Next Article in Journal
Colloid Carcinoma of the Pancreas with a Series of Radiological and Pathological Studies for Diagnosis: A Case Report
Next Article in Special Issue
A Rare Case of Radiation-Induced Liver Disease in Treated Abdominal Lymphoma Showing High [18F]FDG Avidity and Low EOB Uptake Proportional to the Irradiation Dose
Previous Article in Journal
CT- and MRI-Based 3D Reconstruction of Knee Joint to Assess Cartilage and Bone
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Combined 68Ga DOTA-Peptides and 18F FDG PET/CT in the Evaluation of Gastroenteropancreatic Neuroendocrine Neoplasms

by
Chalermrat Kaewput
1,* and
Sobhan Vinjamuri
2
1
Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
2
Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool L7 8XP, UK
*
Author to whom correspondence should be addressed.
Diagnostics 2022, 12(2), 280; https://doi.org/10.3390/diagnostics12020280
Submission received: 30 December 2021 / Revised: 19 January 2022 / Accepted: 20 January 2022 / Published: 22 January 2022
(This article belongs to the Special Issue Image-Guided Cancer Diagnosis and Therapy)

Abstract

:
This review article summarizes the role of combined 68Ga DOTA-peptides and 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) in the evaluation of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). Patients with GEP-NENs can initially present themselves to a gastroenterologist or endocrinologist rather than cancer specialist; hence, it is vital for a wider group of clinicians to be familiar with the range of tests available for the evaluation of these patients. The role of PET scanning by using 68Ga DOTA-peptides has a high sensitivity in the diagnosis of GEP-NENs and to guide patient selection for treatment with somatostatin analogues (SSA) and/or peptide receptor radionuclide therapy (PRRT). The loss of somatostatin receptor (SSTR) expression was found to be associated with an increased glucose metabolism in cells. However, the routine use of SSTR targeted radiotracers in combination with 18F-FDG to evaluate glucose utilization in GEP-NENs is still debatable. In our opinion, in patients with NENs, 18F-FDG PET should be performed in the case of a negative or slightly positive 68Ga DOTA-peptides PET scan for assessing the dedifferentiation status, to guide correct therapeutic strategy and to evaluate the prognosis. The approach of combined receptor and metabolic imaging can improve diagnostic accuracy, especially considering the heterogeneity of these lesions. Therefore, 68Ga DOTA-peptides and 18F-FDG PET should be considered complementary in patients with GEP-NENs.

1. Introduction

Neuroendocrine neoplasms (NENs) are epithelial neoplasms that arise from cells of the endocrine and nervous systems, with distinctive differentiation, and could occur in the various organ but more frequently from the gastrointestinal tract, followed by the lung. In the past, these tumors were thought to be extremely rare, but many recent studies have shown an increasing incidence and prevalence, and the incidence of GEP-NENs has increased more than six-fold between 1997 and 2012 [1,2,3]. This increase is partly due to better diagnostic methods and greater awareness of the disease.
Most NENs express somatostatin receptors (SSTR), which could be used as a target for radionuclides imaging and treatment. Somatostatin (SST) is a cyclic peptide hormone, also known as somatotropin release inhibiting factor (SRIF). It was originally found to be a growth hormone inhibitor, but it is currently known to be involved in inhibiting various metabolic processes related to neurotransmitters and endocrine secretions, as well as controlling exocrine secretions [4]. SSTRs are widely expressed throughout the body, with five subtypes reported (SSTR 1–5). For most NENs, SSTR2 is the most predominantly expressed receptor subtype [5]. Innovations in somatostatin receptor-based technology have pushed forward molecular diagnostics and treatment of NENs, markedly improving the quality of life of many patients.
GEP-NENs are NENs occurring at various anatomical sites of the gastrointestinal tract and pancreas, and they comprise a heterogeneous group of epithelial neoplasms with a wide range of aggressiveness. According to the WHO 2019 classification system, GEP-NENs are classified according to the Ki-67 index and mitotic count. An important feature of the new classification is a difference between differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs), since both share common expression of neuroendocrine markers. These dichotomous morphological subdivisions into NETs and NECs are also differentiated by genetic evidence at specific anatomic sites, clinical, epidemiologic, histological differences, and prognosis. NETs are classified as G1, G2, or G3 in many organ systems, and they rely on the mitotic count and/or Ki-67 labeling index, and/or the presence of necrosis. The distinction of well-differentiated NET G3 from NEC, previously applicable only for Pan-NENs, is now used for gastrointestinal NENs in the revised WHO classification for GEP-NENs, as described in Table 1 [6].
NETs arising at various anatomical areas of the gastrointestinal tract display tumor identities that vary in their physiology and clinical presentation (Table 2). Specific staining for peptide hormones could be used to confirm the origin of clinical symptomatology, but there is no definite agreement between immunohistochemistry (IHC) and symptoms, since bioactive compounds can be synthesized without secretion or non-functioning NENs (NF-NENs).
There are several prognostic predictors of NENs, including pathological factors (cytology and Ki-67 index) and biological factors (chromogranin A) [8]. In general, NENs have a wide range of cell differentiation. The presence of receptors at the cell surface appears to depend on the differentiation of tumor cell, with well-differentiated tumors showing a higher affinity for somatostatin [9].
Imaging acts as a major role in evaluating NENs. These include detecting, staging, evaluating the response, and prognosis [10]. Relying on the site of tumor origin, NENs might show site-specific symptoms. Due to these variations, NENs patients can present with various stage, and over 50% have metastases at the time of diagnosis. Functioning NENs tend to present earlier than nonfunctioning NENs. Therefore, the imaging modalities need to be very different, and they need to be tailor-made for each patient depending on their clinical presentation.

1.1. Conventional Imaging

Conventional imaging (CI) methods, including contrast-enhanced CT scans and MRI, are usually used as the primary imaging methods for patients presenting with signs or symptoms suggestive of GEP-NENs, both for detecting the site of primary tumor and possible metastatic sites. These modalities are most useful for staging and surgical planning, due to the great anatomic detail of the tumors and their surrounding structures. Most GEP-NENs are shown as contrast-enhancing lesions during the arterial phase on CT or MRI. MRI is an ideal imaging modality for differentiating between benign and malignant hepatic lesions. MRI seems to have higher sensitivity than CT in the diagnosis of liver and bone-marrow metastases, but both types of imaging techniques may miss small lymph node metastases. The sensitivity of techniques can also be variable for either recurrent or metastatic malignancy. This is due to the fact that these techniques primarily detect change in anatomy rather than function, and, therefore, they are considered to have moderate specificity [11].
According to diagnostic and therapeutic guidelines for GEP-NENs, the sensitivity of CT imaging in the diagnosis of pancreatic tumors is approximately 70%, and specificity is approximately 90%. In addition, CT has high sensitivity and specificity in the detection of metastases. The sensitivity is 82% and the specificity is 92% in liver metastases, while the sensitivity is 60–70% and specificity is 87–100% for lymph nodes metastases [12,13]. CT is preferable for imaging the lungs as compared to MRI, because it has a better spatial resolution. It is considered as the most effective imaging method for the detection of small bowel NENs. The disadvantage of contrast-enhanced CT is its poor sensitivity to detect small lesions less than 1 cm and for bone metastases (sensitivity about 50%). Recent studies have reported that MRI has more than 80% sensitivity to detect hepatic metastases [14,15,16].

1.2. 111In-Pentetreotide

Functional imaging studies based on the expression of SSTRs play an important role in evaluating patients with suspected GEP-NENs and high density of SSTRs. CT and MRI allow for anatomical characterization, but they cannot provide accurate functional information and are often insufficient for the diagnosis of GEP-NENs if the lesions are small and cover various anatomical locations. Somatostatin receptor scintigraphy (SRS) can provide important functional data. Thus, SRS shows good accuracy for whole-body imaging and has been routinely used for the diagnosis and follow-up of GEP-NENs [17].
SRS using the somatostatin analog 111In-pentetreotide (111In-DTPA-D-Phe-conjugate of octreotide; Octreoscan®) was the first radiotracer agent to be approved for NENs in 1989 [18]. The radiotracer preferentially binds to SSTR subtypes 2, 3 and 5, especially 2, and has proven to be an essential diagnostic functional imaging for the diagnosis, staging and follow-up of NENs, both pulmonary and, in particular, GEP-NENs. The added value of a whole body assessment in one setting is particularly advantageous.
111In-Pentetreotide scans are routinely obtained at 4 and 24 h after the intravenous administration of the radiotracer, and image acquisition is ideally performed with a double-headed gamma camera equipped with a medium-energy collimator [18]. However, in spite of the advantage over CI, the principal limitation of using 111In-Pentetreotide is the low spatial resolution of the gamma camera that significantly affects the assessment of small lesions, particularly in organs with a high physiological uptake (such as the liver). The use of SPECT/CT imaging has markedly improved the accuracy of 111In-Pentetreotide, increasing its spatial resolution and the anatomic localization of pathologic sites of increased uptake due to CT co-registration, with decreased false positive results and its capacity to provide cross-sectional scintigraphic images [19,20,21,22,23].
111In-Pentetreotide, acquired as SPECT/CT, shows higher sensitivity and accuracy values than CI and can be still used for routine diagnostic imaging of non-functioning NEN-GEPs, with significant impact on treatment planning. The added value of SPECT/CT over CI was 35.6%. SPECT/CT corrected CI classification and patient management in 27.9% of cases, while it down-staged the disease better than CI in 9.6% of cases. However, the combined use of CI and functional imaging, using 111In-Pentetreotide SPECT/CT, still leaves a big gap in the diagnostic pathway [24]. Therefore, newer, higher-affinity SST analogs, labeled with radioisotopes with better resolution and dosimetry, such as 68Ga, which is a positron emitter, have been considered as promising SSTR imaging agents.

1.3. 68Ga-DOTA-Conjugated Peptide-Binding SSTR PET/CT

68Ga-DOTA-conjugated peptide-binding SSTR-based imaging techniques are acknowledged as a new category of radiopharmaceuticals that has recently been instructed into routine clinical practice, including DOTA-D Phe1-Tyr3-octreotide (DOTATOC), DOTA 1-Nal3-octreotide (DOTANOC), and DOTA-D Phe1-Tyr3-Thr8-octreotide (DOTATATE) [25]. Although all of these radiopeptides bind to SSTR2, each has various affinities for other SSTR subtypes. In addition to the greatest affinity to SSTR2, 68Ga-DOTA-NOC also reveals a good affinity for SSTR3 and SSTR5; 68Ga-DOTA-TOC also shows a high affinity for SSTR5, but less affinity compared to DOTA-NOC, whereas 68Ga DOTA-TATE predominantly binds to SSTR2 [26].
Table 3 lists the frequency of overexpression of different SSTR subtypes in different clinical presentations [27].
68Ga DOTA conjugated peptide-binding SSTR is generally used for primary tumor localization, including metastatic detection (staging) and evaluation of residual, recurrent, or progressive disease (restaging) [28,29] all the way through to the selection of patients for PRRT [30]. PRRT can successfully control symptoms due to hypersecretion of hormones and has also been displayed to improve OS in progressive or symptomatic NETs patients [31]. The mechanism of uptake is based on increased SSTR expression in neuroendocrine cells and can be detected on SSTR scanning [32]. The previous studies revealed statistically significant decreased tumor uptake of 68Ga-DOTA-peptides PET in poorly differentiated NETs, and its uptake might be related to aggressive behavior and might lead to poorer prognosis [33]. However, some cases in this study were low-grade NENs that showed low tumor uptake on 68Ga-DOTA-peptides PET/CT scans. It is possible that the FNA site may not fully reflect the true pathological grade of a patient with heterogeneity of cellular differentiation within the same tumor mass, and this may also reflect the potential ability of 68Ga-labeled SSTR PET/CT to map these cellular characteristics (Figure 1) [33,34].
The recommended indications for patients with NENs according to EANM Guidelines for 68Ga-DOTA-conjugated somatostatin PET/CT imaging [35] are as follows:
  • Localized primary tumors and detect sites of metastasis (staging);
  • Monitoring patients with known disease in the detection of residual, recurrent or progressive disease (re-staging);
  • Determine SSTR status (patients with SSTR positive tumors are more likely to respond to targeted therapy with SSA);
  • Select patients with metastasis for PRRT (177Lu or 90Y–DOTA-labelled peptides).
68Ga-DOTA-conjugated peptide-binding SSTR is significantly superior to 111In-Pentetreotide in both diagnostic accuracy and its therapeutic impact. Available evidence also supports the concept that 68Ga-DOTA-conjugated peptide-binding SSTR imaging often demonstrates tumor uptake in some patients with negative or equivocal 111In-Pentetreotide scans, and it identifies patients who may benefit from PRRT [36].
A prospective study of 131 GEP-NENs patients determined the superiority of 68Ga-DOTATATE PET/CT over 111In-pentetreotide SPECT/CT, CT, and/or MRI. 68Ga-DOTATATE PET/CT detected 95.1% of lesions, while CI detected 45.3% of lesions, and 111In-pentetreotide SPECT/CT detected only 30.9% of lesions with statistically significant differences between all imaging modalities. In the subgroup of patients with carcinoid symptoms but negative biochemical testing, 68Ga-DOTATATE PET/CT detected lesions in 65.2% of patients. Of note, 40% of the lesions detected by 68Ga-DOTATATE PET/CT were missed on CI or 111In-pentetreotide SPECT/CT. Additionally, based on the findings from 68Ga-DOTATATE PET/CT, there was a significant change in the patient management in approximately a third of the patients (32.8%) [37].
Binnebeek et al. revealed that significantly more lesions were detected on 68Ga-DOTATOC-PET/CT as compared to 111In-pentetreotide scans. The sensitivity for PET/CT was 99.9% (95% CI, 99.3–100.0), and for SPECT, it was 60.0% (95% CI, 48.5–70.2). The organ-by-organ analysis showed that the PET was most frequently visualized in the liver and skeleton. They concluded that 68Ga-DOTATOC-PET/CT is superior for detecting NENs compared to 111In-pentetreotide SPECT [38]. Additionally, treatment was changed in more than one-third of patients with SSTR PET/CT rather than 111In-pentetreotide scan [39].

1.4. 18F-FDG PET/CT

18F-FDG is used to evaluate glucose metabolism, and it is the most commonly used radiotracer in PET imaging. It is not currently routinely used for NENs imaging, but recent experience has suggested that it can provide complementary information. 18F-FDG PET/CT has a limited advantage in well-differentiated NETs, since most NENs frequently have near normal glucose metabolism [40]. 18F-FDG exploits the increased glycolytic activity of tumor cells, as glycolytic activity seems to be low in most NENs. In fact, it can differentiate between slowly proliferative and aggressive tumors [41,42]. Additionally, 18F-FDG PET/CT scans can allow prognostic prediction, including overall survival (OS) and progression free survival (PFS). Finally, it could potentially identify those who are not responding earlier, characterize early progression, and change therapy if required.
Assessment of glucose metabolism by 18F-FDG may be helpful in the diagnosis of high-risk aggressive disease associated with poor outcomes. 18F-FDG PET may still an essential modality in the management of NENs patients, due to its better prognostic value and greater sensitivity to identify the extent of disease, particularly in aggressive high-grade tumors [43] Bahri H et al. revealed that the median PFS and OS are significantly greater in patients with a negative 18F-FDG PET finding (OS nearly 120 months) than in patients with a positive 18F-FDG PET finding (only 15 months) [44]. Ezziddin et al. [45] retrospectively reviewed data from 89 patients with metastatic GEP-NENs and classified three different prognostic groups according to FDG uptake: mG1, tumor-to-liver ratio of maximum SUV ≤ 1.0; mG2, 1.0–2.3; and mG3, >2.3. These groups were correlated with significantly different OS (not more than 114 vs. 55 vs. 13 months, respectively). As mentioned, 18F-FDG PET must not only be used in staging but also for grading whole-body imaging modality; positive 18F-FDG PET lesions have a significant correlation with prognosis, irrespective of SSTR expression.
The recommended indications for 18F-FDG PET/CT in patients with NENs according to EANM Guideline for 68Ga-DOTA-conjugated somatostatin PET/CT imaging [35] are as follows:
  • Localization of NECs and high-grade poorly differentiated NETs with aggressive behavior;
  • Prediction of prognosis;
  • Localization of synchrone/metachrone non-NEN malignancy.

1.5. Diagnostic Value of Combined 68Ga SSTR and 18F-FDG PET in GEP-NENs

The tumor grade indicates the tumor’s biological aggressiveness, which is recognized as a strong predictor for prognosis in many tumors. The NETs grading system is based on proliferation rate, which is determined by the percentage of tumor cells immunolabeled for the Ki-67 antigen positive (Ki-67 index) or the number of mitoses per 10 high-power microscopic fields or 2 mm2 (mitotic rate) [46]. Patients with high-grade tumors have high turnover with highly aggressive features and may predict rapid progression, while lower-grade patients may be able to live with a stable disease for a long time. It is essential to differentiate between rapidly progressing tumors and relatively stable tumors, because the treatment approaches for aggressive tumors might have long-term toxicity and only modest efficacy.
Due to the range of SSTR expressions between well-differentiated and poorly differentiated GEP-NENs, 18F-FDG PET/CT and 68Ga-DOTA-peptide PET/CT can have a complementary role in the imaging of NENs. Well-differentiated tumors tend to have greater 68Ga-DOTA-peptide avidity and lower FDG avidity (Figure 2). Conversely, poorly differentiated tumors tend to have lower 68Ga-DOTA-peptide avidity and greater FDG avidity [32,47]. These characteristics are referred to as the “flip-flop” phenomenon that is caused by greater SSTR expression and decreased glycolytic activity in well-differentiated tumors, and conversely decreased SSTR expression and greater glycolytic activity in poorly differentiated tumors [48].
The use of 68Ga DOTA-peptides PET in combination with 18F-FDG PET in GEP-NENs patients provides complementary information and could also be used for the whole-body characterization of tumor heterogeneity, which cannot be assessed with tumor biopsy alone. The combined PET/CT approach can identify the highest-grade and most aggressive lesions by localizing the most highly positive 18F-FDG and negative SSTRs. This whole-body evaluation of tumor glycolytic activity, together with SSTR expression, can help in classification of patients and selection of candidates or SSA therapy or PRRT, and can have a significant impact on therapeutic options. In addition, the combined PET/CT approach scan may detect changes that may occur in molecular heterogeneity of the disease over time (e.g., dedifferentiation of lesions), thereby enabling the timely adjustment of treatment plans [49].
According to 2020 ESMO Clinical Practice Guidelines for the diagnosis, treatment, and follow-up in GEP NENs, it is recommended that whole-body SSTR imaging should be part of the tumor staging, preoperative imaging, and restaging. 68Ga-SSTR PET/CT is recommended, but if it is not available, then SSTR SPECT can be used, although it is much less sensitive. MRI is preferable to CT to detect hepatic, pancreatic, brain, and bone lesions, whereas CT should be used to evaluate lung lesions. The use of 18F-FDG-PET is optional in GEP-NENs and should be used individually by balancing potential advantages with costs [7].
Although the definite role of combining 68Ga SSTR and 18F-FDG PET in NENs has not been established, several studies have shown favorable results by using both studies in the diagnosis, as well as prognosis and outcome prediction.
Kayani et al. [50] revealed in a retrospective study in 38 NETs patients that the sensitivity for the detection of tumors of 68Ga-DOTATATE PET/CT alone was 82%, and that of 18F-FDG PET/CT alone was 66%, while the sensitivity of combined dual PET/CT was more than 90%. The 68Ga-DOTATATE uptake was significantly higher than 18F-FDG in low-grade NENs (SUV 29 vs. 2.9, p < 0.001), whereas there was significantly higher 18F-FDG uptake in high-grade NENs over 68Ga-DOTATATE (SUV 11.7 vs. 4.4). A significant correlation with prominent tumor uptake of 68Ga-DOTATATE or 18F-FDG and tumor grade on histology was reported. In a systematic review of Muffatti et al. [51], they also showed similar results at initial diagnosis and restaging after treatment. The sensitivity of 68Ga DOTA-peptide PET and 18F-FDG PET was 93% and 58%, respectively, whereas the sensitivity of dual tracer PET was higher with the level of 94%.
The 68Ga-DOTA-peptide PET/CT is an advantage imaging modality for NENs and seems to surpass 18F-FDG for imaging well-differentiated NETs. However, functional imaging with dual PET tracer has the ability for more complete tumor evaluation, especially in intermediate- and high-grade tumors (Figure 3).
The study of Naswa N et al. suggested the combined using dual PET tracer because SSTR PET/CT overcomes the low sensitivity of 18F-FDG PET/CT, and 18F-FDG PET/CT overcomes the low specificity of SSTR PET/CT when the lesions do not concentrate 68Ga-DOTA-NOC. Moreover, the combined dual PET studies could determine the appropriate treatment regimen in NENs patients (octreotide therapy or conventional chemotherapy) [52].
The concept of combining the two PET tracers can be based on cell differentiation, and the SUVmax of both PET/CT studies in the context of Ki-67. Zhang et al. [53] revealed a negative correlation between the SUVmax of 68Ga-DOTATATE and Ki-67 (r = −0.415; p ≤ 0.001; in addition, a positive correlation between Ki-67 and SUVmax of 18F-FDG was reported (r = 0.683; p ≤ 0.001). They suggested that 68Ga-DOTATATE PET should be performed in patients with well-differentiated NETs, and 18F-FDG would be appropriate for patients with Ki-67 ≥ 10%.
A low SUVmax of 68Ga-DOTA-peptide PET is associated with poor prognosis for PFS and OS in NETs patients. In well-differentiated NETs, there is a better prognosis compared with other grades of NETs. The SUVmax of 68Ga-DOTA-peptide PET can be used as a prognosis predictor [54]. Lee et al. reported that a low SUVmax on SSTR PET/CT independently predicts early failure with SSA monotherapy in well-differentiated grade 1/2 GEP-NET. The SUVmax of SSTR PET predicted treatment failure with sensitivity of 39% and specificity of 98%. A SSTR SUVmax below 18.35 was associated with a shorter PFS. In multivariate analysis, a low SSTR SUVmax was the only predictor of early treatment failure with a hazard ratio (HR) of 6.85. Using SSTR PET/CT has a very high specificity in identified patients who do not receive the benefit from SSA treatment [55]. However, in high-grade NETs, it is also necessary to evaluate with 68Ga-DOTA-peptide PET prior to PRRT treatment, due to evidence that high-grade NETs patients were positive on both 68Ga-DOTA-peptide and 18F FDG PET. You et al. revealed that all patients with high-grade poorly differentiated NETs had positive SSTR PET/CT, and their findings were not different from FDG PET/CT in identifying >10 lesions or distant disease [56].
In the study of Cingarlini et al. [57], the SUVmax of 68Ga-DOTA-TOC was significantly higher than the SUVmax of 18F-FDG in grade 1/2 pancreatic NET; however, the SUVmax of 18F-FDG was higher in G2 than G1. Similarly, Chen et al. [58] reported that SUVs from 18F-FDG PET patients with G2/G3 NET were significantly higher than in patients with G1, which is lower than in the latter subgroup.
The study by Nilica et al. [59] revealed that the variation of SUVmax in FDG PET study between baseline and follow-up period was associated with prognosis. When SUVmax was constant or slightly changed, it was associated with a good prognosis. An increased SUVmax of an FDG value above 40% is associated with a poor prognosis.
The FDG PET assessment is also useful to predict treatment response to 177Lu-PRRT in NETs patients. Severi et al. suggested that no patients with FDG-PET-negative had disease progression at the first follow-up after 177Lu-PRRT. Grade 2 NET with FDG PET-positive (SUV cutoff >2.5) were often correlated with more aggressive tumors. PET-positive lesions with grade 2 NET (about 32%) did not respond to PRRT alone, so they may benefit from a more intensive treatment strategy, such as combined PRRT and chemotherapy [41].
The results of Panagoitidis revealed a change in the treatment plan in 84 NETs patients (80.8%). In 22 patients (21.1%), the decision was changed on the basis of 18F-FDG results; in 32 (30.8%), on both scan results; and in 50 (48.1%), on the 68Ga-DOTATATE results only. The OS declined rapidly with an increase in grade (p = 0.001) at approximately 91 months for G1, 59 months for G2, and 48 months for G3 [60].
FDG PET should be performed during follow-up, especially if there are signs of progression on other imaging modalities. Nilica et al. found that patients who were FDG-negative at baseline but FDG-positive during follow-up after treatment strongly correlate with an increased risk of progression. Then, patients might develop 18F-FDG-positive lesions during follow-up. These findings support the advantage of routinely repeating 18F-FDG PET in the long-term follow-up of NENs [59]. Binderup et al. revealed that, during the one-year follow-up, 14 patients died. Thirteen of the 57 (23%) FDG-PET-positive patients died compared to 1 in 41 (2%) FDG-PET-negative patients. A positive FDG-PET finding was correlated with significantly higher risk of death with HR of 10.3. The FDG-PET-positive cases had significantly lower PFS compared with the FDG-PET-negative cases with HR of 9.4. The only predictor of PFS was SUVmax of >3, with an HR of 8.4 [61].
Based on the current review study of Evangelista et al., they also suggest that combined 68Ga-DOTA peptides and 18F-FDG should be regarded as the initial imaging method for clinically suspicious NET or NET of unknown primary origin [62]. The retrospective study of Partelli et al. of 49 patients with a cytologically and/or histologically proven diagnosis of pancreatic NET (pNET) who performed both 68Ga-DOTANOC and 18F-FDG PET/CT on the same day reported that the dual-tracer PET/CT imaging approach resulted in a sensitivity of 100% for the diagnosis of pNET.
In spite of the fact that dual-tracer PET/CT revealed different tumor grading in the same lesions, it was not reported as having a significant impact on the treatment decision, except in NENs with Ki-67 > 10 [63].
Table 4 the role of SSTR PET and FDG PET in GEP-NENs.

2. Dual PET Imaging Grading System

2.1. The NETPET Score

In 2017, Chan et al. developed a scoring system known as the “NETPET score”, whereby results from the combined approach using 68Ga-DOTA-peptides and 18F-FDG PET in the workup of patients with metastatic NENs were incorporated into a 0–5 category scale (P0–P5) [47]. According to this system, P1 indicates purely SSTRs-avid disease without 18F-FDG activity in any lesion, P5 indicates the presence of significant 18F-FDG-positive and no SSTRs-positive lesion, P0 indicates a normal scan on both 18F-FDG and 68Ga-DOTA-peptide PET, and P2–P4 indicate patients with lesions aviding both tracers with different intensity (as the score increases from 2 to 4, 18F-FDG uptake increases and 68Ga-DOTA-peptide uptake decreases). They developed this PET-based scoring system by retrospectively reviewing 62 pathologically proven metastatic NET patients (mostly GEP NENs), who performed both 18F-FDG and 68Ga-DOTATATE PET/CT studies within 31 days. The authors visually compared uptake of the 68Ga-DOTATATE PET and FDG PET by using the SUVmax scale from 0 to 15 for 68Ga-DOTATATE and 0 to 7 for FDG PET as the values used for reporting in clinical practice. This study revealed that the OS was significantly correlated with the NETPET score (p = 0.0018). SSTR PET SUVmax might predict the response to PRRT, even though FDG-positive/SSTR-negative lesions may respond to PRRT, as well. This study suggested that subjects with NET-PET scores of P4 and P5 (significant FDG-positive/negative SSTR disease) might not obtain adequate tumor control from only PRRT and should receive systemic chemotherapy alternately, and this hypothesis should be researched in further studies.
In 2021, Chan et al. also revealed comparable results in a retrospective multicenter study (2011–2018) of 38 patients with bronchial NENs who had both 18F-FDG and 68Ga-DOTATATE PET within 60 days of each other. The NETPET scores and histology were significantly correlated with OS in univariate analyses (p = 0.003 and p = 0.01, respectively). In the multivariate analysis, only the NETPET score showed significance (p = 0.03). The NETPET scores were significantly correlated with histological grade (p = 0.006, chi-squared test). They concluded that NETPET score is a prognostic biomarker in bronchial NENs, as well as GEP NENs. Whilst it needs to be validated in prospective studies, it holds significant importance as a biomarker for a wide variety of NENs [64].
The NETPET scoring system is quite interesting because the results of both scans are summarized in a single parameter. Because it is a retrospective analysis, therefore, it is not possible to analyze the reasons leading to imaging with the dual tracer, type of treatment received, and the impact on the result obtained. Therefore, these findings require prospective validation. Although at present it might be too early to use NETPET score in routine clinical practice, the current studies have suggested several points for future research. Beyond confirmatory prospective studies, the benefit of dual PET imaging should be investigated in changing management decisions based on the high cost of imaging, and also of treatment [65].

2.2. Three-Scale Grading System

In 2020, Karfis et al. [66] developed a PET-based scoring scheme in a retrospective study of 85 metastatic GEP-NENs patients that included information from SSTR PET and FDG PET and was undertaken within a period of 3 months of each other. They grouped the study population into three different imaging groups (C1, all lesions were FDG negative/SSTR positive; C2, one or more lesions were FDG positive, while all of them were SSTR positive; and C3, one or more lesions were FDG positive lesions, while at least one of them was SSTR negative). They compared the prognostic value of this PET-based grading scheme with histological grade based on Ki-67. The histological grade showed no statistically significant difference in median PFS between G1 and G2 (p = 0.34) but was significant between G2 and G3 (p < 0.001), whereas the median PFS was significantly higher in C1 compared to C2 and in C2 compared to C3. They concluded that the combined 68Ga-DOTATATE/18F-FDG PET imaging stratification represents the accurate phenotype of GEP-NENs at any time of disease and has a high prognostic value when compared to histological grade classification. Its value as a prognostic imaging biomarker would be further corroborated within prospective study and multicenter testing to establish inter-rater reliability.
Although the new PET-based scoring system currently requires further validation with prospective studies including a large number of patients, the study also supports the use of the dual PET/CT imaging approach as a biomarker in prognostication and treatment selection, especially in patients with GEP-NETs.

3. Economic Benefits and Cost-Effectiveness

Currently, GEP-NENs represent a potentially important healthcare burden because they are usually diagnosed incidentally or when the patient has symptoms associated with hormone overproduction. However, GEP-NENs are heterogeneous tumors with non-specific symptoms, resulting in extensive healthcare use and delayed diagnosis affecting survival. In a global online survey of 1928 NENs patients, the average time reported from onset of the first symptom to diagnosis was 52 months, with nearly 30% of responders reporting waiting 5 years or more for an officially NENs diagnosis [67].
The previous study of Shen C. et al. [68] identified 9319 elderly patients diagnosed with NENs (mostly GEP-NENs) between 2003 and 2011, based on the Surveillance, Epidemiology and End Results (SEER) Medicare. They examined the patients’ conditions that may be related to NENs, resource use, and expense during the year prior to diagnosis. They reported that NENs patients tend to be diagnosed with various conditions, such as hypertension (64% vs. 53%), abdominal pain (22% vs. 8%), heart failure (12% vs. 8%), diarrhea (6% vs. 2%), peripheral edema (5% vs. 4%), and irritable bowel syndrome (1% vs. 0.5%), as compared to the non-cancerous control group. They also had much higher resource usage, including the average number of outpatient visits (22 vs. 17), percentage of ER visits (21% vs. 12%), and hospitalizations (28% vs. 17%). Similarly, NEN patients had significantly higher average total ($14,600 vs. $9500), outpatient ($6000 vs. $4200), and inpatient costs ($8600 vs. $5200). This study found that NEN patients require higher use of resources and costs. This information can inform the allocation of resources tailored to NENs needs.
Nowadays, it is accepted that PET/CT can provide value for money in the management of patients with NENs by using a more precise staging capability as compared to conventional imaging. This can result in cost reduction by preventing unnecessary or ineffective treatment and the associated side effects. PET/CT can predict outcomes and helps the clinician to identify patients who require more or less aggressive treatment, thereby improving outcomes. PET/CT reduces the likelihood of pretreatment progression, thus leading to more appropriate treatment, and it may increase the chance of disease cure. The previous study of Froelich MF et al. [69] analyzed the cost-effectiveness of 68Ga-DOTA-TATE PET/CT compared to 111In-pentetreotide SPECT/CT and to CT alone in NENs detection. In the base-case investigation, 68Ga-DOTA-TATE PET/CT ended up with a total cost of $88,000, while CT ended up with a total cost of approximately $89,000. SPECT/CT ended up with a total cost of approximately $90,000. Thus, CT and SPECT/CT were occupied by 68Ga-DOTA-TATE PET/CT. The use of PET/CT remains a cost-effective strategy. This is due to the reduced cost of treatment associated with timely detection. Additional costs of 68Ga-DOTA-TATE PET/CT compared to CT alone are reasonable in terms of treatment cost-saving and better outcome. The previous study of Schreiter et al. [70] revealed comparable results in 51 GEP-NENs patients who underwent 68Ga-DOTATOC PET/CT (n = 29) or 111In-DTPA-octreotide (n = 22). The results revealed 68Ga-DOTATOC PET/CT provided total costs (cost of equipment, personnel, and material) of approximately 550€, while the total cost of 111In-DTPA-octreotide is approximately 830€. They concluded that 68Ga-DOTATOC PET/CT was considerably cheaper than 111In-DTPA-octreotide in both material and personnel costs. In addition, using 68Ga-DOTATOC PET/CT may also have the potential to reduce the need for additional investigations, such as CT or MRI, thereby reducing the consequential costs.
Despite a large amount of works in the published literature in the NENs area, there is a lack of economic research data and specific comparative results relevant to 18F-FDG PET imaging in NENs.

4. Conclusions

Dual-tracer imaging with 68Ga-DOTA-peptide and 18F-FDG PET/CT appears to be a reasonable alternative to tissue sampling, because of the ability to reflect two different aspects of tumor biology, including SSTR expression and glucose metabolism. Therefore, imaging with dual tracers is recommended for well-differentiated GEP-NENs with Ki-67 ≥ 10%, providing information for treatment selection of SSA, PRRT, and chemotherapy. The combined dual-tracer PET is useful for precise clinical management and also shows a clear linear correlation between SUVmax and Ki-67 index.
With references to the heterogeneous expression and complementary findings to histopathology, we suggest that dual tracer PET should be permitted in the evaluation of patients at the time of diagnosis and during post-treatment follow-up in biological behavior assessment and prognosis. Although there is still very limited evidence to support the routine use of combined 68Ga-DOTA-peptide and 18F-FDG PET in the initial workup, dual-tracer PET might play a key role in the characterization of GEP-NENs patients, especially in patients affected with advanced tumors with a high disease burden. They should be performed both in the initial staging and during follow-up. The actual value of a combined PET approach is that it demonstrates the heterogeneity of GEP-NENs in both low-grade and advanced tumors. A single-core biopsy may not accurately evaluate the complexity and the heterogeneity of GEP-NENs, and, in some cases, it may underestimate the tumor grade. In addition, PET/CT is considered the most economical approach for diagnosis and treatment planning of GEP-NENs.

Author Contributions

Conceptualization, C.K. and S.V.; methodology, C.K.; formal analysis, C.K.; investigation, C.K.; resources, C.K. and S.V.; data curation, C.K.; writing—original draft preparation, C.K.; writing—review and editing, C.K. and S.V.; visualization, S.V.; supervision, S.V.; project administration, C.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dasari, A.; Shen, C.; Halperin, D.; Zhao, B.; Zhou, S.; Xu, Y.; Shih, T.; Yao, J.C. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients with Neuroendocrine Tumors in the United States. JAMA Oncol. 2017, 3, 1335–1342. [Google Scholar] [CrossRef] [PubMed]
  2. Mocellin, S.; Nitti, D. Gastrointestinal carcinoid: Epidemiological and survival evidence from a large population-based study (n = 25,531). Ann. Oncol. 2013, 24, 3040–3044. [Google Scholar] [CrossRef] [PubMed]
  3. Tsikitis, V.L.; Wertheim, B.C.; Guerrero, M.A. Trends of incidence and survival of gastrointestinal neuroendocrine tumors in the United States: A seer analysis. J. Cancer 2012, 3, 292–302. [Google Scholar] [CrossRef] [PubMed]
  4. Eychenne, R.; Bouvry, C.; Bourgeois, M.; Loyer, P.; Benoist, E.; Lepareur, N. Overview of Radiolabeled Somatostatin Analogs for Cancer Imaging and Therapy. Molecules 2020, 25, 4012. [Google Scholar] [CrossRef] [PubMed]
  5. Reubi, J.C.; Waser, B. Concomitant expression of several peptide receptors in neuroendocrine tumours: Molecular basis for in vivo multireceptor tumour targeting. Eur. J. Nucl. Med. Mol. Imaging 2003, 30, 781–793. [Google Scholar] [CrossRef]
  6. Nagtegaal, I.D.; Odze, R.D.; Klimstra, D.; Paradis, V.; Rugge, M.; Schirmacher, P.; Washington, K.M.; Carneiro, F.; Cree, I.A. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020, 76, 182–188. [Google Scholar] [CrossRef] [Green Version]
  7. Pavel, M.; Öberg, K.; Falconi, M.; Krenning, E.P.; Sundin, A.; Perren, A.; Berruti, A. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Pract. Guidel. Ann. Oncol. 2020, 31, 844–860. [Google Scholar] [CrossRef]
  8. Söreide, J.A.; van Heerden, J.A.; Thompson, G.B.; Schleck, C.; Ilstrup, D.M.; Churchward, M. Gastrointestinal carcinoid tumors: Long-term prognosis for surgically treated patients. World J. Surg. 2000, 24, 1431–1436. [Google Scholar] [CrossRef]
  9. Ezziddin, S.; Logvinski, T.; Yong-Hing, C.; Ahmadzadehfar, H.; Fischer, H.P.; Palmedo, H.; Bucerius, J.; Reinhardt, M.J.; Biersack, H.J. Factors predicting tracer uptake in somatostatin receptor and MIBG scintigraphy of metastatic gastroenteropancreatic neuroendocrine tumors. J. Nucl. Med. 2006, 47, 223–233. [Google Scholar]
  10. Dromain, C.; Déandréis, D.; Scoazec, J.Y.; Goere, D.; Ducreux, M.; Baudin, E.; Tselikas, L. Imaging of neuroendocrine tumors of the pancreas. Diagn. Interv. Imaging 2016, 97, 1241–1257. [Google Scholar] [CrossRef]
  11. Bushnell, D.L.; Baum, R.P. Standard Imaging Techniques for Neuroendocrine Tumors. Endocrinol. Metab. Clin. N. Am. 2011, 40, 153–162. [Google Scholar] [CrossRef] [PubMed]
  12. Poltorak-Szymczak, G.; Budlewski, T.; Furmanek, M.I.; Wierzba, W.; Sklinda, K.; Walecki, J.; Mruk, B. Radiological Imaging of Gastro Entero-Pancreatic Neuroendocrine Tumors. The Review of Current Literature Emphasizing the Diagnostic Value of Chosen Imaging Methods. Front. Oncol. 2021, 11, 670233. [Google Scholar] [CrossRef] [PubMed]
  13. Sundin, A.; Arnold, R.; Baudin, E.; Cwikla, J.B.; Eriksson, B.; Fanti, S.; Fazio, N.; Giammarile, F.; Hicks, R.J.; Kjaer, A.; et al. Antibes Consensus Conference Participants. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Radiological. Nucl. Med. Hybrid. Imaging Neuroendocrinol. 2017, 105, 212–244. [Google Scholar]
  14. Lee, L.; Ito, T.; Jensen, R.T. Imaging of Pancreatic Neuroendocrine Tumors: Recent Advances, Current Status, and Controversies. Expert Rev. Anticancer Ther. 2018, 18, 837–880. [Google Scholar] [CrossRef] [PubMed]
  15. Sundin, A.; Wills, M.; Rockall, A. Radiological Imaging: Computed Tomography, Magnetic Resonance Imaging and Ultrasonography. Front. Horm. Res. 2015, 44, 58–72. [Google Scholar] [PubMed]
  16. Schraml, C.; Schwenzer, N.F.; Sperling, O.; Aschoff, P.; Lichy, M.P.; Müller, M.; Brendle, C.; Werner, M.K.; Claussen, C.D.; Pfannenberg, C. Staging of neuroendocrine tumours: Comparison of [68Ga]DOTATOC multiphase PET/CT and whole-body MRI. Cancer Imaging 2013, 13, 63–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Cives, M.; Strosberg, J.R. Gastroenteropancreatic Neuroendocrine Tumors. CA Cancer J. Clin. 2018, 68, 471–487. [Google Scholar] [CrossRef]
  18. Bombardieri, E.; Aktolun, C.; Baum, R.P.; Bishof-Delaloye, A.; Buscombe, J.; Chatal, J.F.; Maffioli, L.; Moncayo, R.; Mortelmans, L.; Reske, S.N. Oncology Committee of the EANM 111In-pentetreotide scintigraphy; procedure guidelines for tumor imaging. Eur. J. Nucl. Med. Mol. Imaging 2003, 30, 140–147. [Google Scholar]
  19. Kanzaki, T.; Takahashi, Y.; Higuchi, T.; Zhang, X.; Mogi, N.; Suto, T.; Tsushima, Y. Evaluation of a Correction Method for 111In-Pentetreotide SPECT Imaging of Gastroenteropancreatic Neuroendocrine Tumors. J. Nucl. Med. Technol. 2020, 48, 326–330. [Google Scholar] [CrossRef]
  20. Schillaci, O.; Danieli, R.; Manni, C.; Simonetti, G. Is SPECT/CT with a hybrid camera useful to improve scintigraphic imaging interpretation? Nucl. Med. Commun. 2004, 25, 705–710. [Google Scholar] [CrossRef] [Green Version]
  21. Schillaci, O.; Spanu, A.; Scopinaro, F.; Falchi, A.; Danieli, R.; Marongiu, P.; Pisu, N.; Madeddu, G.; Delle Fave, G.; Madeddu, G. Somatostatin receptor scintigraphy in liver metastasis detection from gastroenteropancreatic neuroendocrine tumors. J. Nucl. Med. 2003, 44, 359–368. [Google Scholar]
  22. Castaldi, P.; Rufini, V.; Treglia, G.; Bruno, I.; Perotti, G.; Stifano, G.; Barbaro, B.; Giordano, A. Impact of 111In-DTPA-octreotide SPECT/CT fusion images in the management of neuroendocrine tumours. Radiol. Med. 2008, 113, 1056–1067. [Google Scholar] [CrossRef] [PubMed]
  23. Hu, Y.; Ye, Z.; Wang, F.; Qin, Y.; Xu, X.; Yu, X.; Ji, S. Role of Somatostatin Receptor in Pancreatic Neuroendocrine Tumor Development, Diagnosis, and Therapy. Front. Endocrinol. 2021, 12, 679000. [Google Scholar] [CrossRef] [PubMed]
  24. Spanu, A.; Schillaci, O.; Piras, B.; Calvisi, D.F.; Falchi, A.; Danieli, R.; Nuvoli, S.; Dore, F.; Madeddu, G. Non-functioning gastroenteropancreatic (GEP) tumors: A 111 In-Pentetreotide SPECT/CT diagnostic study. Am. J. Nucl. Med. Mol. Imaging 2017, 7, 181–194. [Google Scholar]
  25. Kwekkeboom, D.J.; Kam, B.L.; van Essen, M.; Teunissen, J.J.; van Eijck, C.H.; Valkema, R.; De Jong, M.; de Herder, W.W.; Krenning, E.P. Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr. Relat. Cancer 2010, 17, 53–73. [Google Scholar] [CrossRef] [Green Version]
  26. Reubi, J.C. Peptide receptors as molecular targets for cancer diagnosis and therapy. Endocr. Rev. 2003, 24, 389–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Oberg, K.; Kvols, L.; Caplin, M.; Fave, G.D.; Herder, D.W.; Rindi, G.; Ruszniewski, P.; Woltering, E.A.; Wiedenmann, B. Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann. Oncol. 2004, 15, 966–973. [Google Scholar] [CrossRef]
  28. Virgolini, I.; Ambrosini, V.; Bomanji, J.B.; Baum, R.P.; Fanti, S.; Gabriel, M.; Papathanasiou, N.D.; Pepe, G.; Oyen, W.; De Cristoforo, C.; et al. Procedure guidelines for PET/CT tumour imaging with 68Ga-DOTAconjugated peptides: 68Ga-DOTA-TOC, 68Ga-DOTA-NOC, 68Ga-DOTATATE. Eur. J. Nucl. Med. Mol. Imaging 2010, 37, 2004–2010. [Google Scholar] [CrossRef]
  29. Ambrosini, V.; Campana, D.; Bodei, L.; Nanni, C.; Castellucci, P.; Allegri, V.; Montini, G.C.; Tomassetti, P.; Paganelli, G.; Fanti, S. 68Ga-DOTANOC PET/CT clinical impact in patients with neuroendocrine tumors. J. Nucl. Med. 2010, 51, 669–673. [Google Scholar] [CrossRef] [Green Version]
  30. Mojtahedi, A.; Thamake, S.; Tworowska, I.; Ranganathan, D.; Delpassand, E.S. The value of 68Ga-DOTATATE PET/CT in diagnosis and management of neuroendocrine tumors compared to current FDA approved imaging modalities: A review of literature. Am. J. Nucl. Med. Mol. Imaging 2014, 4, 426–434. [Google Scholar]
  31. Vinjamuri, S.; Gilbert, T.M.; Banks, M.; McKane, G.; Maltby, P.; Poston, G.; Weissman, H.; Palmer, D.H.; Vora, J.; Pritchard, D.M.; et al. Peptide receptor radionuclide therapy with 90Y-DOTATATE/90Y-DOTATOC in patients with progressive metastatic neuroendocrine tumours: Assessment of response, survival and toxicity. Br. J. Cancer 2013, 108, 1440–1448. [Google Scholar] [CrossRef]
  32. Garin, E.; Le Jeune, F.; Devillers, A.; Cuggia, M.; de Lajarte-Thirouard, A.S.; Bouriel, C.; Boucher, E.; Raoul, J.L. Predictive value of 18F-FDG PET and somatostatin receptor scintigraphy in patients with metastatic endocrine tumors. J. Nucl. Med. 2009, 50, 858–864. [Google Scholar] [CrossRef] [Green Version]
  33. Kaewput, C.; Suppiah, S.; Vinjamuri, S. Correlation between Standardized Uptake Value of 68Ga-DOTA-NOC Positron Emission Tomography/Computed Tomography and Pathological Classification of Neuroendocrine Tumor. World J. Nucl. Med. 2018, 17, 34–40. [Google Scholar] [CrossRef]
  34. Efficacy of Galium-68 DOTATATE PET/CT in the Detection of Metastasis Rate of Well-Differentiated Gastroenteropancreatic Neuroendocrine Tumors. Asia Ocean J. Nucl. Med. Biol. Spring 2019, 7, 141–148.
  35. Bozkurt, M.F.; Virgolini, I.; Balogova, S.; Beheshti, M.; Rubello, D.; Decristoforo, C.; Ambrosini, V.; Kjaer, A.; Delgado-Bolton, R.; Kunikowska, J.; et al. Guideline for PET/CT imaging of neuroendocrine neoplasms with 68Ga-DOTA-conjugated somatostatin receptor targeting peptides and 18F-DOPA. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 1588–1601. [Google Scholar] [CrossRef]
  36. Deppen, S.A.; Blume, J.; Bobbey, A.J.; Shah, C.; Graham, M.M.; Lee, P.; Delbeke, D.; Walker, R.C. 68Ga-DOTATATE compared with 111InDTPA-octreotide and conventional imaging for pulmonary and gastroenteropancreatic neuroendocrine tumors: A systematic review and meta-analysis. J. Nucl. Med. 2016, 57, 872–878. [Google Scholar] [CrossRef] [Green Version]
  37. Sadowski, S.M.; Neychev, V.; Millo, C.; Shih, J.; Nilubol, N.; Herscovitch, P.; Pacak, K.; Marx, S.J.; Kebebew, E. Prospective study of 68Ga-DOTATATE positron emission tomography/computed tomography for detecting gastroenteropancreatic neuroendocrine tumors and unknown primary sites. J. Clin. Oncol. 2016, 34, 588–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Binnebeek, S.V.; Vanbilloen, B.; Baete, K.; Terwinghe, C.; Koole, M.; Mottaghy, F.M.; Clement, P.M.; Mortelmans, L.; Bogaerts, K.; Haustermans, K.; et al. Comparison of diagnostic accuracy of 111In-pentetreotide SPECT and 68Ga-DOTATOC PET/CT: A lesion-by-lesion analysis in patients with metastatic neuroendocrine tumours. Eur. Radiol. Vol. 2016, 26, 900–909. [Google Scholar] [CrossRef] [PubMed]
  39. Barrio, M.; Czernin, J.; Fanti, S.; Ambrosini, V.; Binse, I.; Du, L.; Eiber, M.; Herrmann, K.; Fendler, W.P. The Impact of Somatostatin Receptor-Directed PET/CT on the Management of Patients With Neuroendocrine Tumor: A Systematic Review and Meta-Analysis. J. Nucl. Med. 2017, 58, 756–761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Belhocine, T.; Foidart, J.; Rigo, P.; Najjar, F.; Thiry, A.; Quatresooz, P.; Hustinx, R. Fluorodeoxyglucose positron emission tomography and somatostatin receptor scintigraphy for diagnosing and staging carcinoid tumours: Correlations with the pathological indexes p53 and Ki-67. Nucl. Med. Commun. 2002, 23, 727–734. [Google Scholar] [CrossRef] [PubMed]
  41. Severi, S.; Nanni, O.; Bodei, L.; Sansovini, M.; Ianniello, A.; Nicoletti, S.; Scarpi, E.; Matteucci, F.; Gilardi, L.; Paganelli, G. Role of 18FDG PET/CT in patients treated with 177Lu DOTATATE for advanced differentiated neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 881. [Google Scholar] [CrossRef] [PubMed]
  42. Delpassand, E.S.; Samarghandi, A.; Mourtada, J.S.; Zamanian, S.; Espenan, G.D.; Sharif, R.; MacKenzie, S.; Kosari, K.; Barakat, O.; Naqvi, S.; et al. Long Term Survival, Toxicity Profile, and role of 18F-FDG PET/CT scan in Patients with Progressive Neuroendocrine Tumors Following Peptide Receptor Radionuclide Therapy with High Activity In 111 Pentetreotide. Theranostics 2012, 2, 472–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Panagiotidis, E.; Bomanji, J. Role of 18F-fluorodeoxyglucose PET in the study of neuroendocrine tumors. PET Clin. 2014, 9, 43–55. [Google Scholar] [CrossRef] [PubMed]
  44. Bahri, H.; Laurence, L.; Edeline, J.; Leghzali, H.; Devillers, A.; Raoul, J.L.; Cuggia, M.; Mesbah, H.; Clement, B.; Boucher, E.; et al. High prognostic value of 18F-FDG PET for metastatic gastroenteropancreatic neuroendocrine tumors: A long-term evaluation. J. Nucl. Med. 2014, 55, 1786–1790. [Google Scholar] [CrossRef] [Green Version]
  45. Ezziddin, S.; Adler, L.; Sabet, A.; Pöppel, T.D.; Grabellus, F.; Yüce, A.; Fischer, H.P.; Simon, B.; Höller, T.; Biersack, H.J.; et al. Prognostic stratification of metastatic gastroenteropancreatic neuroendocrine neoplasms by 18F-FDG PET: Feasibility of a metabolic grading system. J. Nucl. Med. 2014, 55, 1260–1266. [Google Scholar] [CrossRef] [Green Version]
  46. Klimstra, D.S.; Modlin, I.R.; Coppola, D.; Lloyd, R.V.; Suster, S. The pathologic classification of neuroendocrine tumors: A review of nomenclature, grading, and staging systems. Pancreas 2010, 39, 707–712. [Google Scholar] [CrossRef]
  47. Chan, D.L.; Pavlakis, N.; Schembri, G.P.; Bernard, E.J.; Hsiao, E.; Hayes, A.; Barnes, T.; Diakos, C.; Khasraw, M.; Samra, J.; et al. Dual somatostatin receptor/FDG PET/CT Imaging in metastatic neuroendocrine tumours: Proposal for a novel grading scheme with prognostic significance. Theranostics 2017, 7, 1149–1158. [Google Scholar] [CrossRef] [PubMed]
  48. Deroose, C.M.; Hindié, E.; Kebebew, E.; Goichot, B.; Pacak, K.; Taïeb, D.; Imperiale, A. Molecular imaging of gastroenteropancreatic neuroendocrine tumors: Current status and future directions. J. Nucl. Med. 2016, 57, 1949–1956. [Google Scholar] [CrossRef] [Green Version]
  49. Papadakis, G.Z.; Karantanas, A.H.; Marias, K.; Millo, C. Current status and future prospects of PET-imaging applications in patients with gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs). Eur. J. Radiol. 2021, 143, 109932. [Google Scholar] [CrossRef]
  50. Kayani, I.; Bomanji, J.B.; Groves, A.; Conway, G.; Gacinovic, S.; Win, T.; Dickson, J.; Caplin, M.; Ell, P.J. Functional imaging of neuroendocrine tumors with combined PET/CT using 68Ga-DOTATATE (DOTA DPhe1,Tyr3octreotate) and 18F-FDG. Cancer 2008, 112, 2447–2455. [Google Scholar] [CrossRef]
  51. Muffatti, F.; Partelli, S.; Cirocchi, R.; Andreasi, V.; Mapelli, P.; Picchio, M.; Gianolli, L.; Falconi, M. Combined 68Ga-DOTA-peptides and 18F-FDG PET in the diagnostic work-up of neuroendocrine neoplasms (NEN). Clin. Transl. Imaging 2019, 7, 181–188. [Google Scholar] [CrossRef]
  52. Naswa, N.; Sharma, P.; Gupta, S.K.; Karunanithi, S.; Reddy, R.M.; Patnecha, M.; Lata, S.; Kumar, R.; Malhotra, A.; Bal, C. Dual tracer functional imaging of gastroenteropancreatic neuroendocrine tumors using 68Ga-DOTA-NOC PET-CT and 18F-FDG PET-CT: Competitive or complimentary? Clin. Nucl. Med. 2014, 39, e27–e34. [Google Scholar] [CrossRef]
  53. Zhang, P.; Yu, J.; Li, J.; Shen, L.; Li, N.; Zhu, H.; Zhai, S.; Zhang, Y.; Yang, Z.; Lu, M. Clinical and Prognostic Value of PET/CT Imaging with Combination of 68Ga-DOTATATE and 18F-FDG in Gastroenteropancreatic Neuroendocrine Neoplasms. Contrast Media. Mol. Imaging 2018, 26, 2340389. [Google Scholar] [CrossRef] [Green Version]
  54. Lee, D.Y.; Kim, Y. Prognostic Value of Maximum Standardized Uptake Value in 68Ga-Somatostatin Receptor Positron Emission Tomography for Neuroendocrine Tumors. A Systematic Review and Meta-analysis. Clin. Nucl. Med. 2019, 44, 777–783. [Google Scholar] [CrossRef] [PubMed]
  55. Lee, H.; Eads, J.R.; Pryma, D.A. 68Ga-DOTATATE Positron Emission Tomography-Computed Tomography Quantification Predicts Response to Somatostatin Analog Therapy in Gastroenteropancreatic Neuroendocrine Tumors. Oncologist 2020, 25, 1–9. [Google Scholar] [CrossRef] [PubMed]
  56. You, H.; Kandathil, A.; Beg, M.; Blanche, L.; Kazmib, S.; Subramaniam, R.M. 68Ga-DOTATATE PET/CT and 18F-FDG PET/CT in the evaluation of low and intermediate versus high-grade neuroendocrine tumors. Nucl. Med. Commun. 2020, 41, 1060–1065. [Google Scholar] [CrossRef] [PubMed]
  57. Cingarlini, S.; Ortolani, S.; Salgarello, M.; Butturini, G.; Malpaga, A.; Malfatti, V.; D’Onofrio, M.; Davì, M.V.; Vallerio, P.; Ruzzenente, A.; et al. Role of Combined 68Ga-DOTATOC and 18F-FDG Positron Emission Tomography/Computed Tomography in the Diagnostic Workup of Pancreas Neuroendocrine Tumors Implications for Managing Surgical Decision. Pancreas 2017, 46, 42–47. [Google Scholar] [CrossRef] [PubMed]
  58. Chen, S.-H.; Chang, Y.-C.; Hwang, T.-L.; Chen, J.-S.; Chou, W.-C.; Hsieh, C.-H.; Yeh, T.-S.; Hsu, J.-T.; Yeh, C.-N.; Tseng, J.-H.; et al. 68Ga-DOTATOC and 18F-FDG PET/CT for identifying the primary lesions of suspected and metastatic neuroendocrine tumors: A prospective study in Taiwan. J. Med. Assoc. 2018, 117, 480–487. [Google Scholar] [CrossRef]
  59. Nilica, B.; Waitz, D.; Stevanovic, V.; Uprimny, C.; Kendler, D.; Buxbaum, S.; Warwitz, B.; Gerardo, L.; Henninger, B.; Virgolini, I.; et al. Direct comparison of 68Ga-DOTATOC and 18F-FDG PET/CT in the follow-up of patients with neuroendocrine tumour treated with the first full peptide receptor radionuclide therapy cycle. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 1585–1592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Panagiotidis, E.; Alshammari, A.; Michopoulou, S.; Skoura, E.; Naik, K.; Maragkoudakis, E.; Mohmaduvesh, M.; Al-Harbi, M.; Belda, M.; Caplin, M.E.; et al. Comparison of the impact of 68Ga-DOTATATE and 18F-FDG PET/CT on clinical management in patients with neuroendocrine tumors. J. Nucl. Med. 2018, 58, 91–96. [Google Scholar] [CrossRef] [Green Version]
  61. Binderup, T.; Knigge, U.; Loft, A.; Federspiel, B.; Kjaer, A. 18F-Fluorodeoxyglucose Positron Emission Tomography Predicts Survival of Patients with Neuroendocrine Tumors. Clin. Cancer Res. 2010, 16, 978–985. [Google Scholar] [CrossRef] [Green Version]
  62. Evangelista, L.; Ravelli, I.; Bignotto, A.; Cecchin, D.; Zucchetta, P. 68Ga-DOTA-peptides and 18F-FDG PET/CT in patients with neuroendocrine tumor: A review. Clin. Imaging 2020, 67, 113–116. [Google Scholar] [CrossRef]
  63. Partelli, S.; Rinzivillo, M.; Maurizi, A.; Panzuto, F.; Salgarello, M.; Polenta, V.; Delle Fave, G.; Falconi, M. The role of combined 68Ga-DOTANOC and 18FDG PET/CT in the management of patients with pancreatic neuroendocrine tumors. Neuroendocrinology 2014, 100, 293–299. [Google Scholar] [CrossRef]
  64. Chan, D.L.; Ulaner, G.A.; Pattison, D.; Wyld, D.; Ladwa, R.; Kirchner, J.; Li, B.T.; Lai, W.V.; Pavlakis, N.; Roach, P.J.; et al. Dual Positron Emission Tomography imaging in bronchial neuroendocrine neoplasms (NENs): The NETPET score as a prognostic biomarker. J. Nucl. Med. 2021, 62, 1278–1284. [Google Scholar] [CrossRef] [PubMed]
  65. Hindié, E. The NETPET Score: Combining FDG and Somatostatin Receptor Imaging for Optimal Management of Patients with Metastatic Well-Differentiated Neuroendocrine Tumors. Theranostics 2017, 7, 1159–1163. [Google Scholar] [CrossRef] [PubMed]
  66. Karfis, I.; Marin, G.; Levillain, H.; Drisis, S.; Muteganya, R.; Critchi, G.; Taraji-Schiltz, L.; Guix, C.A.; Shaza, L.; Elbachiri, M.; et al. Prognostic value of a three-scale grading system based on combining molecular imaging with 68Ga-DOTATATE and 18F-FDG PET/CT in patients with metastatic gastroenteropancreatic neuroendocrine neoplasias. Oncotarget 2020, 11, 589–599. [Google Scholar] [CrossRef] [Green Version]
  67. Singh, S.; Granberg, D.; Wolin, E.; Warner, R.; Sissons, M.; Kolarova, T.; Goldstein, G.; Pavel, M.; Öberg, K.; Leyden, J. Patient-Reported Burden of a Neuroendocrine Tumor (NET) Diagnosis: Results From the First Global Survey of Patients With NETs. J. Glob. Oncol. 2017, 3, 43–53. [Google Scholar] [CrossRef] [PubMed]
  68. Shen, C.; Dasari, A.; Xu, Y.; Zhou, S.; Gu, D.; Chu, Y.; Halperin, D.M.; Shih, Y.T.; Yao, J.C. Pre-existing Symptoms and Healthcare Utilization Prior to Diagnosis of Neuroendocrine Tumors: A SEER-Medicare Database Study. Sci. Rep. 2018, 8, 16863. [Google Scholar] [CrossRef] [PubMed]
  69. Froelich, M.F.; Schnitzer, M.L.; Holzgreve, A.; Gassert, F.G.; Gresser, E.; Overhoff, D.; Schwarze, V.; Fabritius, M.P.; Nörenberg, D.; von Münchhausen, N.; et al. Cost-Effectiveness Analysis of 68Ga DOTA-TATE PET/CT, 111In-Pentetreotide SPECT/CT and CT for Diagnostic Workup of Neuroendocrine Tumors. Diagnostics 2021, 11, 334. [Google Scholar] [CrossRef]
  70. Schreiter, N.F.; Brenner, W.; Nogami, M.; Buchert, R.; Huppertz, A.; Pape, U.; Prasad, V.; Hamm, B.; Maurer, M.H. Cost comparison of 111In-DTPA-octreotide scintigraphy and 68Ga-DOTATOC PET/CT for staging enteropancreatic neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2012, 39, 72–82. [Google Scholar] [CrossRef]
Figure 1. A 41-year-old male with recurrent NENs at the duodenum. 68Ga-DOTA-NOC PET/CT shows abnormal focal tracer uptake at the duodenum (SUVmax of 5.9), without other definite evidence of abnormal tracer uptake. Ki-67 index from endoscopic fine-needle aspiration of duodenum was 1%. It is possible that the FNA site may not fully reflect the true pathological grade of a patient with heterogeneity of cellular differentiation.
Figure 1. A 41-year-old male with recurrent NENs at the duodenum. 68Ga-DOTA-NOC PET/CT shows abnormal focal tracer uptake at the duodenum (SUVmax of 5.9), without other definite evidence of abnormal tracer uptake. Ki-67 index from endoscopic fine-needle aspiration of duodenum was 1%. It is possible that the FNA site may not fully reflect the true pathological grade of a patient with heterogeneity of cellular differentiation.
Diagnostics 12 00280 g001
Figure 2. A patient with pathological proven pancreatic NENs (Ki-67 of 7%). 68Ga-DOTA-NOC PET image ((A), arrow) reveals 3 foci increased radiotracer uptake at the pancreas and also seen in axial 68Ga-DOTA-NOC PET/CT image ((B), arrow). However, the axial 18F-FDG PET/CT image (C) shows no abnormal FDG uptake in the pancreas at the same level.
Figure 2. A patient with pathological proven pancreatic NENs (Ki-67 of 7%). 68Ga-DOTA-NOC PET image ((A), arrow) reveals 3 foci increased radiotracer uptake at the pancreas and also seen in axial 68Ga-DOTA-NOC PET/CT image ((B), arrow). However, the axial 18F-FDG PET/CT image (C) shows no abnormal FDG uptake in the pancreas at the same level.
Diagnostics 12 00280 g002
Figure 3. A patient with pathological proven pancreatic NENs (G3, Ki-67 of 30%). Axial 68Ga-DOTA-NOC PET/CT image ((A), arrow) reveals intense radiotracer uptake at the body of the pancreas and is also seen in axial 18F-FDG PET/CT image ((B), arrow). The value of combined PET studies is to demonstrate the heterogeneity of GEP-NENs, especially in high-grade NENs, and could determine the appropriate therapy.
Figure 3. A patient with pathological proven pancreatic NENs (G3, Ki-67 of 30%). Axial 68Ga-DOTA-NOC PET/CT image ((A), arrow) reveals intense radiotracer uptake at the body of the pancreas and is also seen in axial 18F-FDG PET/CT image ((B), arrow). The value of combined PET studies is to demonstrate the heterogeneity of GEP-NENs, especially in high-grade NENs, and could determine the appropriate therapy.
Diagnostics 12 00280 g003
Table 1. WHO 2019 classification for GEP-NENs [6].
Table 1. WHO 2019 classification for GEP-NENs [6].
TerminologyDifferentiationGradeMitotic Count (2 mm2) aKi-67 Index (%)
NET, G1Well differentiationG1<2<3
NET, G2G22–203–20
NET, G3G3>20>20
NECPoorly differentiationG3>20>20
  • Small-cell
  • Large-cell
MiNENWell or poorly differentiationVariableVariableVariable
MiNEN, mixed neuroendocrine–non-neuroendocrine neoplasm; a 10 HPF = 2 mm2, at least 40 fields evaluated in areas of highest mitotic density.
Table 2. Clinical presentation of GEP-NENs by site of origin and by hormone presentation [7].
Table 2. Clinical presentation of GEP-NENs by site of origin and by hormone presentation [7].
TypeFrequencySymptomsSecretory Product
Intestinal NENs50% of GEP-NENs
  • With carcinoid syndrome
20%FlushingProstaglandin, Tachykinin, substance P
DiarrheaSerotonin
Endocardial fibrosisSerotonin
WheezingHistamine, kinins, CgA
  • Without carcinoid syndrome
80%Unspecific abdominal painCgA
Pan-NENs30% of GEP-NENs
  • Functioning
10–30%Zollinger-Ellison syndromeGastrin
HypoglycemiaInsulin
Necrolytic erythema, HyperglycemiaGlucagon
WDHA syndromeVIP
Diabetes, gallstones, diarrheaSomatostatin
Cushing syndromeCRH, ACTH
AcromegalyGHRH, GH
HypercalcemiaPTHrP
FlushingCalcitonin
DiarrheaSerotonin, CgA
  • Non-functioning
70–90%Unspecific abdominal painCgA
Rarely jaundice (cholestasis and cholelithiasis)Pancreatic polypeptide
ACTH, adrenocorticotropic hormone; CgA, chromogranin; CRH, corticotropin-releasing hormone; PTHrP, parathyroid-hormone-related peptide; VIP, vasoactive intestinal peptide; WDHA syndrome, watery diarrhea–hypokalemia–achlorhydria syndrome.
Table 3. Expression of somatostatin receptors in different GEP-NENs (%).
Table 3. Expression of somatostatin receptors in different GEP-NENs (%).
Tumor TypesReceptor Subtypes
SSTR 1SSTR 2SSTR 3SSTR 4SSTR 5
All GEP-NENs6886469357
Gastrinoma3350178350
Insulinoma331003310067
Glucagonoma67100676767
VIPoma100100100100100
Mid-Gut NENs8095653575
Table 4. Role of SSTR PET and 18F FDG PET in GEP-NENs.
Table 4. Role of SSTR PET and 18F FDG PET in GEP-NENs.
RoleInformation
68Ga DOTA-peptide PET/CT
  • Diagnosis (initial staging)
  • Localize primary tumor and detect metastatic site
  • High sensitivity in the detection of well-differentiated and/or low-grade NENs (G1,G2)
  • Pretreatment evaluation
  • Determine SSTR status (positive SSTR patients are more likely to respond to targeted SSA treatment or PRRT)
  • Surveillance after treatment (re-staging)
  • Useful in monitoring patients with known disease in the detection of residual, recurrent, or progressive disease
  • Assessment of treatment response
  • Useful for monitoring response to therapy (surgery, radiotherapy, chemotherapy, and PRRT)
  • Prognosis
  • Well-differentiated NETs tend to have higher SUV on 68Ga-DOTA peptide PET with favorable prognosis, while lower SUV of 68Ga-DOTA-peptide PET may be associated with aggressive behavior and lead to poor prognosis
18F FDG PET/CT
  • Diagnosis (initial staging)
  • High sensitivity in the detection of poorly differentiated and/or high-grade NETs (NEC) or G3 with negative SSTR imaging
  • Pretreatment evaluation
  • High FDG uptake is recommended for chemotherapy after SSA therapy or PRRT
  • It is useful in delineating disease extent, particularly in aggressive and high-grade tumors
  • Surveillance after treatment/re-staging
  • Repeating 18F-FDG PET in the long-term follow-up, especially if signs of progression in other imaging methods are detected
  • Patient with FDG-negative initially but FDG-positive during follow-up after treatment strongly correlates with a higher risk of progression
  • Assessment of treatment response
  • Useful for predicting response after PRRT; FDG-PET-negative after treatment tends to have a good response, but PET-positive were frequently associated with more aggressive disease and did not respond to PRRT; they may benefit from more intensive treatment, for example, the combined chemotherapy
  • Prognosis
  • A positive FDG-PET result was associated with significantly higher mortality risk and significantly lower PFS compared to the FDG-PET-negative result
ombined 68Ga DOTA-peptides and 18F FDG PET/CT
  • Diagnosis (initial staging)
  • Should be considered in the patient with clinically suspected GEP-NENs or NET of unknown primary origin (regarding the heterogeneous expression and complementary findings to histopathology)
  • Pretreatment evaluation
  • Increased sensitivity in the detection of lesions as compared to only 68Ga-DOTA-peptide or FDG PET study alone
  • Assessment of treatment response
  • Combination of 2 studies can decide the mode of appropriate therapy (octreotide therapy or conventional chemotherapy)
  • Prognosis
  • Higher 68Ga-DOTA-peptide avidity and lower FDG avidity had favorable prognosis, while lower 68Ga-DOTA-peptide avidity and greater FDG avidity had poor prognosis
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kaewput, C.; Vinjamuri, S. Role of Combined 68Ga DOTA-Peptides and 18F FDG PET/CT in the Evaluation of Gastroenteropancreatic Neuroendocrine Neoplasms. Diagnostics 2022, 12, 280. https://doi.org/10.3390/diagnostics12020280

AMA Style

Kaewput C, Vinjamuri S. Role of Combined 68Ga DOTA-Peptides and 18F FDG PET/CT in the Evaluation of Gastroenteropancreatic Neuroendocrine Neoplasms. Diagnostics. 2022; 12(2):280. https://doi.org/10.3390/diagnostics12020280

Chicago/Turabian Style

Kaewput, Chalermrat, and Sobhan Vinjamuri. 2022. "Role of Combined 68Ga DOTA-Peptides and 18F FDG PET/CT in the Evaluation of Gastroenteropancreatic Neuroendocrine Neoplasms" Diagnostics 12, no. 2: 280. https://doi.org/10.3390/diagnostics12020280

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

Article Metrics

Back to TopTop