Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Clin Pharmacol Ther ; 113(2): 260-274, 2023 02.
Article in English | MEDLINE | ID: mdl-35373336

ABSTRACT

The combined use of diagnostic and therapeutic radioligands with the same molecular target, also known as theranostics, enables accurate patient selection, targeted therapy, and prediction of treatment response. Radioiodine, bone-seeking radioligands and norepinephrine analogs have been used for many years for diagnostic imaging and radioligand therapy of thyroid carcinoma, bone metastases, pheochromocytoma, paraganglioma, and neuroblastoma, respectively. In recent years, radiolabeled somatostatin analogs and prostate-specific membrane antigen ligands have shown clinical efficacy in the treatment of neuroendocrine tumors and prostate cancer, respectively. Several candidate compounds are targeting novel theranostic targets such as fibroblast activation protein, C-X-C chemokine receptor 4, and gastrin-releasing peptide receptor. In addition, several strategies to improve efficacy of radioligand therapy are being evaluated, including dosimetry-based dose optimization, multireceptor targeting, upregulation of target receptors, radiosensitization, pharmacogenomics, and radiation genomics. Design and evaluation of novel radioligands and optimization of dose and dose schedules, within the complex context of individualized multimodal cancer treatment, requires a multidisciplinary approach that includes clinical pharmacology. Significant increases in the use of these radiopharmaceuticals in routine oncological practice can be expected, which will have major impact on patient care as well as (radio)pharmacy utilization.


Subject(s)
Neuroendocrine Tumors , Prostatic Neoplasms , Male , Humans , Iodine Radioisotopes/therapeutic use , Somatostatin , Neuroendocrine Tumors/drug therapy , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/therapeutic use
3.
Eur J Endocrinol ; 181(1): 45-53, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31067510

ABSTRACT

OBJECTIVES: Inoperable or metastatic paragangliomas (PGLs) and malignant pheochromocytomas (PCCs) are rare tumours with limited options for systemic treatment. Aim of this study was to assess the safety and efficacy of the radiolabelled somatostatin analogue (177LutetiumDOTA0-Tyr3)octreotate (177Lu-DOTATATE) for the treatment of PGLs and PCCs. METHODS: Patients with histologically proven inoperable or malignant PGLs and PCCs treated with 177Lu-DOTATATE at our centre were retrospectively analysed. Patients were treated with up to four cycles of 177Lu-DOTATATE with an intended dose of 7.4 Gb per cycle. Response was assessed with use of RECIST 1.1. RESULTS: Thirty patients were included: 17 with parasympathetic, 10 with sympathetic PGLs and 3 with PCCs. Grade 3/4 subacute haematotoxicity occurred in 6 (20%) of patients. A reversible subacute adverse event due to cardiac failure following possible catecholamine release occurred in two patients. Best tumour response was partial response in 7 (23%) and stable disease in 20 (67%), whereas 3 (10%) patients had progressive disease. In 20 patients with baseline disease progression, tumour control was observed in 17 (85%); the median progression-free survival was 91 months in patients with parasympathetic PGLs, 13 months in patients with sympathetic PGLs and 10 months in patients with metastatic PCCs. CONCLUSION: This study suggests that PRRT with 177Lu-DOTATATE is a safe and effective treatment option for patients with inoperable or malignant PGL and PCC.


Subject(s)
Adrenal Gland Neoplasms/radiotherapy , Octreotide/analogs & derivatives , Organometallic Compounds/therapeutic use , Paraganglioma/radiotherapy , Pheochromocytoma/radiotherapy , Radioisotopes/therapeutic use , Adult , Aged , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Octreotide/therapeutic use , Radiation Dosage , Receptors, Peptide/radiation effects , Retrospective Studies , Treatment Outcome
4.
J Clin Endocrinol Metab ; 104(4): 1336-1344, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30566620

ABSTRACT

PURPOSE: Peptide receptor radionuclide therapy (PRRT) with the radiolabeled somatostatin analogue [Lutetium-177-DOTA0-Tyr3]octreotate (177Lu-DOTATATE) is widely applied for inoperable metastatic small intestinal and nonfunctioning pancreatic neuroendocrine tumors (pNETs). The aim of this study is to describe the safety and efficacy of the treatment of functioning pNETs. METHODS: Patients were treated with up to four cycles of 177Lu-DOTATATE with an intended dose of 7.4 Gbq per cycle. Radiological (Response Evaluation Criteria in Solid Tumors 1.1), symptomatic, and biochemical response were analyzed retrospectively for all patients with a functioning pNET (insulinoma, gastrinoma, VIPoma, and glucagonoma) treated with 177Lu-DOTATATE. Quality of life (QOL) was assessed with the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-Core Module questionnaire. RESULTS: Thirty-four patients with a metastatic functioning pNET (European Neuroendocrine Tumor Society grade 1 or 2) were included: 14 insulinomas, 5 VIPomas, 7 gastrinomas, and 8 glucagonomas. Subacute hematological toxicity, grade 3 or 4 occurred in 4 patients (12%) and a hormonal crisis in 3 patients (9%). PRRT resulted in partial or complete response in 59% of patients and the disease control rate was 78% in patients with baseline progression. 71% of patients with uncontrolled symptoms had a reduction of symptoms and a more than 80% decrease of circulating hormone levels was measured during follow-up. After PRRT, median progression-free survival was 18.1 months (interquartile range: 3.3 to 35.7) with a concurrent increase in QOL. CONCLUSION: Treatment with 177Lu-DOTATATE is a safe and effective therapy resulting in radiological, symptomatic and biochemical response in a high percentage of patients with metastatic functioning pNETs. Hormonal crises occur relatively frequent and preventive therapy should be considered before and/or during PRRT.


Subject(s)
Coordination Complexes/administration & dosage , Lutetium/administration & dosage , Neuroendocrine Tumors/radiotherapy , Octreotide/analogs & derivatives , Pancreatic Neoplasms/radiotherapy , Radioisotopes/administration & dosage , Adult , Aged , Coordination Complexes/adverse effects , Female , Gastrins/blood , Gastrins/metabolism , Glucagon/blood , Glucagon/metabolism , Humans , Insulin/blood , Insulin/metabolism , Lutetium/adverse effects , Male , Middle Aged , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/pathology , Octreotide/administration & dosage , Octreotide/adverse effects , Pancreas/metabolism , Pancreas/pathology , Pancreas/radiation effects , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Quality of Life , Radiation Dosage , Radioisotopes/adverse effects , Response Evaluation Criteria in Solid Tumors , Retrospective Studies , Vasoactive Intestinal Peptide/blood , Vasoactive Intestinal Peptide/metabolism
5.
BMC Gastroenterol ; 18(1): 84, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29902988

ABSTRACT

BACKGROUND: Neuroendocrine tumours (NET) consist of a heterogeneous group of neoplasms with various organs of origin. At diagnosis 21% of the patients with a Grade 1 NET and 30% with a Grade 2 NET have distant metastases. Treatment with peptide receptor radionuclide therapy (PRRT) shows a high objective response rate and long median survival after treatment. However, complete remission is almost never achieved. The liver is the most commonly affected organ in metastatic disease and is the most incriminating factor for patient survival. Additional treatment of liver disease after PRRT may improve outcome in NET patients. Radioembolization is an established therapy for liver metastasis. To investigate this hypothesis, a phase 2 study was initiated to assess effectiveness and toxicity of holmium-166 radioembolization (166Ho-RE) after PRRT with lutetium-177 (177Lu)-DOTATATE. METHODS: The HEPAR PLUS trial ("Holmium Embolization Particles for Arterial Radiotherapy Plus 177 Lu-DOTATATE in Salvage NET patients") is a single centre, interventional, non-randomized, non-comparative, open label study. In this phase 2 study 30-48 patients with > 3 measurable liver metastases according to RECIST 1.1 will receive additional 166Ho-RE within 20 weeks after the 4th and last cycle of PRRT with 7.4 GBq 177Lu-DOTATATE. Primary objectives are to assess tumour response, complete and partial response according to RECIST 1.1, and toxicity, based on CTCAE v4.03, 3 months after 166Ho-RE. Secondary endpoints include biochemical response, quality of life, biodistribution and dosimetry. DISCUSSION: This is the first prospective study to combine PRRT with 177Lu-DOTATATE and additional 166Ho-RE in metastatic NET. A radiation boost on intrahepatic disease using 166Ho-RE may lead to an improved response rate without significant additional side-effects. TRIAL REGISTRATION: Clinicaltrials.gov NCT02067988 , 13 February 2014. Protocol version: 6, 30 november 2016.


Subject(s)
Antineoplastic Agents/therapeutic use , Embolization, Therapeutic/methods , Holmium/therapeutic use , Liver Neoplasms/therapy , Neuroendocrine Tumors/therapy , Octreotide/analogs & derivatives , Organometallic Compounds/therapeutic use , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Biomarkers, Tumor , Combined Modality Therapy , Holmium/adverse effects , Humans , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/secondary , Neoplasm Metastasis , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/secondary , Octreotide/therapeutic use , Quality of Life , Radioisotopes/adverse effects , Radiopharmaceuticals/adverse effects , Remission Induction , Survival Analysis
6.
J Nucl Med ; 59(3): 452-458, 2018 03.
Article in English | MEDLINE | ID: mdl-28775205

ABSTRACT

Peptide receptor radionuclide therapy (PRRT) may induce long-term toxicity to the bone marrow (BM). The aim of this study was to analyze persistent hematologic dysfunction (PHD) after PRRT with 177Lu-DOTATATE in patients with gastroenteropancreatic neuroendocrine tumors (GEP NETs). Methods: The incidence and course of PHD were analyzed in 274 GEP NET patients from a group of 367 patients with somatostatin receptor-positive tumors. PHD was defined as diagnosis of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML), myeloproliferative neoplasm (MPN), MDS/MPN, or otherwise unexplained cytopenia (for >6 mo). Using data from The Netherlands Cancer Registry, the expected number of hematopoietic neoplasms (MDS, AML, MPN, and MDS/MPN) was calculated and adjusted for sex, age, and follow-up period. The following risk factors were assessed: sex, age over 70 y, bone metastasis, prior chemotherapy, prior external-beam radiotherapy, uptake on the [111In-DTPA0]octreotide scan, tumor load, grade 3-4 hematologic toxicity during treatment, estimated absorbed BM dose, elevated plasma chromogranin A level, baseline blood counts, and renal function. Results: Eleven (4%) of the 274 patients had PHD after treatment with 177Lu-DOTATATE: 8 patients (2.9%) developed a hematopoietic neoplasm (4 MDS, 1 AML, 1 MPN, and 2 MDS/MPN) and 3 patients (1.1%) developed BM failure characterized by cytopenia and BM aplasia. The median latency period at diagnosis (or first suspicion of a PHD) was 41 mo (range, 15-84 mo). The expected number of hematopoietic neoplasms based on The Netherlands Cancer Registry data was 3.0, resulting in a relative risk of 2.7 (95% confidence interval, 0.7-10.0). No risk factors for PHD could be identified for the GEP NET patients, not even bone metastasis or estimated BM dose. Seven patients with PHD developed anemia in combination with a rise in mean corpuscular volume. Conclusion: The prevalence of PHD after PRRT with 177Lu-DOTATATE was 4% in our patient population. The median time at which PHD developed was 41 mo after the first PRRT cycle. The relative risk for developing a hematopoietic neoplasm was 2.7. No risk factors were found for the development of PHD in GEP NET patients.


Subject(s)
Intestinal Neoplasms/physiopathology , Intestinal Neoplasms/radiotherapy , Neuroendocrine Tumors/physiopathology , Neuroendocrine Tumors/radiotherapy , Octreotide/analogs & derivatives , Organometallic Compounds/therapeutic use , Pancreatic Neoplasms/physiopathology , Pancreatic Neoplasms/radiotherapy , Receptors, Peptide/metabolism , Stomach Neoplasms/physiopathology , Stomach Neoplasms/radiotherapy , Aged , Female , Hematology , Humans , Incidence , Male , Middle Aged , Octreotide/therapeutic use
7.
Clin Cancer Res ; 23(16): 4617-4624, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28428192

ABSTRACT

Purpose: Bronchial and gastroenteropancreatic neuroendocrine tumors (NET) are slow-growing tumors, which frequently express somatostatin receptors on their cell membranes. These receptors are targets for therapy with Lutetium-177-labeled somatostatin analogues. We have treated over 1,200 patients with peptide receptor radionuclide therapy (PRRT) with [177Lu-DOTA0,Tyr3]octreotate (177Lu-DOTATATE) since the year 2000 and present the results on efficacy, survival, and toxicity of this therapy.Experimental Design: For safety analysis, 610 patients treated with a cumulative dose of at least 100 mCi (3.7 GBq) 177Lu-DOTATATE were included. A subgroup of 443 Dutch patients who were treated with a cumulative dose of at least 600 mCi (22.2 GBq) 177Lu-DOTATATE before 2013 was further analyzed for efficacy and survival.Results: The objective response rate of the total group of patients was 39%. Stable disease was reached in 43% of patients. Progression-free survival (PFS) and overall survival (OS) for all NET patients were 29 months [95% confidence interval (CI), 26-33 months] and 63 months (95% CI, 55-72 months). Long-term toxicity included acute leukemia in four patients (0.7%) and myelodysplastic syndrome in nine patients (1.5%). No therapy-related long-term renal or hepatic failure occurred.Conclusions: PRRT with 177Lu-DOTATATE is a favorable therapeutic option in patients with metastatic bronchial and gastroenteropancreatic NETs that express somatostatin receptors. PRRT with 177Lu-DOTATATE is safe with few side-effects and shows good response rates with PFS of 29 months and OS of 63 months. Clin Cancer Res; 23(16); 4617-24. ©2017 AACR.


Subject(s)
Bronchial Neoplasms/radiotherapy , Intestinal Neoplasms/radiotherapy , Neuroendocrine Tumors/radiotherapy , Octreotide/analogs & derivatives , Organometallic Compounds/therapeutic use , Pancreatic Neoplasms/radiotherapy , Stomach Neoplasms/radiotherapy , Acute Disease , Adult , Aged , Aged, 80 and over , Female , Humans , Kaplan-Meier Estimate , Leukemia/etiology , Male , Middle Aged , Myelodysplastic Syndromes/etiology , Octreotide/adverse effects , Octreotide/therapeutic use , Organometallic Compounds/adverse effects , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/therapeutic use , Time Factors , Treatment Outcome
8.
Endocr Relat Cancer ; 24(5): 243-251, 2017 05.
Article in English | MEDLINE | ID: mdl-28320783

ABSTRACT

Peptide receptor radionuclide therapy (PRRT) with [177Lu-DOTA0,Tyr3]octreotate (177Lu-DOTATATE) is a treatment with good results in patients with metastatic gastroenteropancreatic neuroendocrine tumours (GEPNETs). However, there are some pitfalls that should be taken into consideration when evaluating the treatment response after PRRT. 354 Dutch patients with GEPNETs who were treated with 177Lu-DOTATATE between March 2000 and December 2011 were retrospectively selected. Liver function parameters and chromogranin A were measured before each therapy and in follow-up. Anatomical imaging was performed before therapy and in follow-up. An increase in aminotransferases by ≥20% compared to baseline was observed in 83 of 351 patients (24%). In patients with an objective response (OR) and stable disease (SD) this increase was observed in 71/297 (24%) and in patients with progressive disease (PD) it was observed in 12/54 patients (22%). An increase in chromogranin A by ≥20% compared to baseline was observed in 76 patients (29%). This was present in 34% of patients who eventually had PD and 27% of patients who had OR/SD. In 70% of patients this tumour marker returned to baseline levels after therapy. An increase in liver enzymes and chromogranin A is not uncommon after PRRT. In the vast majority of patients this will resolve in follow-up. Clinicians should be aware that these changes may occur due to radiation-induced inflammation or disease progression and that repeated measurements over time are necessary to differentiate between the two.


Subject(s)
Biomarkers, Tumor , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/radiotherapy , Octreotide/analogs & derivatives , Organometallic Compounds/therapeutic use , Receptors, Peptide/therapeutic use , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/standards , Female , Humans , Male , Middle Aged , Monitoring, Physiologic/methods , Monitoring, Physiologic/standards , Neuroendocrine Tumors/pathology , Octreotide/chemistry , Octreotide/therapeutic use , Organometallic Compounds/chemistry , Predictive Value of Tests , Prognosis , Receptors, Peptide/chemistry , Retrospective Studies , Young Adult
9.
Eur J Nucl Med Mol Imaging ; 43(10): 1802-11, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27160225

ABSTRACT

PURPOSE: After peptide receptor radionuclide therapy (PRRT), renal toxicity may occur, particular in PRRT with (90)Y-labelled somatostatin analogues. Risk factors have been identified for increased probability of developing renal toxicity after PRRT, including hypertension, diabetes and age. We investigated the renal function over time, the incidence of nephrotoxicity and associated risk factors in patients treated with PRRT with [(177)Lu-DOTA(0),Tyr(3)]-Octreotate ((177)Lu-Octreotate). Also, radiation dose to the kidneys was evaluated and compared with the accepted dose limits in external beam radiotherapy and PRRT with (90)Y-radiolabelled somatostatin analogues. METHODS: The annual decrease in creatinine clearance (CLR) was determined in 209 Dutch patients and the incidence of grade 3 or 4 renal toxicity (according to CTCAE v4.03) was evaluated in 323 patients. Risk factors were analysed using a nonlinear mixed effects regression model. Also, radiation doses to the kidneys were calculated and their association with high annual decrease in renal function were analysed. RESULTS: Of the 323 patients, 3 (1 %) developed (subacute) renal toxicity grade 2 (increase in serum creatinine >1.5 - 3.0 times baseline or upper limit of normal). No subacute grade 3 or 4 nephrotoxicity was observed. The estimated average baseline CLR (± SD) was 108 ± 5 ml/min and the estimated average annual decrease in CLR (± SD) was 3.4 ± 0.4 %. None of the risk factors (hypertension, diabetes, high cumulative injected activity, radiation dose to the kidneys and CTCAE grade) at baseline had a significant effect on renal function over time. The mean absorbed kidney dose in 228 patients was 20.1 ± 4.9 Gy. CONCLUSION: Nephrotoxicity in patients treated with (177)Lu-octreotate was low. No (sub)acute grade 3 or 4 renal toxicity occurred and none of the patients had an annual decrease in renal function of >20 %. No risk factors for renal toxicity could be identified. Our data support the idea that the radiation dose threshold, adopted from external beam radiotherapy and PRRT with (90)Y-labelled somatostatin analogues, does not seem valid for PRRT with (177)Lu-octreotate.


Subject(s)
Coordination Complexes/therapeutic use , Kidney Diseases/mortality , Neoplasms/mortality , Neoplasms/radiotherapy , Octreotide/analogs & derivatives , Radiation Injuries/mortality , Radiotherapy/mortality , Aged , Aged, 80 and over , Comorbidity , Female , Humans , Incidence , Male , Molecular Targeted Therapy/mortality , Netherlands/epidemiology , Octreotide/therapeutic use , Radiopharmaceuticals/therapeutic use , Radiotherapy Dosage , Risk Factors , Survival Rate
10.
Best Pract Res Clin Endocrinol Metab ; 30(1): 103-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26971847

ABSTRACT

In the past decades, the number of neuroendocrine tumours that are detected is increasing. A relative new and promising therapy for patients with metastasised or inoperable disease is peptide receptor radionuclide therapy (PRRT). This therapy involves an infusion of somatostatin analogues linked to radionuclides like Yttrium-90 or Lutetium-177. Objective response rates are reported in 15-35%. Response rates may vary between type of tumour and radionuclide. Besides the objective response rate, overall survival and progression free survival increase significantly. Also, the quality of life improves as well. Serious side-affects are rare. PRRT is usually well tolerated, also in patients with extensive metastasised disease. Recent studies combined PRRT with other types of therapies. Unfortunately no randomised trials comparing these strategies are available. In the future, more research is needed to evaluate the best therapy combinations or sequence of therapies.


Subject(s)
Neuroendocrine Tumors/radiotherapy , Radiopharmaceuticals/therapeutic use , Radiotherapy/methods , Receptors, Peptide/metabolism , Humans , Octreotide/adverse effects , Octreotide/therapeutic use , Radiopharmaceuticals/adverse effects , Radiotherapy/adverse effects
11.
Eur J Nucl Med Mol Imaging ; 43(3): 453-63, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26419852

ABSTRACT

PURPOSE: In peptide receptor radionuclide therapy (PRRT), the bone marrow (BM) is one of the dose-limiting organs. The accepted dose limit for BM is 2 Gy, adopted from (131)I treatment. We investigated the incidence and duration of haematological toxicity and its risk factors in patients treated with PRRT with (177)Lu-DOTA(0)-Tyr(3)-octreotate ((177)Lu-DOTATATE). Also, absorbed BM dose estimates were evaluated and compared with the accepted 2 Gy dose limit. METHODS: The incidence and duration of grade 3 or 4 haematological toxicity (according to CTCAE v3.0) and risk factors were analysed. Mean BM dose per unit (gigabecquerels) of administered radioactivity was calculated and the correlations between doses to the BM and haematological risk factors were determined. RESULTS: Haematological toxicity (grade 3/4) occurred in 34 (11 %) of 320 patients. In 15 of the 34 patients, this lasted more than 6 months or blood transfusions were required. Risk factors significantly associated with haematological toxicity were: poor renal function, white blood cell (WBC) count <4.0 × 10(9)/l, age over 70 years, extensive tumour mass and high tumour uptake on the OctreoScan. Previous chemotherapy was not associated. The mean BM dose per administered activity in 23 evaluable patients was 67 ± 7 mGy/GBq, resulting in a mean BM dose of 2 Gy in patients who received four cycles of 7.4 GBq (177)Lu-DOTATATE. Significant correlations between (cumulative) BM dose and platelet and WBC counts were found in a selected group of patients. CONCLUSION: The incidence of subacute haematological toxicity after PRRT with (177)Lu-DOTATATE is acceptable (11 %). Patients with impaired renal function, low WBC count, extensive tumour mass, high tumour uptake on the OctreoScan and/or advanced age are more likely to develop grade 3/4 haematological toxicity. The BM dose limit of 2 Gy, adopted from (131)I, seems not to be valid for PRRT with (177)Lu-DOTATATE.


Subject(s)
Lutetium/chemistry , Neuroendocrine Tumors/radiotherapy , Octreotide/analogs & derivatives , Organometallic Compounds/chemistry , Radioisotopes/chemistry , Receptors, Peptide/chemistry , Aged , Bone Marrow/radiation effects , Female , Humans , Iodine/chemistry , Male , Middle Aged , Netherlands , Neuroendocrine Tumors/mortality , Octreotide/adverse effects , Octreotide/chemistry , Organometallic Compounds/adverse effects , Prognosis , Prospective Studies , Radiation Injuries/etiology , Radiation Injuries/prevention & control , Radioisotopes/adverse effects , Radiometry , Radiopharmaceuticals/therapeutic use , Risk Factors , Treatment Outcome
12.
Front Horm Res ; 44: 73-87, 2015.
Article in English | MEDLINE | ID: mdl-26303705

ABSTRACT

An important role is reserved for nuclear imaging techniques in the imaging of neuroendocrine tumors (NETs). Somatostatin receptor scintigraphy (SRS) with (111)In-DTPA-octreotide is currently the most important tracer in the diagnosis, staging and selection for peptide receptor radionuclide therapy (PRRT). In the past decade, different positron-emitting tomography (PET) tracers have been developed. The largest group is the (68)Gallium-labeled somatostatin analogs ((68)Ga-SSA). Several studies have demonstrated their superiority compared to SRS in sensitivity and specificity. Furthermore, patient comfort and effective dose are favorable for (68)Ga-SSA. Other PET targets like ß-[(11)C]-5-hydroxy-L-tryptophan ((11)C-5-HTP) and 6-(18)F-L-3,4-dihydroxyphenylalanine ((18)F-DOPA) were developed recently. For insulinomas, glucagon-like peptide-1 receptor imaging is a promising new technique. The evaluation of response after PRRT and other therapies is a challenge. Currently, the official follow-up is performed with radiological imaging techniques. The role of nuclear medicine may increase with the newest tracers for PET. In this review, the different nuclear imaging techniques and tracers for the imaging of NETs will be discussed.


Subject(s)
Neuroendocrine Tumors/diagnostic imaging , Nuclear Medicine/methods , Positron-Emission Tomography/methods , Radioactive Tracers , Humans
13.
Clin Transl Imaging ; 2: 55-66, 2014.
Article in English | MEDLINE | ID: mdl-24765618

ABSTRACT

Peptide receptor radionuclide therapy (PRRT) has been shown to be an effective treatment for neuroendocrine tumors (NETs) if curative surgery is not an option. A majority of NETs abundantly express somatostatin receptors. Consequently, following administration of somatostatin (SST) analogs labeled with γ-emitting radionuclides, these tumors can be imaged for diagnosis, staging or follow-up purposes. Furthermore, when ß-emitting radionuclides are used, radiolabeled peptides (radiopeptides) can also be used for the treatment for NET patients. Even though excellent results have been achieved with PRRT, complete responses are still rare, which means that there is room for improvement. In this review, we highlight some of the directions currently under investigation in pilot clinical studies or in preclinical development to achieve this goal. Although randomized clinical trials are still lacking, early studies have shown that tumor response might be improved by application of other radionuclides, such as α-emitters or radionuclide combinations, or by adjustment of radiopeptide administration routes. Individualized dosimetry and better insight into tumor and normal organ radiation doses may allow adjustment of the amount of administered activity per cycle or the number of treatment cycles, resulting in more personalized treatment schedules. Other options include the application of novel (radiolabeled) SST analogs with improved tumor uptake and radionuclide retention time, or a combination of PRRT with other systemic therapies, such as chemotherapy or treatment with radio sensitizers. Though promising directions appear to bring improvements of PRRT within reach, additional research (including randomized clinical trials) is needed to achieve such improvements.

14.
Eur J Nucl Med Mol Imaging ; 40(12): 1843-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23877632

ABSTRACT

PURPOSE: The aim of this study was to explore the possible mechanisms involved in an observed decline in serum calcium levels in patients with a neuroendocrine tumour (NET) treated with [(177)Lu-DOTA(0),Tyr(3)]octreotate ((177)Lu-octreotate). METHODS: In 47 patients with NET who were normocalcaemic at baseline, serum calcium, albumin, creatinine, alkaline phosphatase, gamma glutamyl transpeptidase, magnesium, phosphate and 25-hydroxyvitamin D were prospectively analysed at baseline and up to 6 months after treatment. Parathyroid hormone (PTH), 1,25-dihydroxyvitamin D3, type 1 aminoterminal propeptide of procollagen, bone-specific alkaline phosphatase, carboxyterminal crosslinking telopeptide of bone collagen, collagen type I crosslinked N-telopeptide, and creatinine and calcium in 24-h urine samples, were evaluated at baseline and at 3 and 6 months. Another 153 patients with NET were included in a retrospective study to estimate the occurrence of hypocalcaemia in a larger patient group. RESULTS: In the prospectively included patients, the mean serum calcium level decreased significantly after treatment (2.31 ± 0.01 to 2.26 ± 0.02 mmol/l, p = 0.02). Eight patients (17%) showed a marked decrease in serum calcium levels with a nadir of ≤ 2.10 mmol/l. In five patients (11%), calcium substitution therapy was prescribed. PTH increased significantly (5.9 ± 0.6 to 6.7 ± 0.8 pmol/l, p = 0.02), presumably in response to the decreasing serum calcium levels. 25-Hydroxyvitamin D remained stable after treatment. Creatinine levels increased significantly (73 ± 3 to 77 ± 3 µmol/l, p = 0.01), but not enough to explain the hypocalcaemia. Phosphate levels remained unaffected. In the retrospectively analysed patients, the mean serum calcium level decreased significantly from 2.33 ± 0.01 at baseline to a nadir of 2.24 ± 0.01 mmol/l at 18 months after treatment (p < 0.001). Of the 153 patients, 33 (22%) showed a serum calcium nadir of ≤ 2.10 mmol/l, and 11 (7%) received calcium substitution therapy. CONCLUSION: The mean serum calcium level decreased significantly after treatment with (177)Lu-octreotate, resulting in mild hypocalcaemia in about 20% of patients. We excluded several potential causes of this hypocalcaemia, so the cause remains unknown. Serum calcium levels should be monitored after peptide receptor radionuclide therapy, and calcium substitution therapy should be initiated if appropriate.


Subject(s)
Hypocalcemia/blood , Hypocalcemia/chemically induced , Octreotide/analogs & derivatives , Organometallic Compounds/adverse effects , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Calcium/blood , Female , Humans , Male , Middle Aged , Neuroendocrine Tumors/radiotherapy , Octreotide/adverse effects , Octreotide/therapeutic use , Organometallic Compounds/therapeutic use
15.
Neuroendocrinology ; 97(1): 74-85, 2013.
Article in English | MEDLINE | ID: mdl-22237390

ABSTRACT

The primary treatment of gastroenteropancreatic neuroendocrine tumors (GEPNETs) is surgery with curative intent or debulking of the tumor mass. In case of metastatic disease, cytoreductive options are limited. A relatively new therapeutic modality, peptide receptor radionuclide therapy (PRRT) with radiolabeled somatostatin analogs, is currently available in a number of mostly European centers. Complete and partial responses obtained after treatment with [90Y-DOTA0,Tyr3]octreotide are in the same range as after treatment with [177Lu-DOTA0,Tyr3]octreotate (i.e. 10-30%). However, significant nephrotoxicity has been observed after treatment with [90Y-DOTA0,Tyr3]octreotide. Options to improve PRRT may include combinations of radioactive labeled somatostatin analogs, intra-arterial administration, and the use of radiosensitizing drugs combined with PRRT. Other therapeutic applications of PRRT may include additional therapy cycles in patients with progressive disease after benefit from initial therapy, PRRT in adjuvant or neoadjuvant setting, or PRRT combined with new targeted therapies, such as sunitinib or everolimus. Randomized clinical trials comparing PRRT with other treatment modalities, or comparing various radioactive labeled somatostatin analogs should be undertaken to determine the best treatment options and treatment sequelae for patients with GEPNETs.


Subject(s)
Carcinoma, Neuroendocrine/therapy , Gastrointestinal Neoplasms/therapy , Pancreatic Neoplasms/therapy , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Receptors, Peptide/metabolism , Humans
16.
Clin Nucl Med ; 37(8): 781-2, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22785510

ABSTRACT

The normal scintigraphic features of 111In pentetreotide include uptake in the pituitary, thyroid, liver, spleen, kidneys, urinary bladder, gallbladder, and bowel. In premenopausal women, faint activity can be seen in the breasts. We present 2 cases of symmetrical, markedly increased mammary uptake. Our hypothesis to explain this pattern of uptake is an upregulation of the somatostatin type 2 receptor caused by high blood levels of estradiol during pregnancy. Estradiol levels during the menstrual cycle could therefore explain the variable degree of uptake commonly seen in premenopausal women.


Subject(s)
Breast/diagnostic imaging , Somatostatin/analogs & derivatives , Adult , Female , Gamma Cameras , Humans , Radionuclide Imaging , Receptors, Somatostatin/metabolism
17.
Best Pract Res Clin Gastroenterol ; 26(6): 867-81, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23582925

ABSTRACT

Peptide receptor radionuclide therapy (PRRT) with radiolabelled somatostatin analogues plays an increasing role in the treatment of patients with inoperable or metastasised gatroenteropancreatic neuroendocrine tumours (GEP-NETs). (90)Y-DOTATOC and (177)Lu-DOTATATE are the most used radiopeptides for PRRT with comparable tumour response rates (about 15-35%). The side effects of this therapy are few and mild. However, amino acids should be used for kidney protection, especially during infusion of (90)Y-DOTATOC. Options to improve PRRT may include combinations of radioactive labelled somatostatin analogues and the use of radiosensitising drugs combined with PRRT. Other therapeutic applications of PRRT may include intra-arterial administration, neo-adjuvant treatment and additional PRRT cycles in patients with progressive disease, who have benefited from initial therapy. Considering the mild side-effects, PRRT may well become the first-line therapy in patients with metastasised or inoperable GEP-NETs if more widespread use of PRRT can be accomplished.


Subject(s)
Intestinal Neoplasms/radiotherapy , Neuroendocrine Tumors/radiotherapy , Radiopharmaceuticals/therapeutic use , Somatostatin/analogs & derivatives , Stomach Neoplasms/radiotherapy , Heterocyclic Compounds/therapeutic use , Heterocyclic Compounds, 1-Ring/therapeutic use , Humans , Intestinal Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Octreotide/analogs & derivatives , Octreotide/therapeutic use , Organometallic Compounds/therapeutic use , Peptides, Cyclic/therapeutic use , Receptors, Somatostatin/metabolism , Retreatment , Somatostatin/metabolism , Stomach Neoplasms/metabolism , Yttrium Radioisotopes/therapeutic use
18.
Endocr Relat Cancer ; 18 Suppl 1: S27-51, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22005114

ABSTRACT

Nuclear medicine plays a pivotal role in the imaging and treatment of neuroendocrine tumours (NETs). Somatostatin receptor scintigraphy (SRS) with [(111)In-DTPA(0)]octreotide has proven its role in the diagnosis and staging of gastroenteropancreatic NETs (GEP-NETs). New techniques in somatostatin receptor imaging include the use of different radiolabelled somatostatin analogues with higher affinity and different affinity profiles to the somatostatin receptor subtypes. Most of these analogues can also be labelled with positron-emitting radionuclides that are being used in positron emission tomography imaging. The latter imaging modality, especially in the combination with computed tomography, is of interest because of encouraging results in terms of improved imaging quality and detection capabilities. Considerable advances have been made in the imaging of NETs, but to find the ideal imaging method with increased sensitivity and better topographic localisation of the primary and metastatic disease remains the ultimate goal of research. This review provides an overview of the currently used imaging modalities and ongoing developments in the imaging of NETs, with the emphasis on nuclear medicine and puts them in perspective of clinical practice. The advantage of SRS over other imaging modalities in GEP-NETs is that it can be used to select patients with sufficient uptake for treatment with radiolabelled somatostatin analogues. Peptide receptor radionuclide therapy (PRRT) is a promising new tool in the management of patients with inoperable or metastasised NETs as it can induce symptomatic improvement with all Indium-111, Yttrium-90 or Lutetium-177-labelled somatostatin analogues. The results that were obtained with [(90)Y-DOTA(0),Tyr(3)]octreotide and [(177)Lu-DOTA(0),Tyr(3)]octreotate are even more encouraging in terms of objective tumour responses with tumour regression and documented prolonged time to progression. In the largest group of patients receiving PRRT, treated with [(177)Lu-DOTA(0),Tyr(3)]octreotate, a survival benefit of several years compared with historical controls has been reported.


Subject(s)
Gastrointestinal Neoplasms/diagnostic imaging , Neuroendocrine Tumors/diagnostic imaging , Pancreatic Neoplasms/diagnostic imaging , 3-Iodobenzylguanidine/analogs & derivatives , 3-Iodobenzylguanidine/therapeutic use , Antineoplastic Agents/therapeutic use , Gastrointestinal Neoplasms/radiotherapy , Humans , Ligands , Magnetic Resonance Imaging , Neuroendocrine Tumors/radiotherapy , Octreotide/analogs & derivatives , Pancreatic Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Radionuclide Imaging , Receptors, Somatostatin/metabolism , Tomography, X-Ray Computed
19.
Semin Nucl Med ; 40(3): 209-18, 2010 May.
Article in English | MEDLINE | ID: mdl-20350630

ABSTRACT

In the 1980s, the (111)In-labeled somatostatin analog OctreoScan (Covidien, Hazelwood, MO) was developed for imaging of somatostatin receptor subtype 2 (sst(2)) overexpressing tumors. On the basis of this success, peptide receptor radionuclide therapy (PRRT) was developed using similar somatostatin analogs with different therapeutic radionuclides. Clinical application of PRRT demonstrated impressive results on tumor response, overall survival, and quality of life in patients with gastroenteropancreatic neuroendocrine tumors. The peptides 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA), Tyr(3)-octreotate (DOTATATE) and DOTA, Tyr(3)-octreotide (DOTATOC) (brand name Onalta), predominantly targeting sst(2), have been granted Orphan Drug status by the European Medicines Agency and the US Food and Drug Administration for application in PRRT. Besides somatostatin receptor-targeting peptides, multiple other radiopeptide analogs were developed targeting several other receptors overexpressed on various tumors. Some of these peptide analogs, including cholecystokinin, gastrin, gastrin-releasing peptide, arginine-glycine-aspartate (RGD)-peptides, and glucagon-like peptide 1 analogs appeared very promising in preclinical and clinical imaging and PRRT studies. Although the success of PRRT with radiolabeled somatostatin analogs has been established, there is still room for improvement. The therapeutic window of PRRT could be enlarged by the use of new and improved targeting compounds, of which new antagonists with excellent tumor to background ratios are very promising. Furthermore, locoregional administration, improved healthy tissue protection, and combination treatment can be applied to increase the effectiveness of PRRT. Combination treatment might include cocktails of different peptide analogs of different therapeutic radionuclides and of radiolabeled peptides with chemotherapeutic or radiosensitizing agents. This review summarizes results of PRRT and describes clinical and preclinical studies regarding PRRT optimizing strategies.


Subject(s)
Neoplasms/metabolism , Neoplasms/radiotherapy , Peptides/pharmacokinetics , Peptides/therapeutic use , Radioisotopes/therapeutic use , Receptors, Peptide/metabolism , Alpha Particles , Animals , Clinical Trials as Topic , Drug Delivery Systems , Drug Evaluation, Preclinical , Humans , Isotope Labeling/methods , Peptides/chemistry , Radioisotopes/chemistry
20.
Semin Nucl Med ; 40(2): 78-88, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20113677

ABSTRACT

Somatostatin receptor imaging with [(111)In-DTPA(0))octreotide has proven its role in the diagnosis and staging of gastroenteropancreatic neuroendocrine tumors. Treatment with radiolabeled somatostatin analogues is a promising new tool in the management of patients with inoperable or metastasized, well-differentiated neuroendocrine tumors. Symptomatic improvement may occur with all (111)In, (90)Y, or (177)Lu-labeled somatostatin analogues that have been used for peptide receptor radionuclide therapy. The results that were obtained with [(90)Y-DOTA(0), Tyr(3)]octreotide and [(177)Lu-DOTA(0), Tyr(3)]octreotate are very encouraging in terms of tumor regression. Also, if kidney protective agents are used, the side effects of this therapy are few and mild, and the median duration of the therapy response for these radiopharmaceuticals is 30 and 40 months, respectively. The patients' self-assessed quality of life increases significantly after treatment with [(177)Lu-DOTA(0), Tyr(3)]octreotate. Finally, compared with historical controls, there is a benefit in overall survival of several years from time of diagnosis in patients treated with [(177)Lu-DOTA(0), Tyr(3)]octreotate. These data compare favorably with the limited number of alternative treatment approaches. If more widespread use of peptide receptor radionuclide therapy can be guaranteed, such therapy may well become the therapy of first choice in patients with metastasized or inoperable gastroenteropancreatic neuroendocrine tumors.


Subject(s)
Neuroendocrine Tumors/radiotherapy , Radiotherapy/methods , Receptors, Somatostatin/therapeutic use , Animals , Humans , Neuroendocrine Tumors/drug therapy , Quality of Life , Radiotherapy/adverse effects , Survival Analysis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...