Karan S.
Tanwar
ab and
Mukesh K.
Pandey
*acd
aDivision of Nuclear Medicine, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN-55905, USA. E-mail: pandey.mukesh@mayo.edu; Tel: +1-507-538-4221
bPost Graduate Institute of Medical Education and Research, Chandigarh-160012, India
cMayo Clinic Comprehensive Cancer Center, Rochester, MN-55905, USA
dDepartment of Pharmacology, Mayo Clinic, Rochester, MN-55905, USA
First published on 29th July 2025
This review delves into the realm of gallium-68 (68Ga)-labeled radiopharmaceuticals. Over the last decade, 68Ga-labeled radiopharmaceuticals have gained prominence and shown tremendous growth in both preclinical evaluation and clinical translation due to their accessibility, favourable physical properties, and simple chemistry. Despite the high positron emission energy of 836 keV, it has been extensively used in diagnostic imaging. The present review aims to elucidate the status of various potential and clinically relevant 68Ga-labeled radiopharmaceuticals, as well as their preclinical and clinical stages. In this review article, we briefly discuss the physical characteristics, mode of production, suitable bifunctional chelators, and radiolabeling chemistry of 68Ga. Various 68Ga-labeled radiopharmaceuticals are discussed with respect to their biological targets, preclinical results, clinical results if available, and their current state of readiness for clinical translation and application in humans.
These limitations have resulted in a demand to explore alternative radioisotopes that are easy to produce, diverse and possess feasible aqueous medium chemistry, while maintaining good PET imaging characteristics.
68Ga has emerged as a potent radionuclide with this versatility. In 1966, Gottschalk et al. introduced 68Ga for the first time in the clinical space, investigating its role in studies of the central nervous system.3 However, the breakthrough in the clinical employment of 68Ga appeared with the advent of the 68Ge/68Ga generator, which provides 68Ga in the form of [68Ga]GaCl3 instead of [68Ga]Ga-EDTA. This generator facilitates easy availability and cost-effective commercial access to 68Ga, enabling simple labeling chemistry in an aqueous medium (suitable for small molecules, peptides, proteins, macromolecules, and nanoparticles).4 In addition, the development of various commercially available macrocyclic chelators and introduction of lutetium-177 (177Lu) as a theranostic pair escalated the clinical utility of 68Ga. In the past few years, several 68Ga-labeled radiopharmaceuticals have been successfully employed in the field of oncology, cardiology, pulmonology, infection, and inflammation imaging. Also, various target-specific (receptors, enzymes, antigens, and small effector) and biological mechanism-specific (proliferation, apoptosis, hypoxia, glycolysis, and angiogenesis) 68Ga-labeled radiopharmaceuticals have been explored. This radionuclide continues to play a potent role in the progression of nuclear medicine globally in clinics and research. The present review article focuses on 68Ga-based radiopharmaceuticals explored at the preclinical and clinical stages in the last decade (up to 2023).
Recent advancements in the development of molecular imaging probes in PET have made it possible to image and quantify cells undergoing apoptosis. Various PET imaging probes targeting the cell membrane, cell membrane acidification, membrane asymmetry, caspase substrates or inhibitors have been explored for apoptosis imaging in different disease conditions. In 2011, Bauwens et al. demonstrated the binding of two 68Ga-DOTA-maleimide-labeled annexin A5 analogues (Cys2- and Cys165-AnxA5) to apoptotic cells in a mouse model. PET/MRI images of both tracers demonstrated their high uptake in the liver and spleen of anti-Fas-treated mice but low uptake in the tumor. According to the authors, the reason for the high discrepancy in the uptake of both tracers between the apoptotic liver and apoptotic tumor is due to the death-induced heterogenicity in the tumor. Based on the results, these tracers cannot be used as tumor imaging agents in conventional PET imaging.29 However, they may serve as potential apoptosis imaging agents in systemic organs, atherosclerosis, acute myocardial infarction, etc.
Vascular cell adhesion molecule-1 (VCAM-1) is a cell surface-expressed transmembrane glycoprotein of the immunoglobulin family. It is closely related to the development of various tumor diseases, i.e., breast cancer,30 ovarian cancer,31 and renal cell carcinoma,32 leading to tumor metastasis. Several tumor tissues are known to be affected by its anti-apoptotic actions. Upregulation and downregulation of VCAM-1 expression in various cancer conditions give a better understanding of tumor growth and tumor cell destruction, respectively. Zhang et al. developed a novel 68Ga-labeled radiopharmaceutical, [68Ga]Ga-NOTA-VCAM-1scvf, to study the expression of VCAM-1 in tumors. The radiolabeling yield and radiochemical purity (RCP) of [68Ga]Ga-NOTA-VCAM-1scvf were found to be 96.92% ± 2.02% and 96.97% ± 2.13%, with a molar activity of 10.17 ± 1.07 MBq per nmol. The serum stability test of [68Ga]Ga-NOTA-VCAM-1scvf revealed >90% RCP at 3.0 h. In vitro immunofluorescence staining performed in B16F10 and A375m cells showed high fluorescence intensity in VCAM-1 expressing B16F10 cells. Also, microPET/CT imaging showed its high uptake and retention in a B16F10 xenograft tumor. The variation in uptake was also seen with the effect of LY2409881, an IKKβ inhibitor, which can cause apoptosis of VCAM1 expressing cells, of the growth of B16F10 melanoma. The results suggest that [68Ga]Ga-NOTA-VCAM-1scvf can specifically bind to tumors with overexpression of VCAM-1 with good retention.33
Apoptosis involves the expression of phosphatidylethanolamine (PE) on the cell membrane surface. PE is a promising biological target for apoptosis imaging. To target PE, duramycin has been investigated given that it demonstrates high affinity and specificity towards PE. It is a small cyclic polypeptide containing 19 amino acids. It has a low molecular weight with favourable characteristics such as stability against enzymatic hydrolysis and ease of conjugation and labeling. Considering these characteristics, the potential of duramycin has been explored for the in vivo imaging of cell death.34 The effectiveness of [68Ga]Ga-NODAGA-duramycin in identifying chemotherapy-induced organ damage in mice was examined by Rix et al. utilizing PET/CT. In the case of [68Ga]Ga-NODAGA-duramycin, the radiochemical yield and RCP were both greater than 95%. The binding specificity of [68Ga]Ga-NODAGA-duramycin towards damaged cells was demonstrated using an in vitro competitive binding assay. [68Ga]Ga-NODAGA-duramycin uptake was assessed in different cell lines in the presence of unlabeled duramycin, which demonstrated a significant reduction in tracer uptake (p < 0.001), confirming its competitive specific binding. Additionally, the PET/CT images showed the uptake of [68Ga]Ga-NODAGA-duramycin in the toxicity-induced organs, which was also confirmed by the immunohistochemistry and blood parameter analysis, thereby endorsing PE-mediated apoptosis imaging.35
[68Ga]Ga-NODAGA-4-(N-(S-glutathionylacetyl)amino)-phenylarsonous acid (GSAO), also known as [68Ga]Ga-cell death indicator (CDI), is another tracer that has been assessed for cell death imaging. GSAO is a trivalent arsenic-based peptide, which enters dying cells via anionic transporters and forms a complex with heat shock 90 proteins (hsp90). A preclinical study performed by Shon et al. demonstrated the favourable pharmacokinetic, imaging characteristics, and radiation dosimetry of a novel tracer.36 Further, to facilitate clinical translation, Shon et al. also conducted a first in human studies using [68Ga]Ga-CDI for imaging tumor cell death. A total of 5 patients with squamous cell carcinoma (SSC) of the ovary, serous carcinoma of the ovary, SSC of the oesophagus, ductal breast carcinoma, and adenocarcinoma were investigated. All the patients showed no adverse side effects upon the intravenous administration of [68Ga]Ga-CDI. PET imaging showed faster clearance of the tracer via the kidneys, where 39% ± 18% of the total radioactivity was excreted within 90 min post injection. In the remaining organs, the tracer demonstrated low-level physiological uptake and varying tumoral uptake depending on the histology of the tumor. Its uptake was observed in all cancer types, with the highest uptake observed in metastatic cutaneous SCC (SUVmax = 6.5), followed by SCC of the oesophagus (SUVmax = 5.7), adenocarcinoma (SUVmax = 4.4), ovarian carcinoma (SUVmax = 2.7) and the least in breast carcinoma (SUVmax = 2.4). The whole-body average effective dose delivered by the tracer was found to be even marginally lower than the dose delivered by [68Ga]Ga-PSMA and [68Ga]Ga-DOTA-TATE.37 However, more clinical trials are needed to assess the potential of [68Ga]Ga-CDI.
Neurodegenerative diseases such as Alzheimer's and Parkinson's disease result from programmed cell death, i.e., apoptosis of neurons. Generally, apoptosis follows extrinsic and intrinsic pathways, which eventually merge into a caspase cascade. Hence, caspase-3 has been identified as a suitable target for apoptosis imaging. Recently, various markers have been engineered to track the activity of caspase-3, which either binds to or is cleaved by active/cleaved caspase-3 (CC3). Ostapchenko et al. created a bifunctional (fluorescence and positron emission) brain-penetrating 68Ga-labeled tracer molecule to accumulate in vivo in cells with increased activity of CC3. The design of the [68Ga]Ga-TC3-OGDOTA tracer includes 68Ga complexation of the DOTA chelator, which is conjugated to the CC3 cleavable DEVD (a peptide sequence) and fluorescent Oregon Green label. The in vitro cell culture study showed the accumulation of [68Ga]Ga-TC3-OGDOTA in 88% of the cortical neuronal cells exhibiting an apoptotic nuclear morphology after treatment with camptothecin (CPT) toxicity compared to about 90–91% accumulation in apoptotic cells exposed to oxygen–glucose deprivation (OGD) and β-amyloid oligomer (AβO). In vivo PET revealed the accumulation of [68Ga]Ga-TC3-OGDOTA in the brain of mouse models of stroke or Alzheimer's disease.38
Recently, a preclinical study was conducted using the 68Ga-labeled C2A domain of synaptotagmin-I ([68Ga]Ga-C2Am) to determine the extent of cell death following treatment. The RCP for [68Ga]Ga-C2Am radiolabeling in a single pot was >95%. In vivo imaging (at 2 h) performed 24 h after treatment revealed the uptake of [68Ga]Ga-C2Am with the tumor to muscle ratio of 2.3 ± 0.4. The tracer showed the ability to evaluate the tumor response following treatment.39
Among the mentioned tracers in Table 1, [68Ga]Ga-CDI is the only tracer that has reached clinical trials. This tracer needs further evaluation regarding the optimization of imaging time post-treatment and detection of different forms of cell death.
Tracer | Structure | Status | Ref. |
---|---|---|---|
[68Ga]Ga-Annexin A5 | Not available | Preclinical phase | Bauwens et al., 201129 |
[68Ga]Ga-NOTA-VCAM-1scvf |
![]() |
Preclinical phase | Zhang et al., 201833 |
[68Ga]Ga-NODAGA-Duramycin | Not available | Preclinical phase | Rix et al., 202035 |
[68Ga]Ga-TC3-OGDOTA |
![]() |
Preclinical phase | Ostapchenko et al., 201938 |
[68Ga]Ga-CDI |
![]() |
Clinical phase | Shon et al., 202236 |
[68Ga]Ga-C2Am |
![]() |
Preclinical phase | Bulat et al., 202339 |
These different tracers exhibit variable affinities towards the different SSTR subtypes, demonstrating different sensitivities toward lesion detection in various pathological conditions. Various comparative studies have been conducted to determine the diagnostic superiority of one tracer over another. One of the comparison studies conducted on 18 gastroenteropancreatic neuroendocrine tumor (GEP NET) patients showed the lesion detection supremacy of [68Ga]Ga-DOTA-NOC over [68Ga]Ga-DOTA-TATE.51 However, a similar study conducted on 20 NET patients presented an equivocal response of both tracers in detecting the number of lesions.52 Thus, researchers are still exploring ideal tracers exhibiting affinity towards all the subtypes of SSTRs.
To target tumors with heterogeneous somatostatin receptor expression, various pasireotide derivatives, [68Ga]Ga-DOTA-PA1,53 [68Ga]Ga-DOTA-SOM230,54 and [68Ga]Ga-DOTA-ST8950,55 have been developed and evaluated preclinically. Similar to octreotide, pasireotide is a somatostatin analogue; however, it targets several SST subtypes, including SST1, SST2, SST3, and SST5. In vitro cellular uptake, in vivo animal biodistribution and microPET imaging of [68Ga]Ga-DOTA-PA1 has shown its potential for targeting multi-receptor (SSTR1, SSTR2, SSTR3, and SSTR5) expressing tumors.53 Conversely, [68Ga]Ga-DOTA-ST8950 has shown specificity towards tumors expressing SSTR2 and SSTR5.55 These tracers possess the capacity to become useful clinical tracers targeting tumors with heterogeneous receptor expression. However, further evaluation is required to clinically translate 68Ga-labeled pasireotide-based radiopharmaceuticals.
Liu et al. synthesized a precursor called benereotide as a potential targeting vector for SSTR positive tumors. It is a large 18-membered lactam and a novel SST analogue exhibiting higher affinities towards subtypes of SSTR and higher stability compared to octreotide. Using a DOTA derivative chelator, benereotide was conjugated, resulting in the highest radiolabeling yield of 60% (pH 3.5, incubated at 100 °C for 20 min). The RCP obtained was about 98%, and the tracer showed excellent stability in different buffers. The highest tracer uptake was noted in the liver and blood with values of 13.87% ± 0.05% ID per g and 4.91% ± 0.39% ID per g at 1.5 h, respectively. MicroPET imaging performed at 4 h resulted in excellent images of the tumor with a high tumor to background ratio.56 However, no further study has been conducted to evaluate its clinical utility.
SSTR imaging helps to identify patients suitable for PRRT, providing more personalized care. A prominent characteristic of SSTR imaging and therapy is the internalization and tumor retention of the agonist tracers. Contrary to this, Ginj et al. introduced a paradigm shift in 2006 by stating that SSTR antagonists may exhibit a superior performance to the present agonists despite their lack of internalization.57 Among the different investigated antagonists, JR11 (Cpa-c(dCys-Aph(Hor)-dAph(Cbm)-Lys-Thr-Cys)-dTyr-NH2)) showed the highest affinity towards the SSTR subtype 2. Fani et al. evaluated the effect of metal chelation on the binding affinity of SST antagonists. Their findings indicated that the DOTA-JR11 precursor had a greater binding affinity (IC50 0.72 ± 0.12 nM) for SSTR2 than the radiolabeled [68Ga]Ga-DOTA-JR11 (IC50 29 ± 2.7 nM). Interestingly, the binding affinity was restored once the chelator was changed to NODAGA from DOTA (IC50 values for NODAGA-JR11 and [68Ga]Ga-NODAGA-JR11 were 4.1 ± 0.2 nM and 1.2 ± 0.2 nM, respectively). Compared with [68Ga]Ga-DOTA-TATE, [68Ga]Ga-DOTA-JR11 (also known as [68Ga]Ga-OPS202 or [68Ga]Ga-IPN01070) demonstrated a notably high tumor uptake (about 1.3-times higher), despite having a lower affinity for SSTR2. Similarly, [68Ga]Ga-NODAGA-JR11 demonstrated higher tumor uptake (nearly 1.7 times) despite its lower affinity for SSTR2 than [68Ga]Ga-DOTA-TATE.58 Thus, this study revealed the encouraging prospective of antagonists for the imaging and therapy of SSTR expressing tumors.
To further unfold the potential of antagonist SST analogues, Fani et al. synthesized another promising SST antagonist peptide, p-Cl-Phe-cyclo(D-Cys-Tyr-D-4-amino-Phe(carbamoyl)-Lys-Thr-Cys)D-Tyr-NH2, also known as LM3,59 which selectively binds to SSTR 2. NODAGA conjugated LM3 complexed with 68Ga achieved more than 95% RCP and a molar activity of 120 MBq per nmol with a labeling yield of >97%. Cell culture studies showed that natGa-NODAGA-LM3 selectively targeted SSTR2, with IC50 values of >1000 nM for human SSTR1, SSTR3, SSTR4, and SSTR5 receptors and 1.3 ± 0.3 for SSTR2. In addition, it was observed that natGa-NODAGA-LM3 at a higher concentration (1000 nM) did not induce receptor internalization in human embryonic kidney (HEK)-SST2 cells. An animal biodistribution study performed on nude mice bearing HEK-sst2 xenografts showed the significant SSTR2 tumor uptake of [68Ga]Ga-NODAGA-LM3 (37.27% ± 5.49% injected dose per gram (% ID per g) at 1 h). Also, [68Ga]Ga-NODAGA-LM3 demonstrated 30% higher tumor uptake than [68Ga]Ga-DOTA-LM3. Zhu et al. presented a case study using [68Ga]Ga-DOTANOC and [68Ga]Ga-NODAGA-LM3 in the case of pancreatic NET and extensive tumor thrombosis in the portal venous system. According to their observation, both tracers revealed similar patterns with high tumor affinity and tumor-to-background ratio.60 Due to its promising results in preclinical studies, Zhang et al. performed a case study with [68Ga]Ga-DOTA-LM3 in a patient with a [68Ga]Ga-DOTATOC negative scan in high-grade liver metastases of a pancreatic neuroendocrine neoplasm.61,62 After performing a diagnostic [68Ga]Ga-DOTA-LM3 scan, intra-arterial [177Lu]Lu-DOTA-LM3 therapy was successfully given to the patient. The patient was in complete remission after receiving three cycles of intra-arterial PRRT.61 Based on these assuring results, a preliminary clinical study was recently performed on patients with well-differentiated NETs. This study was conducted to evaluate the safety, biodistribution, dosimetry (phase I), and diagnostic efficacy (phase II) of [68Ga]Ga-NODAGA-LM3 and [68Ga]Ga-DOTA-LM3 in patients with well-differentiated NETs. PET/CT images revealed the significantly lower uptake of [68Ga]Ga-DOTA-LM3 in comparison to [68Ga]Ga-NODAGA-LM3 in the pituitary, parotids, liver, spleen, pancreas, adrenal, stomach, small intestine, and kidneys. The whole-body effective dose was 0.026 ± 0.003 mSv per MBq for [68Ga]Ga-NODAGA-LM3 and 0.025 ± 0.002 mSv per MBq for [68Ga]Ga-DOTA-LM3. Similar to the study by Fani et al., the radiolabeled antagonists showed long tumor retention despite their minimal internalization.
The structures of all the somatostatin receptor-targeting tracers are listed in Table 2.
Tracer | Target | Nature | Structure | IC50 for SSTR2 | Status |
---|---|---|---|---|---|
[68Ga]Ga-DOTA-NOC | SSTR2, SSTR3 and SSTR5 | Agonist |
![]() |
1.9 ± 0.4 nM50 | Clinical Phase |
[68Ga]Ga-DOTA-TOC | SSTR2 and SSTR5 | Agonist |
![]() |
2.5 ± 0.5 nM49 | Clinical phase |
[68Ga]Ga-DOTA-TATE | SSTR2 | Agonist |
![]() |
0.2 ± 0.04 nM49 | Clinical phase |
[68Ga]Ga-DOTA-PA1 | SSTR1, SSTR2, SSTR3 and SSTR5 | Agonist |
![]() |
17.8 ± 3.2 nM (Kd value)53 | Pre-clinical phase |
[68Ga]Ga-DOTA-ST8950 | SSTR2 and SSTR5 | Agonist |
![]() |
0.32 nM55 | Pre-clinical phase |
[68Ga]Ga-DOTA-Benereotide | SSTR1, SSTR2, SSTR3 and SSTR5 | Agonist |
![]() |
Not reported56 | Pre-clinical phase |
[68Ga]Ga-NODAGA-JR11 | SSTR2 | Antagonist |
![]() |
1.2 ± 0.2 nM63 | Clinical phase |
[68Ga]Ga-NODAGA-LM3 | SSTR2 | Antagonist |
![]() |
1.3 ± 0.3 nM59 | Clinical phase |
Tracer | Structure | Status | Ref. |
---|---|---|---|
[68Ga]Ga-PSMA-11 |
![]() |
Clinical phase | Eder et al., 201279 |
[68Ga]Ga-PSMA-617 |
![]() |
Clinical phase | Benešova et al., 201573 |
[68Ga]Ga-P16-093 |
![]() |
Clinical phase | Zha et al., 201874 |
[68Ga]Ga-NGUL |
![]() |
Clinical phase | Moon et al., 201880 |
[68Ga]Ga-PSMA-I&T |
![]() |
Clinical phase | Weineisen et al., 201577 |
[68Ga]Ga-NO3A-DM1-Lys-Urea-Glu |
![]() |
Pre-clinical phase | Kumar et al., 201678 |
PSMA is a transmembrane glycoprotein having three subunits, an internal unit of 19 amino acids, a transmembrane unit of 24 amino acids and an external unit of 707 amino acids.65 Being highly expressed on the surface of prostate cancer cells, PSMA has been considered the best-targeting antigen for imaging.66
Afshar et al. used 68Ga-labeled PSMA ligands in humans to perform PET imaging for the diagnosis of prostate cancer. This study showed that [68Ga]Ga-HBED-CC-PSMA-11 can detect prostate cancer relapses by targeting the extracellular domain of PSMA.67 Since then, many comparative studies have been conducted to evaluate its superiority over existing tracers for prostate cancer imaging.68–72 [68Ga]Ga-HBED-CC-PSMA-11 demonstrated a high detection rate at very low PSA levels in almost all the studies. As a result, [68Ga]Ga-HBED-CC-PSMA-11, which received FDA approval in 2020, has emerged as the most popular tracer for prostate cancer imaging. HBED-CC exhibits several ideal characteristics as a chelator for 68Ga complexation; however, it does not form a stable complex with therapeutic isotopes such as 177Lu, 225Ac and 213Bi, which are considered therapeutic pairs of 68Ga. This led to the development of PSMA-617 with the same pharmacophore (Lys-u-Glu), in conjugation with a DOTA chelator for PSMA-targeted therapies.73 The research on more effective PSMA-targeting agents is still very active, and recently many novel tracers have surfaced with better pharmacokinetics and greater affinity towards PSMA-expressing prostate cancer.
Zha et al. synthesized a novel PSMA-targeting tracer, i.e., [68Ga]Ga-Glu-NH-CO-NH-Lys(Ahx)-linker-HBED-CC conjugated with O-(carboxymethyl)-L-tyrosine, also known as [68Ga]Ga-P16-093.74 Clinical studies have been conducted based on the promising preclinical results of [68Ga]Ga-P16-093. Recently, a head-to-head comparison study of [68Ga]Ga-P16-093 and [68Ga]Ga-PSMA-617 was conducted to evaluate the superiority of diagnosing recurrent prostate cancer. This study revealed the significant accumulation of [68Ga]Ga-P16-093 in tumoral sites in comparison to [68Ga]Ga-PSMA-617 (SUVmax of 7.88 ± 5.26 vs. 6.01 ± 3.88, respectively). The authors also observed the high detection capability, less blood pool retention and lower bladder uptake of [68Ga]Ga-P16-093.75 This promising tracer is currently under phase 1 clinical trials.
[68Ga]Ga-NOTA Glu-Urea-Lys (NGUL), another intriguing PSMA-targeting tracer, has been developed and evaluated in patients with metastatic prostate cancer. This tracer could detect all the primary as well as metastatic lesions identified with [68Ga]Ga-PSMA-11 but with far less accumulation in normal organs and a quicker rate of urine clearance. In contrast to [68Ga]Ga-PSMA-11, a relatively low tumor to background ratio was observed.76
In 2015, Weineisen et al. introduced another PSMA inhibitor, PSMA I & T (DOTAGA-(I-y)fk(Sub-KuE)), where I & T refer to imaging and therapy, respectively. It was synthesized, radiolabeled with 68Ga and evaluated preclinically. Its promising preclinical results led to its further use in humans as a proof of concept. The RCY and RCP obtained after the fully automated synthesis of [68Ga]Ga-PSMA I & T were 67% ± 10% (non-decay corrected) and 98% ± 2%, respectively. It was found to be safe for human administration given that no adverse side effects showed up in the first patient. PET/CT images of a metastatic CRPC patient revealed significant tracer uptake in multiple abdominal lymph nodes, liver metastasis and bone lesions with an average lesion to background SUVmax ratio of 17.6, 20.7 and 35.2, respectively. The physiological tracer uptake was found in the salivary glands, kidneys, liver, spleen and proximal segments of the small intestine. PSMA I & T has emerged as a potential theranostic PSMA targeting inhibitor, which can be radiolabeled with 68Ga as well as 177Lu.77
Exploring the theranostic application of PSMA targeting, a novel small molecule drug conjugate was radiolabeled with 68Ga, [68Ga]Ga-NO3A-DM1-Lys-Urea-Glu, for simultaneous imaging and therapy. It is composed of 3 units, as follows: (a) a PSMA binding motif (Lys-u-Glu), (b) a chemotherapy drug, maytansine (DM1), and (c) a chelator (NO3A) for complexation with 68Ga. PET/CT images of small animals with a PSMA-positive PC3-PIP tumor showed the significant uptake (4.30% ± 0.20% ID per g at 1 h) of [68Ga]Ga-NO3A-DM1-Lys-Urea-Glu.78 The PSMA-targeted chemo theranostic concept has shown promising results; however, further optimization of the tracer is ongoing.
The newly developed PSMA-targeting tracers have emerged as potential competitors to the existing [68Ga]Ga-PSMA-11. Among them, [68Ga]Ga-P16-093 had shown superior tumor detection potential in comparison to the [68Ga]Ga-PSMA-11, but [68Ga]Ga-P16-093 has yet to be examined in different clinical scenarios of prostate cancer in a head-to-head comparison.
[68Ga]Ga-FAPI derivatives | Structure | Radiolabeling conditions | IC50 | Status |
---|---|---|---|---|
[68Ga]Ga-FAPI-2 |
![]() |
Temperature: 95 °C | 21 nM86 | Clinical phase |
Incubation: 10 min | ||||
[68Ga]Ga-FAPI-4 |
![]() |
Temperature: 95 °C | 6.5 nM86 | Clinical phase |
Incubation: 10 min | ||||
[68Ga]Ga-FAPI-21 |
![]() |
Temperature: 95 °C | 6.7 nM84 | Pre-clinical phase |
Incubation: 10 min | ||||
[68Ga]Ga-FAPI-46 |
![]() |
Temperature: 95 °C | 2.06 ± 1.84 nM92 | Clinical phase |
Incubation: 10 min | ||||
[68Ga]Ga-FAPI-55 |
![]() |
Temperature: 95 °C | 5.4 nM84 | Pre-clinical phase |
Incubation: 10 min | ||||
[68Ga]Ga-FAPI-74 |
![]() |
Room temperature | Not reported99,100 | Pre-clinical phase |
Incubation: 15 min | ||||
[68Ga]Ga-FAP-2286 |
![]() |
Temperature: 95 °C | 3.4 ± 1.1 nM95 | Clinical phase |
pH: 4–4.5 | ||||
Incubation: 15 min | ||||
[68Ga]Ga-2P(FAPI)2 |
![]() |
Temperature: 100 °C | 3.68 ± 1.82 nM92 | Clinical phase |
pH: 3.3–3.6 | ||||
Incubation: 15 min | ||||
[68Ga]Ga-SA.FAPI |
![]() |
Temperature: 95 °C | 1.4 ± 0.2 nM101 | Clinical phase |
Incubation: 15 min | ||||
[68Ga]Ga-MHLL1 |
![]() |
Temperature: 100 °C | 212 nM98 | Pre-clinical phase |
Incubation: 5–7 min |
Conversely, FAPI-02 showed much better pharmacokinetics such as slow clearance, rapid internalization, and high tumor uptake in both tumor xenografts and epithelial carcinoma patients.85 Hence, FAPI-02 was investigated further, and based on it many compounds were synthesized to optimize the tracer uptake and retention in tumor tissues.86 Among the various FAP-targeting compounds, FAPI-02, FAPI-04, FAPI-21, FAPI-46, FAPI-55, FAPI-74, SA.FAPI and FAP-2286 have been radiolabeled with 68Ga and further investigated in clinical applications.84,87 Among the modified derivatives of FAPI-02, FAPI-04 showed the most promising results due to its prolonged tumor retention, allowing improved signal-noise ratios.84,88,89 Besides FAPI-04, FAPI-46 and SA.FAPI have emerged as potential clinical tracers with even further improved tumor retention.90,91 However, to realize their therapeutic application, further optimizations are in progress.
Recently, Zhao et al. synthesized a FAPI dimer radiolabeled with 68Ga and performed a preclinical evaluation together with a pilot clinical study. The study revealed the better pharmacokinetics of [68Ga]Ga-DOTA-2P(FAPI)2 than the monomer [68Ga]Ga-FAPI-46. This study showed the potential of the tracer for diagnostic imaging as well as targeted therapy of FAP-expressing tumors.92 A clinical study has been registered to assess the clinical utility of [68Ga]Ga-DOTA-2P(FAPI)2 in various cancer conditions (ClinicalTrials.gov NCT number: NCT04941872).
Further extending the applications of FAP inhibitors, Rosenkrans et al. evaluated [68Ga]Ga-FAPI-46 preclinically in pulmonary fibrosis-induced mice. Significant tracer uptake was observed in the fibrosis-induced mice at 7 days (0.33% ± 0.09% IA per cc) and 14 days (1.01% ± 0.12% IA per cc). The biodistribution results were consistent with the histology.93 Recently, a clinical study was carried out by Luo et al. in 20 patients of rheumatoid arthritis. In total, 244 affected joints were detected using [68Ga]Ga-FAPI. Among the 244 detected joints, 20 affected joints were not even shown in the [18F]FDG PET/CT images (ClinicalTrials.gov NCT number: NCT04514614).94 Thus, this study revealed the great diagnostic potential of [68Ga]Ga-FAPI in detecting rheumatoid arthritis.
The pharmacokinetic profile of the majority of FAPIs shows their fast tumor clearance, which results in compromised therapeutic efficacy. Consequently, a novel cyclic peptide-based FAP-targeting tracer, FAP-2286, has been developed and radiolabeled with both 68Ga and 177Lu to address the low tumor retention of FAP inhibitor-based compounds. In vitro analysis and in vivo animal imaging of FAP-2286 have shown excellent results, favouring its clinical translation. The PET/CT images of a pilot human study performed in 11 patients revealed the theranostic potential of FAP-2286.95 A phase 1/2 study in advanced solid tumors is under clinical trials (ClinicalTrials.gov NCT number: NCT04939610). Recently, an updated extensive review article was published, covering the existing literature on [68Ga]Ga-FAPI and its future prospects.96
Also, non-oncological applications of FAP imaging are also in demand, especially in cardiology. It has been identified that FAP is highly expressed in the early week after the onset of myocardial infarction (MI).97 This makes FAP imaging of activated myofibroblasts in myocardial injury or MI possible. Therefore, a study of fibroblast activity after MI was performed using [68Ga]Ga-FAPI-04,97 and recently with the novel tracer [68Ga]Ga-MHLL1.98 Langer et al. synthesized a novel FAP-specific compound, i.e., MHLL1 radiolabeled with 68Ga, and performed in vivo PET imaging in a mouse model together with an in vitro cellular study. The RCY and RCP obtained in the radiolabeled [68Ga]Ga-MHLL1 were 74.4% ± 2.86%, and 71.2% ± 12.9%. The radiolabeled product was stable in phosphate buffered saline and human serum by >80% at 180 min. The in vitro analysis performed in transfected HT1080 cells expressing human FAP and murine FAP showed the specificity of [68Ga]Ga-MHLL1 with an IC50 of 212 nM and 142 nM, respectively. In vivo serial PET imaging on day-2 and day-7 revealed tracer accumulation in the infarcted region of the heart. Ex vivo autoradiography and histology of the excised heart confirmed the accumulation of the tracer in the infarcted region at day-7 after MI. According to the authors, further evaluation and optimization of the tracer are needed.98
To date, the US-FDA has not approved any FAPI compounds. However, recently, a company has submitted an IND application to the US-FDA for a phase II clinical trial of pancreatic adenocarcinoma patients with [68Ga]Ga-FAPI-46 (ClinicalTrials.gov NCT number: NCT05262855). Besides [68Ga]Ga-FAPI-04, [68Ga]Ga-FAPI-46 is presently the most clinically tested FAPI derivative.
In 2013, Beylergil et al. developed a fragment of trastuzumab, i.e., F(ab)2, and labeled it with 68Ga to image and quantitatively analyse HER2 expression in breast cancer patients. The authors observed the favourable pharmacokinetics and radiation dosimetry of [68Ga]Ga-DOTA-F(ab)2-trastuzumab. However, in many of the tumoral sites, minimal or no uptake of the tracer was observed. Thus, the authors inferred that the tracer is safe for human administration but requires evaluation in a better-defined group of patients, immunoreactivity and optimization in the mass of the antibody.
Ueda et al. developed a 68Ga-labeled anti-HER2 single-chain Fv (scFv) fragment to evaluate the HER2 expression in a tumor model. Anti-HER2-scFv was conjugated to the p-isothiocyanatobenzyl derivative of deferoxamine (Df-p-SCN) chelator instead of DOTA because of its ability to form a complex at room temperature (important for heat-sensitive antibodies/antibody fragments). The RCY and RCP obtained were 86% and >98%, respectively. In vitro analysis performed using 68Ga revealed the binding affinity of [68Ga]Ga-Df-anti-HER2 scFv to be 9.94 ± 1.36 nM. The biodistribution study and PET imaging of [68Ga]Ga-Df-anti-HER2 scFv showed its significant accumulation in HER2-positive xenografts.
The third generation of monoclonal antibodies, i.e., having two heavy chains with only a single variable domain (VHH, ∼15 kDa), are also known as nanobodies. A HER2 nanobody was first used clinically in 2014 (Phase I) to detect primary and metastatic HER2-expressing breast tumors.110 Based on its promising results, a phase I/phase II trial on breast cancer patients involving brain metastases is ongoing using [68Ga]Ga-NOTA-Anti-HER2 VHH1.111
The first clinically investigated affibody molecule labeled with 68Ga was ABY-002. Breast cancer patients tolerated this molecule well and its pharmacokinetics showed rapid excretion from normal tissues.105 A preclinical study of 68Ga-labeled ABY-002 affibody was carried out by Tolmachev et al., demonstrating its specific binding to HER-2 expressing cells. In vivo imaging showed the specific targeting of SKOV-3 (a human epithelial ovarian cancer cell line derived from the ascitic fluid of a patient with ovarian adenocarcinoma) xenografts with high contrast. The tracer showed reasonable clearance from the blood, lungs, gastrointestinal, and muscle at 2 h post injection.112 However, significant tracer accumulation was noted in the liver and kidneys. This led to the development of a re-engineered second-generation derivative, i.e., ABY-025. Initially, ABY-025 was radiolabeled with a SPECT radioisotope (111In), but due to its limited ability to resolve small lesions and its low SPECT/CT resolution, 68Ga was explored for PET imaging. A clinical study was performed to study the HER2 expression in 16 patients of known metastatic breast cancer by administering the 68Ga-labeled ABY-025 affibody. This study revealed tracer uptake in all HER-2 positive lesions at 1 h, which further increased at 4 h. In contrast, the HER-2-negative lesions showed consistent minimal tracer uptake from 1 h to 4 h. This study also demonstrated the PET SUV correlation with biopsy HER-2 scores (r = 0.91, p < 0.001). In addition, the tracer helped in converting the disease status from HER-2 negative to HER-2 positive in 3 patients, and further the course of treatment changed.113 Another HER-2-targeting affibody, [68Ga]Ga-NOTA-MAL-Cys-MZHER2:342, was clinically tested in 2 patients. PET/CT images revealed the detection of HER-2-positive primary tumors with the SUVmax of 2.16 ± 0.27 and HER-2-negative primary tumor with the SUVmax of 0.32 ± 0.05.114 Miao et al. conducted a clinical study in 24 histopathologically confirmed breast cancer patients with [68Ga]Ga-NOTA-MAL-MZHER2, which demonstrated the sensitivity and specificity of 91.7% and 84.6%, respectively, in differentiating HER-2-expressing breast cancer.115
The need for size optimization of HER2-specific targeting antibodies has led to the development of engineered scaffold proteins. The albumin binding domain (ABD) of streptococcal protein G has been used as a scaffold for developing HER2 targeting tracers. Various ABD-derived targeting proteins, commonly known as ADAPTs, i.e., ABD-derived affinity proteins (∼5 kDa), have been developed and utilized in molecular imaging. Among the ADAPTs, ADAPT6 was conjugated with a maleimido-derivative of DOTA, DOTA-C-(HE)3-ADAPT6, and radiolabeled with 68Ga and used as a HER2-specific targeting novel tracer for imaging. The radiolabeling yield obtained was 98.3% ± 1.7%, with more than 95% RCP. In vivo PET imaging of a human tumor xenograft in mice using [68Ga]Ga-DOTA-C-(HE)3-ADAPT6 showed significant tracer uptake in the tumor site with a high tumor to normal tissue ratio. This study unveiled the potential of small-sized engineered scaffold proteins for non-invasive PET imaging.116
Another member of the EGFR family, HER3, playing a potential role in targeted therapy resistance, has also been explored as a suitable target. A novel peptide, HER3P1, was synthesized and radiolabeled with 68Ga to quantify the heterogeneous temporal and spatial expression of HER3. Larimer et al. synthesized this novel peptide by performing phage display selection and performed its in vitro characterization, developed a tumor model and performed in vivo PET imaging.117 The competitive cell binding assay revealed the high specificity of [68Ga]Ga-NOTA-HER3P1 towards moderately HER3-expressing MDA-MB-453 cells compared to low HER3-expressing HCC-1954 cells. The RCP obtained was greater than 95% with a specific activity of 296 ± 25.9 MBq per mg of the labeled peptide. The PET imaging of 22RV1 (human prostate castration-resistant cancer cell line) tumor-bearing mice showed its uptake in the tumor with a tumor-to background ratio in the range of 1.59–3.32, together with its uptake in the kidneys and bladder, showing the standard excretion route. The quantification of HER3 expression performed by PET imaging of [68Ga]Ga-NOTA-HER3P1 was highly correlated with the Western blotting quantification. The authors stated that this novel peptide has potential for visualising and quantifying HER3 expression, but increasing its tumor uptake still requires further optimization. [68Ga]Ga-NODAGA-(HE)3-ZHER3 (an affibody tracer) and [68Ga]Ga-DFO-seribantumab-F(ab′)2 have also been recently evaluated for targeting HER3 receptors.118,119
Affibodies targeting HER-2 expression have shown superiority over full antibody and antibody fragments because of their reduced size and better pharmacokinetics. As a result, there are numerous active clinical trials involving affibodies.
Tracer | Structure | Status | Ref. |
---|---|---|---|
[68Ga]Ga-RM1 |
![]() |
Clinical phase | Mansi et al., 2009125 |
[68Ga]Ga-RM2 |
![]() |
Clinical phase | Mansi et al., 2010135 |
[68Ga]Ga-AMBA |
![]() |
Pre-clinical phase | Mansi et al., 2009125 |
[68Ga]Ga-RM26 |
![]() |
Clinical phase | Varasteh et al., 2013136 |
[68Ga]Ga-SB3 |
![]() |
Clinical phase | Maina et al., 2015130 |
[68Ga]Ga-NeoBOMB1 |
![]() |
Clinical phase | Nock et al., 2016132 |
[68Ga]Ga-MJ9 |
![]() |
Clinical phase | Gourni et al., 2014133 |
Several GRPR antagonist-based radiolabeled analogues have been developed, characterized, and studied preclinically as well as clinically for different clinical conditions. The first GRPR antagonist studied was [68Ga]Ga-RM1 (DOTA-Gly-4-aminobenzoyl-JMV594), which showed preclinical dominance over the GRPR agonist [68Ga]Ga-AMBA (DOTA-Gly-4-aminobenzoyl-BBN(7–14)).125 This led to the further development of antagonistic peptides, and in 2010, Mansi et al. developed the DOTA-conjugated bombesin antagonist analogue RM2 (DOTA-4-amino-1-carboxymethyl-piperidine-D-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2), also known as BAY86-7548, targeting GRPR-positive tumors. It emerged as a promising agent, which was evaluated clinically in healthy volunteers and GRPR-expressing prostate cancer patients. The tracer was found to be safe for human administration with an estimated whole body effective dose of 0.051 mSv per MBq, which is comparable but slightly higher than that of the routinely used 68Ga labeled radiopharmaceuticals. In another study conducted on 14 prostate cancer patients, this tracer revealed the sensitivity, specificity and accuracy of 88%, 81% and 83%, respectively, in detecting primary prostate cancer.126,127
In the case of another GRPR antagonist, JMV594 derivative RM26 (NOTA-PEG3-JMV594), several studies have demonstrated the specific uptake of [68Ga]Ga-RM26 in GRPR-positive breast and prostate lesions, together with metastatic sites.128,129 On comparing the PET/CT images of [68Ga]Ga-RM26 with [68Ga]Ga-BBN in 22 prostate cancer patients, Zhang et al. observed the superiority of [68Ga]Ga-RM26 in detecting primary lesions as well as metastatic lesions over [68Ga]Ga-BBN.129 Recently, a phase 1 clinical trial of [68Ga]Ga-NOTA-PEG2-RM26 has been registered to evaluate the diagnostic efficacy in GRPR-expressing prostate cancer patients (ClinicalTrials.gov NCT number: NCT06147362).
In 2015, a DOTA-conjugated GRPR antagonist, SB3 (DOTA-p-aminomethylaniline-diglycolic acid-DPhe-Gln-Trp-Ala-Val-Gly-His-Leu-NHEt), was radiolabeled with 68Ga and clinically investigated for the first time in patients with advanced prostate cancer and breast cancer.130 A phase 1 clinical study involving 10 prostate cancer patients was also carried out, which revealed that the sensitivity of this tracer in detecting GRPR-positive lesions is 88%.131 Further extending the theranostic application of SB3, it was also radiolabeled with 111In (for SPECT) and 177Lu (for therapy). However, the respective radioligands showed less affinity towards GRPR as well as low biological stability. As a consequence, the clinical applications of SB3 were limited. Later, the same group introduced another novel GRPR antagonist, i.e., NeoBOMB1, by replacing the C-terminal -His12-Leu13-NHEt of SB3 with -His12-NH-CH[CH2-CH(CH3)2]2. This modification was required to increase the theranostic potential of the GRPR antagonist. Nock et al. performed in vitro assays on human androgen-independent prostate adenocarcinoma PC-3 cells expressing GRPR receptors. Their study showed the high affinity of NeoBOMB1 and natGa-bound NeoBOMB1 towards GRPR receptors expressed on the cell membrane of PC3 cells. The authors also mentioned the low internalization of the tracer, exhibiting its antagonistic nature. An in vivo study was performed in PC3 xenograft–bearing mice, which showed the stability of [68Ga]Ga-NeoBOMB1 in mice (>90% at 30 min post injection), and its biodistribution was observed in the PC3 xenograft and the pancreas, which is a GRPR-rich organ. Further translating into humans, four patients with prostate adenocarcinoma were administered [68Ga]Ga-NeoBOMB1 and PET/CT scans were performed. The tracer was well tolerated by the patients and no side effects were observed. The scans showed high uptake of [68Ga]Ga-NeoBOMB1 in primary and metastatic lesions, and also detected osseous (bone) and liver micrometastasis.132
Another novel GRPR antagonist, MJ9, Pip-D-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2 (Pip, 4-amino-1-carboxymethyl-piperidine), has been developed and investigated. NODAGA- and NOTA-conjugated MJ9 were radiolabeled with 68Ga, and greater than 98% labeling yield was obtained with a specific activity of 130 GBq per mol and 60 GBq per mol, respectively. The IC50 value obtained for natGa-NODAGA-MJ9 and natGa-NOTA-MJ9 was 2.1 ± 0.3 nmol L−1 and 0.5 ± 0.1 nmol L−1, respectively. The biodistribution study revealed the fast clearance of both 68Ga-labeled tracers with high GRPR-positive tumor uptake at 2 h post injection (approximately 20% IA per g). Also, the GRPR-expressing organ, the pancreas, showed a high tracer uptake of nearly 40% IA per g at 2 h post injection. Small animal PET imaging showed the uptake of both 68Ga-labeled tracers in the induced tumor at 1 h post injection, together with their significant uptake in the pancreas and intestine.133 To assess the radiation dosimetry, which is particularly important for these compounds due to their non-target organ uptake, Gnesin et al. carried out a first-in-human study utilizing [68Ga]Ga-NODAGA-MJ9 in 5 prostate adenocarcinoma patients. The pancreas, being an organ expressing the highest GRPR, received the maximum absorbed dose of 260 μGy per MBq, followed by the urinary bladder, small intestine, and kidneys (69.8, 38.8 and 34.8 μGy per MBq, respectively). The effective dose delivered to the patients assuming a 30 min voiding cycle was 17 μSv per MBq, which was found to be in a similar range to other 68Ga-labeled analogues.134
Generally, GRPR-targeting antagonist tracers have been used in small clinical investigations. Thus, the potential of these 68Ga-labeled antagonist tracers needs to be evaluated in a large population study.
Alshoukr et al. synthesized two novel DOTA-conjugated neurotensin analogues and radiolabeled them with 68Ga, [68Ga]Ga-DOTA-NT-20.3 and [68Ga]Ga-DOTA-LB119, to study NTR positive tumors. The in vitro assay on an NTR1-positive HT-29 cell line (human colon adenocarcinoma cells) revealed the IC50 of 14 ± 2 nM and 7.5 ± 0.7 nM for [68Ga]Ga-DOTA-NT-20.3 and [68Ga]Ga-DOTA-LB119, respectively. Based on the biodistribution and PET imaging of nude mice grafted with HT29 cells, [68Ga]Ga-DOTA-NT-20.3 showed superiority over [68Ga]Ga-DOTA-LB119 given that it also detected small tumors.145 This preliminary study demonstrated the potential of [68Ga]Ga-DOTA-NT-20.3 as an imaging agent for NTR-positive tumors. Based on the promising results of [68Ga]Ga-DOTA-NT-20.3, the same group conducted another preclinical study to discriminate human pancreatic ductal adenocarcinoma (PDAC) from pancreatitis. Preclinical PET imaging, biodistribution, blocking and histology examination in an animal model showed the ability of this tracer to discriminate human PDAC from pancreatitis.146 Maschauer et al. developed different neurotensin peptide derivatives based on the sequence NLys-Lys-Pro-Tyr-Tle-Leu, radiolabeled with 18F and 68Ga for PET imaging. In vitro and in vivo results demonstrated the receptor affinity for NTR1 (19–110 nM) and their high tumor uptake in HT29-tumor bearing mice, respectively. Among the different 68Ga-labeled peptides, [68Ga]Ga-8 (NT derivative with a metabolically stable peptide sequence of NLys8-Lys9-Pro10-Tyr11-Tle12-Leu13) showed promising results for further evaluation.147 Recently, another preclinical study was carried out by Wu et al. to explore [68Ga]Ga-DOTA-NT-20.3 in PSMA-negative prostate cancer. It was successfully synthesized with an RCY and RCP of 88.07% ± 1.26% and 99%, respectively. The in vitro analysis demonstrated the affinity of [68Ga]Ga-DOTA-NT-20.3 towards NTR1 (IC50 7.59 ± 0.41 nM). The in vivo biodistribution and imaging of [68Ga]Ga-DOTA-NT-20.3 revealed its significant tumor uptake (4.95% ± 0.67% ID per g at 1 h) and high contrast images.148
Bodin et al. devised, synthesized, and preclinically assessed [68Ga]Ga-JMV 7488, a new neurotensin analogue that targets NTR2. The results showed the utility of [68Ga]Ga-JMV 7488 in targeting NTR2-expressing cancer conditions, particularly in breast cancer with low FDG avidity, prostate cancer with negative PSMA expression and colorectal cancer.149
All the tracers that target NTR expression are in preclinical stage and revealed only mild to moderate uptake. Therefore, the search for suitable NTR targeting tracers for clinical application is still ongoing.
Tracer | Structure | Status | Ref. |
---|---|---|---|
[68Ga]Ga-CPCR4-2 |
![]() |
Clinical phase | Demmer164 and Gourni et al., 2011165 |
[68Ga]Ga-DOTA-4-FBn-TN14003 |
![]() |
Pre-clinical phase | Hennrich et al., 2011166 |
[68Ga]Ga-NOTA-CCIC16 |
![]() |
Pre-clinical phase | George et al., 2013167 |
[68Ga]Ga-NOTA-NFB |
![]() |
Clinical phase | Jacobson et al., 2011168 |
[68Ga]Ga-Pentixafor |
![]() |
Clinical phase | Vag et al., 2016172 |
Wang et al. translated a 68Ga-labeled CXCR4 antagonist, i.e., [68Ga]Ga-NOTA-NFB, based on its promising pre-clinical results.168 This tracer showed good pharmacokinetics with rapid blood clearance and high target to non-target ratio. The glioma tissues showed significant tracer accumulation (SUVmax of 4.11 ± 2.90), which was consistent with the overexpression seen in the histopathological labeling of the excised tumor tissues. Zhang et al. tested [68Ga]Ga-NOTA-NFB in breast cancer patients expressing chemokine receptors. This tracer picked all the primary lesions (SUVmax of 3.78 ± 2.03) and 11 positive lymph nodes (SUVmax of 2.85 ± 1.86) out of 14 positive on the [18F]FDG scan. Also, it demonstrated potential for both screening for CXCR4 treatment and imaging patients with breast cancer.169 However, additional clinical testing is required to clinically establish the [68Ga]Ga-NOTA-NFB tracer.170
Following the successful preclinical investigations and optimization conducted by Wester's group, [68Ga]Ga-CPCR4.2, also referred to as [68Ga]Ga-pentixafor, showed excellent pharmacokinetics in a preliminary clinical study in patients with hematologic malignancies. This tracer demonstrated rapid clearance and low background uptake, together with substantial accumulation in the tumors.164,165,171 Further exploring the clinical applications of [68Ga]Ga-pentixafor, Vag and co-workers performed a clinical study involving 21 patients diagnosed with various solid tumor types, namely pancreatic cancer, laryngeal cancer, non–small cell lung cancer (NSCLC), prostate cancer, melanoma, breast cancer, hepatocellular carcinoma, glioblastoma, sarcoma, and cancer of unknown primary. The maximum accumulation of [68Ga]Ga-pentixafor yielding a high tumor-to-background contrast was noted in patients with NSCLC and cancer of unknown primary (SUVmax of 10.9 and 13.8, respectively).172
A pilot study involving 15 patients of suspected glioblastoma or recurrent glioblastoma was conducted to assess the sensitivity of CXCR4-targeting [68Ga]Ga-pentixafor. Among 15 patients, 13 demonstrated [68Ga]Ga-pentixafor uptake. Semiquantitative analysis in comparison to [18F]FET showed the lower SUVmax and SUVmean of [68Ga]Ga-pentixafor (5.3 ± 2.3 and 3.9 ± 2.0; 4.4 ± 2.0 and 3.0 ± 1.5, respectively) and higher tumor to background ratio. Thus, it has emerged as a potential tracer for identifying suitable patients for targeted therapy.173 Further, several insightful clinical studies have been performed to explore the clinical utilities of [68Ga]Ga-pentixafor in solid and advanced hematological cancers.174–178 Due to its demonstrated diagnostic utility, [68Ga]Ga-pentixafor has been utilized to identify patients suitable for 177Lu-Pentixather therapy. To facilitate easy clinical translation, the radiochemical synthesis of [68Ga]Ga-pentixafor has also been developed in parallel and optimised with automated synthesis modules.179,180
Among the above-mentioned CXCR4-targeting tracers, only [68Ga]Ga-NOTA-NFB and [68Ga]Ga-pentixafor have been tested clinically tested. [68Ga]Ga-NOTA-NFB has been clinically tested in patients with glioma and breast cancer, whereas [68Ga]Ga-pentixafor has been tested in various solid and hematological cancer conditions.
Different derivatives of 68Ga-labeled exendin-4, i.e., [68Ga]Ga-Ex4NOD12, [68Ga]Ga-Ex4NOD27 and [68Ga]Ga-Ex4NOD40 (Table 7), were also developed and investigated preclinically to assess their effect on binding and pharmacokinetics of the peptide. Compared to unmodified exendin-4 (IC50: 4.1 nM), all the modified derivatives showed reduced binding affinities (IC50 values for natGa-Ex4NOD12, natGa-Ex4NOD27, and natGa-Ex4NOD40 were 29 nM, 53 nM, and 54 nM, respectively) for CHL-GLP-1R-positive cells. The in vivo biodistribution of [68Ga]Ga-Ex4NOD12 and [68Ga]Ga-Ex4NOD40 showed similar pharmacokinetics, whereas [68Ga]Ga-Ex4NOD27 demonstrated slightly inferior results.188
Tracer | Structure | Status | Ref. |
---|---|---|---|
[68Ga]Ga-DO3A-VS-Cys40-Exendin-4 |
![]() |
Pre-clinical phase | Velikyan et al., 2017198 |
[68Ga]Ga-Ex4NOD12 |
![]() |
Pre-clinical phase | Jodal et al., 2014188 |
[68Ga]Ga-Ex4NOD27 |
![]() |
Pre-clinical phase | Jodal et al., 2014188 |
[68Ga]Ga-Ex4NOD40 |
![]() |
Pre-clinical phase | Jodal et al., 2014188 |
[68Ga]Ga-DO3A-Tuna-2 |
![]() |
Clinical phase | Wagner et al., 2020197 |
Based on further promising preclinical studies,185,186,189,19068Ga labeled exendin-4 was translated into human studies involving patients with GLP-1R expressing insulinomas.191–195 However, GLP-1R imaging has a major drawback, its expression in malignant insulinomas is much less compared to benign insulinomas,196 making GLP-1R imaging less sensitive.
Another novel tracer, i.e., [68Ga]Ga-DO3A-Tuna-2, has been explored preclinically to target glucagon receptors/GLP1R, and owing to the promising results obtained in in vitro and in vivo examinations, it has been translated to human clinical trials.197
The treatment management of type 2 diabetes (T2D) involves targeting glucagon receptors/GLP1R. Based on this concept, [68Ga]Ga-DO3A-Tuna-2 was utilised for the monitoring of T2D disease status by targeting glucagon receptors/GLP1R. A recent study demonstrated the GMP-compliant production of [68Ga]Ga-DO3A-Tuna-2,198 and the tracer was applied to quantify glucagon receptors/GLP-1R in humans. The reproducible GMP-compliant production of [68Ga]Ga-DO3A-Tuna-2 revealed the non-decay corrected RCY of 45.2% ± 2.5% and RCP of 98.9% ± 0.6%. High-quality PET/CT images were obtained by administering the tracer activity of 0.47 ± 0.04 MBq per kg body weight. In PET/CT images, the biodistribution of the tracer showed specifically high uptake in the liver due to the high expression of glucagon receptors on hepatocytes.199
Another clinical study has been performed to quantitatively assess the dual receptor targeting (GLP1R and Glucagon receptors) of SAR425899 in T2D patients using [68Ga]Ga-DO3A-Tuna-2 and [68Ga]Ga-DO3A-Exendin4.200 This study is part of an ongoing clinical trial (ClinicalTrials.gov NCT number: NCT03350191). Many more clinical studies are anticipated considering the potential of [68Ga]Ga-DO3A-Tuna-2.
At present, the most clinically explored tracer for GLP-1R imaging is [68Ga]Ga-NODAGA-exendin-4.
Wei et al. investigated [68Ga]Ga-CHX-A′′-Re(Arg11)CCMSH, a novel α-MSH peptide analogue, in B16/F1 melanoma-bearing mice. The radiolabeling efficiency obtained for [68Ga]Ga-CHX-A′′-Re(Arg11)CCMSH was >95% with the specific activity of 1.85 GBq per μg. The biodistribution study demonstrated rapid tumor uptake with 2.68% ± 0.69% ID per g at 2 h post injection. PET imaging revealed the specificity of the tracer by showing melanoma tumor uptake in the shoulder of the mice (non-blocked) and no uptake in the tumor site of the mice given 20 μg of non-radiolabeled NDP peptide (blocked).201
In a recent clinical study, [68Ga]Ga-VMT02 was used to target MC1R in patients with stage IV melanoma. In the 3 h PET images, the tracer showed the best tumor to background ratio. [68Ga]Ga-VMT02 has demonstrated the potential to identify individuals who may respond well to [212Pb]Pb-VMT01 alpha-emission therapy.202
Two 68Ga-labeled glucosamine derivatives, i.e., DOTA-RMX-GC-BNCO and DOTA-RMX-GC-MFCO, targeting the glucose transporter have also been explored for imaging metastatic melanoma. Both conjugates were radiolabeled and found to be stable in FBS and PBS up to 4 h. PET imaging showed the favourable pharmacokinetics of [68Ga]Ga-DOTA-RMX-GC-BNCO of about >5% ID per g of tumor tissue, rapid renal clearance and no retention in other organs at 60 min post injection.203
Gai et al. synthesized 1,4,7-triazacyclononane (TACN)-based bifunctional chelators with phosphonic acid arms, i.e., p-SCN-PhPr-NE2A1P and p-SCN-PhPr-NE2P1A, and conjugated them with a peptidomimetic ligand, LLP2A-PEG4. Among them, [68Ga]Ga-NE2P1A-PEG4-LLP2A, a very late antigen-4 (VLA-4)-targeting ligand, also known as α4β1, was radiolabeled and evaluated in mice bearing B16F10 tumor xenografts. The radiolabeling yield was quantitative (30% ± 4%), and less than 5% dissociation was observed up to 2 h in the serum stability test. The biodistribution study and PET imaging revealed high tumor uptake with specificity towards VLA-4 receptors overexpressed in melanoma.204
Another MC1R targeting tracer, [68Ga]Ga-DOTA-GGNle-CycMSHhex, has been preclinically evaluated and further advanced for clinical testing. Yang et al. evaluated the specificity of [68Ga]Ga-DOTA-GGNle-CycMSHhex towards MC1R in different cell lines (murine and human melanoma cells) and tumor models. In vitro and in vivo examinations showed the MC1R-specific targeting properties of [68Ga]Ga-DOTA-GGNle-CycMSHhex. [68Ga]Ga-DOTA-GGNle-CycMSHhex was further tested in two metastatic melanoma patients and demonstrated significant uptake in the metastatic lesions but needs more clinical studies to fully evaluate its clinical potential.205
Among the many investigated tracers, [68Ga]Ga-VMT02 and [68Ga]Ga-DOTA-GGNle-CycMSHhex have emerged as promising tracers for targeting MC1R-expressing tumors in patients with metastatic melanoma.
Various cyclic peptides (monomeric as well as multimeric) containing RGD as a sequence have been developed, e.g., cyclo-[RGDfK], cyclo-[RGDyK], cyclo-[RGDfE], cyclo-[RGDf(NMe)V], E[c(RGDfK)]2, and E[c(RGDyK)]2, and optimized to increase the tracer uptake in αvβ3 integrin-expressing tumors.207–210 [68Ga]Ga-RGD peptide derivatives specifically targeting αvβ3 integrin receptors have been further evaluated preclinically and clinically in various disease conditions such as thyroid cancer with Tg elevated negative iodine scan (TENIS), atherosclerotic plaques, myocardial infarction, breast cancer, glioblastoma, lung cancer, and chondroblastic osteosarcoma. These studies unveiled that with an increase in peptide multiplicity (especially in tetramers), significant tracer uptake in normal organs was observed, and consequently the desired target to background ratio was not obtained. Additionally, tetramers are not cost effective. Hence, dimeric cyclic RGD peptides have been found to be optimal for αvβ3 integrin-expressing tumor imaging.211
Besides αvβ3, many integrin subtypes, i.e., αvβ6, αvβ8, and α5β1, have been found to be overexpressed in different cancer conditions. Thus, to target these other integrins and increase the affinity of ligands, various monomers and trimers of integrin ligands with varying affinities have been evaluated preclinically and clinically (Table 8). Recently, Quigley et al. preclinically evaluated [68Ga]Ga-Trivehexin (a trimer with the peptide sequence cyclo[YRGDLAYp(NMe)K]), targeting the αvβ6 subtype of integrins, and also demonstrated the PET/CT imaging of head and neck and pancreatic cancer in humans. The radiolabeling resulted in the RCY of >95% and RCP of >99%. An in vitro assay was performed on αvβ6-positive H2009 cell lines, which showed its high affinity towards αvβ6 integrins (IC50 = 0.047 nM). The animal biodistribution and animal PET imaging showed high specific tracer uptake at the tumor site with low background uptake. These promising results were a driving force to perform human administration of [68Ga]Ga-Trivehexin for PET/CT imaging of 4 patients with head and neck and pancreatic cancer. The results of human imaging showed significant high tracer uptake in the metastatic lesions (SUVmax in the range of 6–14) together with excretory organs, i.e., kidneys and bladder. Low-grade uptake was also noted in the tumor-associated inflammatory sites. No other uptake was noted in any organs except excretory organs and the tumor site. This makes it a promising imaging agent for tumors expressing integrins.212
Tracer | Structure | Status | IC50 |
---|---|---|---|
[68Ga]Ga-DOTA-RGD |
![]() |
Clinical phase | 5.5 ± 1.6 nM207 |
[68Ga]Ga-BNOTA-PRGD2 |
![]() |
Clinical phase | 82.7 nM232 |
[68Ga]Ga-NOTA-c[RGDyK] |
![]() |
Pre-clinical phase | 3.6 ± 2.4 nM (Ki value)233 |
[68Ga]Ga-Avebetrin |
![]() |
Pre-clinical phase | 0.22 nM234 |
[68Ga]Ga-NODAGA-FR366 |
![]() |
Pre-clinical phase | 1.3 ± 0.08 nM235,236 |
[68Ga]Ga-Aquibeprin |
![]() |
Pre-clinical phase | 0.08 nM234 |
[68Ga]Ga-TRAP-AvB8 |
![]() |
Pre-clinical phase | 38 nM237 |
[68Ga]Ga-Triveoctin |
![]() |
Clinical phase | 5.7 nM238 |
[68Ga]Ga-TRAP-SDM17 |
![]() |
Pre-clinical phase | 7.4 nM239 |
[68Ga]Ga-TRAP(SDM17)3 |
![]() |
Pre-clinical phase | 0.26 nM239 |
[68Ga]Ga-Avebehexin |
![]() |
Pre-clinical phase | 0.26 nM240 |
[68Ga]Ga-Trivehexin |
![]() |
Clinical phase | 0.047 nM212 |
[68Ga]Ga-IAC |
![]() |
Clinical phase | Not reported216 |
[68Ga]Ga-NODAGA-c(NGR) |
![]() |
Pre-clinical phase | Not reported226 |
[68Ga]Ga-Cycratide |
![]() |
Clinical phase | 20.17 ± 1.68 nM241 |
Another potent 68Ga-labeled peptide, i.e., 4-[2-(3,4,5,6-tetrahydropyrimidine-2-ylamino)ethyloxy]benzoyl-2-[N-(3-amino-neopenta-1-carbamyl)]-aminoethylsulfonyl-amino-β-alanine (IAC), has been evaluated clinically to target the αvβ3 integrin subtype for angiogenesis imaging. This peptide has been conjugated with various chelating agents to increase its tumor retention and clearance from the body.213 Multiple studies are ongoing to evaluate the role of IAC as a theranostic molecule for several aggressive tumors.214–216 A phase I/II clinical trial is ongoing involving a multicentre interventional study (ClinicalTrials.gov NCT number: NCT04480619) using [68Ga]Ga-PEG-αvβ3-IAC.
Studies have shown the overexpression of the aminopeptidase N receptor (APN), also known as CD13, in angiogenic blood vessels. Therefore, it has been considered a suitable target for the detection and treatment of APN-expressing tumors. Various cancer conditions, such as melanoma, prostate, lung and ovarian cancer, have shown overexpression of APN.217 Pasqualini et al. developed a molecule containing an asparagine-glycine-arginine tripeptide sequence (NGR), which is highly specific to APN/CD13 receptors.218 Since then, various studies evaluating APN targeting NGR-based 68Ga-labeled tracers have been performed. It was found that NGR-based peptides have three-times the detection efficiency of neoangiogenic vessels than RGD peptides.219 As a result, several linear and cyclic NGR based peptides have been developed, studied and radiolabeled with 68Ga.220–227 Recently, Yang et al. synthesized and evaluated a cyclic NGR dimer peptide, [68Ga]Ga-DOTA-c(NGR)2, to study the CD13 expression in an ovarian cancer xenograft. The biodistribution study showed the excellent pharmacokinetics of the tracer with a high tumor-to-background ratio of 10.30 ± 0.26 at 1 h imaging. The tracer showed reduced uptake when the cold peptide was coinjected, which is attributed to the specificity of the tracer towards CD13.228 Alternatively, the NGR sequence tends to undergo deamidation of the asparagine (Asn) side chain,229 which can result in the formation of isoaspartic acid (isoAsp) and Asn residues.230 Therefore, to overcome the stability issues of NGR analogues, other peptides targeting the neoangiogenic process such as APN-selective LN peptide (YEVGHRC) and VEGFR-1-selective APRPG peptide, were preclinically evaluated.231
The search for a suitable tracer that targets pan integrins is still underway. However, [68Ga]Ga-Trivehexin and [68Ga]Ga-IAC are under evaluation clinically.
Further research towards improving tumor targeting, reducing kidney uptake and increasing the in vivo enzymatic stability of the MG analogue led to the development of another novel MG analogue, DOTA-D-Glu-Ala-Tyr-Gly-Trp-(N-Me)Nle-Asp-1-Nal-NH2 (DOTA-MGS5) (Table 9). Klingler et al. synthesized this novel CCK2-specific peptide and performed radiolabeling with 111In, 68Ga and 177Lu to carry out an extensive preclinical study. The radiolabeling of DOTA-MGS5 with 68Ga resulted in 95% RCP with a molar activity of 60 GBq per mmol. The labeled product was stable in PBS, saline and human serum, with more than 95% of the radiolabeled product intact in human serum for up to 4 h. The cellular study revealed more than 50–60% uptake of [68Ga]Ga-DOTA-MGS5 in A431-CCK2R cells after 2 h incubation, which was more than that of [68Ga]Ga-DOTA-PP-F11N (15–30%). Also, a blocking study with human gastrin revealed a decrease in uptake, i.e., below 1.5%, indicating the high specificity of the tracer towards CCK2 receptors. An animal biodistribution study revealed the physiological uptake of [68Ga]Ga-DOTA-MGS5 in the blood, heart, spleen, lung, and liver with slightly higher uptake in the CCK2-expressing spleen and stomach. Also, significant tracer accumulation at 1 h was noted in A431-CCK2R tumor-bearing animals (23.25% ± 4.70% IA per g).249 This tracer was further evaluated by Hörmann et al., and to translate it into clinics, cassette-based automated synthesis was performed.250 Furthermore, a case study involving [68Ga]Ga-DOTA-MGS5 vs. [18F]FDOPA in a 75-year-old patient suffering from MTC was reported. The PET/CT images showed fewer lymph node and bone lesions picked up by [68Ga]Ga-DOTA-MGS5 compared to [18F]FDOPA. However, more liver lesions with high SUV values were detected by [68Ga]Ga-DOTA-MGS5.251 Another preliminary clinical study was carried out by Guggenberg et al. in 6 patients of MTC using [68Ga]Ga-DOTA-MGS5. The tracer detected 87 lesions in total (50 bone lesions, 27 liver lesions, 8 lymph nodes and 2 local lesions) with higher uptake at 2 h than at 1 h (mean SUVmax, 7.2 vs. 6.0, respectively).252 A phase I/IIa clinical trial of [68Ga]Ga-DOTA-MGS5 is ongoing in patients with advanced NET, MTC, and gastroenteropancreatic and bronchopulmonary NET (EudraCT: 2020-003932-26). Another minigastrin analogue, i.e., [68Ga]Ga-DOTA-MGS8, has been developed, synthesized with an automated module and evaluated preclinically by the same group.253 The promising results of this preclinical study encouraged researchers to evaluate it further to translate it into clinics. There are review articles covering the development of CCK2 receptor imaging using various novel radiopharmaceuticals.254,255
The most clinically studied CCK2 receptor-targeting tracer is [68Ga]Ga-DOTA-MGS5 but additional clinical studies are ongoing using this tracer to optimize its tumor targeting, improve its pharmacokinetics and enhance its in vivo stability.
Radiopharmaceutical | Structure | Status | Ref. |
---|---|---|---|
[68Ga]Ga-P15-041 |
![]() |
Clinical Phase | Doot et al., 2020257 and Zha et al., 2020256 |
[68Ga]Ga-DOTA-PAM |
![]() |
Pre-clinical phase | Meckel et al., 2016292 |
[68Ga]Ga-DOTA-ZOL |
![]() |
Clinical phase | Meckel et al., 2016292 |
[68Ga]Ga-DOTA-BPAPD |
![]() |
Pre-clinical phase | Meckel et al., 2013293 |
[68Ga]Ga-DOTA-BPPED |
![]() |
Pre-clinical phase | Meckel et al., 2013293 |
[68Ga]Ga-NO2AP-BP |
![]() |
Clinical phase | Holub et al., 2014294 |
[68Ga]Ga-DOTA-BPAMD |
![]() |
Clinical phase | Fellner et al., 2010295 and Wu et al., 2016296 |
[68Ga]Ga-PhenA-BPAMD |
![]() |
Clinical phase | Fellner et al., 2010295 and Wu et al., 2016296 |
[68Ga]Ga-PhenA-HBP |
![]() |
Pre-clinical phase | Wu et al., 2016296 |
[68Ga]Ga-DOTA-(D-Asp)n |
![]() |
Pre-clinical phase | Ogawa et al., 2017297 |
[68Ga]Ga-EDTMP |
![]() |
Clinical phase | Toegel et al., 2008298 |
[68Ga]Ga-NOTA-BP |
![]() |
Pre-clinical phase | Suzuki et al., 2011299 and Holub et al., 2014294 |
[68Ga]Ga-DOTA-BP |
![]() |
Pre-clinical phase | Suzuki et al., 2011299 and Holub et al., 2014294 |
[68Ga]Ga-NOTAM-BP |
![]() |
Pre-clinical phase | Holub et al., 2014294 |
[68Ga]Ga-TRAP-(MDP)3 |
![]() |
Pre-clinical phase | Notni et al., 2012300 |
[68Ga]Ga-TRAP-(PDP)3 |
![]() |
Pre-clinical phase | Notni et al., 2012300 |
Multiple myeloma (MM) presents uniform overexpression of CD38 glycoprotein, which is considered the best target for therapeutic and imaging approaches. Pursuing this approach, a CD38-targeting single domain antibody i.e., Nb1053, has been developed. The binding moiety in Nb1053 is similar to that present in daratumumab, an approved monoclonal antibody for treating MMs. Wang et al. developed a novel [68Ga]Ga-NOTA-Nb1053 immunoPET imaging tracer and performed its characterization in preclinical MM and lymphoma models. The RCY and RCP obtained in radiolabeling were 47.84% ± 12.2%, and >99%, respectively. Biodistribution and PET imaging revealed the diagnostic potential of [68Ga]Ga-NOTA-Nb1053 over [18F]FDG in mice bearing subcutaneous MM.1S xenografts and orthotopic MM.1S. The authors demonstrated a significant reduction in renal uptake upon premedication with sodium maleate. The preclinical results of this novel tracer favor its clinical translation. However, before translating it to clinics, the high renal uptake and low tumor retention of [68Ga]Ga-NOTA-Nb1053 need further evaluation.259
Targeted molecular imaging of the immune checkpoint, i.e., programmed death ligand-1 (PD-L1), and its receptor, i.e., programmed death-1 (PD-1), utilizing nanobody-based radiotracers, i.e., [68Ga]Ga-NOTA-mal-hPD-L1 Nb and [68Ga]Ga-NOTA-Nb109, has emerged as a potential tool to assay the expression of PD-L1/PD-1. Initially, [68Ga]Ga-NOTA-mal-hPD-L1 Nb was introduced by Bridoux et al.260 and showed the feasibility of PD-L1 imaging, and subsequently [68Ga]Ga-NOTA-Nb109 showed the potential of nanobody-based PET imaging.261,262 Thereafter, various preclinical studies have been performed to target PD-L1/PD-1.262–264 Jinquan et al. used a custom-synthesized PD-L1-binding peptide, i.e., WL12 (a 12-mer anti-PD-L1 peptide) and radiolabeled it with 68Ga after conjugation with the NOTA chelator. The RCP obtained was >99% with a molar activity of 27.8–111.2 GBq per μmol. In vitro cellular and in vivo animal studies were performed together with a first in the human administration of [68Ga]Ga-WL12 to assess the feasibility of clinical translation of this tracer for PD-L1 imaging. The study showed significant tracer uptake at primary and metastatic sites (SUVmax in the range of 2.0–6.0).265 This tracer is currently under a phase 1 clinical trial involving patients with gastrointestinal tumors (ClinicalTrials.gov NCT number NCT04629326). Ma et al. preclinically evaluated a novel 68Ga labeled nanobody, [68Ga]Ga-THP-APN09, for the assessment of PD-L1 quantification and biodistribution in tumor-bearing mice. In addition, rapid one-step radiolabeling with no further purification was demonstrated, and further a pilot clinical study involving 9 NSCLC patients was performed (ClinicalTrials.gov NCT Number: NCT05156515).266 Another study of PD-L1 imaging is in an early phase 1 clinical trial (ClinicalTrials.gov NCT Number: NCT05490264). This study involves nanobody based tracer, [68Ga]Ga-NOTA-SNA002, for the PET imaging of patients with solid tumors.
Other potential biological targets have also been explored for immuno PET imaging; human CD8+ and CD8+ T cells have been targeted by various 68Ga-labeled nanobodies to assess their distribution in the tumor environment. Different derivatives of SNA006 nanobodies, i.e., SNA006a, SNA006c and SNA006d, were synthesized and conjugated with p-SCN-Bn-NOTA, and subsequently radiolabeled with 68Ga. This in vitro cellular study and in vivo PET imaging revealed the excellent characteristics of [68Ga]Ga-NOTA-SNA006a for the specific detection of CD8+ and CD8+ T cells in tumor lesions as well as across the whole body.267 The SNA006 nanobody-based tracer [68Ga]Ga-NODAGA-SNA006 is in an early phase 1 clinical trial (ClinicalTrials.gov NCT number: NCT05126927).
Another immune checkpoint explored for immuno PET imaging is the T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain (TIGIT) receptor. A novel peptide antagonist was synthesized and radiolabeled with 68Ga, i.e., [68Ga]Ga-NOTA-GP12, to evaluate the expression of TIGIT in cancers. Wang et al. developed this tracer and performed in vitro, in vivo and in human PET imaging of TIGIT expression in 2 patients with non-small cell lung carcinoma. The novel tracer was safe and demonstrated that uptake in the primary and metastatic lesions was consistent with the uptake of [18F]FDG.268 Thus, to establish this promising tracer clinically, more clinical studies must be performed to unfold its potential.
The tumor microenvironment involving cancer-associated fibroblasts, cancer-associated adipocytes, immune cells, macrophages and endothelial cells, plays an essential role in the development and growth of tumors. An alteration in any of the processes of tumor microenvironment makes it a suitable cancer-specific target. Tumor-associated macrophages (TAM) have been identified as a potential target to study their expression in cancer conditions. Recently, several 68Ga-labeled radiopharmaceuticals, i.e., [68Ga]Ga-NOTA-anti-MMR-sdAb and [68Ga]Ga-mannosylated serum albumin (MSA)-targeting macrophage mannose receptors (MMR) expressing TAM,269,270 [68Ga]Ga-NOTA-COG1410 targeting triggering receptor expressed on myeloid cells 2 (TREM2) of TAM,271 and [68Ga]Ga-DOTA-M2pep targeting M2 macrophage type of TAM,272 have emerged as potential TAM-specific preclinical imaging tracers. However, recently, a different side chain antibody-based 68Ga labeled tracer has undergone phase I/II clinical trials involving patients with different solid tumors (ClinicalTrials.gov NCT number: NCT04168528).
Another biological target for molecular imaging identified in the tumor microenvironment is extra domain-B fibronectin (EDB-F), an oncoprotein overexpressed in pancreatic cancer conditions. To perform the early diagnosis of pancreatic cancer, a ZD2 peptide (Thr-Val-Arg-Thr-Ser-Ala-Asp) specific to EDB-F was synthesized, radiolabeled with 68Ga and in vitro and animal in vivo studies performed. The preclinical results obtained were promising for the early detection of pancreatic cancer.273
A variety of PET and SPECT-based folate conjugates has been developed to target folate receptor expression.274 In 2014, Aljammaz et al. preclinically evaluated 68Ga-labeled NOTA-folate and NOTAM-folate conjugates for targeted imaging of folate receptor-expressing cancers such as ovarian cancer. This study demonstrated the easy and efficient radiolabeling of the tracer with a short synthesis time. The RCY and RCP obtained for the 68Ga-labeled NOTA- and NOTAM-folate conjugates were greater than 95% (decay corrected) and 98%, respectively. The in vitro cell binding affinities of the [68Ga]Ga-NOTA-folate and [68Ga]Ga-NOTAM-folate conjugates were found to be 54.36 ± 6.31 nM and 78.01 ± 13.0 nM, respectively. The in vivo biodistribution study showed significant tracer uptake in nude mice bearing a human KB xenograft owing to the specificity of the tracer towards folate positive tumors.275 Since then, no clinical studies have been performed to evaluate the potential of [68Ga]Ga-NOTA-folate. However, recently, another interesting preclinical study was performed to unfold the potential of [68Ga]Ga-NOTA-folate for macrophage associated inflammation imaging.276
The cell cycle is an essential phenomenon that every cell undergoes for reproduction and function, which is disturbed in many cancer conditions. This has helped researchers look for potential biological targets that can facilitate the diagnosis and treatment of cancer. Consequently, cyclin-dependent kinase 4 and 6 (CDK4/6) have been identified as suitable targets due to their overexpression in various cancers, especially breast cancer.277 Recently, a novel tracer, i.e., [68Ga]Ga-DOTA-palbociclib, was synthesized and preclinically evaluated for CDK4/6 positive tumors. The RCP obtained for [68Ga]Ga-DOTA-palbociclib was >95%, and this compound was found to be stable in the in vitro and in vivo analysis. It also showed excellent pharmacokinetics, together with significant uptake in the tumor lesion with low muscle background uptake. This tracer can potentially identify suitable patients for CDK4/6 inhibitor-based treatment.278
A recent study for the early detection of hepatocellular carcinoma (HCC) using the [68Ga]Ga-NOTA-G2 immunoPET imaging tracer targeting glypican 3 (GPC3), a proteoglycan, was performed by An et al. This study showed the superiority of the novel [68Ga]Ga-NOTA-G2 over the traditional [18F]-FDG in delineating subcutaneous HCC lesions.279
Among the various overexpressed biomarkers, metastasis-associated lung adenocarcinoma transcript-1 (MALAT-1) has also been found to be overexpressed in many different cancer conditions such as breast cancer, gastric cancer, and HCC. As a potential target for diagnostic imaging, a novel tracer, i.e., [68Ga]Ga-MALAT-1 ASO (antisense oligodeoxynucleotide), was developed to image malignant tumors with a high expression of MALAT-1. The study revealed excellent radiolabeling with >98% RCY and >99% RCP. In vitro analysis showed its specific binding to MHCC-LM3 cells, and significant tracer uptake (3.04% ± 0.11% ID per g) was noted in tumor-bearing mice in the biodistribution study. A similar finding was observed in microPET images, confirming the specific uptake of the tracer in the tumor-induced mouse models.280 This study showed promising results for this tracer. However, further optimization is needed given that the kidneys showed intense tracer uptake.
Further extending the role of molecular imaging, an interesting novel radiopharmaceutical has been developed i.e., 68Ga-labeled “magnetic-near infrared persistent luminescent hybrid mesoporous nanoparticles”, to perform multimodal imaging together with guided chemotherapy and photodynamic therapy. The study showed the great potential of this tracer as a one-stop nanotheranostic tool.281
Another novel tracer, i.e., [68Ga]Ga-DOTA-ethylenediamine-phenylboronic acid ([68Ga]Ga-DOTA-en-pba), was developed and evaluated preclinically for sialic acid overexpression in cancer cells. The authors performed in vitro studies on Mel-C and B16-F10 melanoma cells and found high specificity towards B16-F10 melanoma cells owing to sialic acid overexpression. Similar results were found in the ex vivo Mel-C and B16-F10 melanoma tumor model, confirming the specificity of the tracer for sialic acid overexpression.282 However, to translate it into clinics, this tracer needs further evaluation.
Urokinase-type plasminogen activator receptors (uPARs), an established cancer biomarker, have become a key focus for cancer diagnosis and treatment. To target uPARs, which are overexpressed in several cancer conditions such as breast cancer, prostate cancer, and urinary bladder cancer, a novel peptide-based tracer, i.e., [68Ga]Ga-NOTA-AE105, was developed, and currently is in a clinical phase.283,284
Substance P (SP) has surfaced in response to the demand for a possible neuropeptide that can be used as a therapeutic carrier and diagnostic biomarker in inoperable cancer conditions such as GBM. It is a small undecapeptide belonging to the family of tachykinins.285 It is known to bind the transmembrane neurokinin type 1 receptors (NK-1R). The first clinical study using SP was in 2006.286 However, it has regained the attention from researchers owing to the emergence of suitable alpha emitters. Recently, [68Ga]Ga-DOTA-[Thi8,Met(O2)11]-substance P ([68Ga]Ga-DOTA-SP) has been clinically employed for the assessment of its biodistribution in GBM patients overexpressing NK-1R. PET/CT images allowed visualization of tracer distribution in the brain and body. Only 5% of the injected activity was found in the bladder.287 Hence, [68Ga]Ga-DOTA-SP has emerged as an important diagnostic agent in the follow up of locoregional therapy of gliomas. Understanding the need for readily available tracers, Suthiram et al. demonstrated facile radiolabeling using a kit-like formulation of DOTA-[Thi8,Met(O2)11]-substance P.288
Gonadotrophin-releasing hormone (GnRH)-based analogues have been synthesized and radiolabeled with 68Ga i.e., [68Ga]Ga-DOTA-GnRH-I, to image reproductive system-associated cancers. The predominant subtypes of the GnRH receptor system expressed in different reproductive system tumors are GnRH receptor I (GnRHR-I) and GnRH receptor II (GnRHR-II). In 2008, Schottelius et al. showed that tracer uptake in GnRHR-positive OVCAR-3 xenografted nude mice was not significant. This study presented the challenges of the suitability of the tracer to target the GnRHR-system.289 Another study was reported in 2016 by Zoghi et al., in which they performed radiolabeling, characterization and preclinical evaluation of [68Ga]Ga-triptorelin (TRP) for GnRHR. This tracer demonstrated significant uptake in GnRHR-positive 4T1 tumor-bearing mice between 30–120 min post injection. At 60 min, the tumor to blood and tumor to muscle ratios were found to be 28 and >50, respectively.290 This study presented the possibility of utilizing GnRHR peptide for diagnostic and therapeutic applications. The same group also radiolabeled a derivative of TRP with 177Lu to evaluate it preclinically.291 However, in recent times, no study has been published related to GnRHR-targeting 68Ga-labeled peptides.
Radiopharmaceutical | Structure | Application | Status |
---|---|---|---|
[68Ga]Ga-NO2A-[Nle13]-Col |
![]() |
Liver fibrosis251 | Pre-clinical |
[68Ga]Ga-BOT5035 |
![]() |
Liver fibrosis252 | Pre-clinical |
[68Ga]Ga-NEB |
![]() |
Lymphatic disorders, Chylous fistula303–305 | Clinical phase |
[68Ga]Ga-H2CHXdedpa |
![]() |
MPI324 | Pre-clinical phase |
[68Ga]Ga-EDTA |
![]() |
Renal imaging261 | Clinical phase |
[68Ga]Ga-DTPA |
![]() |
Renal imaging325 | Clinical phase |
[68Ga]Ga-NOTA |
![]() |
Renal imaging262 | Pre-clinical phase |
[68Ga]Ga-DOTA |
![]() |
Renal imaging316 | Clinical Phase |
[68Ga]Ga-IRDye800-tilmanocept |
![]() |
Renal imaging317 | Pre-clinical Phase |
[68Ga]Ga-HBED-CC-DiAsp |
![]() |
Renal imaging318 | Pre-clinical Phase |
[68Ga]Ga-GSA |
![]() |
Liver function322 | Clinical phase |
[68Ga]Ga-NOTA-insulin |
![]() |
Alzheimer's disease323 | Pre-clinical phase |
Velikyan et al. also demonstrated the fully automated GMP production of [68Ga]Ga-BOT5035, which specifically targets platelet-derived growth factor receptor β (PDGFR-β) overexpressed in fibrotic pathologies. The RCY and RCP obtained in the GMP-compliant automated production were 43.7% ± 7.6% and 97.7% ± 0.4%, respectively. The biodistribution study revealed faster clearance of [68Ga]Ga-BOT5035 in most organs, including blood. The dosimetry study demonstrated that the effective dose delivered from 75 MBq of [68Ga]Ga-BOT5035 was 2.9 mSv in male rats and 3.4 mSv in female rats.302
Clinical studies have shown that 68Ga-labeled 1,4,7-triazacyclononane-N,N′,N′′-triacetic acid (NOTA) conjugated with truncated Evan blue (NEB), i.e., [68Ga]Ga-NEB, plays an essential role in the evaluation of lymphatic disorders.303,304 This tracer showed uptake in the lymphatic channels and lymph nodes in all the patients. According to the comparison of [99mTc]Tc-SC (sulfur colloid) lymphoscintigraphy and [68Ga]Ga-NEB, it was found that more information with respect to the detection of leak sites and lesions was received in 5 out of 13 patients in the [68Ga]Ga-NEB PET/CT images. Also, the acquisition time for [68Ga]Ga-NEB was much shorter. Other recently explored non-oncological applications of [68Ga]Ga-NEB include diagnosis of chylous fistula.305
Another tracer that has been evaluated for blood pool and lymph PET imaging is 68Ga-labeled maleimide ([68Ga]Ga-DM). The RCY and RCP obtained for this tracer was >90% and >95%, respectively. The partition coefficient determined for it was −3.15 ± 0.08. In vitro analysis showed the effective binding of [68Ga]Ga-DM to albumin with a binding fraction of over 70%. MicroPET imaging showed the ability of this tracer to detect transient micro bleeding in rats.306
The advantages of PET imaging over conventional SPECT or SPECT/CT imaging have led to the substitution of 99mTc with 68Ga in pulmonary ventilation/perfusion (V/Q) imaging. Ventilation imaging using [68Ga]Ga-carbon nanoparticles307 (Galligas) and perfusion imaging using [68Ga]Ga-macroaggregated albumin ([68Ga]Ga-MAA) were reported. In 2011, a pilot study was performed on patients with suspicion of pulmonary embolism (PE) by Hofman et al. The image quality obtained in PET/CT was superior to conventional SPECT in all the patients. The diagnosis made in PET/CT based on 68Ga-labeled tracers was consistent with the conventional V/Q imaging based on 99mTc tracers.308 The promising results of this study encouraged the further evaluation and optimization of Galligas and [68Ga]Ga-MAA. Since then, various studies have been conducted to justify the potential of Galligas and [68Ga]Ga-MAA PET/CT imaging in various oncological and non-oncological clinical conditions over conventional V/Q imaging.309–312 A recent study revealed that MAA particles do not discriminate between different isotopes when forming bonds. This allows researchers to investigate the radiolabeling potential of MAA particles with other isotopes.
A clinical study was conducted using [68Ga]Ga-NODAGA-ZOL to determine the correlation between tracer uptake in atherosclerosis plaques and cardiovascular risk profile patients. A positive correlation was observed between the tracer uptake of atherosclerosis plaques and atherogenic risk factors.313
Various 68Ga-labeled tracers for renal imaging have been identified, developed and under investigation. These tracers include [68Ga]Ga-EDTA, [68Ga]Ga-DTPA, [68Ga]Ga-NOTA, [68Ga]Ga-DOTA, [68Ga]Ga-IRDye800-tilmanocept and [68Ga]Ga-HBED-CC-DiAsp. Among them, [68Ga]Ga-EDTA has shown superior results, and has also been evaluated in humans for glomerular filtration rate (GFR) assessment. According to the authors, the GFR obtained with [68Ga]Ga-EDTA was consistent with the [51Cr]Cr-EDTA results.314 [68Ga]Ga-NOTA has also shown promising results in head-to-head comparison studies between [51Cr]Cr-EDTA and [68Ga]Ga-NOTA for GFR estimation in mice.315 However, the translation of [68Ga]Ga-NOTA into humans is yet to be evaluated. Recently, David et al. performed dynamic renal imaging in humans using [68Ga]Ga-DOTA and inferred that it can be used as a suitable alternative to conventional scintigraphy.316
Quin et al. introduced the novel 68Ga-labeled tilmanocept, which specifically binds to CD206 present in mesangial cells (a biomarker of diabetic nephropathy). This preclinical study showed the potential of [68Ga]Ga-IRDye800-tilmanocept in assessing GFR non-invasively through mesangial cell uptake.317 Another tracer, i.e., [68Ga]Ga-HBED-CC-DiAsp, was recently studied preclinically. The introduction of two aspartic acids in the HBED-CC chelator increased its hydrophilicity, which led to rapid renal clearance. Hence, this tracer showed promising results in GFR estimation.318
Similar to renal imaging, there are different 68Ga-labeled tracers, i.e., [68Ga]Ga-neolactosylated human serum albumin (LSA),319 [68Ga]Ga-NOTA-hexavalent lactoside (HL),320 and [68Ga]Ga-galactosyl human serum albumin (GSA),321,322 which have been studied for liver function imaging by targeting the asialoglycoprotein receptor expressed by hepatocytes. However, despite their promising preclinical and clinical results, they have not been translated into routine clinical studies because they showed no additional advantage over existing conventional techniques.
Taubel et al. developed a novel tracer i.e., [68Ga]Ga-NOTA-insulin, and preclinically evaluated the role of insulin in Alzheimer's disease (AD). In vivo biodistribution and micro PET imaging demonstrated significant tracer uptake in different brain regions (the cortex, thalamus, brain stem, and cerebellum) of AD mice compared to normal mice.323
Generally, infection is described as the invasion and growth of pathogens or microbes inside the body. It is caused by bacteria, viruses, fungi, parasites, or prions, and its prognosis becomes poorer in cases of unclear or no diagnosis, which can lead to sepsis or death. Therefore, it becomes important to diagnose and discriminate the cause of infection early. Presently available diagnostic tools such as blood investigations (WBC counts and C-reactive protein) and radiological examinations (X-ray, MRI, CT, and ultrasound), being nonspecific and anatomical imaging, pose challenges in determining the type of infection. Hence, the treatment management suffers significantly. With the introduction of hybrid imaging modalities such as PET-CT, PET-MRI and SPECT-CT, the early diagnosis of infection has become feasible, aided by the introduction of many novel specific radiopharmaceuticals, as shown in Table 12.
Radiopharmaceutical | Structure | Clinical Indication | Status |
---|---|---|---|
[68Ga]Ga-DOTA-ciprofloxacin |
![]() |
Staphylococcus aureus | Pre-clinical Phase332 |
[68Ga]Ga-CP-PA-SCN-BZ-DOTA |
![]() |
Staphylococcus aureus | Pre-clinical Phase331 |
[68Ga]Ga-CP-PA-SCN-BZ-NOTA |
![]() |
Staphylococcus aureus | Pre-clinical Phase331 |
[68Ga]Ga-ciprofloxacin-desferrichrome |
![]() |
Pseudomonas aeruginosa | Pre-clinical Phase353 |
Staphylococcus aureus | |||
and Klebsiella pneumoniae | |||
[68Ga]Ga-A8-K-DOTA |
![]() |
Staphylococcus aureus | Pre-clinical Phase338 |
[68Ga]Ga-A9-K-DOTA |
![]() |
||
[68Ga]Ga-A11-GSGK-DOTA |
![]() |
||
[68Ga]Ga-NOTA-UBI29-41 |
![]() |
Staphylococcus aureus | Clinical Phase354 |
[68Ga]Ga-NOTA-UBI30-41 |
![]() |
||
[68Ga]Ga-NOTA-UBI31-38 |
![]() |
||
[68Ga]Ga-DOTA-TBIA101 |
![]() |
E. coli | Pre-clinical Phase341 |
[68Ga]Ga-NODAGA-CDP1 |
![]() |
E. coli, Staphylococcus aureus, and Mycobacterium tuberculosis | Pre-clinical Phase344 |
[68Ga]Ga-TAFC |
![]() |
Aspergillus fumigatus | Pre-clinical Phase355,356 |
[68Ga]Ga-FC |
![]() |
||
[68Ga]Ga-FOXE |
![]() |
||
[68Ga]Ga-DFO-B |
![]() |
P. aeruginosa and S. aureus | Pre-clinical Phase357 |
[68Ga]Ga-pyoverdine |
![]() |
P. aeruginosa | Pre-clinical Phase358 |
[68Ga]Ga-Ornibactin |
![]() |
Burkholderia cepacia | Pre-clinical Phase359 |
[68Ga]Ga-citrate |
![]() |
Osteomyelitis, diskitis, intra-abdominal infection, tuberculosis, interstitial nephritis | Clinical Phase347,348 |
Inflammation is a physiological phenomenon, which is necessary for healing of wounds, protection from foreign bodies, and maintaining tissue homeostasis. However, severe inflammation can cause pathological alterations and even become a disease itself. It has been seen that inflammation is associated with many serious clinical conditions, including tumor progression. Hence, its accurate diagnosis and corresponding effective treatment are needed. In recent times, with the emergence of novel specific radiopharmaceuticals, the molecular imaging and therapy field has shown some strides in the management of inflammatory diseases. The biggest challenge in molecular imaging is to employ radiopharmaceuticals that can differentiate between infection and inflammation.
The first gallium isotope used for infection and inflammation imaging was Ga-67 (SPECT) as [67Ga]Ga-citrate, and it is still in clinical practice owing to its clinical applications in lung infections, sarcoidosis, tuberculosis, acute/chronic osteomyelitis, and retroperitoneal fibrosis.326 However, it has shortcomings, i.e., non-specific tumor accumulation, radiation burden to non-target organs, and availability. Hence, other gallium tracer options have been explored by the researchers.
The mechanism of action of antimicrobials enables them to specifically act on microbes.327 Utilizing this ‘infection-specific’ property, the following 68Ga-labeled tracers have been developed.
Ciprofloxacin is the most commonly employed fluoroquinolone-based antibiotic drug to treat Gram-positive and Gram-negative bacterial infections.328–330 Satapati et al. synthesized and radiolabeled ciprofloxacin analogues with 68Ga to evaluate their efficacy in imaging bacterial infections in rats. The RCY obtained for both conjugates was >90%, together with in vitro stability for 4 h. Based on bacterial binding and biodistribution studies, they inferred that both antibiotic conjugates can differentiate between bacterial infection and inflammation.331 Recently, a comparative study was conducted between 99mTc and 68Ga-labeled ciprofloxacin conjugates to study their physicochemical (stability and lipophilicity) and biological (binding study to Staphylococcus aureus and Pseudomonas aeruginosa) properties.332
Tracers based on antimicrobial proteins and peptides have shown superiority over antibiotic analogues because of their higher specificity for infection. The most promising results have been demonstrated by the antimicrobial peptide ubiquicidin (UBI) labeled with 68Ga. Several UBI-derived antimicrobial peptides were investigated by Welling et al. and it was inferred that the peptide fragment (29–41) of UBI with the amino acid sequence Thr-Gly-Arg-Ala-Lys-Arg-Arg-Met-Gln-Tyr-Asn-Arg-Arg has potential to differentiate infection from inflammation.333 Since then, various UBI-based derivatives, namely NOTA-UBI29-41, NOTA-UBI30-41, and NOTA-UBI31-38, have been radiolabeled with 68Ga to evaluate their efficacy in detecting infection.334–337 Bhatt et al. performed a preliminary clinical evaluation of [68Ga]Ga-NOTA-UBI31-38 in three patients with suspected infection and found that it gets accumulated at sites of infection. PET-CT images showed high target-to-background ratios in the order of 2.6 and 2.1 in patients with lung and knee infections, respectively. It was inferred from their study that [68Ga]Ga-NOTA-UBI31-38 is specific for Staphylococcus aureus infection.334 Hence, for [68Ga]Ga-NOTA-UBI31-38 to become a standard part of clinical practice in the case of Staphylococcus aureus infection, more clinical research is required.
Staphylococcus aureus is well known to form a biofilm, which often leads to serious life-threatening bacteraemia and sepsis. Various radiopharmaceuticals specific to bacteria in biofilms have been developed and studied to discriminate between infection and inflammation. Nielsen et al. evaluated 68Ga-labeled phage-display selected peptides ([68Ga]Ga-DOTA-A8-K, [68Ga]Ga-DOTA-A9-K and [68Ga]Ga-DOTA-A11-GSGK) for positron emission tomography imaging of Staphylococcus aureus biofilm-associated infections. Among them, [68Ga]Ga-DOTA-A9-K outperformed and showed favourable characteristic features of being a novel bacteria-specific imaging tracer.338 However, Afzelius et al. conducted a study that refutes the previously cited findings. In a juvenile pig model, osteomyelitis bone lesions caused by Staphylococcus aureus did not exhibit any [68Ga]Ga-DOTA-A9-K uptake.339
Depsipeptides are naturally occurring peptides containing more than one ester bond in their cyclic structure. The derivatives of depsipeptides exhibit different biological activities such as antiviral, antimicrobial, insecticidal, antitumor, tumor promotive, immunosuppressive, and anti-inflammatory.340 Based on the antimicrobial action of depsipeptides, Mokaleng et al. developed and radiolabeled a depsipeptide derivative, i.e., [68Ga]Ga-DOTA-TBIA101, to study E. coli-infection in a BALB/c mouse model. The authors reported a low target-to-non-target uptake ratio and fast renal clearance in PET/CT images.341 Another study was performed using [68Ga]Ga-DOTA-TBIA101 by Ebenhan et al. to evaluate its diagnostic potential as an infection imaging agent in rabbits with various disease models, i.e., positive Staphylococcus aureus infection, sterile inflammation and Mycobacterium tuberculosis. Images showed high uptake at the sterile inflammation and Mycobacterium tuberculosis sites.342
Another tracer based on LL37, i.e., an antimicrobial peptide with a 37 amino acid chain, has been explored as a potential imaging agent for the diagnosis of infection. Primary studies revealed the selectivity of [68Ga]Ga-CDP1 towards bacterial infection.343,344 However, further studies must be carried out to determine the utility of [68Ga]Ga-CDP1.
Besides these tracers, antimicrobial peptide fragment-based tracers, i.e., [68Ga]Ga-GF-17 (peptide sequence-GFKRIVQRIKDFLRNLV-NH2) and [68Ga]Ga-RAWVAWR-NH2, specific to E. coli and S. aureus, have also been evaluated in animals.345
Various novel siderophore-based tracers such as [68Ga]Ga-desferri-triacetylfusarinine C (TAFC), [68Ga]Ga-desferri-ferricrocin (FC), [68Ga]Ga-ferrioxamine E (FOXE), [68Ga]Ga-fusarinine C (FUS), [68Ga]Ga-desferrioxamine B (DFO-B), [68Ga]Ga-pyoverdine and [68Ga]Ga-ornibactin have also been developed and evaluated in animals to determine their utility in different infection conditions. Peukert et al. also developed, optimized and radiolabeled artificial siderophores with 68Ga for bacterial infection imaging. A total of 11 cyclen-based siderophores with different binding sites for iron and 68Ga was developed. Based on the in vitro and in vivo analysis, [68Ga]7 and [68Ga]15 were found to be the most suitable for imaging. Both tracers showed significant uptake in mice infected with Escherichia coli.346
In 1971, [67Ga]Ga-citrate was used as an infection and inflammation imaging agent. More recent advancements in PET imaging and the easy availability of 68Ga via a generator have led to the initiation of [68Ga]Ga-citrate into clinical studies. Among the listed 68Ga-labeled tracers in Table 12, [68Ga]Ga-citrate is the only tracer incorporated into clinical applications given that it exhibits high diagnostic accuracy of more than 90% for osteomyelitis and diskitis.347,348 The diverse applications of [68Ga]Ga-citrate in the field of molecular imaging include skeletal muscle infection and inflammation, abdominal infection, inflammatory bowel disease,349 tuberculosis,350 atherosclerosis,351 and contagious disease imaging.
In 2011, Kumar et al. identified the detection ability of [68Ga]Ga-apo-transferrin for Staphylococcus aureus infection induced in rat models. Specific uptake of [68Ga]Ga-apo-transferrin in the lesions was observed, and no uptake of [68Ga]GaCl3 was observed in the lesions up to 2 h post injection.352
To target these biomarkers, various 68Ga-labeled imaging agents have been investigated preclinically and clinically. Many of these tracers were reviewed extensively by Velikyan in 2018,360 and recently, a review article covering all the 68Ga-labeled tracers for imaging inflammatory disorders was published.361
Another interesting novel radiopharmaceutical, i.e., [68Ga]Ga-DOTA-HsTX1[R14A], has been recently investigated preclinically in neuroinflammatory disease conditions overexpressing the voltage-gated potassium channel, i.e., Kv1.3. These channels have been found to be overexpressed in the post-mortem brain of patients with Alzheimer's and Parkinson's disease.362,363 HsTX1[R14A] is an analogue of HsTX1, which is a C-terminally amidated peptide isolated from scorpion venom. In mice treated with lipopolysaccharide (LPS), PET/CT images of [68Ga]Ga-DOTA-HsTX1[R14A] biodistribution revealed its uptake in the inflamed brain, cerebellum, and joints. In contrast, the mice given saline treatment showed no tracer uptake in their joints or brains. The tracer exhibited sensitivity to Kv1.3, which was elevated because of the LPS treatment. Thus, this study successfully demonstrated to potential of this tracer to diagnose disease conditions overexpressing Kv1.3.364
Tracer | Structure | Status | Ref. |
---|---|---|---|
[68Ga]Ga-iPSMA-BN |
![]() |
Clinical Phase | Eder et al., 2014366 |
[68Ga]Ga-NOTA-DUPA-RM26 |
![]() |
Pre-clinical Phase | Mitran et al., 2019370 |
[68Ga]Ga-NOTA-BBN-RGD |
![]() |
Clinical Phase | Liu et al., 2009377 |
[68Ga]Ga-NOTA-3P-TATE-RGD |
![]() |
Clinical Phase | Zheng et al., 2019371 |
[68Ga]Ga-FAPI-RGD |
![]() |
Clinical Phase | Zang et al., 2022373 |
[68Ga]Ga-NOTA-RGD-GE11 |
![]() |
Pre-clinical Phase | Yu et al., 2015374 |
[68Ga]Ga-NGR-RGD |
![]() |
Pre-clinical Phase | Gai et al., 2020378 |
Another bispecific heterodimer radioligand evaluated for prostate cancer imaging is [68Ga]Ga-NOTA-DUPA-RM26. Its structure consists of both a PSMA-targeting moiety, i.e., DUPA (2-[3-(1,3-dicarboxy propyl)ureido]pentanedioic acid) and a GRPR-targeting moiety, i.e., RM26 (D-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2 ([D-Phe6, Sta13, Leu14]-bombesin(6–14)). Labeling of the heterodimer with 68Ga exhibited >98% RCY with high stability in the serum. The in vivo biodistribution revealed physiological tracer uptake in GRPR-expressing organs with minimal uptake in PSMA-expressing organs, and considerably higher tumor uptake in mice bearing PC3-PIP cells (at 1 h post injection 8% ± 2% ID per g). High tumor to non-tumor activity was observed at 3 h post injection. In vivo imaging showed a similar trend to that observed in the in vivo biodistribution study. This study demonstrated the excellent potential of [68Ga]Ga-NOTA-DUPA-RM26 for targeting GRPR and PSMA-expressing prostate cancer.370 However, this tracer needs further evaluation before being translated into clinics.
In 2016, a clinical trial study was registered involving [68Ga]Ga-NOTA-BBN-RGD as an imaging agent for prostate cancer patients (ClinicalTrials.gov NCT number: NCT02747290). The results of this study have yet to be published. Recently, a clinical trial was registered (ClinicalTrials.gov NCT number: NCT05549024) to evaluate a novel bispecific heterodimer radioligand, [68Ga]Ga-RM26-RGD, as a potential imaging agent for GRPR- and αvβ3-positive tumors.
One more dual-targeting tracer, i.e., [68Ga]Ga-NOTA-3P-TATE-RGD, has undergone clinical evaluation.371,372 It outperformed the conventional [68Ga]Ga-DOTA-TATE in detecting liver lesions and metastatic lesions.372
Another significant novel tracer, [68Ga]Ga-FAPI-RGD, which targets both FAP and αvβ3, has been developed. Zang et al. synthesized this tracer using the quinolone-based FAPI-02 as one arm and cyclic RGDfK as the other arm, coupled to the NOTA chelator. The authors assessed the specificity, radiation dosimetry, in vivo pharmacological behaviour, and preclinical imaging of this tracer. The same team also carried out a pilot human trial with 6 patients to further translate it into clinics. The tracer demonstrated faster clearance from the urinary tract and prolonged tumor retention. The effective dose delivered by [68Ga]Ga-FAPI-RGD was found to be 19.4 μSv per MBq. The PET/CT images revealed a higher target-to-background ratio, which further increased with time, resulting in good image quality for clinical interpretation. No significant difference was observed pertaining to SUVmax of the primary tumor and metastatic sites in 5 out of 6 patients. However, in one patient of renal carcinoma, high uptake of [68Ga]Ga-FAPI-RGD was observed compared to [18F]FDG.373 A phase I/II clinical study has been registered with [68Ga]Ga-FAPI-RGD involving various cancer patients (ClinicalTrials.gov NCT number: NCT05515783).
To image tumors that express αvβ3 and EGFR, Yu et al. successfully synthesized 68Ga-labeled NOTA-RGD-GE11, a bispecific heterodimeric ligand.374In vitro study demonstrated the binding affinity of NOTA-RGD-GE11 towards both αvβ3 and EGFR. The in vivo biodistribution and PET imaging studies of [68Ga]Ga-NOTA-RGD-GE11 showed the higher tumor uptake and tumor to muscle ratio, 3.446% ± 0.548% ID per g and 4.397% ± 0.972%, respectively, at 2 h post injection. The results of the bispecific heterodimer ligand were superior to that of the [68Ga]Ga-NOTA-RGD and [68Ga]Ga-NOTA-GE11 monomeric ligands.375 However, the tracer has not yet been translated into human imaging.
Long et al. preclinically assessed a bispecific heterodimer radioligand, [68Ga]Ga-NGR-RGD, in ovarian cancer xenografts with the aim of targeting CD13 and αvβ3. The results showed that in comparison to [18F]FDG, there was a high tumor-to-background ratio, minimal inflammatory uptake, and a high tumor targeting efficacy. Thus, this tracer presents potential for further evaluation.376
This journal is © The Royal Society of Chemistry 2025 |