[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (58)

Search Parameters:
Keywords = 177Lu-DOTA-TATE

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 1217 KiB  
Article
Hematological Side Effects of 177Lu-DOTA-TATE Therapy in Patients with NENs
by Luciano Carideo, Rosaria Meucci, Giuseppe Campagna, Vincenzo Marcello Russo, Enrico D’Ippolito, Maria Rinzivillo, Francesco Panzuto and Daniela Prosperi
Hemato 2025, 6(1), 1; https://doi.org/10.3390/hemato6010001 - 30 Dec 2024
Viewed by 323
Abstract
Background/Objectives: Lutathera® ([177Lu]Lu-DOTA-TATE) is the first radiolabelled somatostatin (SST) analog approved for the treatment of patients with well-differentiated (G1 and G2) unresectable or metastatic gastro-entero-pancreatic neuro-endocrine-neoplasms (GEP-NENs). The bone marrow and kidneys are critical organs for RLT with [ [...] Read more.
Background/Objectives: Lutathera® ([177Lu]Lu-DOTA-TATE) is the first radiolabelled somatostatin (SST) analog approved for the treatment of patients with well-differentiated (G1 and G2) unresectable or metastatic gastro-entero-pancreatic neuro-endocrine-neoplasms (GEP-NENs). The bone marrow and kidneys are critical organs for RLT with [177Lu]Lu-DOTA-TATE. Our purpose was to evaluate hematological and renal toxicity in 29 patients (18 males, 11 females) treated with Lutathera®. Methods: According to standard protocols, four cycles of (177Lu)Lu-DOTA-TATE were administered every eight/nine weeks. Patients received pre-medication with anti-emetic and anti-acid drugs and a slow amino acid infusion for renal protection. Blood count and serum creatinine data were collected at three time points: before the first cycle, after the second cycle, and at the end of treatment. Results: We found that almost all hematological parameters significantly decreased between the baseline and/or interim and post-therapy evaluation, although without a clinical impact. The presence of total tumor load or bone metastases had no influence on these findings, while male patients showed less hematological toxicity than females. Conversely, creatinine levels did not vary during treatment. Conclusions: Our study confirms that [177Lu]Lu-DOTATATE RLT is safe and well tolerated despite some minor (grade 1) hematological toxicity. Full article
(This article belongs to the Section Radiolabeled Blood Elements and Other Imaging Modalities)
Show Figures

Figure 1

Figure 1
<p>Trend of red blood cells in 29 patients.</p>
Full article ">Figure 2
<p>Trend of platelet count in 29 patients.</p>
Full article ">Figure 3
<p>Trend of white blood cells in 29 patients.</p>
Full article ">
12 pages, 1555 KiB  
Article
Peptide Receptor Radionuclide Therapy Using 90Y- and 177Lu-DOTATATE Modulating Atherosclerotic Plaque Inflammation: Longitudinal Monitoring by 68Ga-DOTATATE Positron Emissions Tomography/Computer Tomography
by German Rubinstein, Harun Ilhan, Peter Bartenstein, Sebastian Lehner, Marcus Hacker, Andrei Todica, Mathias Johannes Zacherl and Maximilian Fischer
Diagnostics 2024, 14(22), 2486; https://doi.org/10.3390/diagnostics14222486 - 7 Nov 2024
Viewed by 646
Abstract
Background: Atherosclerosis and its sequels, such as coronary artery disease and cerebrovascular stroke, still represent global health burdens. The pathogenesis of atherosclerosis consists of growing calcified plaques in the arterial wall and is accompanied by inflammatory processes, which are not entirely understood. This [...] Read more.
Background: Atherosclerosis and its sequels, such as coronary artery disease and cerebrovascular stroke, still represent global health burdens. The pathogenesis of atherosclerosis consists of growing calcified plaques in the arterial wall and is accompanied by inflammatory processes, which are not entirely understood. This study aims to evaluate the effect of peptide receptor radionuclide therapy (PRRT) using 90Y- and 177Lu-DOTATATE on atherosclerotic plaque inflammation. Methods: Atherosclerotic plaques in 57 cancer patients receiving PRRT using 90Y- and 177Lu-DOTATATE were longitudinally monitored by 68Ga-DOTATATE PET/CT. The target-to-background ratio (TBR) and overall vessel uptake (OVU) were measured in eight distinct arterial regions (ascending aorta, aortic arch, descending aorta, abdominal aorta, both iliac arteries, and both carotid arteries) to monitor calcified plaques. Results: PET/CT analysis shows a positive correlation between calcified plaque scores and the 68Ga-DOTATATE overall vessel uptake (OVU) in cancer patients. After PRRT, an initially high OVU was observed to decrease in the therapy group compared to the control group. An excellent correlation could be shown for each target-to-background ratio (TBR) to the OVU, especially the ascending aorta. Conclusions: The ascending aorta could present a future reference for estimating generalized atherosclerotic inflammatory processes. PRRT might represent a therapeutic approach to modulating atherosclerotic plaques. Full article
(This article belongs to the Section Medical Imaging and Theranostics)
Show Figures

Figure 1

Figure 1
<p>PET/CT image analysis. (<b>A</b>) Example of modified calcified plaque (CP) scoring system. Representative images show scores of 0 (no calcified plaque), 1 (calcified plaque involving 10% of vessel circumference), 2 (calcified plaque involving 10% to 25%), 3 (calcified plaque involving 25% to 50%), and 4 (calcified plaque involving &gt;50%). (<b>B</b>) Representative image for target-to-background (TBR) calculation method. ROIs were drawn in the center of the vena cava inferior (blue) and arterial wall (red) for SUV evaluation.</p>
Full article ">Figure 2
<p>Correlation analysis of CP score, age, and OVU in the first PET/CT scan prior to PRRT therapy. <span class="html-italic">N</span> = 57.</p>
Full article ">Figure 3
<p>Boxplots illustrating the therapy group’s longitudinal assessment of the OVU, including the first to third <sup>68</sup>Ga-DOTATATE scan.</p>
Full article ">Figure 4
<p>Boxplot illustration of the OVU course in therapy and control group in all three PET/CTs. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
13 pages, 679 KiB  
Article
Adverse Events of Radioligand Therapy in Patients with Progressive Neuroendocrine Neoplasms: The Biggest Eastern European Prospective Study
by Adam Daniel Durma, Marek Saracyn, Maciej Kołodziej, Katarzyna Jóźwik-Plebanek, Dorota Brodowska-Kania, Beata Dmochowska, Adrianna Mróz, Beata Kos-Kudła and Grzegorz Kamiński
Cancers 2024, 16(20), 3509; https://doi.org/10.3390/cancers16203509 - 17 Oct 2024
Viewed by 725
Abstract
Background: Neuroendocrine neoplasms (NENs) are neoplastic tumors developing in every part of the body, mainly in the gastrointestinal tract and pancreas. Their treatment involves the surgical removal of the tumor and its metastasis, long-acting somatostatin analogs, chemotherapy, targeted therapy, and radioligand therapy (RLT). [...] Read more.
Background: Neuroendocrine neoplasms (NENs) are neoplastic tumors developing in every part of the body, mainly in the gastrointestinal tract and pancreas. Their treatment involves the surgical removal of the tumor and its metastasis, long-acting somatostatin analogs, chemotherapy, targeted therapy, and radioligand therapy (RLT). Materials and Methods: A total of 127 patients with progressive neuroendocrine neoplasms underwent RLT—4 courses, administered every 10 weeks—with the use of 7.4 GBq [177Lu]Lu-DOTA-TATE or tandem therapy with 1.85 GBq [177Lu]Lu-DOTA-TATE and 1.85 GBq [90Y]Y-DOTA-TATE. Assessment of short- and long-term complications, as well as the calculation of progression-free survival (PFS) and overall survival (OS) were performed. Results: RLT caused a statistically but not clinically significant decrease in blood morphology parameters during both short- and long-term observations. Glomerular filtration rate (GFR) significantly decreased only in a long-term observation after RLT; however, it was clinically acceptable. Computed predictions of progression-free survival (PFS) and overall survival (OS) indicated that five years post-RLT, there is a 74% chance of patients surviving, with only a 58.5% likelihood of disease progression. Conclusions: Computed predictions of PFS and OS confirmed treatment efficiency and good patient survival. RLT should be considered a safe and reliable line of treatment for patients with progressive NENs as it causes only a low number of low-grade adverse events. Full article
(This article belongs to the Special Issue Radioligand Therapy (RLT) in Neuroendocrine Neoplasms)
Show Figures

Figure 1

Figure 1
<p>Overall survival probability calculations.</p>
Full article ">Figure 2
<p>Progression-free survival probability calculations.</p>
Full article ">
13 pages, 2268 KiB  
Article
The Safety and Efficacy of Peptide Receptor Radionuclide Therapy for Gastro-Entero-Pancreatic Neuroendocrine Tumors: A Single Center Experience
by Leandra Piscopo, Emilia Zampella, Fabio Volpe, Valeria Gaudieri, Carmela Nappi, Erica Di Donna, Stefania Clemente, Antonio Varallo, Mariano Scaglione, Alberto Cuocolo and Michele Klain
Curr. Oncol. 2024, 31(9), 5617-5629; https://doi.org/10.3390/curroncol31090416 - 18 Sep 2024
Viewed by 881
Abstract
The aim of the present study was to evaluate the safety and efficacy of radionuclide therapy with [177Lu]Lu-DOTA-TATE according to our single center experience at the University of Naples Federico II. For the present analysis, we considered 21 patients with progressive, [...] Read more.
The aim of the present study was to evaluate the safety and efficacy of radionuclide therapy with [177Lu]Lu-DOTA-TATE according to our single center experience at the University of Naples Federico II. For the present analysis, we considered 21 patients with progressive, advanced, well-differentiated G1 and G2 in patients with gastro-entero-pancreatic (GEP) neuroendocrine tumors (NETs) treated with [177Lu]Lu-DOTA-TATE according to the decisions of a multidisciplinary team. All patients underwent four cycles of 7–8 GBq of [177Lu]Lu-DOTA-TATE every 8 weeks. A whole-body scan (WBS) was performed 4, 48, and 168 h after each treatment. The dosimetry towards the organ at risk and target lesions was calculated. For each patient, renal and bone marrow parameters were evaluated before, during, and 3 months after the end of the treatment. Follow-up data were obtained and RECIST criteria were considered as the endpoint. Among 21 patients enrolled (mean age 65 ± 9 years); 17 (81%) were men and the small intestine was the most frequent location of disease (n = 12). A mild albeit significant variation (p < 0.05) in both platelets and white blood cell counts among all time points was observed, despite it disappearing 3 months after the end of the therapy. According to the RECIST criteria, 11 (55%) patients had a partial response to therapy and 8 (40%) had stable disease. Only one (5%) patient had disease progression 4 months after treatment. Our data confirm that [177Lu]Lu-DOTA is safe and effective in controlling the burden disease of G1/G2 GEP-NETs patients. Full article
(This article belongs to the Special Issue Application of Nuclear Medicine in Cancer Diagnosis and Treatment)
Show Figures

Figure 1

Figure 1
<p>Comparison of white blood cells, hemoglobin, and platelets dosage before each PRRT cycle and 3 months after the treatment.</p>
Full article ">Figure 1 Cont.
<p>Comparison of white blood cells, hemoglobin, and platelets dosage before each PRRT cycle and 3 months after the treatment.</p>
Full article ">Figure 2
<p>Distribution of response to therapy in the entire population according to RECIST criteria.</p>
Full article ">Figure 3
<p>MIP views and transaxial fusion images of <sup>68</sup>Ga-DOTATOC PET/CT scans performed before PRRT (<b>A</b>), WBS scans after [<sup>177</sup>Lu]Lu-DOTA-TATE (<b>B</b>), and MIP views and transaxial fusion images of a post-therapy <sup>68</sup>Ga-DOTATOC PET/CT scan (<b>C</b>) in a patient with metastases to the liver and abdominal lymph nodes from pancreatic NET. The focal uptake on liver and lymph node metastases is reduced on the post-therapy images.</p>
Full article ">Figure 3 Cont.
<p>MIP views and transaxial fusion images of <sup>68</sup>Ga-DOTATOC PET/CT scans performed before PRRT (<b>A</b>), WBS scans after [<sup>177</sup>Lu]Lu-DOTA-TATE (<b>B</b>), and MIP views and transaxial fusion images of a post-therapy <sup>68</sup>Ga-DOTATOC PET/CT scan (<b>C</b>) in a patient with metastases to the liver and abdominal lymph nodes from pancreatic NET. The focal uptake on liver and lymph node metastases is reduced on the post-therapy images.</p>
Full article ">Figure 4
<p>MIP views and transaxial fusion images of <sup>68</sup>Ga-DOTATOC PET/CT scans performed before PRRT (<b>A</b>), WBS scans after [<sup>177</sup>Lu]Lu-DOTA-TATE (<b>B</b>), and MIP views and transaxial fusion images of a post-therapy <sup>68</sup>Ga-DOTATOC PET/CT scan (<b>C</b>) in a patient with metastases to the bone, liver and abdominal lymph nodes from ileal G1 NET. The uptake on bone, liver and lymph node metastases is stable in the post-therapy images.</p>
Full article ">Figure 4 Cont.
<p>MIP views and transaxial fusion images of <sup>68</sup>Ga-DOTATOC PET/CT scans performed before PRRT (<b>A</b>), WBS scans after [<sup>177</sup>Lu]Lu-DOTA-TATE (<b>B</b>), and MIP views and transaxial fusion images of a post-therapy <sup>68</sup>Ga-DOTATOC PET/CT scan (<b>C</b>) in a patient with metastases to the bone, liver and abdominal lymph nodes from ileal G1 NET. The uptake on bone, liver and lymph node metastases is stable in the post-therapy images.</p>
Full article ">
8 pages, 1496 KiB  
Article
Safety and Efficacy of Peptide Receptor Radionuclide Therapy (PRRT) Following Bland Embolization for Metastatic Neuroendocrine Tumors
by Adam Alayli, Hoang Ngo, Dhiraj Sikaria, Altan Ahmed, Elias Salloum, Jonathan R. Strosberg, Taymeyah E. Al-Toubah, Bela Kis, Mintallah Haider and Ghassan El-Haddad
Cancers 2024, 16(15), 2703; https://doi.org/10.3390/cancers16152703 - 30 Jul 2024
Cited by 1 | Viewed by 1009
Abstract
Rationale: Evaluating the long-term safety and efficacy of peptide receptor radionuclide therapy (PRRT) in patients with metastatic neuroendocrine tumors (mNETs) who have undergone prior bland hepatic transarterial embolization (TAE). Methods: Retrospective review of mNET patients who received PRRT with 177Lu-DOTATATE [...] Read more.
Rationale: Evaluating the long-term safety and efficacy of peptide receptor radionuclide therapy (PRRT) in patients with metastatic neuroendocrine tumors (mNETs) who have undergone prior bland hepatic transarterial embolization (TAE). Methods: Retrospective review of mNET patients who received PRRT with 177Lu-DOTATATE between 4/2018 and 02/2022 with and without prior TAE. The most recent clinical, imaging, and laboratory findings, including hepatic Common Terminology Criteria for Adverse Events v5.0, were compared to pre-PRRT. Results: 171 patients (95 M, 76 F, median age = 66) with mNET of different primary sites (9 foregut, 100 midgut, 9 hindgut, 44 pancreas, 9 unknown) received at least 1 cycle of PRRT with at least 6 months of follow-up, 110 of whom were embolization-naïve and 61 who had prior TAE. The median follow up was 22 months (range: 6–43). Patients with prior TAE had higher liver tumor burden on average than patients without prior TAE; however, the difference was not statistically significant (p = 0.06). There was no significant difference in the rates of G3 or G4 hepatotoxicity (p = 0.548 and p = 0.999, respectively) in patients who underwent prior TAE and those who were TAE-naïve. The hepatic progression-free survival was 22.9 months in TAE-naïve patients and 25.7, 20.2, and 12.8 months in patients with 1, 2, and 3 prior TAE treatments, respectively. Conclusion: Peptide receptor radionuclide therapy following transarterial bland embolization for mNET is safe and effective. Full article
(This article belongs to the Special Issue Updates in Neuroendocrine Neoplasms)
Show Figures

Figure 1

Figure 1
<p>Flowchart showing patient selection.</p>
Full article ">Figure 2
<p>Kaplan–Meier curve showing hepatic progression-free survival.</p>
Full article ">Figure 3
<p>Kaplan–Meier curve showing global progression-free survival.</p>
Full article ">
16 pages, 4878 KiB  
Article
Investigating the Radiobiological Response to Peptide Receptor Radionuclide Therapy Using Patient-Derived Meningioma Spheroids
by Thom G. A. Reuvers, Vivian Grandia, Renata M. C. Brandt, Majd Arab, Sybren L. N. Maas, Eelke M. Bos and Julie Nonnekens
Cancers 2024, 16(14), 2515; https://doi.org/10.3390/cancers16142515 - 11 Jul 2024
Cited by 1 | Viewed by 1218
Abstract
Peptide receptor radionuclide therapy (PRRT) using 177Lu-DOTA-TATE has recently been evaluated for the treatment of meningioma patients. However, current knowledge of the underlying radiation biology is limited, in part due to the lack of appropriate in vitro models. Here, we demonstrate proof-of-concept [...] Read more.
Peptide receptor radionuclide therapy (PRRT) using 177Lu-DOTA-TATE has recently been evaluated for the treatment of meningioma patients. However, current knowledge of the underlying radiation biology is limited, in part due to the lack of appropriate in vitro models. Here, we demonstrate proof-of-concept of a meningioma patient-derived 3D culture model to assess the short-term response to radiation therapies such as PRRT and external beam radiotherapy (EBRT). We established short-term cultures (1 week) for 16 meningiomas with high efficiency and yield. In general, meningioma spheroids retained characteristics of the parental tumor during the initial days of culturing. For a subset of tumors, clear changes towards a more aggressive phenotype were visible over time, indicating that the culture method induced dedifferentiation of meningioma cells. To assess PRRT efficacy, we demonstrated specific uptake of 177Lu-DOTA-TATE via somatostatin receptor subtype 2 (SSTR2), which was highly overexpressed in the majority of tumor samples. PRRT induced DNA damage which was detectable for an extended timeframe as compared to EBRT. Interestingly, levels of DNA damage in spheroids after PRRT correlated with SSTR2-expression levels of parental tumors. Our patient-derived meningioma culture model can be used to assess the short-term response to PRRT and EBRT in radiobiological studies. Further improvement of this model should pave the way towards the development of a relevant culture model for assessment of the long-term response to radiation and, potentially, individual patient responses to PRRT and EBRT. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

Figure 1
<p>Generation of meningioma tumor tissue-originated spheroid model. Serum-free = growth factor-supplemented and serum = FCS-supplemented medium. (<b>A</b>) General overview of establishment of spheroid model from moment of resection to culturing. (<b>B</b>) Morphology of cultured meningioma fragments in different medium types directly after dissociation, at day 1 and day 7 of culturing. (<b>C</b>) Live/dead staining of meningioma fragments at day 1 and day 7 of culturing, as assessed by calcein AM (<b>middle</b> panel, green, live cells) and BOBO-3 Iodide (<b>right</b> panel, red, dead cells) staining. Images for a negative control (fragments treated with 70% ethanol), staining negative for Calcein AM and positive for BOBO-3-Iodide are shown in the <b>bottom</b> panel.</p>
Full article ">Figure 2
<p>Assessment of spheroid phenotype by H&amp;E and IHC for Ki-67 and SSTR2 compared to parental tumors. H&amp;E staining (<b>top</b> panel) and IHC staining for Ki-67 (<b>middle</b> panel) and SSTR2 (<b>bottom</b> panel) for parental tumor tissue and spheroids cultured for 1, 3, or 7 days. Example images are shown for spheroids derived from MO-10 (<b>A</b>) and MO-02 (<b>B</b>), only for spheroids cultured in FCS-containing medium. For results for spheroids cultured in growth factor-supplemented medium, see <a href="#app1-cancers-16-02515" class="html-app">Supplementary Figure S1</a>.</p>
Full article ">Figure 3
<p>Binding of <sup>177</sup>Lu-DOTA-TATE to meningioma spheroids. (<b>A</b>) Uptake assay determining quantity of radioactivity in counts per minute (cpm) in meningioma spheroids after <sup>177</sup>Lu-DOTA-TATE treatment with (black bars) or without (grey bars) co-treatment with 100× molar excess of DOTA-TATE. For each meningioma, data are shown for spheroids incubated with 0.1 and 1 MBq/mL <sup>177</sup>Lu-DOTA-TATE. (<b>B</b>) Representative tile scan overview from MO-10 of IF staining for SSTR2 (green) for control spheroids, spheroids treated with PRRT (1 MBq/mL) or spheroids treated with PRRT co-incubated with DOTA-TATE. DAPI (blue) was used as a nuclear counterstain. Directly after treatment with either PRRT or PRRT and DOTA-TATE, loss of membranous signal and intracellular accumulation of SSTR2 can be seen throughout the spheroid. White squares indicate the zoomed-in regions, shown right from each corresponding tile scan for the peripheral (1) or core (2) region of the spheroid.</p>
Full article ">Figure 4
<p>Assessment of DNA damage in meningioma spheroids after EBRT and PRRT. (<b>A</b>,<b>D</b>) IHC staining for SSTR2 in parental tumor tissue for a meningioma staining strongly (MO-14, (<b>A</b>) and weakly (MO-12 (<b>D</b>)) SSTR2-positive. (<b>B</b>,<b>E</b>) Representative images of 53BP1 IF staining (red) used for the quantifications in (<b>C</b>,<b>F</b>). Examples are shown for EBRT (2 Gy)- and PRRT (1 MBq/mL)-treated spheroids at both day 0 and day 2 post-therapy. DAPI (blue) was used as a nuclear counterstain. (<b>C</b>,<b>F</b>) Quantification of the number of 53BP1 foci per nucleus after EBRT, PRRT, or PRRT co-incubated with DOTA-TATE at day 0 or day 2 post-therapy for MO-14 (<b>C</b>) and MO-12 (<b>F</b>). Black horizontal bars indicate the mean. ns = not significant; *** <span class="html-italic">p</span> ≤ 0.001; **** <span class="html-italic">p</span> ≤ 0.0001. Line graphs on the right show the mean number of 53BP1 foci for each condition. Quantification for other meningioma samples can be found in <a href="#app1-cancers-16-02515" class="html-app">Supplementary Figure S2</a>.</p>
Full article ">
11 pages, 2769 KiB  
Article
Effects of Peptide Receptor Radiotherapy in Patients with Advanced Paraganglioma and Pheochromocytoma: A Nation-Wide Cohort Study
by Linda Skibsted Kornerup, Mikkel Andreassen, Ulrich Knigge, Anne Kirstine Arveschoug, Per Løgstup Poulsen, Andreas Kjær, Peter Sandor Oturai, Henning Grønbæk and Gitte Dam
Cancers 2024, 16(7), 1349; https://doi.org/10.3390/cancers16071349 - 29 Mar 2024
Cited by 2 | Viewed by 1369
Abstract
Introduction: Pheochromocytomas and paragangliomas are rare neuroendocrine tumours that originate from chromaffin cells within the adrenal medulla or extra-adrenal sympathetic ganglia. Management of disseminated or metastatic pheochromocytomas and paragangliomas continues to pose challenges and relies on limited evidence. Method: In this study, we [...] Read more.
Introduction: Pheochromocytomas and paragangliomas are rare neuroendocrine tumours that originate from chromaffin cells within the adrenal medulla or extra-adrenal sympathetic ganglia. Management of disseminated or metastatic pheochromocytomas and paragangliomas continues to pose challenges and relies on limited evidence. Method: In this study, we report retrospective data on median overall survival (OS) and median progression-free survival (PFS) for all Danish patients treated with peptide receptor radionuclide therapy (PRRT) with 177Lu-Dotatate or 90Y-Dotatate over the past 15 years. One standard treatment of PRRT consisted of 4 consecutive cycles with 8–14-week intervals. Results: We included 28 patients; 10 were diagnosed with pheochromocytoma and 18 with paraganglioma. Median age at first PRRT was 47 (IQR 15–76) years. The median follow-up time was 31 (IQR 17–37) months. Eight patients died during follow-up. Median OS was 72 months, and 5-year survival was 65% with no difference between pheochromocytoma and paraganglioma. Patients with germline mutations had better survival than patients without mutations (p = 0.041). Median PFS after the first cycle of PRRT was 30 months. For patients who previously received systemic treatment, the median PFS was 19 months, compared with 32 months for patients with no previous systemic treatment (p = 0.083). Conclusions: The median OS of around 6 years and median PFS of around 2.5 years found in this study are comparable to those reported in previous studies employing PRRT. Based on historical data, the efficacy of PRRT may be superior to 131I-MIBG therapy, and targeted therapy with sunitinib and PRRT might therefore be considered as first-line treatment in this patient group. Full article
(This article belongs to the Special Issue Neuroendocrine Tumors: From Diagnosis to Therapy)
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier survival plots displaying median overall survival after the first PRRT cycle for; (<b>A</b>) all patients; and (<b>B</b>) stratified by diagnosis (paraganglioma; pheochromocytoma).</p>
Full article ">Figure 2
<p>Kaplan–Meier survival plots displaying median overall survival after the first PRRT cycle stratified by germline mutations.</p>
Full article ">Figure 3
<p>Kaplan–Meier survival plots displaying median progression-free survival for; (<b>A</b>) all patients; and (<b>B</b>) stratified by diagnosis (paraganglioma; pheochromocytoma).</p>
Full article ">
20 pages, 2079 KiB  
Article
Amide-to-Triazole Switch in Somatostatin-14-Based Radioligands: Impact on Receptor Affinity and In Vivo Stability
by Xabier Guarrochena, Panagiotis Kanellopoulos, Anna Stingeder, Lisa-Maria Rečnik, Irene V. J. Feiner, Marie Brandt, Wolfgang Kandioller, Theodosia Maina, Berthold A. Nock and Thomas L. Mindt
Pharmaceutics 2024, 16(3), 392; https://doi.org/10.3390/pharmaceutics16030392 - 13 Mar 2024
Cited by 1 | Viewed by 1736
Abstract
The use of metabolically stabilized, radiolabeled somatostatin (SST) analogs ([68Ga]Ga/[177Lu]Lu-DOTA-TATE/TOC/NOC) is well established in nuclear medicine. Despite the pivotal role of these radioligands in the diagnosis and therapy of neuroendocrine tumors (NETs), their inability to interact with all five [...] Read more.
The use of metabolically stabilized, radiolabeled somatostatin (SST) analogs ([68Ga]Ga/[177Lu]Lu-DOTA-TATE/TOC/NOC) is well established in nuclear medicine. Despite the pivotal role of these radioligands in the diagnosis and therapy of neuroendocrine tumors (NETs), their inability to interact with all five somatostatin receptors (SST1–5R) limits their clinical potential. [111In]In-AT2S is a radiolabeled DOTA-conjugate derived from the parent peptide SST-14 that exhibits high binding affinity to all SSTR subtypes, but its poor metabolic stability represents a serious disadvantage for clinical use. In order to address this issue, we have replaced strategic trans-amide bonds of [111In]In-AT2S with metabolically stable 1,4-disubstituted 1,2,3-triazole bioisosteres. From the five cyclic triazolo-peptidomimetics investigated, only [111In]In-XG1 combined a preserved nanomolar affinity for the SST1,2,3,5R subtypes in vitro and an improved stability in vivo (up to 17% of intact peptide 5 min postinjection (pi) versus 6% for [111In]In-AT2S). The involvement of neprilysin (NEP) in the metabolism of [111In]In-XG1 was confirmed by coadministration of Entresto®, a registered antihypertensive drug, in vivo releasing the selective and potent NEP-inhibitor sacubitrilat. A pilot SPECT/CT imaging study conducted in mice bearing hSST2R-positive xenografts failed to visualize the xenografts due to the pronounced kidney uptake (>200% injected activity (IA)/g at 4 h pi), likely the result of the formation of cationic metabolites. To corroborate the imaging data, the tumors and the kidneys were excised and analyzed with a γ-counter. Even if receptor-specific tumor uptake for [111In]In-XG1 could be confirmed (1.61% IA/g), further optimization is required to improve its pharmacokinetic properties for radiotracer development. Full article
Show Figures

Figure 1

Figure 1
<p>Structures of: (<b>a</b>) DOTA-TOC (R<sub>1</sub> = 4-hydroxyphenyl/R<sub>2</sub> = CH<sub>2</sub>OH), DOTA-TATE (R<sub>1</sub> = 4-hydroxyphenyl/R<sub>2</sub> = COOH) and DOTA-NOC (R<sub>1</sub> = 1-naphthyl/R<sub>2</sub> = CH<sub>2</sub>OH); (<b>b</b>), DOTA-KE108, (<b>c</b>) DOTA-SOM230; (<b>d</b>) AT2S. The DOTA chelator is indicated in a green color.</p>
Full article ">Figure 2
<p>Structures of: (<b>a</b>) the reference compound [<sup>111</sup>In]In-AT2S; (<b>b</b>) the triazolo-peptidomimetic [<sup>111</sup>In]In-XG1 (<b>b</b>). The bonds highlighted in colors were subjected to the amide-to-triazole substitution (<a href="#pharmaceutics-16-00392-t001" class="html-table">Table 1</a>). The olive-green <span class="html-italic">trans</span>-amide bonds correspond to the reported positions cleaved by NEP and the turquoise position is an adjacent bond to a cleavage site.</p>
Full article ">Figure 3
<p>In vitro characterization of (<b>a</b>,<b>b</b>) [<sup>111</sup>In]In-AT2S and (<b>c</b>,<b>d</b>) [<sup>111</sup>In]In-XG1 in SST<sub>2</sub>R-expressing AR42J cells where cell binding and internalization kinetics were studied (<b>a</b>,<b>c</b>, n = 3 in triplicates) and (<b>b</b>,<b>d</b>) the K<sub>D</sub> were determined in saturation binding assays (n = 2–3 in triplicates) using GraphPad Prism 8.0. Data points show mean values ± standard deviation (SD). NSB stands for nonspecific binding.</p>
Full article ">Figure 4
<p>Displacement of [<sup>125</sup>I][I-Tyr<sup>25</sup>]LTT-SST-28 from hSST<sub>1</sub>R (yellow line), hSST<sub>2</sub>R (green line), hSST<sub>3</sub>R (blue line) and hSST<sub>5</sub>R (gray line)-expressing membranes at increasing concentrations of <sup>nat</sup>In-XG1; by using GraphPad Prism 8.0., data points show mean values ± SD from four experiments performed in triplicate.</p>
Full article ">Figure 5
<p>In vivo stability of (<b>a</b>) [<sup>111</sup>In]In-XG1. Color-coded structures of [<sup>111</sup>In]In-AT2S (black), [<sup>111</sup>In]In-XG1 (olive-green), the suggested major metabolite of [<sup>111</sup>In]In-XG1 (turquoise) and hydrophilic radiometabolites (yellow). The amino acids depicted in red represent an hydrolyzed amide bond. Analysis of blood sample 5 min pi of (<b>b</b>) [<sup>111</sup>In]In-AT2S (6% intact peptide), (<b>c</b>) [<sup>111</sup>In]In-XG1 (17% of intact peptide) and (<b>d</b>) [<sup>111</sup>In]In-XG1 from a mouse pretreated with Entresto<sup>®</sup> (75% of intact peptide). Three mice were used per experiment.</p>
Full article ">Figure 6
<p>Static whole-body SPECT/CT images of SCID mice bearing HEK293-hSST<sub>2</sub>R tumors in their left flank (green arrows) and hSST<sub>2</sub>R-negative wtHEK293 control tumors in their right flank (orange arrow) at 4 h pi of [<sup>111</sup>In]In-XG1: (<b>a</b>) without pretreatment with Entresto<sup>®</sup> or (<b>b</b>) 30 min following oral administration of Entresto<sup>®</sup>. The kidneys are indicated with blue arrows and the color bars represent the difference in accumulated activity (purple being the lowest and white the highest level of accumulation).</p>
Full article ">Scheme 1
<p>Synthesis of the 1,4-Tz-containing peptidomimetic XG1 with the side-chains of the cysteine residues used in the cyclization in magenta and the Tz in olive-green. CTC refers to the 2-chlorotrityl resin.</p>
Full article ">
13 pages, 1295 KiB  
Article
Prediction of 177Lu-DOTATATE Therapy Outcomes in Neuroendocrine Tumor Patients Using Semi-Automatic Tumor Delineation on 68Ga-DOTATATE PET/CT
by Hwan Lee, Sarit T. Kipnis, Remy Niman, Sophia R. O’Brien, Jennifer R. Eads, Bryson W. Katona and Daniel A. Pryma
Cancers 2024, 16(1), 200; https://doi.org/10.3390/cancers16010200 - 31 Dec 2023
Cited by 5 | Viewed by 2026
Abstract
Background: Treatment of metastatic neuroendocrine tumors (NET) with 177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) results in favorable response only in a subset of patients. We investigated the prognostic value of quantitative pre-treatment semi-automatic 68Ga-DOTATATE PET/CT analysis in NET patients treated with [...] Read more.
Background: Treatment of metastatic neuroendocrine tumors (NET) with 177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) results in favorable response only in a subset of patients. We investigated the prognostic value of quantitative pre-treatment semi-automatic 68Ga-DOTATATE PET/CT analysis in NET patients treated with PRRT. Methods: The medical records of 94 NET patients who received at least one cycle of PRRT at a single institution were retrospectively reviewed. On each pre-treatment 68Ga-DOTATATE PET/CT, the total tumor volume (TTV), maximum tumor standardized uptake value for the patient (SUVmax), and average uptake in the lesion with the lowest radiotracer uptake (SUVmin) were determined with a semi-automatic tumor delineation method. Progression-free survival (PFS) and overall survival (OS) among the patients were compared based on optimal cutoff values for the imaging parameters. Results: On Kaplan–Meier analysis and univariate Cox regression, significantly shorter PFS was observed in patients with lower SUVmax, lower SUVmin, and higher TTV. On multivariate Cox regression, lower SUVmin and higher TTV remained predictive of shorter PFS. Only higher TTV was found to be predictive of shorter OS on Kaplan–Meier and Cox regression analyses. In a post hoc Kaplan–Meier analysis, patients with at least one high-risk feature (low SUVmin or high TTV) showed shorter PFS and OS, which may be the most convenient parameter to measure in clinical practice. Conclusions: The tumor volume and lowest lesion uptake on 68Ga-DOTATATE PET/CT can predict disease progression following PRRT in NET patients, with the former also predictive of overall survival. NET patients at risk for poor outcomes following PRRT can be identified with semi-automated quantitative analysis of 68Ga-DOTATATE PET/CT. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Cancers)
Show Figures

Figure 1

Figure 1
<p>A coronal maximum intensity projection image of <sup>68</sup>Ga-DOTATATE PET showing an example of the semi-automatic tumor delineation in red.</p>
Full article ">Figure 2
<p>Kaplan–Meier plot of progression-free survival and overall survival in 94 NET patients who received PRRT. Median progression-free survival was 21.0 months and median overall survival was not reached.</p>
Full article ">Figure 3
<p>Kaplan–Meier plots of post-PRRT progression-free survival based on pre-treatment <sup>68</sup>Ga-DOTATATE PET/CT parameters. Progression-free survival was significantly shorter in the (<b>A</b>) low SUVmax (<span class="html-italic">p</span> = 0.032), (<b>B</b>) low SUVmin (<span class="html-italic">p</span> = 0.006), and (<b>C</b>) high TTV (<span class="html-italic">p</span> = 0.034) groups. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Kaplan–Meier plots of post-PRRT overall survival based on pre-treatment <sup>68</sup>Ga-DOTATATE PET/CT parameters. (<b>A</b>) Low SUVmax (<span class="html-italic">p</span> = 0.40) and (<b>B</b>) low SUVmin (<span class="html-italic">p</span> = 0.22) were not predictive of mortality risk, but patients with (<b>C</b>) higher TTV showed significantly shorter overall survival (<span class="html-italic">p</span> &lt; 0.001). *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Kaplan–Meier plots of post-PRRT survival based on high vs. low-risk stratification using pre-treatment <sup>68</sup>Ga-DOTATATE PET/CT. High-risk patients showed significantly shorter (<b>A</b>) progression-free survival (<span class="html-italic">p</span> = 0.001) and (<b>B</b>) overall survival (<span class="html-italic">p</span> = 0.006). ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
21 pages, 2502 KiB  
Article
Epidemiology of Neuroendocrine Neoplasms and Results of Their Treatment with [177Lu]Lu-DOTA-TATE or [177Lu]Lu-DOTA-TATE and [90Y]Y-DOTA-TATE—A Six-Year Experience in High-Reference Polish Neuroendocrine Neoplasm Center
by Adam Daniel Durma, Marek Saracyn, Maciej Kołodziej, Katarzyna Jóźwik-Plebanek, Beata Dmochowska, Waldemar Kapusta, Wawrzyniec Żmudzki, Adrianna Mróz, Beata Kos-Kudła and Grzegorz Kamiński
Cancers 2023, 15(22), 5466; https://doi.org/10.3390/cancers15225466 - 18 Nov 2023
Cited by 5 | Viewed by 3371
Abstract
Neuroendocrine neoplasms (NENs) are a group of neoplasms arising from neuroendocrine cells. The worldwide incidence and prevalence of the NENs are estimated to be 6/100,000 and 35/100,000, respectively. Those numbers are increasing every decade, requiring higher and higher diagnosis and treatment costs. Radioligand [...] Read more.
Neuroendocrine neoplasms (NENs) are a group of neoplasms arising from neuroendocrine cells. The worldwide incidence and prevalence of the NENs are estimated to be 6/100,000 and 35/100,000, respectively. Those numbers are increasing every decade, requiring higher and higher diagnosis and treatment costs. Radioligand therapy (RLT) using beta-emitting radioisotopes is an efficient and relatively safe method of treatment, typically used as a second-line treatment. RLT tolerability is higher than other available pharmacotherapies (chemotherapy or tyrosine kinase inhibitors). Recent studies show an increase in overall survival among patients treated with RLT. The present study aimed to learn the epidemiology of NENs in Poland and assess the effectiveness of RLT in a high-reference center. A prospective analysis of 167 patients treated with RLT in one of Poland’s highest-reference NEN centers was performed. The analysis covered 66 months of observation (1 December 2017–30 May 2023), during which 479 RLT single administrations of radioisotope were given. The standard procedure was to give four courses of [177Lu]Lu-DOTA-TATE alone, or tandem therapy—[177Lu]Lu-DOTA-TATE and [90Y]Y-DOTA-TATE. Grading analysis showed that most patients had non-functioning G2 NEN with a mean Ki-67 of 6.05% (SD ± 6.41). The most common primary tumor location was the pancreas. Over two-thirds of patients did undergo surgery due to primary tumors or distant metastases. The majority of patients were using lanreotide as a chronically injected somatostatin analog. Median progression-free survival (PFS) on somatostatin analogs was 21.0 (IQR = 29.0) months. Directly after the last course of RLT, disease stabilization was noted in 69.46% of patients, partial regression was noted in 20.36% of patients, complete regression was noted in 0.60% of patients, and progression was noted in 9.58% of patients. In long-term follow-up, the median observation time among patients who underwent four treatment cycles (n = 108) was 29.8 (IQR = 23.9) months. Stabilization of the disease was observed in 55.56% of the patients and progression was observed in 26.85% of the patients, while 17.59% of patients died. Median PFS was 29.3 (IQR 23.9), and the median OS was 34.0 months (IQR 16.0). The mean age of NEN diagnosis is the sixth decade of life. It takes almost three years from NEN diagnosis to the start of RLT. In long-term observation, RLT leads to disease stabilization in over half of the patients with progressive disease. No differences in PFS or OS depend on the radioisotope used for RLT. In Poland, organized coordination of NEN treatment in high-reference centers ensures the continuity of patient care. Full article
(This article belongs to the Special Issue Advances in Neuroendocrine Neoplasms)
Show Figures

Figure 1

Figure 1
<p>Gender distribution in NEN grading subgroups.</p>
Full article ">Figure 2
<p>Distribution of primary tumor localization.</p>
Full article ">Figure 3
<p>Distribution of primary tumor localization in locations other than GEP-NEN.</p>
Full article ">Figure 4
<p>Radioisotope distribution in different NEN grading (in black boxes: number of patients).</p>
Full article ">Figure 5
<p>Non-surgical NEN tumor location.</p>
Full article ">Figure 6
<p>Detailed distribution of somatostatin analogs used in the study group (in black boxes: number of patients).</p>
Full article ">Figure 7
<p>Details of comorbidities (=metabolic risk factors) in the study group (white text—number of patients).</p>
Full article ">Figure 8
<p>Map of Poland with marked referral regions. Number of patients referred from the center located in this region.</p>
Full article ">Figure 9
<p>Kaplan–Meyer plot of the study group depending on primary tumor location.</p>
Full article ">
18 pages, 2590 KiB  
Review
Lung Carcinoids: A Comprehensive Review for Clinicians
by Dan Granberg, Carl Christofer Juhlin, Henrik Falhammar and Elham Hedayati
Cancers 2023, 15(22), 5440; https://doi.org/10.3390/cancers15225440 - 16 Nov 2023
Cited by 6 | Viewed by 3518
Abstract
Lung carcinoids are neuroendocrine tumors, categorized as typical or atypical carcinoids based on their histological appearance. While most of these tumors are slow-growing neoplasms, they still possess malignant potential. Many patients are diagnosed incidentally on chest X-rays or CT scans. Presenting symptoms include [...] Read more.
Lung carcinoids are neuroendocrine tumors, categorized as typical or atypical carcinoids based on their histological appearance. While most of these tumors are slow-growing neoplasms, they still possess malignant potential. Many patients are diagnosed incidentally on chest X-rays or CT scans. Presenting symptoms include cough, hemoptysis, wheezing, dyspnea, and recurrent pneumonia. Endocrine symptoms, such as carcinoid syndrome or ectopic Cushing’s syndrome, are rare. Surgery is the primary treatment and should be considered in all patients with localized disease, even when thoracic lymph node metastases are present. Patients with distant metastases may be treated with somatostatin analogues, chemotherapy, preferably temozolomide-based, mTOR inhibitors, or peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE. Most patients have an excellent prognosis. Poor prognostic factors include atypical histology and lymph node metastases at diagnosis. Long-term follow-up is mandatory since metastases may occur late. Full article
Show Figures

Figure 1

Figure 1
<p>Histological and immunohistochemical hallmarks of a typical pulmonary carcinoid. (<b>A</b>) Low-power (×40 magnification) overview of a hematoxylin–eosin-stained slide depicting an endobronchial tumorous mass. (<b>B</b>) High-power magnification (×200) of tumor cells in cord-like arrangements. Note the lack of nuclear atypia, mitotic figures, and necrosis. (<b>C</b>) Typical carcinoids are diffusely positive for chromogranin A and synaptophysin; the latter staining is depicted here.</p>
Full article ">Figure 2
<p>Atypical carcinoid. (<b>A</b>) Hematoxylin–eosin-stained tumor cells at ×400 magnification with slightly irregular tumor nuclei; several mitotic figures are noted (in circles). (<b>B</b>) The Ki-67 index was 15% (×200 magnification). Although not diagnostic, the Ki-67 index is usually much higher in atypical carcinoids compared with typical ones.</p>
Full article ">Figure 3
<p>Carcinoid in the upper left lobe strongly positive on <sup>68</sup>Ga-DOTATOC PET/CT (<b>left</b>) and negative on FDG PET/CT (<b>right</b>).</p>
Full article ">Figure 4
<p>Carcinoid filling almost the whole right main bronchus.</p>
Full article ">
20 pages, 319 KiB  
Review
Monitoring and Surveillance of Patients with Gastroenteropancreatic Neuroendocrine Tumors Undergoing Radioligand Therapy
by Thorvardur R. Halfdanarson, Nadine Mallak, Scott Paulson, Chandrikha Chandrasekharan, Mona Natwa, Ayse Tuba Kendi and Hagen F. Kennecke
Cancers 2023, 15(19), 4836; https://doi.org/10.3390/cancers15194836 - 2 Oct 2023
Cited by 1 | Viewed by 2355
Abstract
Radioligand therapy (RLT) with [177Lu]Lu-DOTA-TATE is a standard of care for adult patients with somatostatin-receptor (SSTR)-positive gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Taking advantage of this precision nuclear medicine approach requires diligent monitoring and surveillance, from the use of diagnostic SSTR-targeted radioligand imaging [...] Read more.
Radioligand therapy (RLT) with [177Lu]Lu-DOTA-TATE is a standard of care for adult patients with somatostatin-receptor (SSTR)-positive gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Taking advantage of this precision nuclear medicine approach requires diligent monitoring and surveillance, from the use of diagnostic SSTR-targeted radioligand imaging for the selection of patients through treatment and assessments of response. Published evidence-based guidelines assist the multidisciplinary healthcare team by providing acceptable approaches to care; however, the sheer heterogeneity of GEP-NETs can make these frameworks difficult to apply in individual clinical circumstances. There are also contradictions in the literature regarding the utility of novel approaches in monitoring and surveilling patients with GEP-NETs receiving RLT. This article discusses the emerging evidence on imaging, clinical biochemistry, and tumor assessment criteria in the management of patients receiving RLT for GEP-NETs; additionally, it documents our own best practices. This allows us to offer practical guidance on how to effectively implement monitoring and surveillance measures to aid patient-tailored clinical decision-making. Full article
(This article belongs to the Section Cancer Therapy)
13 pages, 1655 KiB  
Article
Radioligand Therapy with [177Lu]Lu-DOTA-TATE or [177Lu]Lu-DOTA-TATE and [90Y]Y-DOTA-TATE in Patients with Neuroendocrine Neoplasms of Unknown Locations, or Locations Other Than the Midgut and Pancreas as Primaries in a G1, G2 and G3 Grade
by Adam Daniel Durma, Marek Saracyn, Maciej Kołodziej, Katarzyna Jóźwik-Plebanek, Beata Dmochowska, Adrianna Mróz, Wawrzyniec Żmudzki and Grzegorz Kamiński
Pharmaceuticals 2023, 16(9), 1205; https://doi.org/10.3390/ph16091205 - 24 Aug 2023
Viewed by 1894
Abstract
Background: Neuroendocrine neoplasms (NENs) are a rare group of tumors with a different clinical course, prognosis and location. Radioligand therapy (RLT) can be used as a first or second line of treatment. It is registered in gastroenteropancreatic NENs (GEP-NENs) as grades G1 and [...] Read more.
Background: Neuroendocrine neoplasms (NENs) are a rare group of tumors with a different clinical course, prognosis and location. Radioligand therapy (RLT) can be used as a first or second line of treatment. It is registered in gastroenteropancreatic NENs (GEP-NENs) as grades G1 and G2. Tumors with an unknown point of origin, diagnosed outside the gastrointestinal tract and pancreas (non-GEP) or at the G3 grade, remain in the “grey area” of treatment. Materials and Methods: Analysis of 51 patients with NENs who underwent RLT in a single highest reference center from 2018 to 2023 was performed. Treatment was administrated to the patients with neoplasms of unknown origin, non-GEP-NENs, and ones with G3 grade. In total, 35 patients received 177-Lutetium (7.4 GBq), while 16 received 177-Lutetium and 90-Yttrium with equal activities (1.85 + 1.85 GBq). Results: The progression-free survival (PFS) before RLT qualification was 34.39 ± 35.88 months for the whole study group. In subgroups of patients with an unknown tumor location (n = 25), the median PFS was 19 months (IQR = 23), with “other” locations (n = 21) at 31 months (IQR = 28), and with NEN G3 (n = 7) at 18 months (IQR = 40). After RLT, disease stabilization or regression was observed in 42 (87.5% of) patients. RLT did not cause statistical changes in creatinine or GFR values. Hematological parameters (RBC, WBC, PLT, HGB) as well as chromogranin A concentration decreased significantly. There were no statistical differences between both subgroups regarding the type of radioisotope (177-Lutetium vs. 177-Lutetium and 90-Yttrium). After RLT in long-term observation, the median observation time (OT) was 14 months (IQR = 18 months). In patients with progression (n = 8), the median PFS was 20 months (IQR = 16 months), while in patients with confirmed death (n = 9), the median overall survival (OS) was 8 months (IQR = 14 months). Conclusions: Our study showed that 87.5% of NEN patients with unknown origin, non-GEP-NENs, and those with GEP-NEN G3 grade had benefited from the radioligand therapy. There were no significantly negative impacts on renal parameters. The decrease of bone marrow parameters was acceptable in relation to beneficial disease course. The decrease of chromogranin concentration was confirmed as a predictive factor for disease stabilization or regression. Full article
Show Figures

Figure 1

Figure 1
<p>Age distribution of the study group.</p>
Full article ">Figure 2
<p>Gender and NEN grading distribution of the study group.</p>
Full article ">Figure 3
<p>Tumor locations in the study group.</p>
Full article ">Figure 4
<p>Epidemiological analysis of the study group.</p>
Full article ">Figure 5
<p>Treatment analysis of the study group.</p>
Full article ">Figure 6
<p>Long-term observation data of the group—percentage of treatment outcome.</p>
Full article ">
13 pages, 3484 KiB  
Article
Recombinant α1-Microglobulin (rA1M) Protects against Hematopoietic and Renal Toxicity, Alone and in Combination with Amino Acids, in a 177Lu-DOTATATE Mouse Radiation Model
by Abdul Ghani Alattar, Amanda Kristiansson, Helena Karlsson, Suvi Vallius, Jonas Ahlstedt, Eva Forssell-Aronsson, Bo Åkerström, Sven-Erik Strand, Johan Flygare and Magnus Gram
Biomolecules 2023, 13(6), 928; https://doi.org/10.3390/biom13060928 - 1 Jun 2023
Cited by 2 | Viewed by 1974
Abstract
177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) is used clinically to treat metastasized or unresectable neuroendocrine tumors (NETs). Although 177Lu-DOTATATE is mostly well tolerated in patients, bone marrow suppression and long-term renal toxicity are still side effects that should be considered. Amino [...] Read more.
177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) is used clinically to treat metastasized or unresectable neuroendocrine tumors (NETs). Although 177Lu-DOTATATE is mostly well tolerated in patients, bone marrow suppression and long-term renal toxicity are still side effects that should be considered. Amino acids are often used to minimize renal radiotoxicity, however, they are associated with nausea and vomiting in patients. α1-microglobulin (A1M) is an antioxidant with heme- and radical-scavenging abilities. A recombinant form (rA1M) has previously been shown to be renoprotective in preclinical models, including in PRRT-induced kidney damage. Here, we further investigated rA1M’s renal protective effect in a mouse 177Lu-DOTATATE model in terms of administration route and dosing regimen and as a combined therapy with amino acids (Vamin). Moreover, we investigated the protective effect of rA1M on peripheral blood and bone marrow cells, as well as circulatory biomarkers. Intravenous (i.v.) administration of rA1M reduced albuminuria levels and circulatory levels of the oxidative stress-related protein fibroblast growth factor-21 (FGF-21). Dual injections of rA1M (i.e., at 0 and 24 h post-177Lu-DOTATATE administration) preserved bone marrow cellularity and peripheral blood reticulocytes. Administration of Vamin, alone or in combination with rA1M, did not show any protection of bone marrow cellularity or peripheral reticulocytes. In conclusion, this study suggests that rA1M, administered i.v. for two consecutive days in conjunction with 177Lu-DOTATATE, may reduce hematopoietic and kidney toxicity during PRRT with 177Lu-DOTATATE. Full article
Show Figures

Figure 1

Figure 1
<p>Pharmacokinetic study of rA1M. Plasma concentration of rA1M in female BALB/cJBomTac mice (<span class="html-italic">n</span> = 3–4/timepoint) after s.c. (20 mg/kg, (<b>A</b>)), i.v. (5 mg/kg, (<b>B</b>)), or i.p. (20 mg/kg, (<b>C</b>)) administration. In (<b>D</b>) all three administration routes are presented. Data is presented as mean ± SEM.</p>
Full article ">Figure 2
<p>Evaluation of the protective effects of i.v. vs. s.c. administered rA1M. Albumin levels in urine (<b>A</b>) and plasma FGF-21 (<b>B</b>) were measured 4 days after dosing in animals receiving NaCl and the vehicle (i.v. or s.c., volume corresponding to that of 150 MBq <sup>177</sup>Lu-DOTATATE or rA1M injections, respectively), 150 MBq <sup>177</sup>Lu-DOTATATE with vehicle (i.v. or s.c., volume corresponding to that of rA1M injections), or with rA1M (i.v., 5 mg/kg or s.c., 20 mg/mg). Data is presented as scatter plots with mean ± SEM. Statistical comparison between groups was made with one-way ANOVA with a Šídák’s multiple comparisons post-hoc test. Comparison was made between groups with the same administration route and between rA1M i.v. and s.c. Only significant differences are presented in the figure. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Evaluation of protective effect of different dosing regimen of rA1M. Urine albumin levels, corrected for creatinine (<b>A</b>), circulatory reticulocytes (<b>B</b>), representative image of bone marrow isolated single cells, showing red pellet as an effect of the irradiation treatment on depletion of white marrow (<b>C</b>), and bone marrow cellularity (<b>D</b>) were measured 4 days after dosing in animals receiving NaCl and the vehicle (volume corresponding to that of 150 MBq <sup>177</sup>Lu-DOTATATE or one dose of rA1M injection), 150 MBq <sup>177</sup>Lu-DOTATATE with vehicle (volume corresponding to that of one dose of rA1M injection), or with rA1M (1, 5 or 2 × 5 mg/kg). The representative gating strategies for flowcytometric evaluation are presented in <a href="#app1-biomolecules-13-00928" class="html-app">Supplementary Figure S3</a>. Data is presented as scatter plots with mean ± SEM. Statistical comparison between groups was made with one-way ANOVA with a Šídák’s multiple comparisons post hoc test. Comparison was made between <sup>177</sup>Lu-DOTATATE + vehicle and all groups in addition to between the different doses of rA1M. Only significant differences are presented in the figure. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Impact of amino acid co-administration on rA1M biodistribution. Animals were injected with <sup>125</sup>I-labelled rA1M alone (<span class="html-italic">n</span> = 8), or in combination with amino acids (Vamin, <span class="html-italic">n</span> = 8). Four (4) animals from each group were sacrificed after 10 (<b>A</b>) or 60 min (<b>B</b>). Organ specific uptake values were calculated as percent injected activity per gram tissue (%IA/g) in kidney, liver, spleen, and femur. Data is presented in bar graphs with mean ± SEM. No statistical differences between the two groups were detected following comparison between groups with a two-way ANOVA.</p>
Full article ">Figure 5
<p>Impact of amino acid administration on the protective effects of rA1M. Urine albumin levels, corrected for creatinine (<b>A</b>), circulatory reticulocytes (<b>B</b>), and bone marrow cellularity (<b>C</b>) were measured 4 days after dosing in animals receiving NaCl and vehicle (volume corresponding to that of 150 MBq <sup>177</sup>Lu-DOTATATE or rA1M injections, respectively), 150 MBq <sup>177</sup>Lu-DOTATATE with the vehicle (volume corresponding to that of rA1M injections), the vehicle (volume corresponding to that of rA1M injections) and Vamin (35 mg/200 μL, administered i.p.), rA1M (2 × 5 mg/kg) or Vamin (35 mg/200 μL, administered i.p.), and rA1M (2 × 5 mg/kg). Data is presented as scatter plots with mean ± SEM. Statistical comparison between groups was made with one-way ANOVA with a Šídák’s multiple comparisons post hoc test. Comparison was made between <sup>177</sup>Lu-DOTATATE + vehicle and all groups, and between <sup>177</sup>Lu-DOTATATE + Vamin and rA1M with or without Vamin and between rA1M alone or rA1M + Vamin. Only significant differences are presented in the figure. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
19 pages, 5368 KiB  
Article
Disturbing the Redox Balance Using Buthionine Sulfoximine Radiosensitized Somatostatin Receptor-2 Expressing Pre-Clinical Models to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE
by Wendy Delbart, Gwennaëlle Marin, Basile Stamatopoulos, Roland de Wind, Nicolas Sirtaine, Pieter Demetter, Marie Vercruyssen, Erwin Woff, Ioannis Karfis, Ghanem E. Ghanem, Patrick Flamen and Zéna Wimana
Cancers 2023, 15(8), 2332; https://doi.org/10.3390/cancers15082332 - 17 Apr 2023
Viewed by 2090
Abstract
Peptide receptor radionuclide therapy with 177Lu-DOTATATE improves the outcome of patients with somatostatin receptor (SSTR)-expressing neuroendocrine tumours. Nevertheless, stable disease has been the main response pattern observed, with some rare complete responses. Lu-177 exerts about two-thirds of its biological effects via the [...] Read more.
Peptide receptor radionuclide therapy with 177Lu-DOTATATE improves the outcome of patients with somatostatin receptor (SSTR)-expressing neuroendocrine tumours. Nevertheless, stable disease has been the main response pattern observed, with some rare complete responses. Lu-177 exerts about two-thirds of its biological effects via the indirect effects of ionizing radiation that generate reactive oxygen species, eventually leading to oxidative damage and cell death. This provides a rationale for targeting the antioxidant defence system in combination with 177Lu-DOTATATE. In the present study, the radiosensitizing potential and the safety of depleting glutathione (GSH) levels using buthionine sulfoximine (BSO) during 177Lu-DOTATATE therapy were assessed in vitro and in vivo using a xenograft mouse model. In vitro, the combination resulted in a synergistic effect in cell lines exhibiting a BSO-mediated GSH decrease. In vivo, BSO neither influenced 177Lu-DOTATATE biodistribution nor induced liver, kidney or bone marrow toxicity. In terms of efficacy, the combination resulted in reduced tumour growth and metabolic activity. Our results showed that disturbing the cell redox balance using a GSH synthesis inhibitor increased 177Lu-DOTATATE efficacy without additional toxicity. Targeting the antioxidant defence system opens new safe treatment combination opportunities with 177Lu-DOTATATE. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

Figure 1
<p>Timeline of the in vivo experiments. Female athymic nude mice were subcutaneously injected in the right flank with EJM multiple myeloma cells. Mice were randomized into four treatment groups: (1) control (IV saline injection), (2) BSO (IV saline injection and BSO via drinking water), (3) <sup>177</sup>Lu-DOTATATE (a single IV injection of 30 MBq) and (4) <sup>177</sup>Lu-DOTATATE + BSO (a single IV injection of 30 MBq and BSO via drinking water). Mice had a baseline and a post-treatment <sup>18</sup>F-FDG PET/CT to assess metabolic tumour response. Tumour growth was also followed using a calliper. Another group of mice receiving <sup>177</sup>Lu-DOTATATE alone or in combination with BSO was sacrificed on days 1, 3 and 7 p.i. for ex vivo biodistribution studies and early glutathione quantification. All animals were continuously monitored for body weight. IV = intravenous, p.o. = per os.</p>
Full article ">Figure 2
<p>Effect of BSO and its combination with <sup>177</sup>Lu-DOTATATE on the survival of melanoma (HBL and MM162), multiple myeloma (COLO-677 and EJM) and GEP (MIA-PACA-2 and HT-29) cell lines. Cells were exposed to 5 MBq of <sup>177</sup>Lu-DOTATATE for 4 h. BSO (10<sup>−7</sup>M) was present in the medium from the day before irradiation until cell survival assessment on day 10. Results are expressed as a percentage of the non-treated counterpart and are represented as mean ± SEM (n = 12 from 3 independent experiments). The black dotted line represents 100%. *** <span class="html-italic">p</span> ≤ 0.001; ** <span class="html-italic">p</span> ≤ 0.01; NS = non-statistically significant.</p>
Full article ">Figure 3
<p>Total glutathione in melanoma (HBL and MM162), multiple myeloma (COLO-677 and EJM) and GEP (MIA-PACA-2 and HT-29) cell lines exposed to BSO. Total glutathione quantification was performed using cell lines exposed to 10<sup>−7</sup> M BSO for 18 h. Results are expressed as a percentage of the non-treated counterpart (black dotted line) and are represented as mean ± SEM (n = 6 from 2 independent experiments). *** <span class="html-italic">p</span> ≤ 0.001; ** <span class="html-italic">p</span> ≤ 0.01; * <span class="html-italic">p</span> ≤ 0.05; NS = non-statistically significant.</p>
Full article ">Figure 4
<p>Biodistribution of <sup>177</sup>Lu-DOTATATE at 24, 72 and 168 h p.i. in EJM xenograft mice. Mice in both groups were intravenously injected with 30 MBq of <sup>177</sup>Lu-DOTATATE on day 0. Mice in the combination group (<sup>177</sup>Lu-DOTATATE + BSO) additionally received 10 mM BSO via drinking water, starting on day-1. Results are expressed as the mean of the % injected activity per gram of tissue (%IA/g) ± SD of 4 mice per group.</p>
Full article ">Figure 5
<p><sup>177</sup>Lu-DOTATATE time–activity curves of the liver and kidneys of EJM xenograft mice. Mice in both groups were intravenously injected with 30 MBq of <sup>177</sup>Lu-DOTATATE on day 0. Mice in the combination group (<sup>177</sup>Lu-DOTATATE + BSO) additionally received 10 mM BSO via drinking water, starting on day-1. Solid lines represent the single exponential curve fitting. Dashed lines represent 95% confidence bands. n = 1 to 6 mice per group.</p>
Full article ">Figure 6
<p>Effect of BSO on total glutathione levels in the liver and kidneys. Mice were intravenously injected with 30 MBq of <sup>177</sup>Lu-DOTATATE on day 0. BSO (10 mM) was administered via drinking water starting on day-1. Total glutathione levels in the liver and kidneys after 2, 4, 8 and 14–18 days of continuous BSO exposure. Results are expressed as the mean ± SD of 2 to 5 mice per group (pooled data from mice sacrificed between days 14 and 18). ** <span class="html-italic">p</span> ≤ 0.01; * <span class="html-italic">p</span> ≤ 0.05; NS = non-statistically significant.</p>
Full article ">Figure 7
<p>Effect of BSO on total glutathione levels in EJM tumours. Mice were intravenously injected with 30 MBq of <sup>177</sup>Lu-DOTATATE on day 0. BSO (10 mM) was administered via drinking water starting on day-1. Total glutathione levels in tumours after 2 days (<b>A</b>) and 14–18 days (<b>B</b>) of continuous BSO exposure. Results are expressed as the mean ± SD of 2 to 5 mice per group (pooled data from mice sacrificed between days 14 and 18). *** <span class="html-italic">p</span> ≤ 0.001; ** <span class="html-italic">p</span> ≤ 0.01; NS = non-statistically significant.</p>
Full article ">Figure 8
<p>Effect of <sup>177</sup>Lu-DOTATATE and its combination with BSO on bone marrow. Mice were intravenously injected with 30 MBq of <sup>177</sup>Lu-DOTATATE on day 0. Mice in the combination group (<sup>177</sup>Lu-DOTATATE + BSO) additionally received 10 mM BSO via drinking water starting on day-1. BM (from both femurs) of 3 to 4 mice per group, sacrificed 29 to 38 days post-<sup>177</sup>Lu-DOTATATE injection, were analysed for apoptosis (<b>A</b>) or cytological assessment (<b>B</b>–<b>D</b>). (<b>A</b>) Apoptotic cells were classified into early-apoptotic (Annexin V+/7AAD−) and late-apoptotic/necrotic cells (Annexin V+/7AAD+). Results are expressed as mean ± SEM. The relative proportions of major BM lineages were evaluated: erythroid (<b>B</b>), granulocytic (<b>C</b>) and lymphoid lineages (<b>D</b>). Results are represented as mean ± SD. The dotted lines represent lower and upper normal ranges. NS = non-statistically significant.</p>
Full article ">Figure 9
<p>Representative H&amp;E-stained slides of mice kidneys. (<b>A</b>) Control, (<b>B</b>) BSO (<b>C</b>), <sup>177</sup>Lu-DOTATATE and (<b>D</b>) <sup>177</sup>Lu-DOTATATE + BSO. Magnification 20×. No morphological changes were observed. G = glomeruli.</p>
Full article ">Figure 10
<p>Representative H&amp;E-stained slides of mice liver. (<b>A</b>) Control, (<b>B</b>) BSO, (<b>C</b>) <sup>177</sup>Lu-DOTATATE and (<b>D</b>) <sup>177</sup>Lu-DOTATATE + BSO. Magnification 20×. No morphological changes were observed.</p>
Full article ">Figure 11
<p>Effect of <sup>177</sup>Lu-DOTATATE alone and in combination with BSO on EJM tumours. Mice bearing EJM multiple myeloma tumours were treated on day 28 post-tumour cell inoculation with 30 MBq of <sup>177</sup>Lu-DOTATATE alone or in combination with 10 mM BSO via drinking water, starting on day 27, for 3 weeks. Tumours were measured 3 times per week in 8 to 10 mice per group. (<b>A</b>) Tumour growth over time, represented as mean relative tumour volume ± SEM. (<b>B</b>) Box plot distribution of relative tumour volume (calliper measurement) on day 45. Only significant differences are indicated, where * <span class="html-italic">p</span> ≤ 0.05. (<b>C</b>) Tumour doubling time was obtained by fitting an exponential growth equation from tumour growth measures. (<b>D</b>) Box plot distribution of changes in tumour volume before and after treatment, measured on the CT part of the PET/CT, represented as a fractional increase in tumour volume (ΔVol<sub>T</sub>). Only significant differences are indicated, where * <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 12
<p>Effect of <sup>177</sup>Lu-DOTATATE alone and in combination with BSO on EJM tumours <sup>18</sup>F-FDG metabolic activity. Mice bearing EJM multiple myeloma tumours were treated on day 28 post-tumour cells inoculation with 30 MBq of <sup>177</sup>Lu-DOTATATE alone or in combination with 10 mM BSO via drinking water, starting on day 27, for 3 weeks. Box plot distribution showing the changes in <sup>18</sup>F-FDG uptake before and after treatment (day 45 post-inoculation) represented as fractional increase in total lesion glycolysis (ΔTLG). Only significant differences are indicated, where * <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">
Back to TopTop