Next Article in Journal
Changes in Treatment Patterns and Costs for Lung Cancer Have Not Resulted in Relevant Improvements in Survival: A Population-Based Observational Study in Catalonia
Next Article in Special Issue
Defining MRI Superiority over CT for Colorectal and Neuroendocrine Liver Metastases
Previous Article in Journal
Caerin 1.1/1.9 Enhances Antitumour Immunity by Activating the IFN-α Response Signalling Pathway of Tumour Macrophages
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Clinical Guide to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE in Neuroendocrine Tumor Patients

by
Morticia N. Becx
1,*,
Noémie S. Minczeles
2,3,
Tessa Brabander
2,
Wouter W. de Herder
3,
Julie Nonnekens
1,2,† and
Johannes Hofland
3,†
1
Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
2
Department of Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
3
Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work and share the last authorship.
Cancers 2022, 14(23), 5792; https://doi.org/10.3390/cancers14235792
Submission received: 3 October 2022 / Revised: 18 November 2022 / Accepted: 21 November 2022 / Published: 24 November 2022
(This article belongs to the Special Issue Neuroendocrine Tumors: Treatment and Management Volume II)

Abstract

:

Simple Summary

Peptide receptor radionuclide therapy (PRRT) is one of the treatment options for locally advanced or metastatic gastroenteropancreatic (GEP) neuroendocrine tumors (NETs). This treatment makes use of radioactive labeled somatostatin analogues, with 177Lu-DOTATATE as its established standard. PRRT has positive effects in tumor control and it lowers the risk of disease progression or death. It also improves the quality of life of the patient. Unfortunately, important risk factors for a minority of patients include renal and hematological toxicities. NET is a rare disease and treating patients with PRRT requires clinical expertise. This guide gives an overview of the background of PRRT and the current results in NET patient care.

Abstract

Peptide receptor radionuclide therapy (PRRT) with [177Lu]Lu-[DOTA0,Tyr3]-octreotate (177Lu-DOTATATE) has become an established second- or third-line treatment option for patients with somatostatin receptor (SSTR)-positive advanced well-differentiated gastroenteropancreatic (GEP) neuroendocrine tumors (NETs). Clinical evidence of the efficacy of PRRT in tumor control has been proven and lower risks of disease progression or death are seen combined with an improved quality of life. When appropriate patient selection is performed, PRRT is accompanied by limited risks for renal and hematological toxicities. Treatment of NET patients with PRRT requires dedicated clinical expertise due to the biological characteristics of PRRT and specific characteristics of NET patients. This review provides an overview for clinicians dealing with NET on the history, molecular characteristics, efficacy, toxicity and relevant clinical specifics of PRRT.

1. Introduction

Peptide receptor radionuclide therapy (PRRT) with radiolabeled somatostatin analogs (SSAs) has become an established second- or third-line treatment option for patients with progressive well-differentiated (grade 1–2) gastroenteropancreatic (GEP) neuroendocrine tumors (NETs). Due to the increasing incidence and prevalence of GEP-NETs over recent decades [1] and the development of NET-specific treatments and treatment protocols, there is a growing utilization of such systemic treatment for this advanced patient group. PRRT with [177Lu]Lu-[DOTA0,Tyr3]octreotate (177Lu-DOTATATE) is the first registered theranostic application in the field of NETs [2,3]. With this therapy, radiolabeled SSAs target the somatostatin receptor (SSTR) subtype 2 that is over-expressed on the cancer cell surface [4]. Treatment of NET patients with PRRT requires dedicated clinical expertise due to the biological characteristics of PRRT (for example, binding to SSTR, DNA damage induction capacity and the radioactive component) and specific characteristics of NET patients (such as origin, grade and SSTR expression). Consequently, eligibility of NET patients for PRRT should be discussed in an expert multidisciplinary team. This review provides an overview of the development of PRRT, clinical management of patients undergoing PRRT and key unmet needs for future investigations.

2. Background of PRRT

2.1. Mechanism of Action

PRRT with 177Lu-DOTATATE uses an intravenously administered beta radiation-emitting radiopharmaceutical targeting SSTR on tumor cells. SSTRs are G-protein coupled receptors with five subtypes of which subtype 2 (SSTR2) is the most commonly expressed in NETs, followed by SSTR1 and SSTR5 [5,6]. Healthy non-diseased organs can also express SSTR subtypes, as can be seen in the healthy liver, spleen, pituitary, salivary glands, thyroid, adrenals, kidney, prostate, pancreas, intestine and blood vessels [7,8,9]. The antitumoral effect of PRRT is triggered by radiation-induced DNA damage. After binding to the SSTR2, the radiopharmaceutical agonists are internalized into the tumor cell where the irradiation induces different types of DNA damage such as DNA single strand breaks (SSBs) and DNA double strand breaks (DSBs) of which the latter are the major contributors to tumor cell death induction [10,11] (Figure 1).

2.2. The Choice for 177Lu-DOTATATE

Clinical development of PRRT commenced with SSAs labeled with indium-111, yttrium-90 and lutetium-177 [12]. PRRT with 177Lu-DOTATATE is approved by the Food and Drug Administration (FDA) and European Medicine Agency (EMA) and consists of the SSA octreotate [13] and 1,4,7,10-tetraazacyclotetradecane-1,4,7,10-tetraacetic acid (DOTA), which is the chelator for stable binding of lutetium-177 to octreotate [14]. Lutetium-177 is a medium-energy beta-emitting radionuclide and a low-energy gamma-emitting radionuclide. The gamma emission allows imaging and dosimetry where the beta emission is used for therapeutic purposes. The beta emission of lutetium-177 shows tissue penetration with a maximum range of 2 mm. Detailed information on chemical structure, production and quality control of 177Lu-DOTATATE can be found elsewhere [15]. Indium-111 is much less suitable for therapy compared to lutetium-177, because its auger electrons will not reach the nucleus and will therefore not be able to induce sufficient DNA damage [16]. Another important part of the potential therapeutic effect is the choice of SSA. Octreotate shows a higher affinity to SSTR2 than the earlier developed octreotide. This can explain why the uptake of 177Lu-DOTATATE in the tumor was increased three to four times in comparison to 111In-diethylenetriaminepentaacetic acid (DTPA)-octreotide [17]. Therapeutic activity of 111In-DTPA-octreotide, known for its diagnostic use as OctreoScan®, originally showed promising preliminary results on symptom control in NETs, but radiological tumor regression was rare and high levels of myelotoxicity were observed [18,19]. Given the specific characteristics of the different radionuclides, not only is the variance in efficacy important to take into account when selecting the preferred radionuclide, but also the risk of toxicity has to be taken into account. Less renal toxicity was seen in patients treated with 177Lu-DOTATATE compared to patients treated with [90Y]Y-[DOTA0,Tyr3]octreotide (90Y-DOTATOC). This can be explained by the lower beta-energy and tissue penetration of lutetium-177. Due to the lack of gamma emission, yttrium-90 is also not suitable for scintigraphy with a gamma camera [20], however, positron emission tomography (PET) imaging is possible [21]. 177Lu-DOTATATE can also be used for tumor dosimetry to obtain more knowledge about the tumor-absorbed dose. An indication of the absorbed dose can be obtained by dosimetric calculation on the total body scintigraphy. Quantitative single photon emission computed tomography/computerized tomography (SPECT/CT) can be more precise in the estimation of the tumor-absorbed dose per cycle. However, these scans are more time consuming, especially for dosimetry at multiple time points. A reduction of uptake in the tumor in every subsequent cycle might be an indication of a good tumor response. On the other hand, there is still a large variety between cycles and different tumor sides within the patient [22]. A significant dose–response correlation was found in pancreatic NETs (panNETs) but not in small intestinal NETs (SI-NETs) [23,24]. The different range of the beta particles from lutetium-177 and yttrium-90 could potentially be an advantage in patients with tumors of different sizes, for instance in an induction setting, although further research is needed [25,26].

3. Efficacy of PRRT

3.1. Tumor Control

Clinical evidence of the efficacy of PRRT in tumor control was proven by the phase III NETTER-1 study and several phase II clinical trials [27,28,29]. The NETTER-1 clinical trial compared four cycles of 7.4 GBq (800 mCi total) 177Lu-DOTATATE plus intramuscular long-acting octreotide 30 mg every 4 weeks to a control group which received octreotide 60 mg every 4 weeks in grade 1 and 2 midgut NET patients, progressive on a long-acting SSA. The rate of progression-free survival (PFS) after 20 months was 65% of patients treated with 177Lu-DOTATATE compared to 11% of patients in the control group. The risk of progressive disease (PD) or death was 79% lower in the 177Lu-DOTATATE group and the objective response rate (ORR) was significantly higher in the 177Lu-DOTATATE group (18%) than in the control group (3%). Sixteen percent of the patients treated with 177Lu-DOTATATE showed PD compared to 56% of the control group [27]. After a follow-up of 76 months in the intention-to-treat population, the median overall survival (OS) was 48 months in the 177Lu-DOTATATE group, which did not significantly differ from the median OS of 36 months in the control group. The similarity in OS between both groups may be explained by the fact that 36% of the patients in the control group crossed over to the PRRT arm during follow-up [30]. In the largest phase II prospective trial to date, Brabander et al. included patients treated with PRRT with 177Lu-DOTATATE for GEP-NETs, bronchial NET and NET of unknown primary origin. A median PFS of 29 months and a median OS of 63 months were observed in 443 patients with bronchial and GEP-NETs who received a cumulative activity of 22.2–29.6 GBq (600–800 mCi) 177Lu-DOTATATE. An ORR of 39% after PRRT was found, whereas stable disease (SD) was observed in 43% of the patients [29]. In other phase II trials, retrospective studies and meta-analyses of PRRT with 177Lu-DOTATATE, similar survival and response data were found [31,32,33,34,35,36]. An overview of these results is given in Table 1. Although PRRT with 177Lu-DOTATATE is the best option for treatment of patients with advanced GEP-NETs at this moment, complete response rates are still rare (1–2%) [27,29].

3.2. Symptom Control

PRRT is not only effective in reducing tumor growth, but it can also lead to symptomatic improvement in NET patients, including associated hormonal syndromes [37]. In the NETTER-1 trial, PRRT with 177Lu-DOTATATE provided a significant benefit in quality of life (QoL) compared to octreotide LAR. Time to deterioration (TTD) was significantly longer in the 177Lu-DOTATATE group, with positive outcomes in the domains of global health, physical functioning, diarrhea, pain, body image, disease-related worries and fatigue [38]. In a prospective series of 265 GEP- and bronchial NET patients, QoL, performance status and symptoms (particularly insomnia, appetite loss and diarrhea) improved significantly after PRRT [39]. Importantly, QoL in asymptomatic NET patients did not decline during therapy. In a single center study of 144 patients, a symptomatic response to PRRT with regard to diarrhea, abdominal pain, flushing and fatigue was observed in 70%, 63%, 64% and 53%, respectively [33]. Results are summarized in Table 1. In patients with a functioning NET, PRRT has shown to be an effective treatment regarding symptom control and circulating hormone levels [40,41]. Carcinoid syndrome (CS) is the most prevalent hormonal NET syndrome and is caused by excretion of hormones and amines such as serotonin, histamine, catecholamines, prostaglandins and tachykinins. In a study involving 22 patients with refractory CS who received PRRT for symptomatic control, flushing and bowel movement frequency improved significantly [42]. Two-thirds of the patients who had at least two episodes of flushing per day had a minimal decrease of 50% of these episodes. Of patients with bowel movement frequency of at least four times a day, 47% experienced > 30% decrease, while 29% experienced > 50% decrease [37]. In a systematic review, symptomatic improvement after PRRT was observed in 74% of patients with diarrhea and in 6% of patients with flushing [43]. Together, these results have positioned PRRT with 177Lu-DOTATATE as a viable option for refractory CS [44]. In addition, case reports have shown improvement of symptoms and echocardiographic parameters of carcinoid heart disease after PRRT, which is a severe symptom of CS [45]. Similar positive effects of PRRT on hormonal levels and symptoms have been observed in patients with functioning panNET syndromes, such as insulinoma, gastrinoma, glucagonoma and VIPoma [40].

3.3. Prediction of Response to PRRT

There are a variety of NET subtypes with differences in prognosis and response to PRRT. GEP-NETs show higher PFS (25–30 months) and OS (39–71 months) after PRRT than non GEP-NETs (12–29 months PFS and 39–53 months OS) [28,29]. Within the group of GEP-NETs, panNETs and small intestinal NETs are the most prevalent. After PRRT, a median PFS of 30 months for both groups was observed with a median OS of 71 months in panNETs and 60 months in midgut NETs [29]. The outcome of PRRT has also been associated with the extent of tumor burden and in particular the hepatic tumor load [46]. An extensive tumor burden correlated with an increased rate of PD after PRRT and a significantly shorter OS [29,46]. Patients of whom the liver had over 50% tumor burden showed significantly lower OS and PFS in phase II trials [47,48]. Conversely, in the NETTER-1 trial, total liver tumor burden was found not to correlate with PFS, whereas the presence of at least one large target lesion of > 3 cm was associated negatively with PFS [49]. Additionally, higher uptake on SSTR imaging correlated significantly with better response rates after PRRT [50]. Recently, the cumulative activity of 177Lu-DOTATATE was found to be an independent predictive factor for survival after PRRT. In 130 patients who received a reduced activity of 177Lu-DOTATATE because of disease-unrelated causes (such as bone marrow and renal toxicity), PFS, OS and response rates were all lower as compared to 350 control patients who received 800 mCi 177Lu-DOTATATE [51]. The association between PFS and OS and reduced administered activity of 177Lu-DOTATATE persisted in multivariable Cox regression analyses in which relevant confounders were included. 18F-FDG PET/CT has limited added value in the assessment for PRRT in well-differentiated NETs. Although neuroendocrine neoplasm (NEN) patients with positive uptake on 18F-FDG PET/CT had a significantly shorter PFS than patients with a negative scan [52], this modality is likely more a prognostic than predictive marker [53]. A grading score has been developed to integrate the results of both 18F-FDG PET/CT and SSA PET imaging ranging from non-aggressive somatostatin receptor imaging (SRI)-positive, FDG-negative to aggressive FDG-positive, SRI-negative NETs. This NETPET score was found to be a significant prognostic marker of OS in patients with GEP-NETs or bronchial NETs, but its predictive role in PRRT is unknown [54,55].

3.4. Predictive Markers

The biochemical response to treatment in NETs has often been evaluated by serum chromogranin A (CgA) levels. An increased level is associated with hepatic tumor burden, rapid tumor progression and a shorter OS [56]. In patients who received PRRT, a decrease in CgA of >50% was observed in 25–52% of patients [33,36,57]. Patients with a significant decline in CgA levels after PRRT displayed prolonged PFS and OS [57]. Changes in CgA levels during PRRT should be interpreted with caution though, as an increase in CgA can be caused by both tumor progression and cell damage or lysis by PRRT [58]. The PRRT predictive quotient (PPQ) is a circulating transcript assay that can predict the response of GEP-NETs and bronchopulmonary NETs. A positive PPQ measured before the start of PRRT accurately predicted the outcome of disease control after PRRT in 97% of cases, whereas a negative PPQ was associated with PD after PRRT in 61% of cases at 2 months and 89% at 6–9 months after PRRT [59]. Levels of the NETest, a circulating transcript analysis of 51 genes, during and after PRRT were found to be associated with response according to RECIST on imaging [60]. A lack of decrease in NETest levels at the fourth PRRT cycle signified the advent of PD in advance of imaging, with an overall diagnostic accuracy of 93% to predict response [61].

4. PRRT Protocol

The eligibility of a patient to receive PRRT should be discussed in a multidisciplinary team in a specialized NET center for each individual patient. Although multiple protocols for PRRT with 177Lu-DOTATATE have been proposed, the most commonly used treatment schedule entails four cycles of 7.4 GBq 177Lu-DOTATATE, which is based on the Rotterdam Erasmus MC protocol and also used in the NETTER-1 study [27,29]. The interval between cycles is 6 to 10 weeks. In case of toxicity, this interval can be extended up to 16 weeks [2,3,27]. In parallel with the administration of 177Lu-DOTATATE, an amino acid solution of 2.5% arginine and lysine in 1 L saline is co-infused in order to prevent renal toxicity. This infusion starts approximately 30–60 min before the administration of 177Lu-DOTATATE with a total infusion time of 4 h. The potential volume overload of the amino acid infusion in decompensated carcinoid heart disease patients can generally be managed by a prolonged infusion period or the administration of loop diuretics [62]. The amino acid infusion can cause nausea for which an antiemetic, typically ondansetron or granisetron, should be given prophylactically before the start of the infusion. 177Lu-DOTATATE allows for post-therapy scintigraphy with planar imaging or SPECT/CT. At patient discharge, the radiation exposure should be measured and patients should receive tailored advice on the duration of radiation safety precautions at home, to avoid a high radiation exposure to other people, particularly children and pregnant women. Patients with NET-associated hormonal syndromes who have an indication for continuation of SSA use should adjust the moment of the injections to the PRRT cycles. Long-acting SSA should not be given within 4–6 weeks before a cycle of PRRT because of interference with the radiolabeled SSA. Although there is conflicting evidence from two limited single center studies whether continuation of SSA treatment is beneficial in non-functioning NETs [63,64], this practice is often adopted. If the patient suffers from severe hormonal symptoms, short-acting SSA can be used to bridge this period up till 24 h before PRRT. Radiopharmaceuticals such as 177Lu-DOTATATE need to be administered at specialized facilities by medical personnel trained in radiation safety. These facilities should adhere to national and international regulations on the use of radiopharmaceuticals and be licensed by the regulatory authorities. Depending on local protocol and exposure regulations, PRRT with 177Lu-DOTATATE can be given in an in-patient as well as an out-patient setting. In between cycles, patients should be reviewed for adverse effects, including full blood count and renal and liver function. Response evaluation by cross-sectional imaging is usually performed 2–3 and 6 months after the last cycle of PRRT. Long-term follow-up is determined on an individual basis taking into account the tumor biology and therapeutic response [65]. Pseudo-progression is a phenomenon that should be considered in the response evaluation when an increase in tumor size is seen during or short after treatment with PRRT. Pseudo-progression is probably based on localized, temporarily edema caused by inflammation as a response to PRRT and does not show the actual tumor response to the therapy [58]. When pseudo-progression is suspected, functional imaging (for example, PET/CT) can help differentiate between true progression and pseudo-progression [31].

5. Salvage PRRT

In NET patients who showed tumor response at least 18 months after the first cycle of 177Lu-DOTATATE, re-treatment with PRRT (R-PRRT) with two additional cycles of 7.4 GBq each after renewed PD has shown antitumoral effects. In a meta-analysis on the effect of R-PRRT, the pooled median PFS was 14 months with a pooled median OS of 27 months. Similarly, the pooled ORR was 17% with a disease control rate of 77%. Response rates, PFS and OS were lower than for initial PRRT [66], nonetheless R-PRRT remains a potential option for GEP-NET patients when other systemic treatment options are limited. The limited efficacy of R-PRRT as compared to initial PRRT might be explained by the administration of lower cumulative activity (i.e., generally half of the initial PRRT dosage) [51], the increase in tumor bulk at baseline before R-PRRT and potential changes in the tumor biology, such as a longitudinal increase in Ki-67. In the largest study to date by van der Zwan et al., no difference in toxicity after R-PRRT as compared with initial PRRT was observed, particularly no increased occurrence of nephrotoxicity or significant hematological disease [67]. In cases where R-PRRT has provided additional benefit on tumor response and prolonged PFS, further re-treatment at the time of progression can be considered [67].

6. Patient Selection

PRRT with 177Lu-DOTATATE is registered for patients with GEP-NETs that are progressive on SSA treatment. Non-radiolabeled or ‘cold’ long-acting SSAs such as octreotide LAR and lanreotide are generally considered the first-line systemic treatment option for advanced and metastatic SSTR-positive GEP-NETs, particularly for tumors with a Ki-67 index below 10% [68]. Among the different second-line options in well-differentiated panNETs, 177Lu-DOTATATE seems to compare favorably to targeted therapy (everolimus or sunitinib) or chemotherapy [69]. In the randomized phase II OCLURANDUM trial, PRRT with 177Lu-DOTATATE was associated with a higher median PFS of 20.7 months compared to 11.0 months for sunitinib treatment in patients with advanced, SSTR-positive, progressive panNET [70]. Currently, the randomized phase III COMPETE trial is comparing PRRT with 177Lu-edotreotide with everolimus in advanced, progressive GEP-NET patients. A subset of GEP-NET patients present with extensive tumor bulk or high proliferative rate (Ki-67 index of 10–55%). In these cases, treatment with octreotide LAR or lanreotide has questionable antiproliferative effects [71]. Given its ORR of 39%, which increases to 55% in panNETs [29], 177Lu-DOTATATE can be considered as first-line therapy if response is clinically necessitated [72]. These response rates compare favorably to targeted therapy [73,74,75] and for panNETs appear similar to capecitabine–temozolomide chemotherapy [76]. Poorly differentiated neuroendocrine carcinomas (NECs) and well-differentiated grade 3 NETs are high-grade NENs that display a more aggressive biological behavior than the more common grade 1 and 2 NETs [77]. PRRT is currently not considered a standard treatment option for high-grade NENs [68,78]. The rate of SSTR2 expression in grade 3 NETs ranges from 67–92% and in NECs from 32–50%, compared to a positive expression rate in grade 1 and 2 NETs ranging from 67–96% [5,79,80,81,82]. In a meta-analysis of PRRT comprising four studies, grade 3 NET patients had a median PFS of 19 months and median OS of 44 months after PRRT. The median PFS was 11 and 4 months and the median OS was 22 and 9 months for NEC with a Ki-67 of 21–55% and NEC with a Ki-67 above 55%, respectively [83]. Recent studies implicated that PRRT could be considered in grade 3 GEP-NETs and GEP-NEC with a Ki-67 of 21–55%. Importantly, to qualify for PRRT, uptake in all lesions is required on somatostatin receptor imaging [83].

7. Eligibility Criteria for PRRT

There are several inclusion and exclusion criteria to decide if a patient is eligible for treatment with PRRT. A key criterion for PRRT is the degree of uptake on SSTR imaging which is scored by the Krenning score based on planar 111In-DTPA-octereotide imaging. It was reported that 68Ga-DOTA-SSA PET/CT results in higher Krenning scores than 111In-DTPA-octereotide imaging [84]. The uptake of all tumor lesions should minimally be equal to the physiological uptake in the liver (Krenning score grade 2) on 111In-DTPA-octreotide scintigraphy or higher than the physiological uptake in the liver on 68Ga-DOTA-SSA PET/CT for a patient to be eligible for PRRT. The latter functional imaging is more accurate for detecting SSTR-positive primary tumors and metastases and therefore superior for assessing the total extent of disease [85,86,87]. PRRT is only applicable for patients with a Karnofsky Performance Scale of at least 60 [27]. PRRT is contra-indicated for patients who are pregnant or breastfeeding, patients with severe cardiac impairment (NYHA III or IV) and those with a life expectancy less than 3 months [31]. Since PRRT can induce toxicity, the following pre-treatment laboratory values are required: creatinine clearance > 40 mL/min, hemoglobin levels ≥ 6 mmol/L, leukocytes > 2 × 109/L, platelet count > 75 × 109/L, bilirubin, alanine aminotransferase (ALAT) and aspartate aminotransferase (ASAT) < 3 times the upper limit of normal and albumin > 3 g/dL [2,3].

8. PRRT-Related Toxicity

Adverse events related to PRRT are frequently mild and include nausea, abdominal pain and asthenia [27,50]. Increased hair loss is observed in up to 60% of patients treated with 177Lu-DOTATATE, but this is temporary and seldom leads to baldness [88]. Besides these mild adverse events, PRRT can induce more severe toxicities which can be dose-limiting and adjustments to the treatment schedule might be required [89]. When the toxicity has subsided within 16 weeks after the last dose, guidelines advise to administer half of the original activity of 177Lu-DOTATATE during the next cycle. PRRT should be discontinued when the toxicity persists after 16 weeks or recurs after the half dose [3]. In the NETTER-1 trial, 7% of the patients received a reduced dose because of dose-limiting toxicities [27]. The kidneys and the bone marrow are the critical organs for dose-limiting toxicities.

8.1. Hematological Toxicity

PRRT can induce hematological toxicity through bone marrow radiation. The vast majority of patients only have mild and reversible hematological toxicity with a nadir at 4–6 weeks after administration of PRRT [27,36,50]. However, grade 3 or 4 neutropenia, thrombocytopenia or leukopenia have been observed in, respectively, 1%, 2% and 1% of the patients treated with 177Lu-DOTATATE in the NETTER-1 trial [27]. PRRT-induced severe lymphopenia is the most common hematological toxicity [27,29], but it has not been associated with increased susceptibility for infections [90]. Thrombocytopenia is the most common cause of dose reduction in PRRT, whereas bleeding complications are rare [27]. Caution should be taken in patients with widespread bone metastases (Figure 2A), due to the risk of persistent cytopenia. In the absence of alternative treatment options, PRRT should preferably be initiated at half the regular activity (3.7 GBq) 177Lu-DOTATATE in these patients. Additionally, there is a relevant long-term risk of 2% for the development of myelodysplastic syndrome (MDS) and 1% for acute myeloid leukemia (AML) after PRRT [27,29,91]. Little is known about the pathophysiology of persistent hematological toxicity, but a role for clonal hematopoiesis has been postulated [92]. Known risk factors for severe hematological toxicity include decreased renal function, pre-existent cytopenias, extensive tumor mass, age over 70, extensive bone metastases and pre-treatment with myelotoxic chemotherapy [91,93,94]. Additionally, women are at higher risk for developing subacute grade ≥ 2 thrombocytopenia than men, which was independent from other risk factors in a multivariable analysis [89].

8.2. Nephrotoxicity

Due to SSTR expression in the kidneys and the renal excretion of radiolabeled SSAs, the kidneys receive a high radiation dose during PRRT. Following glomerular filtration, SSAs are re-absorbed in the proximal tubuli of the renal cortex because of active transport mechanisms [95]. By saturating this re-uptake mechanism through the use of lysine and arginine, re-absorption of radiolabeled peptides can be significantly reduced. This results in less radiation-induced nephrotoxicity by a reduction of the absorbed kidney dose up to 40% [96,97]. However, nephrotoxicity after PRRT still occurs such as tubulointerstitial scarring, atrophy and thrombotic microangiopathy [98]. In the NETTER-1 study, grade ≥ 3 renal toxicity was observed in 5% of the 177Lu-DOTATATE group and in 4% of the control group [30]. Bergsma et al. reported an overall creatinine clearance loss of 3.4% 1 year after PRRT. No subacute grade ≥ 3 renal toxicity was seen and 1.5% of the patients showed grade 3 renal toxicity in the long term. However, all these patients had a creatinine clearance of <60 mL/min at baseline [99]. A reduced kidney function can lead to a delayed renal excretion of 177Lu-DOTATATE and this has also been associated with a higher risk of hematological toxicity [95]. Risk factors associated with renal toxicity include age > 60 years, hypertension, diabetes mellitus, pre-existing renal disease, cumulative radiation dose to the kidneys, previous nephrotoxic chemotherapy, tumor or metastases close to the kidney and previous PRRT with 90Y-DOTATOC [100]. Post-renal obstruction can be observed in some GEP-NET patients, particularly in those with retroperitoneal or pelvic metastases, but this can also be caused by the primary tumor, nephrolithiasis and abdominal or retroperitoneal fibrosis [101]. Post-renal obstruction can lead to reduced renal excretion of 177Lu-DOTATATE and thereby increase the exposure of radiation to the kidneys, risking (permanent) deterioration of renal function (Figure 2B) [102,103]. In these patients, it is necessary to correct the hydronephrosis of a functional kidney before the start of PRRT to lower the risk of radiation-induced toxicity, for instance with a double J urethral stent or percutaneous nephrostomy [101,104].

8.3. Hepatotoxicity

Mild and severe hepatotoxicity has been observed in, respectively, 12% and 0.4–2.5% of patients after PRRT [28,33]. Based on clinical experience, liver failure after PRRT can occur in cases of severe (>90%) liver involvement of NET metastases (example of a patient with severe liver involvement with an enlarged risk of liver failure in Figure 2C). Moreover, there is a concern about the cumulative hepatotoxicity in patients treated with a combination of PRRT and radioembolization. In the HEPAR phase I trial, one patient died of liver failure after treatment with a dose of 80 Gy 166Ho-radioembolization [105]. No liver failure was observed in 67 patients undergoing 90Y-radioembolization after PRRT [106,107]. Adherence to eligibility criteria is crucial and in borderline cases a first trial infusion with 3.7 GBq 177Lu-DOTATATE is advised. There was no relevant hepatotoxicity reported in the NETTER-1 trial [27]. Grade 3 and 4 toxicity of aminotransferases was observed in 3% in the phase II trial by Brabander et al. Three of these patients had persisting toxicity after 3 months, but no hepatic failure was observed [29].

9. Hormonal Crisis Due to PRRT

A hormonal crisis can be triggered by PRRT due to excessive release of metabolically active amines or peptides from the tumor. A PRRT-induced hormonal crisis was observed in 1% of patients and can occur during the infusion of the radiolabeled SSA up to 48 h after infusion [108]. Risk factors for a hormonal crisis include CS, elevated 5-hydroxyindoleacetic acid (5-HIAA) and CgA levels, metastatic disease, high tumor burden, higher age and histamine release due to medication as β2 agonist bronchodilators. Prophylactic measures consist of controlling the CS before initiation of PRRT, having a good nutritional status and reducing the time without non-radiolabeled SSAs. When a hormonal crisis occurs, symptom control is necessary. The treatment should be hormone-specific and may also include general measures such as monitoring of vital parameters, saline infusion, correction of electrolyte disturbances and inpatient observation if needed. SSAs can be restarted safely 1 h after infusion of 177Lu-DOTATATE and should be considered in severe cases of SSA-responsive CS, gastrinoma, insulinoma and VIPoma. Post-infusion administration of corticosteroids can be considered in patients at risk for carcinoid crisis or those presenting with hormonal crisis [109].

10. Nervous System Location or Compression

GEP-NETs rarely metastasize to the central nervous system, with an incidence of brain metastases of 1.5–5% in NET patients [110,111]. More frequently, vertebral bone metastases can compress the spinal cord or its nerve roots (Figure 2D) [112]. PRRT can reduce tumor volume and metabolic activity of central nervous system metastases, thereby reducing pain and spinal compression [113]. The radiation-induced damage by PRRT can be accompanied by temporary edema, leading to increased compression of surrounding vital structures and potentially (worsening of) neurological symptoms. In cases of brain metastases or for those patients at risk of spinal cord or nerve compression, prophylactic therapy with corticosteroids can be considered to prevent or minimize treatment-induced edema. Some expert centers have used a one-week regimen of 4 mg dexamethasone two times a day. Caution is required with this treatment since it is possible that the use of corticosteroids negatively influences the uptake of SSA [114].

11. Mesenteric Fibrosis and Mesenteric or Peritoneal Metastases

Another midgut NET-related complication is the presence of mesenteric fibrosis, which occurs in up to half of the patients with a small intestinal NET metastatic to the mesenteric lymph nodes. Mesenteric metastases can induce local fibrosis by the secretion of serotonin and other mediators, inducing a desmoplastic reaction, which subsequently can lead to intestinal obstruction and ischemia (Figure 2E) [115,116]. Peritoneal metastases can lead to adhesions between the abdominal wall and the bowel which can cause intestinal immobility, bowel obstruction or ‘frozen abdomen’ (Figure 2F) [117]. The proportions of patients treated with PRRT who had mesenteric and/or peritoneal metastases ranged from 6–51% [27,117,118]. In a retrospective study of 132 NET patients with a mesenteric mass, 177Lu-DOTATATE resulted in an ORR of this mass in only 4% of the patients [119]. In an analysis of NET patients with mesenteric or peritoneal disease, 23% of patients at high risk for complications experienced at least one episode of bowel obstruction within three months of PRRT. These patients were treated with corticosteroids, surgery and total parenteral nutrition [117]. Although a direct causal relationship has not been established, treating physicians should be aware that PRRT seems to be ineffective in reducing the size of a mesenteric mass while there may be an increased risk of abdominal complications in patients with extensive peritoneal disease or desmoplastic changes associated with mesenteric metastases. A post-infusion trial of corticosteroids can be considered in patients at high risk of these complications [117].

12. Conclusions and Future Directions

PRRT with 177Lu-DOTATATE is a widely applicable therapy for patients with advanced and metastatic NETs. It is clear that PRRT contributes to reduction of tumor growth and stabilization of the disease in a significant proportion of these patients. Next, PRRT improves QOL and reduces symptoms [27,29]. Despite the successes, it is important that clinicians treating NET patients are aware of the toxicities (mainly renal and hematological) associated with PRRT and discuss every patient in a multidisciplinary team where careful selection, preparation and management of these patients before and during therapy with this unique radiopharmaceutical can be discussed. Despites its successes, there still is room for improvement of the efficacy and safety of PRRT. Ongoing research projects are exploring how to improve the current strategies. A combination therapy with radiosensitizers is a way to increase the effect of PRRT. This can be conducted by the inhibition of poly ADP-ribose polymerase (PARP)-1 which is essential for the repair of SSBs [120] or inhibition of heat shock protein 90 (HSP90), a chaperone molecule that plays a role in cell protection and maintenance [121]. Other radionuclides are being studied for potential use in PRRT, such as actinium-225, lead-212 and bismuth-213. These alpha-emitting radionuclides can potentially cause more damage to the tumor cells compared to PRRT with 177Lu-DOTATATE and thus increase treatment efficacy [122,123,124]. Altogether, PRRT with 177Lu-DOTATATE is a very potent treatment modality for patients with progressive well-differentiated (grade 1–2) GEP-NET and there are many different strategies currently being explored that can contribute even to better patient care.

Author Contributions

Conceptualization, M.N.B., J.N. and J.H.; writing—original draft preparation, M.N.B.; writing—review and editing, N.S.M., W.W.d.H., T.B., J.N. and J.H.; supervision, J.N. and J.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Oncode Institute (grant number P2019-0035).

Conflicts of Interest

T.B. has received speaker fees and research support from AAA/Novartis and is on the advisory board, as well as speaker fees from Ipsen. W.W.d.H. had received speaker fees and research support from AAA/Novartis and is on the advisory board, as well as speaker fees of Ipsen and is on the advisory board. J.H. has received speaker fees from Ipsen and received compensation from Novartis and Ipsen for service on advisory boards. All other authors have no conflict of interest to declare.

References

  1. Fraenkel, M.; Kim, M.; Faggiano, A.; De Herder, W.W.; Valk, G.D. Incidence of gastroenteropancreatic neuroendocrine tumours: A systematic review of the literature. Endocr.-Relat. Cancer 2014, 21, R153–R163. [Google Scholar] [CrossRef] [PubMed]
  2. FDA Letter of Approval for LUTATHERA®. 2018. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/208700s000lbl.pdf (accessed on 1 January 2022).
  3. Authorization Details for Lutathera® in Europe. 2018. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/lutathera#authorisation-details-section (accessed on 1 January 2022).
  4. De Herder, W.W.; Hofland, L.J.; van der Lely, A.-J.; Lamberts, S.W.J. Somatostatin receptors in gastroentero-pancreatic neuroendocrine tumours. Endocr.-Relat. Cancer 2003, 10, 451–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Reubi, J.C. Somatostatin and other Peptide receptors as tools for tumor diagnosis and treatment. Neuroendocrinology 2004, 80 (Suppl. S1), 51–56. [Google Scholar] [CrossRef]
  6. Hofland, L.J.; Lamberts, S.W.J. The Pathophysiological Consequences of Somatostatin Receptor Internalization and Resistance. Endocr. Rev. 2003, 24, 28–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Shastry, M.; Kayani, I.; Wild, D.; Caplin, M.; Visvikis, D.; Gacinovic, S.; Reubi, J.C.; Bomanji, J.B. Distribution pattern of 68Ga-DOTATATE in disease-free patients. Nucl. Med. Commun. 2010, 31, 1025–1032. [Google Scholar] [CrossRef]
  8. Siebinga, H.; de Wit-van der Veen, B.J.; Beijnen, J.H.; Stokkel, M.P.M.; Dorlo, T.P.C.; Huitema, A.D.R.; Hendrikx, J. A physiologically based pharmacokinetic (PBPK) model to describe organ distribution of (68)Ga-DOTATATE in patients without neuroendocrine tumors. EJNMMI Res. 2021, 11, 73. [Google Scholar] [CrossRef]
  9. Reubi, J.C.; Waser, B.; Schaer, J.C.; Laissue, J.A. Somatostatin receptor sst1-sst5 expression in normal and neoplastic human tissues using receptor autoradiography with subtype-selective ligands. Eur. J. Nucl. Med. 2001, 28, 836–846. [Google Scholar] [CrossRef]
  10. Tamborino, G.; Nonnekens, J.; De Saint-Hubert, M.; Struelens, L.; Feijtel, D.; de Jong, M.; Konijnenberg, M.W. Dosimetric Evaluation of the Effect of Receptor Heterogeneity on the Therapeutic Efficacy of Peptide Receptor Radionuclide Therapy: Correlation with DNA Damage Induction and In Vivo Survival. J. Nucl. Med. 2022, 63, 100–107. [Google Scholar] [CrossRef]
  11. Ceccaldi, R.; Rondinelli, B.; D’Andrea, A.D. Repair Pathway Choices and Consequences at the Double-Strand Break. Trends Cell Biol. 2016, 26, 52–64. [Google Scholar] [CrossRef] [Green Version]
  12. Kwekkeboom, D.J.; Mueller-Brand, J.; Paganelli, G.; Anthony, L.B.; Pauwels, S.; Kvols, L.K.; O’Dorisio, T.M.; Valkema, R.; Bodei, L.; Chinol, M. Overview of results of peptide receptor radionuclide therapy with 3 radiolabeled somatostatin analogs. J. Nucl. Med. 2005, 46 (Suppl. S1), 62S–66S. [Google Scholar]
  13. Reubi, J.C.; Schar, J.C.; Waser, B.; Wenger, S.; Heppeler, A.; Schmitt, J.S.; Macke, H.R. Affinity profiles for human somatostatin receptor subtypes SST1-SST5 of somatostatin radiotracers selected for scintigraphic and radiotherapeutic use. Eur. J. Nucl. Med. 2000, 27, 273–282. [Google Scholar] [CrossRef]
  14. De Jong, M.; Breeman, W.A.P.; Valkema, R.; Bernard, B.F.; Krenning, E.P. Combination radionuclide therapy using 177Lu-and 90Y-labeled somatostatin analogs. J. Nucl. Med. 2005, 46 (Suppl. S1), 13S–17S. [Google Scholar]
  15. Breeman, W.A.; Chan, H.S.; de Zanger, R.M.; Konijnenberg, M.K.; de Blois, E. Overview of Development and Formulation of 177Lu-DOTA-TATE for PRRT. Curr. Radiopharm. 2016, 9, 8–18. [Google Scholar] [CrossRef]
  16. Teunissen, J.J.M.; Kwekkeboom, D.J.; de Jong, M.; Esser, J.-P.; Valkema, R.; Krenning, E.P. Endocrine tumours of the gastrointestinal tract. Peptide receptor radionuclide therapy. Best Pract. Res. Clin. Gastroenterol. 2005, 19, 595–616. [Google Scholar] [CrossRef] [Green Version]
  17. Kwekkeboom, D.J.; Bakker, W.H.; Kooij, P.P.; Konijnenberg, M.W.; Srinivasan, A.; Erion, J.L.; Schmidt, M.A.; Bugaj, J.L.; de Jong, M.; Krenning, E.P. [177Lu-DOTAOTyr3]octreotate: Comparison with [111In-DTPAo]octreotide in patients. Eur. J. Nucl. Med. 2001, 28, 1319–1325. [Google Scholar] [CrossRef]
  18. Valkema, R.; De Jong, M.; Bakker, W.H.; Breeman, W.A.; Kooij, P.P.; Lugtenburg, P.J.; De Jong, F.H.; Christiansen, A.; Kam, B.L.; De Herder, W.W.; et al. Phase I study of peptide receptor radionuclide therapy with [In-DTPA]octreotide: The Rotterdam experience. Semin. Nucl. Med. 2002, 32, 110–122. [Google Scholar] [CrossRef]
  19. Anthony, L.B.; Woltering, E.A.; Espenan, G.D.; Cronin, M.D.; Maloney, T.J.; McCarthy, K.E. Indium-111-pentetreotide prolongs survival in gastroenteropancreatic malignancies. Semin. Nucl. Med. 2002, 32, 123–132. [Google Scholar] [CrossRef]
  20. Bodei, L.; Cremonesi, M.; Ferrari, M.; Pacifici, M.; Grana, C.M.; Bartolomei, M.; Baio, S.M.; Sansovini, M.; Paganelli, G. Long-term evaluation of renal toxicity after peptide receptor radionuclide therapy with 90Y-DOTATOC and 177Lu-DOTATATE: The role of associated risk factors. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 1847–1856. [Google Scholar] [CrossRef]
  21. D’Arienzo, M. Emission of β+ particles via internal pair production in the 0+–0+ transition of 90Zr: Historical background and current applications in nuclear medicine imaging. Atoms 2013, 1, 2–12. [Google Scholar] [CrossRef]
  22. Roth, D.; Gustafsson, J.; Warfvinge, C.F.; Sundlöv, A.; Åkesson, A.; Tennvall, J.; Gleisner, K.S. Dosimetric Quantities in Neuroendocrine Tumors over Treatment Cycles with (177)Lu-DOTATATE. J. Nucl. Med. 2022, 63, 399–405. [Google Scholar] [CrossRef]
  23. Jahn, U.; Ilan, E.; Sandström, M.; Garske-Román, U.; Lubberink, M.; Sundin, A. 177Lu-DOTATATE Peptide Receptor Radionuclide Therapy: Dose Response in Small Intestinal Neuroendocrine Tumors. Neuroendocrinology 2020, 110, 662–670. [Google Scholar] [CrossRef] [PubMed]
  24. Ilan, E.; Sandström, M.; Wassberg, C.; Sundin, A.; Garske-Román, U.; Eriksson, B.; Granberg, D.; Lubberink, M. Dose response of pancreatic neuroendocrine tumors treated with peptide receptor radionuclide therapy using 177Lu-DOTATATE. J. Nucl. Med. 2015, 56, 177–182. [Google Scholar] [CrossRef] [PubMed]
  25. Kunikowska, J.; Królicki, L.; Hubalewska-Dydejczyk, A.; Mikołajczak, R.; Sowa-Staszczak, A.; Pawlak, D. Clinical results of radionuclide therapy of neuroendocrine tumours with 90Y-DOTATATE and tandem 90Y/177Lu-DOTATATE: Which is a better therapy option? Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 1788–1797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kong, G.; Callahan, J.; Hofman, M.S.; Pattison, D.A.; Akhurst, T.; Michael, M.; Eu, P.; Hicks, R.J. High clinical and morphologic response using (90)Y-DOTA-octreotate sequenced with (177)Lu-DOTA-octreotate induction peptide receptor chemoradionuclide therapy (PRCRT) for bulky neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 476–489. [Google Scholar] [CrossRef]
  27. Strosberg, J.; El-Haddad, G.; Wolin, E.; Hendifar, A.; Yao, J.; Chasen, B.; Mittra, E.; Kunz, P.L.; Kulke, M.H.; Jacene, H.; et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. N. Engl. J. Med. 2017, 376, 125–135. [Google Scholar] [CrossRef]
  28. Lin, E.; Chen, T.; Little, A.; Holliday, L.; Roach, P.; Butler, P.; Hosking, E.; Bailey, E.; Elison, B.; Currow, D. Safety and outcomes of (177) Lu-DOTATATE for neuroendocrine tumours: Experience in New South Wales, Australia. Intern. Med. J. 2019, 49, 1268–1277. [Google Scholar] [CrossRef]
  29. Brabander, T.; van der Zwan, W.A.; Teunissen, J.J.M.; Kam, B.L.R.; Feelders, R.A.; de Herder, W.W.; van Eijck, C.H.J.; Franssen, G.J.H.; Krenning, E.P.; Kwekkeboom, D.J. Long-Term Efficacy, Survival, and Safety of [(177)Lu-DOTA(0),Tyr(3)]octreotate in Patients with Gastroenteropancreatic and Bronchial Neuroendocrine Tumors. Clin. Cancer Res. 2017, 23, 4617–4624. [Google Scholar] [CrossRef] [Green Version]
  30. Strosberg, J.R.; Caplin, M.E.; Kunz, P.L.; Ruszniewski, P.B.; Bodei, L.; Hendifar, A.; Mittra, E.; Wolin, E.M.; Yao, J.C.; Pavel, M.E.; et al. (177)Lu-Dotatate plus long-acting octreotide versus highdose long-acting octreotide in patients with midgut neuroendocrine tumours (NETTER-1): Final overall survival and long-term safety results from an open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 2021, 22, 1752–1763. [Google Scholar] [CrossRef]
  31. Hicks, R.J.; Kwekkeboom, D.J.; Krenning, E.; Bodei, L.; Grozinsky-Glasberg, S.; Arnold, R.; Borbath, I.; Cwikla, J.; Toumpanakis, C.; Kaltsas, G.; et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasia: Peptide Receptor Radionuclide Therapy with Radiolabeled Somatostatin Analogues. Neuroendocrinology 2017, 105, 295–309. [Google Scholar] [CrossRef]
  32. Zhang, J.; Song, Q.; Cai, L.; Xie, Y.; Chen, Y. The efficacy of (177)Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) in patients with metastatic neuroendocrine tumours: A systematic review and meta-analysis. J. Cancer Res. Clin. Oncol. 2020, 146, 1533–1543. [Google Scholar] [CrossRef]
  33. Hamiditabar, M.; Ali, M.; Roys, J.; Wolin, E.M.; O’Dorisio, T.M.; Ranganathan, D.; Tworowska, I.; Strosberg, J.R.; Delpassand, E.S. Peptide Receptor Radionuclide Therapy With 177Lu-Octreotate in Patients with Somatostatin Receptor Expressing Neuroendocrine Tumors: Six Years’ Assessment. Clin. Nucl. Med. 2017, 42, 436–443. [Google Scholar] [CrossRef]
  34. Ezziddin, S.; Khalaf, F.; Vanezi, M.; Haslerud, T.; Mayer, K.; Al Zreiqat, A.; Willinek, W.; Biersack, H.J.; Sabet, A. Outcome of peptide receptor radionuclide therapy with 177Lu-octreotate in advanced grade 1/2 pancreatic neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2014, 41, 925–933. [Google Scholar] [CrossRef]
  35. Sabet, A.; Dautzenberg, K.; Haslerud, T.; Aouf, A.; Sabet, A.; Simon, B.; Mayer, K.; Biersack, H.J.; Ezziddin, S. Specific efficacy of peptide receptor radionuclide therapy with (177)Lu-octreotate in advanced neuroendocrine tumours of the small intestine. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 1238–1246. [Google Scholar] [CrossRef]
  36. Bodei, L.; Cremonesi, M.; Grana, C.M.; Fazio, N.; Iodice, S.; Baio, S.M.; Bartolomei, M.; Lombardo, D.; Ferrari, M.E.; Sansovini, M.; et al. Peptide receptor radionuclide therapy with (1)(7)(7)Lu-DOTATATE: The IEO phase I-II study. Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 2125–2135. [Google Scholar] [CrossRef]
  37. Zandee, W.T.; Brabander, T.; Blazevic, A.; Minczeles, N.S.; Feelders, R.A.; de Herder, W.W.; Hofland, J. Peptide Receptor Radionuclide Therapy With 177Lu-DOTATATE for Symptomatic Control of Refractory Carcinoid Syndrome. J. Clin. Endocrinol. Metab. 2021, 106, e3665–e3672. [Google Scholar] [CrossRef]
  38. Strosberg, J.; Wolin, E.; Chasen, B.; Kulke, M.; Bushnell, D.; Caplin, M.; Baum, R.P.; Kunz, P.; Hobday, T.; Hendifar, A.; et al. Health-Related Quality of Life in Patients with Progressive Midgut Neuroendocrine Tumors Treated with (177)Lu-Dotatate in the Phase III NETTER-1 Trial. J. Clin. Oncol. 2018, 36, 2578–2584. [Google Scholar] [CrossRef]
  39. Khan, S.; Krenning, E.P.; van Essen, M.; Kam, B.L.; Teunissen, J.J.; Kwekkeboom, D.J. Quality of life in 265 patients with gastroenteropancreatic or bronchial neuroendocrine tumors treated with [177Lu-DOTA0,Tyr3]octreotate. J. Nucl. Med. 2011, 52, 1361–1368. [Google Scholar] [CrossRef] [Green Version]
  40. Zandee, W.T.; Brabander, T.; Blazevic, A.; Kam, B.L.R.; Teunissen, J.J.M.; Feelders, R.A.; Hofland, J.; de Herder, W.W. Symptomatic and Radiological Response to 177Lu-DOTATATE for the Treatment of Functioning Pancreatic Neuroendocrine Tumors. J. Clin. Endocrinol. Metab. 2019, 104, 1336–1344. [Google Scholar] [CrossRef]
  41. Seregni, E.; Maccauro, M.; Chiesa, C.; Mariani, L.; Pascali, C.; Mazzaferro, V.; De Braud, F.; Buzzoni, R.; Milione, M.; Lorenzoni, A.; et al. Treatment with tandem [90Y]DOTA-TATE and [177Lu]DOTA-TATE of neuroendocrine tumours refractory to conventional therapy. Eur. J. Nucl. Med. Mol. Imaging 2014, 41, 223–230. [Google Scholar] [CrossRef]
  42. de Herder, W.W. Biochemistry of neuroendocrine tumours. Best. Pract. Res. Clin. Endocrinol. Metab. 2007, 21, 33–41. [Google Scholar] [CrossRef]
  43. Hofland, J.; Herrera-Martinez, A.D.; Zandee, W.T.; de Herder, W.W. Management of carcinoid syndrome: A systematic review and meta-analysis. Endocr.-Relat. Cancer 2019, 26, R145–R156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Grozinsky-Glasberg, S.; Davar, J.; Hofland, J.; Dobson, R.; Prasad, V.; Pascher, A.; Denecke, T.; Tesselaar, M.E.T.; Panzuto, F.; Albåge, A.; et al. European Neuroendocrine Tumor Society (ENETS) 2022 Guidance Paper for Carcinoid Syndrome and Carcinoid Heart Disease. J. Neuroendocrinol. 2022, 34, e13146. [Google Scholar] [CrossRef]
  45. Ramesh, S.; Kudachi, S.; Basu, S. Peptide Receptor Radionuclide Therapy with (177)Lu-DOTATATE in Carcinoid Heart Disease: A Contraindication or a Promising Treatment Approach Bettering Chances for Corrective Surgery? J. Nucl. Med. Technol. 2018, 46, 292–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kwekkeboom, D.J.; Bakker, W.H.; Kam, B.L.; Teunissen, J.J.; Kooij, P.P.; de Herder, W.W.; Feelders, R.A.; van Eijck, C.H.; de Jong, M.; Srinivasan, A.; et al. Treatment of patients with gastro-entero-pancreatic (GEP) tumours with the novel radiolabelled somatostatin analogue [177Lu-DOTA(0),Tyr3]octreotate. Eur. J. Nucl. Med. Mol. Imaging 2003, 30, 417–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Yalchin, M.; Oliveira, A.; Theocharidou, E.; Pencharz, D.; Navalkissoor, S.; Quigley, A.-M.; Walker, M.; Caplin, M.; Toumpanakis, C. The Impact of Radiological Response to Peptide Receptor Radionuclide Therapy on Overall Survival in Patients with Metastatic Midgut Neuroendocrine Tumors. Clin. Nucl. Med. 2017, 42, e135–e141. [Google Scholar] [CrossRef]
  48. Ezziddin, S.; Attassi, M.; Yong-Hing, C.J.; Ahmadzadehfar, H.; Willinek, W.; Grünwald, F.; Guhlke, S.; Biersack, H.-J.; Sabet, A. Predictors of long-term outcome in patients with well-differentiated gastroenteropancreatic neuroendocrine tumors after peptide receptor radionuclide therapy with 177Lu-octreotate. J. Nucl. Med. 2014, 55, 183–190. [Google Scholar] [CrossRef] [Green Version]
  49. Strosberg, J.; Kunz, P.L.; Hendifar, A.; Yao, J.; Bushnell, D.; Kulke, M.H.; Baum, R.P.; Caplin, M.; Ruszniewski, P.; Delpassand, E.; et al. Impact of liver tumour burden, alkaline phosphatase elevation, and target lesion size on treatment outcomes with 177Lu-Dotatate: An analysis of the NETTER-1 study. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 2372–2382. [Google Scholar] [CrossRef] [Green Version]
  50. Kwekkeboom, D.J.; de Herder, W.W.; Kam, B.L.; van Eijck, C.H.; van Essen, M.; Kooij, P.P.; Feelders, R.A.; van Aken, M.O.; Krenning, E.P. Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0,Tyr3]octreotate: Toxicity, efficacy, and survival. J. Clin. Oncol. 2008, 26, 2124–2130. [Google Scholar] [CrossRef] [Green Version]
  51. Minczeles, N.S.; de Herder, W.W.; Feelders, R.A.; Verburg, F.A.; Hofland, J.; Brabander, T. Long-term outcomes of submaximal activities of peptide receptor radionuclide therapy with (177)Lu-DOTATATE in neuroendocrine tumour patients. J. Nucl. Med. 2022, 62, 11. [Google Scholar]
  52. Magi, L.; Prosperi, D.; Lamberti, G.; Marasco, M.; Ambrosini, V.; Rinzivillo, M.; Campana, D.; Gentiloni, G.; Annibale, B.; Signore, A.; et al. Role of [(18)F]FDG PET/CT in the management of G1 gastro-entero-pancreatic neuroendocrine tumors. Endocrine 2022, 76, 484–490. [Google Scholar] [CrossRef]
  53. Ambrosini, V.; Kunikowska, J.; Baudin, E.; Bodei, L.; Bouvier, C.; Capdevila, J.; Cremonesi, M.; de Herder, W.W.; Dromain, C.; Falconi, M. Consensus on molecular imaging and theranostics in neuroendocrine neoplasms. Eur. J. Cancer 2021, 146, 56–73. [Google Scholar] [CrossRef]
  54. 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] [Green Version]
  55. 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 PET Imaging in Bronchial Neuroendocrine Neoplasms: The NETPET Score as a Prognostic Biomarker. J. Nucl. Med. 2021, 62, 1278–1284. [Google Scholar] [CrossRef]
  56. Arnold, R.; Wilke, A.; Rinke, A.; Mayer, C.; Kann, P.H.; Klose, K.J.; Scherag, A.; Hahmann, M.; Muller, H.H.; Barth, P. Plasma chromogranin A as marker for survival in patients with metastatic endocrine gastroenteropancreatic tumors. Clin. Gastroenterol. Hepatol. 2008, 6, 820–827. [Google Scholar] [CrossRef]
  57. Fross-Baron, K.; Garske-Roman, U.; Welin, S.; Granberg, D.; Eriksson, B.; Khan, T.; Sandstrom, M.; Sundin, A. 177Lu-DOTATATE Therapy of Advanced Pancreatic Neuroendocrine Tumors Heavily Pretreated with Chemotherapy: Analysis of Outcome, Safety, and Their Determinants. Neuroendocrinology 2021, 111, 330–343. [Google Scholar] [CrossRef]
  58. Brabander, T.; van der Zwan, W.A.; Teunissen, J.J.M.; Kam, B.L.R.; de Herder, W.W.; Feelders, R.A.; Krenning, E.P.; Kwekkeboom, D.J. Pitfalls in the response evaluation after peptide receptor radionuclide therapy with [(177)Lu-DOTA(0),Tyr(3)]octreotate. Endocr.-Relat. Cancer 2017, 24, 243–251. [Google Scholar] [CrossRef] [Green Version]
  59. Bodei, L.; Kidd, M.S.; Singh, A.; Van Der Zwan, W.A.; Severi, S.; Drozdov, I.A.; Cwikla, J.; Baum, R.P.; Kwekkeboom, D.J.; Paganelli, G. PRRT genomic signature in blood for prediction of 177Lu-octreotate efficacy. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 1155–1169. [Google Scholar] [CrossRef]
  60. Kidd, M.; Drozdov, I.; Modlin, I. Blood and tissue neuroendocrine tumor gene cluster analysis correlate, define hallmarks and predict disease status. Endocr.-Relat. Cancer 2015, 22, 561–575. [Google Scholar] [CrossRef]
  61. Bodei, L.; Kidd, M.S.; Singh, A.; van der Zwan, W.A.; Severi, S.; Drozdov, I.A.; Malczewska, A.; Baum, R.P.; Kwekkeboom, D.J.; Paganelli, G.; et al. PRRT neuroendocrine tumor response monitored using circulating transcript analysis: The NETest. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 895–906. [Google Scholar] [CrossRef]
  62. Bodei, L.; Mueller-Brand, J.; Baum, R.P.; Pavel, M.E.; Hörsch, D.; O’Dorisio, M.S.; Howe, J.R.; Cremonesi, M.; Kwekkeboom, D.J.; Zaknun, J.J. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 800–816. [Google Scholar] [CrossRef]
  63. Yordanova, A.; Wicharz, M.M.; Mayer, K.; Brossart, P.; Gonzalez-Carmona, M.A.; Strassburg, C.P.; Fimmers, R.; Essler, M.; Ahmadzadehfar, H. The Role of Adding Somatostatin Analogues to Peptide Receptor Radionuclide Therapy as a Combination and Maintenance Therapy. Clin. Cancer Res. 2018, 24, 4672–4679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Syguła, A.; Ledwon, A.; Hasse-Lazar, K.; Jurecka-Lubieniecka, B.; Michalik, B.; Paliczka-Cieślik, E.; Zeman, M.; Chmielik, E.; Sczasny, J.; Jarzab, B. In patients with well-differentiated neuroendocrine tumours, there is no apparent benefit of somatostatin analogues after disease control by peptide receptor radionuclide therapy. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 3841–3851. [Google Scholar] [CrossRef] [PubMed]
  65. Zaknun, J.J.; Bodei, L.; Mueller-Brand, J.; Pavel, M.E.; Baum, R.P.; Hörsch, D.; O’Dorisio, M.S.; O’Dorisiol, T.M.; Howe, J.R.; Cremonesi, M. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 800–816. [Google Scholar] [CrossRef]
  66. Kim, Y.I. Salvage peptide receptor radionuclide therapy in patients with progressive neuroendocrine tumors: A systematic review and meta-analysis. Nucl. Med. Commun. 2021, 42, 451–458. [Google Scholar] [CrossRef] [PubMed]
  67. Van der Zwan, W.A.; Brabander, T.; Kam, B.L.R.; Teunissen, J.J.M.; Feelders, R.A.; Hofland, J.; Krenning, E.P.; de Herder, W.W. Salvage peptide receptor radionuclide therapy with [(177)Lu-DOTA,Tyr(3)]octreotate in patients with bronchial and gastroenteropancreatic neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 704–717. [Google Scholar] [CrossRef] [Green Version]
  68. Pavel, M.; Oberg, 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. Ann. Oncol. 2020, 31, 844–860. [Google Scholar] [CrossRef]
  69. Pusceddu, S.; Prinzi, N.; Tafuto, S.; Ibrahim, T.; Filice, A.; Brizzi, M.P.; Panzuto, F.; Baldari, S.; Grana, C.M.; Campana, D.; et al. Association of Upfront Peptide Receptor Radionuclide Therapy with Progression-Free Survival among Patients with Enteropancreatic Neuroendocrine Tumors. JAMA Netw. Open 2022, 5, e220290. [Google Scholar] [CrossRef]
  70. Baudin, E.; Walter, T.; Docao, C.; Haissaguerre, M.; Hadoux, J.; Taieb, D.; Ansquer, C.; Dierickx, L.; De Mestier, L.; Deshayes, E.; et al. First multicentric randomized phase II trial investigating the antitumor efficacy of peptide receptor radionuclide therapy with 177Lutetium—Octreotate (OCLU) in unresectable progressive neuroendocrine pancreatic tumor: Results of the OCLURANDOM trial, on behalf of the ENDOCAN RENATEN network and GTE. Ann. Endocrinol. 2022, 83, 289–290. [Google Scholar]
  71. Merola, E.; Alonso Gordoa, T.; Zhang, P.; Al-Toubah, T.; Pellè, E.; Kolasińska-Ćwikła, A.; Zandee, W.; Laskaratos, F.; Mestier, L.; Lamarca, A. Somatostatin Analogs for Pancreatic Neuroendocrine Tumors: Any Benefit When Ki-67 Is ≥ 10%? Oncologist 2021, 26, 294–301. [Google Scholar] [CrossRef]
  72. Minczeles, N.S.; van Eijck, C.H.J.; van Gils, M.J.; van Velthuysen, M.F.; Nieveen van Dijkum, E.J.M.; Feelders, R.A.; de Herder, W.W.; Brabander, T.; Hofland, J.l. Induction therapy with (177)Lu-DOTATATE procures long-term survival in locally advanced or oligometastatic pancreatic neuroendocrine neoplasm patients. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 3203–3214. [Google Scholar] [CrossRef]
  73. Yao, J.C.; Fazio, N.; Singh, S.; Buzzoni, R.; Carnaghi, C.; Wolin, E.; Tomasek, J.; Raderer, M.; Lahner, H.; Voi, M. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): A randomised, placebo-controlled, phase 3 study. Lancet 2016, 387, 968–977. [Google Scholar] [CrossRef]
  74. Yao, J.C.; Pavel, M.; Lombard-Bohas, C.; Van Cutsem, E.; Voi, M.; Brandt, U.; He, W.; Chen, D.; Capdevila, J.; de Vries, E.G.E.; et al. Everolimus for the Treatment of Advanced Pancreatic Neuroendocrine Tumors: Overall Survival and Circulating Biomarkers from the Randomized, Phase III RADIANT-3 Study. J. Clin. Oncol. 2016, 34, 3906–3913. [Google Scholar] [CrossRef]
  75. Raymond, E.; Dahan, L.; Raoul, J.L.; Bang, Y.J.; Borbath, I.; Lombard-Bohas, C.; Valle, J.; Metrakos, P.; Smith, D.; Vinik, A.; et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N. Engl. J. Med. 2011, 364, 501–513. [Google Scholar] [CrossRef] [Green Version]
  76. Kunz, P.L.; Graham, N.; Catalano, P.J.; Nimeiri, H.; Fisher, G.A.; Longacre, T.A.; Suarez, C.J.; Rubin, D.; Yao, J.C.; Kulke, M.H.; et al. A randomized study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumors: Final analysis of efficacy and evaluation of MGMT (ECOG-ACRIN E2211). J. Clin. Oncol. 2022, 40, 4004. [Google Scholar] [CrossRef]
  77. Sorbye, H.; Baudin, E.; Borbath, I.; Caplin, M.; Chen, J.; Cwikla, J.B.; Frilling, A.; Grossman, A.; Kaltsas, G.; Scarpa, A.; et al. Unmet Needs in High-Grade Gastroenteropancreatic Neuroendocrine Neoplasms (WHO G3). Neuroendocrinology 2019, 108, 54–62. [Google Scholar] [CrossRef]
  78. Pavel, M.; O’Toole, D.; Costa, F.; Capdevila, J.; Gross, D.; Kianmanesh, R.; Krenning, E.; Knigge, U.; Salazar, R.; Pape, U.F.; et al. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology 2016, 103, 172–185. [Google Scholar] [CrossRef]
  79. Vélayoudom-Céphise, F.-L.; Duvillard, P.; Foucan, L.; Hadoux, J.; Chougnet, C.N.; Leboulleux, S.; Malka, D.; Guigay, J.; Goere, D.; Debaere, T. Are G3 ENETS neuroendocrine neoplasms heterogeneous. Endocr.-Relat. Cancer 2013, 20, 649–657. [Google Scholar] [CrossRef] [Green Version]
  80. Srirajaskanthan, R.; Watkins, J.; Marelli, L.; Khan, K.; Caplin, M.E. Expression of Somatostatin and Dopamine 2 Receptors in Neuroendocrine Tumours and the Potential Role for New Biotherapies. Neuroendocrinology 2009, 89, 308–314. [Google Scholar] [CrossRef]
  81. Heetfeld, M.; Chougnet, C.N.; Olsen, I.H.; Rinke, A.; Borbath, I.; Crespo, G.; Barriuso, J.; Pavel, M.; O’Toole, D.; Walter, T.; et al. Characteristics and treatment of patients with G3 gastroenteropancreatic neuroendocrine neoplasms. Endocr.-Relat. Cancer 2015, 22, 657–664. [Google Scholar] [CrossRef] [Green Version]
  82. Raj, N.; Valentino, E.; Capanu, M.; Tang, L.H.; Basturk, O.; Untch, B.R.; Allen, P.J.; Klimstra, D.S.; Reidy-Lagunes, D. Treatment Response and Outcomes of Grade 3 Pancreatic Neuroendocrine Neoplasms Based on Morphology: Well Differentiated Versus Poorly Differentiated. Pancreas 2017, 46, 296–301. [Google Scholar] [CrossRef]
  83. Sorbye, H.; Kong, G.; Grozinsky-Glasberg, S. PRRT in high-grade gastroenteropancreatic neuroendocrine neoplasms (WHO G3). Endocr.-Relat. Cancer 2020, 27, R67–R77. [Google Scholar] [CrossRef] [PubMed]
  84. Hope, T.A.; Calais, J.; Zhang, L.; Dieckmann, W.; Millo, C. (111)In-Pentetreotide Scintigraphy Versus (68)Ga-DOTATATE PET: Impact on Krenning Scores and Effect of Tumor Burden. J. Nucl. Med. 2019, 60, 1266–1269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Gabriel, M.; Decristoforo, C.; Kendler, D.; Dobrozemsky, G.; Heute, D.; Uprimny, C.; Kovacs, P.; Von Guggenberg, E.; Bale, R.; Virgolini, I.J. 68Ga-DOTA-Tyr3-octreotide PET in neuroendocrine tumors: Comparison with somatostatin receptor scintigraphy and CT. J. Nucl. Med. 2007, 48, 508–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. 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,Tyr3-octreotate) and 18F-FDG. Cancer 2008, 112, 2447–2455. [Google Scholar] [CrossRef]
  87. Deppen, S.A.; Blume, J.; Bobbey, A.J.; Shah, C.; Graham, M.M.; Lee, P.; Delbeke, D.; Walker, R.C. 68Ga-DOTATATE Compared with 111In-DTPA-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]
  88. Kwekkeboom, D.J.; Krenning, E.P. Peptide Receptor Radionuclide Therapy in the Treatment of Neuroendocrine Tumors. Hematol. Oncol. Clin. N. Am. 2016, 30, 179–191. [Google Scholar] [CrossRef]
  89. Minczeles, N.S.; de Herder, W.W.; Konijnenberg, M.W.; Feelders, R.A.; Brabander, T.; Hofland, J. Dose-Limiting Bone Marrow Toxicities after Peptide Receptor Radionuclide Therapy Are More Prevalent in Women Than in Men. Clin. Nucl. Med. 2022, 47, 599–605. [Google Scholar] [CrossRef]
  90. Brabander, T.; Hofland, H. Radionuclide therapy in the time of COVID-19. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 2066–2067. [Google Scholar] [CrossRef]
  91. Bodei, L.; Kidd, M.; Paganelli, G.; Grana, C.M.; Drozdov, I.; Cremonesi, M.; Lepensky, C.; Kwekkeboom, D.J.; Baum, R.P.; Krenning, E.P.; et al. Long-term tolerability of PRRT in 807 patients with neuroendocrine tumours: The value and limitations of clinical factors. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 5–19. [Google Scholar] [CrossRef]
  92. Singh, A.; Mencia-Trinchant, N.; Griffiths, E.A.; Altahan, A.; Swaminathan, M.; Gupta, M.; Gravina, M.; Tajammal, R.; Faber, M.G.; Yan, L.; et al. Mutant PPM1D- and TP53-Driven Hematopoiesis Populates the Hematopoietic Compartment in Response to Peptide Receptor Radionuclide Therapy. JCO Precis. Oncol. 2022, 6, e2100309. [Google Scholar] [CrossRef]
  93. Delpassand, E.S.; Samarghandi, A.; Zamanian, S.; Wolin, E.M.; Hamiditabar, M.; Espenan, G.D.; Erion, J.L.; O’Dorisio, T.M.; Kvols, L.K.; Simon, J.; et al. Peptide receptor radionuclide therapy with 177Lu-DOTATATE for patients with somatostatin receptor-expressing neuroendocrine tumors: The first US phase 2 experience. Pancreas 2014, 43, 518–525. [Google Scholar] [CrossRef] [Green Version]
  94. Bergsma, H.; Konijnenberg, M.W.; Kam, B.L.; Teunissen, J.J.; Kooij, P.P.; de Herder, W.W.; Franssen, G.J.; van Eijck, C.H.; Krenning, E.P.; Kwekkeboom, D.J. Subacute haematotoxicity after PRRT with (177)Lu-DOTA-octreotate: Prognostic factors, incidence and course. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 453–463. [Google Scholar] [CrossRef] [Green Version]
  95. Geenen, L.; Nonnekens, J.; Konijnenberg, M.; Baatout, S.; De Jong, M.; Aerts, A. Overcoming nephrotoxicity in peptide receptor radionuclide therapy using [(177)Lu]Lu-DOTA-TATE for the treatment of neuroendocrine tumours. Nucl. Med. Biol. 2021, 102, 1–11. [Google Scholar] [CrossRef]
  96. Rolleman, E.J.; Valkema, R.; de Jong, M.; Kooij, P.P.; Krenning, E.P. Safe and effective inhibition of renal uptake of radiolabelled octreotide by a combination of lysine and arginine. Eur. J. Nucl. Med. Mol. Imaging 2003, 30, 9–15. [Google Scholar] [CrossRef]
  97. De Jong, M.; Krenning, E. New advances in peptide receptor radionuclide therapy. J. Nucl. Med. 2002, 43, 617–620. [Google Scholar]
  98. Moll, S.; Nickeleit, V.; Mueller-Brand, J.; Brunner, F.P.; Maecke, H.R.; Mihatsch, M.J. A new cause of renal thrombotic microangiopathy: Yttrium 90-DOTATOC internal radiotherapy. Am. J. Kidney Dis. 2001, 37, 847–851. [Google Scholar] [CrossRef]
  99. Bergsma, H.; Konijnenberg, M.W.; van der Zwan, W.A.; Kam, B.L.; Teunissen, J.J.; Kooij, P.P.; Mauff, K.A.; Krenning, E.P.; Kwekkeboom, D.J. Nephrotoxicity after PRRT with (177)Lu-DOTA-octreotate. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 1802–1811. [Google Scholar] [CrossRef] [Green Version]
  100. Stolniceanu, C.R.; Nistor, I.; Bilha, S.C.; Constantin, V.; Simona, V.; Matovic, M.; Stefanescu, C.; Covic, A. Nephrotoxicity/renal failure after therapy with 90Yttrium- and 177Lutetium-radiolabeled somatostatin analogs in different types of neuroendocrine tumors: A systematic review. Nucl. Med. Commun. 2020, 41, 601–617. [Google Scholar] [CrossRef]
  101. Daskalakis, K.; Karakatsanis, A.; Stålberg, P.; Norlén, O.; Hellman, P. Clinical signs of fibrosis in small intestinal neuroendocrine tumours. J. Br. Surg. 2017, 104, 69–75. [Google Scholar] [CrossRef]
  102. Hofman, M.S.; Hicks, R.J. (Eds.) Gallium-68 EDTA PET/CT for Renal Imaging. Seminars in Nuclear Medicine; Elsevier: Amsterdam, The Netherlands, 2016. [Google Scholar]
  103. Van Binnebeek, S.; Baete, K.; Vanbilloen, B.; Terwinghe, C.; Koole, M.; Mottaghy, F.M.; Clement, P.M.; Mortelmans, L.; Haustermans, K.; Van Cutsem, E.; et al. Individualized dosimetry-based activity reduction of 90Y-DOTATOC prevents severe and rapid kidney function deterioration from peptide receptor radionuclide therapy. Eur. J. Nucl. Med. Mol. Imaging 2014, 41, 1141–1157. [Google Scholar] [CrossRef] [Green Version]
  104. Krajewski, W.; Wojciechowska, J.; Dembowski, J.; Zdrojowy, R.; Szydełko, T. Hydronephrosis in the course of ureteropelvic junction obstruction: An underestimated problem? Current opinions on the pathogenesis, diagnosis and treatment. Adv. Clin. Exp. Med. 2017, 26, 857–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Smits, M.L.; Nijsen, J.F.; van den Bosch, M.A.; Lam, M.G.; Vente, M.A.; Mali, W.P.; van Het Schip, A.D.; Zonnenberg, B.A. Holmium-166 radioembolisation in patients with unresectable, chemorefractory liver metastases (HEPAR trial): A phase 1, dose-escalation study. Lancet Oncol. 2012, 13, 1025–1034. [Google Scholar] [CrossRef] [PubMed]
  106. Braat, A.; Ahmadzadehfar, H.; Kappadath, S.C.; Stothers, C.L.; Frilling, A.; Deroose, C.M.; Flamen, P.; Brown, D.B.; Sze, D.Y.; Mahvash, A.; et al. Radioembolization with (90)Y Resin Microspheres of Neuroendocrine Liver Metastases after Initial Peptide Receptor Radionuclide Therapy. Cardiovasc. Intervent. Radiol. 2020, 43, 246–253. [Google Scholar]
  107. Ezziddin, S.; Meyer, C.; Kahancova, S.; Haslerud, T.; Willinek, W.; Wilhelm, K.; Biersack, H.-J.; Ahmadzadehfar, H. 90Y Radioembolization after radiation exposure from peptide receptor radionuclide therapy. J. Nucl. Med. 2012, 53, 1663–1669. [Google Scholar] [CrossRef] [Green Version]
  108. De Keizer, B.; van Aken, M.O.; Feelders, R.A.; de Herder, W.W.; Kam, B.L.; van Essen, M.; Krenning, E.P.; Kwekkeboom, D.J. Hormonal crises following receptor radionuclide therapy with the radiolabeled somatostatin analogue [177Lu-DOTA0,Tyr3]octreotate. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 749–755. [Google Scholar] [CrossRef] [Green Version]
  109. Del Olmo-García, M.I.; Muros, M.A.; López-de-la-Torre, M.; Agudelo, M.; Bello, P.; Soriano, J.M.; Merino-Torres, J.F. Prevention and Management of Hormonal Crisis during Theragnosis with LU-DOTA-TATE in Neuroendocrine Tumors. A Systematic Review and Approach Proposal. J. Clin. Med. 2020, 9, 2203. [Google Scholar] [CrossRef]
  110. Hlatky, R.; Suki, D.; Sawaya, R. Carcinoid metastasis to the brain. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2004, 101, 2605–2613. [Google Scholar] [CrossRef]
  111. Pavel, M.; Grossman, A.; Arnold, R.; Perren, A.; Kaltsas, G.; Steinmüller, T.; De Herder, W.; Nikou, G.; Plöckinger, U.; Lopes, J.M. ENETS consensus guidelines for the management of brain, cardiac and ovarian metastases from neuroendocrine tumors. Neuroendocrinology 2010, 91, 326–332. [Google Scholar] [CrossRef] [Green Version]
  112. Scharf, M.; Petry, V.; Daniel, H.; Rinke, A.; Gress, T.M. Bone metastases in patients with neuroendocrine neoplasm: Frequency and clinical, therapeutic, and prognostic relevance. Neuroendocrinology 2018, 106, 30–37. [Google Scholar] [CrossRef]
  113. Cecchin, D.; Schiavi, F.; Fanti, S.; Favero, M.; Manara, R.; Fassina, A.; Briani, C.; Allegri, V.; Sansovini, M.; Bui, F.; et al. Peptide receptor radionuclide therapy in a case of multiple spinal canal and cranial paragangliomas. J. Clin. Oncol. 2011, 29, e171–e174. [Google Scholar] [CrossRef]
  114. De Bruin, C.; Hofland, L.J.; Nieman, L.K.; van Koetsveld, P.M.; Waaijers, A.M.; Sprij-Mooij, D.M.; van Essen, M.; Lamberts, S.W.; de Herder, W.W.; Feelders, R.A. Mifepristone effects on tumor somatostatin receptor expression in two patients with Cushing’s syndrome due to ectopic adrenocorticotropin secretion. J. Clin. Endocrinol. Metab. 2012, 97, 455–462. [Google Scholar] [CrossRef]
  115. Blažević, A.; Hofland, J.; Hofland, L.J.; Feelders, R.A.; de Herder, W.W. Small intestinal neuroendocrine tumours and fibrosis: An entangled conundrum. Endocr.-Relat. Cancer 2018, 25, R115–R130. [Google Scholar] [CrossRef] [Green Version]
  116. Merola, E.; Prasad, V.; Pascher, A.; Pape, U.-F.; Arsenic, R.; Denecke, T.; Fehrenbach, U.; Wiedenmann, B.; Pavel, M.E. Peritoneal carcinomatosis in gastro-entero-pancreatic neuroendocrine neoplasms: Clinical impact and effectiveness of the available therapeutic options. Neuroendocrinology 2020, 110, 517–524. [Google Scholar] [CrossRef]
  117. Strosberg, J.R.; Al-Toubah, T.; Pellè, E.; Smith, J.; Haider, M.; Hutchinson, T.; Fleming, J.B.; El-Haddad, G. Risk of Bowel Obstruction in Patients with Mesenteric or Peritoneal Disease Receiving Peptide Receptor Radionuclide Therapy. J. Nucl. Med. 2021, 62, 69–72. [Google Scholar] [CrossRef]
  118. Paganelli, G.; Sansovini, M.; Ambrosetti, A.; Severi, S.; Monti, M.; Scarpi, E.; Donati, C.; Ianniello, A.; Matteucci, F.; Amadori, D. 177 Lu-Dota-octreotate radionuclide therapy of advanced gastrointestinal neuroendocrine tumors: Results from a phase II study. Eur. J. Nucl. Med. Mol. Imaging 2014, 41, 1845–1851. [Google Scholar] [CrossRef]
  119. Blažević, A.; Brabander, T.; Zandee, W.T.; Hofland, J.; Franssen, G.J.H.; van Velthuysen, M.-L.F.; Feelders, R.A.; De Herder, W.W. Evolution of the mesenteric mass in small intestinal neuroendocrine tumours. Cancers 2021, 13, 443. [Google Scholar] [CrossRef]
  120. Nonnekens, J.; van Kranenburg, M.; Beerens, C.E.; Suker, M.; Doukas, M.; van Eijck, C.H.; de Jong, M.; van Gent, D.C. Potentiation of Peptide Receptor Radionuclide Therapy by the PARP Inhibitor Olaparib. Theranostics 2016, 6, 1821–1832. [Google Scholar] [CrossRef] [Green Version]
  121. Hofving, T.; Sandblom, V.; Arvidsson, Y.; Shubbar, E.; Altiparmak, G.; Swanpalmer, J.; Almobarak, B.; Elf, A.-K.; Johanson, V.; Elias, E. 177Lu-octreotate therapy for neuroendocrine tumours is enhanced by Hsp90 inhibition. Endocr.-Relat. Cancer 2019, 26, 437–449. [Google Scholar] [CrossRef]
  122. Ballal, S.; Yadav, M.P.; Bal, C.; Sahoo, R.K.; Tripathi, M. Broadening horizons with 225Ac-DOTATATE targeted alpha therapy for gastroenteropancreatic neuroendocrine tumour patients stable or refractory to 177Lu-DOTATATE PRRT: First clinical experience on the efficacy and safety. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 934–946. [Google Scholar] [CrossRef]
  123. Kratochwil, C.; Giesel, F.L.; Bruchertseifer, F.; Mier, W.; Apostolidis, C.; Boll, R.; Murphy, K.; Haberkorn, U.; Morgenstern, A. 213Bi-DOTATOC receptor-targeted alpha-radionuclide therapy induces remission in neuroendocrine tumours refractory to beta radiation: A first-in-human experience. Eur. J. Nucl. Med. Mol. Imaging 2014, 41, 2106–2119. [Google Scholar] [CrossRef] [Green Version]
  124. Stallons, T.A.R.; Saidi, A.; Tworowska, I.; Delpassand, E.S.; Torgue, J.J. Preclinical Investigation of 212Pb-DOTAMTATE for Peptide Receptor Radionuclide Therapy in a Neuroendocrine Tumor Model212Pb-DOTAMTATE for Treatment of Neuroendocrine Tumors. Mol. Cancer Ther. 2019, 18, 1012–1021. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanism of action of PRRT with 177Lu-DOTATATE. Intravenous administration of 177Lu-DOTATATE leads to tumor cell binding via SSTR2. After internalization of the radiopharmaceutical–SSTR2 complex, local radiation by beta particles can lead to cell death through the induction of DNA damage (image created with BioRender.com, accessed on 1 September 2022).
Figure 1. Mechanism of action of PRRT with 177Lu-DOTATATE. Intravenous administration of 177Lu-DOTATATE leads to tumor cell binding via SSTR2. After internalization of the radiopharmaceutical–SSTR2 complex, local radiation by beta particles can lead to cell death through the induction of DNA damage (image created with BioRender.com, accessed on 1 September 2022).
Cancers 14 05792 g001
Figure 2. Potential complicated clinical situations for PRRT. (A) 68Ga-DOTATATE PET scan of extensive bone metastases of SI-NET. R-PRRT with reduced activity of 3.7 GBq 177Lu-DOTATATE induced hematoxicity. (B) Hydronephrosis (1) due to obstruction by peritoneal tumor deposit of metastatic panNET (2), leading to accumulation of radioactivity in renal medulla. (C) 68Ga-DOTATATE PET scan showing extensive liver metastases of panNET before treatment with 3.7 GBq 177Lu-DOTATATE, which was well tolerated by the patient. (D) Bone metastasis of panNET with epidural extension at Th12 on MRI. Post-PRRT edema can lead to infringement of the spinal cord. (E) Patient with small bowel NET and metastatic mesenteric mass with desmoplastic reaction, resulting in venous congestion, bowel wall thickening and ascites. The patient suffered from intermittent abdominal pain with temporary aggravation of complaints during PRRT. (F) Contrast enhanced CT scan of peritoneal metastases of a multifocal NET of a patient who developed a paralytic ileus or ‘frozen abdomen’ after PRRT.
Figure 2. Potential complicated clinical situations for PRRT. (A) 68Ga-DOTATATE PET scan of extensive bone metastases of SI-NET. R-PRRT with reduced activity of 3.7 GBq 177Lu-DOTATATE induced hematoxicity. (B) Hydronephrosis (1) due to obstruction by peritoneal tumor deposit of metastatic panNET (2), leading to accumulation of radioactivity in renal medulla. (C) 68Ga-DOTATATE PET scan showing extensive liver metastases of panNET before treatment with 3.7 GBq 177Lu-DOTATATE, which was well tolerated by the patient. (D) Bone metastasis of panNET with epidural extension at Th12 on MRI. Post-PRRT edema can lead to infringement of the spinal cord. (E) Patient with small bowel NET and metastatic mesenteric mass with desmoplastic reaction, resulting in venous congestion, bowel wall thickening and ascites. The patient suffered from intermittent abdominal pain with temporary aggravation of complaints during PRRT. (F) Contrast enhanced CT scan of peritoneal metastases of a multifocal NET of a patient who developed a paralytic ileus or ‘frozen abdomen’ after PRRT.
Cancers 14 05792 g002
Table 1. Overview of tumor control and symptom control following PRRT as described in key phase II and III clinical trials.
Table 1. Overview of tumor control and symptom control following PRRT as described in key phase II and III clinical trials.
Patients, nNET SubtypePFS (Months)mOS (Months)ORRSDPDmTTD QOL Global Health (Months)Overall Symptom Improvement
Strosberg et al. [27,30,38]101SI-NET284818%66% *16%29 m
  Control group SSA100SI-NET8.5363%41%56%6.1 m
Brabander et al. [29]443GEP and bronchial NET296339%43%12%
Bodei et al. [36]51GEP and bronchial NET3668%
at 36 months
55%27%18%
Ezziddin et al. [34]68GEP-NET345372%13%15%
Sabet et al. [35]61SI-NET 44%48%8%
Hamiditabar et al. [33]143NET of all origins 8%46%38% 47%
Khan et al. [39]265GEP and bronchial NET 38–90%
PFS = progression-free survival, mOS = median overall survival, ORR = objective response rate, SD = stable disease, PD = progressive disease, mTTD QOL = median time to deterioration in quality of life, m = months. * Estimated percentage based on calculations.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Becx, M.N.; Minczeles, N.S.; Brabander, T.; de Herder, W.W.; Nonnekens, J.; Hofland, J. A Clinical Guide to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE in Neuroendocrine Tumor Patients. Cancers 2022, 14, 5792. https://doi.org/10.3390/cancers14235792

AMA Style

Becx MN, Minczeles NS, Brabander T, de Herder WW, Nonnekens J, Hofland J. A Clinical Guide to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE in Neuroendocrine Tumor Patients. Cancers. 2022; 14(23):5792. https://doi.org/10.3390/cancers14235792

Chicago/Turabian Style

Becx, Morticia N., Noémie S. Minczeles, Tessa Brabander, Wouter W. de Herder, Julie Nonnekens, and Johannes Hofland. 2022. "A Clinical Guide to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE in Neuroendocrine Tumor Patients" Cancers 14, no. 23: 5792. https://doi.org/10.3390/cancers14235792

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