/kgSafe and well toleratedPET/CTImaging of various cancer types and functions to increase tumor uptake.[159]
N/A: not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rodrigues Toledo, C.; Tantawy, A.A.; Lima Fuscaldi, L.; Malavolta, L.; de Aguiar Ferreira, C. EGFR- and Integrin αVβ3-Targeting Peptides as Potential Radiometal-Labeled Radiopharmaceuticals for Cancer Theranostics. Int. J. Mol. Sci. 2024, 25, 8553. https://doi.org/10.3390/ijms25158553

AMA Style

Rodrigues Toledo C, Tantawy AA, Lima Fuscaldi L, Malavolta L, de Aguiar Ferreira C. EGFR- and Integrin αVβ3-Targeting Peptides as Potential Radiometal-Labeled Radiopharmaceuticals for Cancer Theranostics. International Journal of Molecular Sciences. 2024; 25(15):8553. https://doi.org/10.3390/ijms25158553

Chicago/Turabian Style

Rodrigues Toledo, Cibele, Ahmed A. Tantawy, Leonardo Lima Fuscaldi, Luciana Malavolta, and Carolina de Aguiar Ferreira. 2024. "EGFR- and Integrin αVβ3-Targeting Peptides as Potential Radiometal-Labeled Radiopharmaceuticals for Cancer Theranostics" International Journal of Molecular Sciences 25, no. 15: 8553. https://doi.org/10.3390/ijms25158553

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop