451
|
Aranda-Lara L, Ferro-Flores G, Azorín-Vega E, Ramírez FDM, Jiménez-Mancilla N, Ocampo-García B, Santos-Cuevas C, Isaac-Olivé K. Synthesis and evaluation of Lys¹(α,γ-Folate)Lys³(¹⁷⁷Lu-DOTA)-Bombesin(1-14) as a potential theranostic radiopharmaceutical for breast cancer. Appl Radiat Isot 2015; 107:214-219. [PMID: 26545016 DOI: 10.1016/j.apradiso.2015.10.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 12/22/2022]
Abstract
The aim of this work was to synthesize Lys(1)(α,γ-Folate)-Lys(3)((177)Lu-DOTA)-Bombesin (1-14) ((177)Lu-Folate-BN), as well as to assess its potential for molecular imaging and targeted radiotherapy of breast tumors expressing folate receptors (FR) and gastrin-releasing peptide receptors (GRPR). Radiation absorbed doses of (177)Lu-Folate-BN (74 MBq, i.v.) estimated in athymic mice with T47D-induced breast tumors (positive to FR and GRPR), showed tumor doses of 23.9±2.1 Gy. T47D-tumors were clearly visible (Micro-SPECT/CT images). (177)Lu-Folate-BN demonstrated properties suitable as a theranostic radiopharmaceutical.
Collapse
Affiliation(s)
- Liliana Aranda-Lara
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico; Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca 50180, Mexico
| | - Guillermina Ferro-Flores
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico.
| | - Erika Azorín-Vega
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico
| | - Flor de María Ramírez
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico
| | - Nallely Jiménez-Mancilla
- Catedrática CONACyT, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico
| | - Blanca Ocampo-García
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico
| | - Clara Santos-Cuevas
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Estado de México, Mexico
| | - Keila Isaac-Olivé
- Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca 50180, Mexico
| |
Collapse
|
452
|
Basu S, Ranade R. Favorable Response of Metastatic Merkel Cell Carcinoma to Targeted 177Lu-DOTATATE Therapy: Will PRRT Evolve to Become an Important Approach in Receptor-Positive Cases? J Nucl Med Technol 2015; 44:85-7. [DOI: 10.2967/jnmt.115.163527] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/02/2015] [Indexed: 11/16/2022] Open
|
453
|
Ambrosini V, Campana D, Polverari G, Peterle C, Diodato S, Ricci C, Allegri V, Casadei R, Tomassetti P, Fanti S. Prognostic Value of 68Ga-DOTANOC PET/CT SUVmax in Patients with Neuroendocrine Tumors of the Pancreas. J Nucl Med 2015; 56:1843-8. [DOI: 10.2967/jnumed.115.162719] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/14/2015] [Indexed: 01/19/2023] Open
|
454
|
Mariniello A, Bodei L, Tinelli C, Baio SM, Gilardi L, Colandrea M, Papi S, Valmadre G, Fazio N, Galetta D, Paganelli G, Grana CM. Long-term results of PRRT in advanced bronchopulmonary carcinoid. Eur J Nucl Med Mol Imaging 2015; 43:441-52. [PMID: 26392198 DOI: 10.1007/s00259-015-3190-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/04/2015] [Indexed: 11/30/2022]
Abstract
PURPOSE Peptide receptor radionuclide therapy (PRRT) for the treatment of neuroendocrine tumours (NET) has been explored for almost two decades, but there are still few trials that have exclusively investigated well-differentiated and moderately differentiated NET arising from the respiratory tree. Thus, the aim of this study was to explore the outcome in patients affected by bronchopulmonary carcinoid (BPC) following PRRT. METHODS We retrospectively analysed 114 patients with advanced stage BPC consecutively treated with PRRT at the European Institute of Oncology, Milan, from 1997 to 2012 and followed until October 2014. The objective responses, overall survival (OS) and progression-free survival (PFS) were rated, and three different PRRT protocols ((90)Y-DOTATOC vs. (177)Lu-DOTATATE vs. (90)Y-DOTATOC + (177)Lu-DOTATATE) were compared with regard to their efficacy and tolerability. RESULTS The median OS (evaluated in 94 of the 114 patients) was 58.8 months. The median PFS was 28.0 months. The (177)Lu-DOTATATE protocol resulted in the highest 5-year OS (61.4%). Morphological responses (partial responses + minor responses) were obtained in 26.5% of the cohort and were associated with longer OS and PFS. The (90)Y-DOTATOC + (177)Lu-DOTATATE protocol provided the highest response rate (38.1%). Adverse events were mild in the majority of patients. However, haematological toxicity negatively affected survival. No severe (grade 3/4) serum creatinine increase was observed. Patients treated with (90)Y-DOTATOC alone more frequently showed a mild/moderate decrease in renal function. In patients treated with chemotherapy before PRRT had a shorter OS and PFS, and a higher risk of developing nephrotoxicity. CONCLUSION In a large cohort of patients with advanced BPC treated in a "real-world" scenario and followed up for a median of 45.1 months (range 2-191 months), PRRT proved to be promising in prolonging survival and delaying disease progression. Despite the potential selection biases, considering the risk-benefit ratio, (177)Lu-DOTATATE monotherapy seems the best option for PRRT. Our results indicate that the use of PRRT in earlier stages of the disease could provide a more favorable outcome.
Collapse
Affiliation(s)
- Annapaola Mariniello
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy.
| | - Lisa Bodei
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | - Carmine Tinelli
- Epidemiology and Biometric Unit, IRCCS Foundation Policlinico San Matteo, Pavia, Italy
| | - Silvia Melania Baio
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | - Laura Gilardi
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | - Marzia Colandrea
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | - Stefano Papi
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | | | - Nicola Fazio
- Unit of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, Milan, Italy
| | - Domenico Galetta
- Thoracic Surgery Division, European Institute of Oncology, Milan, Italy
| | - Giovanni Paganelli
- Nuclear Medicine and Radiometabolic Units, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Chiara Maria Grana
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| |
Collapse
|
455
|
Basu S. Fraction, Cycle, or a New Terminology? What Would Be Most Appropriate for Molecularly Targeted Radiotherapy with Unsealed Sources? J Nucl Med Technol 2015; 43:301. [PMID: 26338488 DOI: 10.2967/jnmt.115.164046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre Bhabha Atomic Research Centre Tata Memorial Hospital Annexe Building Jerbai Wadia Rd. Parel, Mumbai, Maharashtra, India 400 012 E-mail:
| |
Collapse
|
456
|
Abstract
BACKGROUND Medullary thyroid cancer (MTC) is a rare but potentially life-threatening disease with limited therapeutic options. As a neuroendocrine tumor, MTC expresses somatostatin receptors, and therefore, somatostatin-labeled radiopharmaceuticals could be used to treat patients with MTC. OBJECTIVE The aims of this study were to evaluate tumor shrinkage after Lu-DOTATATE treatment, to analyze the impact on quality of life as accessed by the SF-36 questionnaire, and to demonstrate a possible prognostic role for In-DTPA-octreotide uptake in patients with MTC. PATIENTS AND METHODS Patients with progressive MTC underwent evaluation using In-DTPA-octreotide. Patients who demonstrated In-DTPA-octreotide uptake were treated with 4 cycles of 200 mCi of Lu-DOTATATE and were evaluated using CT scans over 8 to 12 months of treatment. RESULTS Of the 16 patients initially enrolled, 9 (56.25%) had lesions that were observed in the In-DTPA-octreotide scans and were eligible for therapy with Lu-DOTATATE. Three patients had a partial response, 3 patients were classified as having stable disease and, 1 patient had a progressive disease. All responders indicated improvement in quality of life 6 to 12 months after therapy. CONCLUSIONS Treatment with Lu-DOTATATE seems to be an alternative therapy for somatostatin receptor-positive tumors, with very mild adverse effects and quality-of-life improvement, at least during a short-term period. Further studies are needed to determine long-term benefits and to identify which patients are more likely to respond to this modality of therapy.
Collapse
|
457
|
Delker A, Fendler WP, Kratochwil C, Brunegraf A, Gosewisch A, Gildehaus FJ, Tritschler S, Stief CG, Kopka K, Haberkorn U, Bartenstein P, Böning G. Dosimetry for 177Lu-DKFZ-PSMA-617: a new radiopharmaceutical for the treatment of metastatic prostate cancer. Eur J Nucl Med Mol Imaging 2015; 43:42-51. [DOI: 10.1007/s00259-015-3174-7] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/10/2015] [Indexed: 12/21/2022]
|
458
|
Peptide receptor radionuclide therapy (PRRT): clinical significance of re-treatment? Eur J Nucl Med Mol Imaging 2015; 42:1949-54. [DOI: 10.1007/s00259-015-3153-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
459
|
The efficacy of 177Lu-labelled peptide receptor radionuclide therapy in patients with neuroendocrine tumours: a meta-analysis. Eur J Nucl Med Mol Imaging 2015; 42:1964-70. [DOI: 10.1007/s00259-015-3155-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/24/2015] [Indexed: 12/16/2022]
|
460
|
Medina-García V, Ocampo-García BE, Ferro-Flores G, Santos-Cuevas CL, Aranda-Lara L, García-Becerra R, Ordaz-Rosado D, Melendez-Alafort L. A freeze-dried kit formulation for the preparation of Lys(27)(99mTc-EDDA/HYNIC)-Exendin(9-39)/99mTc-EDDA/HYNIC-Tyr3-Octreotide to detect benign and malignant insulinomas. Nucl Med Biol 2015; 42:911-6. [PMID: 26364504 DOI: 10.1016/j.nucmedbio.2015.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/15/2015] [Accepted: 08/05/2015] [Indexed: 12/25/2022]
Abstract
About 90% of insulinomas are benign and 5%-15% are malignant. Benign insulinomas express the glucagon-like peptide-1 receptor (GLP-1R) and low levels of somatostatin receptors (SSTR), while malignant insulinomas over-express SSTR or GLP-1R in low levels. A kit for the preparation of Lys(27)((99m)Tc-EDDA/HYNIC)-Exendin(9-39)/(99m)Tc-EDDA/HYNIC-Tyr(3)Octreotide was formulated to detect 100% of insulinomas. The formulation showed radiochemical purity of 97±1%, high stability in human serum, and GLP-1R and SSTR affinity. The biodistribution and imaging studies demonstrated properties suitable for its use as a target-specific agent for the simultaneous molecular imaging of GRP-1R- and/or SSTR-positive tumors.
Collapse
Affiliation(s)
- Veronica Medina-García
- Instituto Nacional de Investigaciones Nucleares, Estado de México, 52750, Mexico; Universidad Autónoma del Estado de México, Estado de México, 50180, Mexico
| | | | | | | | - Liliana Aranda-Lara
- Instituto Nacional de Investigaciones Nucleares, Estado de México, 52750, Mexico; Universidad Autónoma del Estado de México, Estado de México, 50180, Mexico
| | - Rocio García-Becerra
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, 14000, Mexico
| | - David Ordaz-Rosado
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, 14000, Mexico
| | | |
Collapse
|
461
|
|
462
|
de Vries EGE, de Jong S, Gietema JA. Molecular Imaging As a Tool for Drug Development and Trial Design. J Clin Oncol 2015; 33:2585-7. [PMID: 26169612 DOI: 10.1200/jco.2015.61.6425] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
| | - Steven de Jong
- University Medical Center Groningen, University of Groningen, the Netherlands
| | - Jourik A Gietema
- University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
463
|
Ljungberg M, Gleisner KS. Hybrid Imaging for Patient-Specific Dosimetry in Radionuclide Therapy. Diagnostics (Basel) 2015; 5:296-317. [PMID: 26854156 PMCID: PMC4665601 DOI: 10.3390/diagnostics5030296] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 11/16/2022] Open
Abstract
Radionuclide therapy aims to treat malignant diseases by systemic administration of radiopharmaceuticals, often using carrier molecules such as peptides and antibodies. The radionuclides used emit electrons or alpha particles as a consequence of radioactive decay, thus leading to local energy deposition. Administration to individual patients can be tailored with regards to the risk of toxicity in normal organs by using absorbed dose planning. The scintillation camera, employed in planar imaging or single-photon emission computed tomography (SPECT), generates images of the spatially and temporally varying activity distribution. Recent commercially available combined SPECT and computed tomography (CT) systems have dramatically increased the possibility of performing accurate dose planning by using the CT information in several steps of the dose-planning calculation chain. This paper discusses the dosimetry chain used for individual absorbed-dose planning and highlights the areas where hybrid imaging makes significant contributions.
Collapse
Affiliation(s)
- Michael Ljungberg
- Department of Medical Radiation Physics, Lund University, 221 85 Lund, Sweden.
| | | |
Collapse
|
464
|
Mulholland N, Chakravartty R, Devlin L, Kalogianni E, Corcoran B, Vivian G. Long-term outcomes of (131)Iodine mIBG therapy in metastatic gastrointestinal pancreatic neuroendocrine tumours: single administration predicts non-responders. Eur J Nucl Med Mol Imaging 2015; 42:2002-12. [PMID: 26142730 DOI: 10.1007/s00259-015-3116-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 06/10/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND (131)Iodine (I131)-metaiodobenzylguanidine (mIBG) is a radionuclide-based treatment option for metastatic gastrointestinal-pancreatic neuroendocrine tumours (GEP NET). This study aimed at identifying prognostic indicators of long-term outcome based on initial evaluation following a first mIBG treatment (7400 MBq) in a patient cohort with such tumours, with a secondary aim of evaluating progression-free survival (PFS) and overall survival (OS) following mIBG therapy. METHODS Retrospective review of the hospital records was performed to identify a cohort of 38 adult patients who underwent (131)Iodine-mIBG therapy over a 9-year period for metastatic GEP NETs and neuroendocrine tumours with an unknown primary. Treatment response was evaluated based on radiological criteria (RECIST1.1), biochemical markers [serum Chromogranin A (CgA)/urinary 5HIAA] and symptomatic response at clinical follow-up, all evaluated at 3-6 months from first mIBG treatment. Progression-free survival (PFS) and overall survival (OS) from the first mIBG treatment were recorded. RESULTS At 3-6 months following a single mIBG therapy, 75%, 67%, and 63% of patients showed either a partial response (PR) or stable disease (SD) on radiological, biochemical, and symptomatic criteria, respectively. Complete response (CR) was not seen in any patient. OS from the date of diagnosis and from the first therapy was 8 years +/-1.1 (95% CI 5.7 to 10.2 years) and 4 years+/-0.69 (95% CI 2.6-5.3 years), respectively. Twenty-nine percent of patients were alive at 10 years. Significant survival advantage was seen in patients with SD/PR as compared to those who had progressive disease (PD) for each of these three criteria. CONCLUSION Biochemical, radiological (RECIST 1.1) and symptomatic assessment of disease status at 3 to 6 months after first I131-mIBG therapy stratifies patients with a poor prognosis. This can be used to identify patients who may benefit from alternative strategies of treatment.
Collapse
Affiliation(s)
- Nicola Mulholland
- Department of Nuclear Medicine, King's College Hospital, Denmark Hill, London, SE5 9RS, UK.
| | - Riddhika Chakravartty
- Department of Nuclear Medicine, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Lindsey Devlin
- Department of Nuclear Medicine, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Eleni Kalogianni
- Department of Nuclear Medicine, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Ben Corcoran
- Department of Nuclear Medicine, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Gillian Vivian
- Department of Nuclear Medicine, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| |
Collapse
|
465
|
Feasibility and utility of re-treatment with 177Lu-DOTATATE in GEP-NENs relapsed after treatment with 90Y-DOTATOC. Eur J Nucl Med Mol Imaging 2015; 42:1955-63. [DOI: 10.1007/s00259-015-3105-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 06/02/2015] [Indexed: 01/30/2023]
|
466
|
Ahlstedt J, Tran TA, Strand F, Holmqvist B, Strand SE, Gram M, Åkerström B. Biodistribution and pharmacokinetics of recombinant α1-microglobulin and its potential use in radioprotection of kidneys. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2015; 5:333-347. [PMID: 26269772 PMCID: PMC4529588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/27/2015] [Indexed: 06/04/2023]
Abstract
Peptide-receptor radionuclide therapy (PRRT) is a systemically administrated molecular targeted radiation therapy for treatment of neuroendocrine tumors. Fifteen years of clinical use show that renal toxicity, due to glomerular filtration of the peptides followed by local generation of highly reactive free radicals, is the main side-effect that limits the maximum activity that can be administrated for efficient therapy. α1-microglobulin (A1M) is an endogenous radical scavenger shown to prevent radiation-induced in vitro cell damage and protect non-irradiated surrounding cells. An important feature of A1M is that, following distribution to the blood, it is equilibrated to the extravascular compartments and filtrated in the kidneys. Aiming at developing renal protection against toxic side-effects of PRRT, we have characterized the pharmacokinetics and biodistribution of intravenously (i.v.) injected (125)I- and non-labelled recombinant human A1M and the (111)In- and fluorescence-labelled somatostatin analogue octreotide. Both molecules were predominantly localized to the kidneys, displaying a prevailing distribution in the cortex. A maximum of 76% of the injected A1M and 46% of the injected octreotide were present per gram kidney tissue at 10 to 20 minutes, respectively, after i.v. injection. Immunohistochemistry and fluorescence microscopy revealed a dominating co-existence of the two substances in proximal tubules, with a cellular co-localization in the epithelial cells. Importantly, analysis of kidney extracts displayed an intact, full-length A1M at least up to 60 minutes post-injection (p.i.). In summary, the results show a highly similar pharmacokinetics and biodistribution of A1M and octreotide, thus enabling the use of A1M to protect the kidneys tissue during PRRT.
Collapse
Affiliation(s)
- Jonas Ahlstedt
- Department of Clinical Sciences in Lund, Section of Medical Radiation Physics, Lund UniversityLund, Sweden
| | - Thuy A Tran
- Lund University Bioimaging Center, Lund UniversityLund, Sweden
| | - Filip Strand
- Department of Clinical Sciences in Lund, Section for Infection Medicine, Lund UniversityLund, Sweden
| | | | - Sven-Erik Strand
- Department of Clinical Sciences in Lund, Section of Medical Radiation Physics, Lund UniversityLund, Sweden
- Lund University Bioimaging Center, Lund UniversityLund, Sweden
| | - Magnus Gram
- Department of Clinical Sciences in Lund, Section for Infection Medicine, Lund UniversityLund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences in Lund, Section for Infection Medicine, Lund UniversityLund, Sweden
| |
Collapse
|
467
|
DNA damage in blood lymphocytes in patients after (177)Lu peptide receptor radionuclide therapy. Eur J Nucl Med Mol Imaging 2015; 42:1739-1749. [PMID: 26048612 PMCID: PMC4554740 DOI: 10.1007/s00259-015-3083-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/05/2015] [Indexed: 01/14/2023]
Abstract
Purpose The aim of the study was to investigate DNA double strand break (DSB) formation and its correlation with the absorbed dose to the blood lymphocytes of patients undergoing their first peptide receptor radionuclide therapy (PRRT) with 177Lu-labelled DOTATATE/DOTATOC. Methods The study group comprised 16 patients receiving their first PRRT. At least six peripheral blood samples were obtained before, and between 0.5 h and 48 h after radionuclide administration. From the time–activity curves of the blood and the whole body, residence times for blood self-irradiation and whole-body irradiation were determined. Peripheral blood lymphocytes were isolated, fixed with ethanol and subjected to immunofluorescence staining for colocalizing γ-H2AX/53BP1 DSB-marking foci. The average number of DSB foci per cell per patient sample was determined as a function of the absorbed dose to the blood and compared with an in vitro calibration curve established in our laboratory with 131I and 177Lu. Results The average number of radiation-induced foci (RIF) per cell increased over the first 5 h after radionuclide administration and decreased thereafter. A linear fit from 0 to 5 h as a function of the absorbed dose to the blood agreed with our in vitro calibration curve. At later time-points the number of RIF decreased, indicating progression of DNA repair. Conclusion Measurements of RIF and the absorbed dose to the blood after systemic administration of 177Lu may be used to obtain data on the individual dose–response relationships in vivo. Individual patient data were characterized by a linear dose-dependent increase and an exponential decay function describing repair. Electronic supplementary material The online version of this article (doi:10.1007/s00259-015-3083-9) contains supplementary material, which is available to authorized users.
Collapse
|
468
|
The nuclear medicine therapy care coordination service: a model for radiologist-driven patient-centered care. Acad Radiol 2015; 22:771-8. [PMID: 25766086 DOI: 10.1016/j.acra.2015.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/27/2015] [Accepted: 01/27/2015] [Indexed: 11/21/2022]
Abstract
We developed a longitudinal care coordination service to proactively deliver high-quality and family-centered care in patients receiving radioiodine therapy for thyroid cancer. In an iterative, multidisciplinary team manner, a pretherapy consultation service, which included scripted interactions, documentation, and checklists for quality control, evolved over time into a robust patient-centered longitudinal care coordination nuclear medicine service. Radiation safety precautions, the rationale for therapy, and management of patient expectations were addressed through the initial consultation, and discharge and posttreatment care were managed during subsequent follow-up. The patient-physician relationship created during longitudinal nuclear medicine therapy care is one tool to help counteract the growing commoditization of radiology. This article describes the process that the nuclear medicine specialists in our department established to enhance radiologist value by providing both exceptional thyroid cancer treatment and continuity of care.
Collapse
|
469
|
Pretherapeutic estimation of kidney function in patients treated with peptide receptor radionuclide therapy: can renal scintigraphy be safely omitted? Nucl Med Commun 2015; 35:1143-9. [PMID: 25171439 DOI: 10.1097/mnm.0000000000000194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE A pretherapeutic assessment of kidney function before peptide receptor radionuclide therapy (PRRT) is considered essential because of the potential renal toxicity associated with PRRT. The aim of this study was to investigate the diagnostic performance of laboratory testing and Tc-mercaptoacetyltriglycine (MAG3) renal scintigraphy with a focus on patients treated with PRRT. MATERIALS AND METHODS From January to December 2013 the kidney function of 152 patients was assessed using laboratory tests [creatinine, blood urea nitrogen (BUN), and glomerular filtration rate (GFR)] and Tc-MAG3 clearance. In 27 patients, kidney function was assessed before PRRT. Results of blood tests and Tc-MAG3 renal scintigraphy, considered the reference standard, were compared in the entire patient cohort (n=152) and in both subgroups (PRRT and non-PRRT) using Student's t-test. The cutoff values for the laboratory tests for the prediction of abnormal Tc-MAG3 clearance were determined by means of receiver operating characteristic analysis. In a further mathematical approach using discriminant analysis, a formula was derived for the prediction of kidney function that included all of the serum parameters. RESULTS In the PRRT subgroup, laboratory test-derived kidney function correlated significantly with Tc-MAG3 clearance (creatinine: r=-0.429, P=0.037; BUN: r=-0.45, P=0.027; GFR: r=0.44, P=0.022). The correlation was confirmed in the non-PRRT subgroup. The receiver operating characteristic analysis for prediction of abnormal Tc-MAG3 clearance resulted in area under the curves of 0.779 for creatinine alone (sensitivity 74.3%, specificity 71.1%; cutoff ≥0.995 mg/dl) and for the combination of creatinine, BUN, and GFR (sensitivity was 74.3% and specificity was 69.3%). CONCLUSION Laboratory tests of kidney function correlate significantly with Tc-MAG3 clearance. Because of the moderate accuracy for laboratory tests, Tc-MAG3 clearance is recommended as a standard test to assess kidney function before PRRT.
Collapse
|
470
|
Banerjee S, Pillai MRA, Knapp FFR. Lutetium-177 therapeutic radiopharmaceuticals: linking chemistry, radiochemistry, and practical applications. Chem Rev 2015; 115:2934-74. [PMID: 25865818 DOI: 10.1021/cr500171e] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sharmila Banerjee
- Radiopharmaceuticals Chemistry Section, Bhabha Atomic Research Centre (BARC), Mumbai 400 085, India.,Molecular Group of Companies, Puthuvype, Ernakulam, Kerala 682 508, India.,Medical Radioisotope Program, Oak Ridge National Laboratory (ORNL), P.O. Box 2008, 1 Bethel Valley Road, Oak Ridge, Tennessee 37830-6229, United States
| | - M R A Pillai
- Radiopharmaceuticals Chemistry Section, Bhabha Atomic Research Centre (BARC), Mumbai 400 085, India.,Molecular Group of Companies, Puthuvype, Ernakulam, Kerala 682 508, India.,Medical Radioisotope Program, Oak Ridge National Laboratory (ORNL), P.O. Box 2008, 1 Bethel Valley Road, Oak Ridge, Tennessee 37830-6229, United States
| | - F F Russ Knapp
- Radiopharmaceuticals Chemistry Section, Bhabha Atomic Research Centre (BARC), Mumbai 400 085, India.,Molecular Group of Companies, Puthuvype, Ernakulam, Kerala 682 508, India.,Medical Radioisotope Program, Oak Ridge National Laboratory (ORNL), P.O. Box 2008, 1 Bethel Valley Road, Oak Ridge, Tennessee 37830-6229, United States
| |
Collapse
|
471
|
Abstract
Peptide receptor radionuclide therapy with (90)Y-peptides is generally well tolerated. Acute side effects are usually mild; some are related to the coadministration of amino acids and others to the radiopeptide itself. Chronic and permanent effects on target organs, particularly kidneys and bone marrow, are generally mild if necessary precautions are taken. The potential risk to kidney and red marrow limits the amount of radioactivity that may be administered. However, when tumor masses are irradiated with adequate doses, volume reduction may be observed. (90)Y-octreotide has been the most used radiopeptide in the first 8 to 10 years of experience.
Collapse
Affiliation(s)
- Lisa Bodei
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Marta Cremonesi
- Division of Medical Physics, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Giovanni Paganelli
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy.
| |
Collapse
|
472
|
Werner RA, Brumberg J, Dierks A, Herrmann K, Biko J, Buck AK, Lapa C. Paralytic Subileus as an Adverse Effect of Amino Acid–Based Nephroprotection in a Patient Undergoing Peptide Receptor Radionuclide Therapy. Clin Nucl Med 2015; 40:263-4. [DOI: 10.1097/rlu.0000000000000624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
473
|
Lopez-Rodriguez V, Gaspar-Carcamo R, Pedraza-Lopez M, Rojas-Calderon E, Arteaga de Murphy C, Ferro-Flores G, Avila-Rodriguez M. Preparation and preclinical evaluation of 66Ga-DOTA-E(c(RGDfK))2 as a potential theranostic radiopharmaceutical. Nucl Med Biol 2015; 42:109-14. [DOI: 10.1016/j.nucmedbio.2014.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 09/26/2014] [Accepted: 09/30/2014] [Indexed: 12/11/2022]
|
474
|
Lapa C, Werner RA, Bluemel C, Lueckerath K, Muegge DO, Strate A, Haenscheid H, Schirbel A, Allen-Auerbach MS, Bundschuh RA, Buck AK, Herrmann K. Prediction of clinically relevant hyperkalemia in patients treated with peptide receptor radionuclide therapy. EJNMMI Res 2014; 4:74. [PMID: 25977880 PMCID: PMC4412196 DOI: 10.1186/s13550-014-0074-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 12/01/2014] [Indexed: 12/04/2022] Open
Abstract
Background Peptide receptor radionuclide therapy (PRRT) is applied in patients with advanced neuroendocrine tumors. Co-infused amino acids (AA) should prevent nephrotoxicity. The aims of this study were to correlate the incidence of AA-induced hyperkalemia (HK) (≥5.0 mmol/l) and to identify predictors of AA-induced severe HK (>6.0). Methods In 38 patients, standard activity of 177Lu-labelled somatostatin analogs was administered. Pre-therapeutic kidney function was assessed by renal scintigraphy and laboratory tests. For kidney protection, AA was co-infused. Biochemical parameters (potassium, glomerular filtration rate, creatinine, blood urea nitrogen (BUN), sodium, phosphate, chloride, and lactate dehydrogenase (LDH)) were obtained prior to 4 and 24 h after the AA infusion. Incidence of HK (≥5.0) was correlated with pre-therapeutic kidney function and serum parameters. Formulas for the prediction of severe hyperkalemia (>6.0) were computed and prospectively validated. Results At 4 h, HK (≥5.0) was present in 94.7% with severe HK (>6.0) in 36.1%. Values normalized after 24 h in 84.2%. Pre-therapeutic kidney function did not correlate with the incidence of severe HK. Increases in K+ were significantly correlated with decreases in phosphate (r = −0.444, p < 0.005) and increases in BUN (r = 0.313, p = 0.056). A baseline BUN of >28 mg/dl had a sensitivity of 84.6% and a specificity of 60.0% (AUC = 0.75) in predicting severe HK of >6.0 (phosphate, AUC = 0.37). Computing of five standard serum parameters (potassium, BUN, sodium, phosphate, LDH) resulted in a sensitivity of 88.9% and a specificity of 79.3% for the prediction of severe HK >6.0 (accuracy = 81.6%). Conclusions A combination of serum parameters predicted prospectively the occurrence of relevant HK with an accuracy of 81.6% underlining its potential utility for identifying ‘high-risk’ patients prone to PRRT.
Collapse
Affiliation(s)
- Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Christina Bluemel
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Katharina Lueckerath
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Dirk O Muegge
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria
| | - Alexander Strate
- Institute of Clinical Chemistry, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Heribert Haenscheid
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Martin S Allen-Auerbach
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles 90095, CA, USA
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Hospital Bonn, Sigmund-Freud-Straße 25, Bonn 53127, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| |
Collapse
|
475
|
D’Huyvetter M, Xavier C, Caveliers V, Lahoutte T, Muyldermans S, Devoogdt N. Radiolabeled nanobodies as theranostic tools in targeted radionuclide therapy of cancer. Expert Opin Drug Deliv 2014; 11:1939-54. [PMID: 25035968 PMCID: PMC4245996 DOI: 10.1517/17425247.2014.941803] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The integration of diagnostic testing for the presence of a molecular target is of interest to predict successful targeted radionuclide therapy (TRNT). This so-called 'theranostic' approach aims to improve personalized treatment based on the molecular characteristics of cancer cells. Moreover, it offers new insights in predicting adverse effects and provides appropriate tools to monitor therapy responses. Recent findings using nanobodies emphasize their potential as theranostic tools in cancer treatment. Nanobodies are recombinant, small antigen-binding fragments that are derived from camelid heavy-chain-only antibodies. AREAS COVERED We review the current status of theranostic approaches in TRNT, with a focus on antibodies, peptides, scaffold proteins and emerging nanobodies. In recent years, nanobodies have been evaluated intensively for molecular imaging. In addition, novel data on TRNT using radiolabeled nanobodies for carcinomas and multiple myeloma highlight their promising opportunities in cancer treatment. EXPERT OPINION We trust that radiolabeled nanobodies will have a future potential as theranostic tools in cancer therapy, both for diagnosis as well as for TRNT.
Collapse
Affiliation(s)
- Matthias D’Huyvetter
- Belgian Nuclear Research Center (SCK·CEN), Radiobiology Unit, Molecular and Cellular Biology Expert Group,
Mol, Belgium
- Vrije Universiteit Brussel (VUB), In vivo Cellular and Molecular Imaging Laboratory (ICMI),
Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Catarina Xavier
- Vrije Universiteit Brussel (VUB), In vivo Cellular and Molecular Imaging Laboratory (ICMI),
Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Vicky Caveliers
- Vrije Universiteit Brussel (VUB), In vivo Cellular and Molecular Imaging Laboratory (ICMI),
Laarbeeklaan 103, 1090 Brussels, Belgium
- UZ Brussel, Department of Nuclear Medicine,
Brussels, Belgium
| | - Tony Lahoutte
- Vrije Universiteit Brussel (VUB), In vivo Cellular and Molecular Imaging Laboratory (ICMI),
Laarbeeklaan 103, 1090 Brussels, Belgium
- UZ Brussel, Department of Nuclear Medicine,
Brussels, Belgium
| | - Serge Muyldermans
- Vrije Universiteit Brussel (VUB), Cellular and Molecular Immunology,
Pleinlaan 2, 1050 Brussels, Belgium+32 2 6291969;
- Vlaams Instituut voor Biotechnologie (VIB), Structural Biology Research Center,
Brussels, Belgium
| | - Nick Devoogdt
- Vrije Universiteit Brussel (VUB), In vivo Cellular and Molecular Imaging Laboratory (ICMI),
Laarbeeklaan 103, 1090 Brussels, Belgium
- Vrije Universiteit Brussel (VUB), Cellular and Molecular Immunology,
Pleinlaan 2, 1050 Brussels, Belgium+32 2 6291969;
| |
Collapse
|
476
|
Chalkia MT, Stefanoyiannis AP, Chatziioannou SN, Round WH, Efstathopoulos EP, Nikiforidis GC. Patient-specific dosimetry in peptide receptor radionuclide therapy: a clinical review. AUSTRALASIAN PHYSICAL & ENGINEERING SCIENCES IN MEDICINE 2014; 38:7-22. [PMID: 25427548 DOI: 10.1007/s13246-014-0312-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 11/06/2014] [Indexed: 12/16/2022]
Abstract
Neuroendocrine tumours (NETs) belong to a relatively rare class of neoplasms. Nonetheless, their prevalence has increased significantly during the last decades. Peptide receptor radionuclide therapy (PRRT) is a relatively new treatment approach for inoperable or metastasised NETs. The therapeutic effect is based on the binding of radiolabelled somatostatin analogue peptides with NETs' somatostatin receptors, resulting in internal irradiation of tumours. Pre-therapeutic patient-specific dosimetry is essential to ensure that a treatment course has high levels of safety and efficacy. This paper reviews the methods applied for PRRT dosimetry, as well as the dosimetric results presented in the literature. Focus is given on data concerning the therapeutic somatostatin analogue radiopeptides (111)In-[DTPA(0),D-Phe(1)]-octreotide ((111)In-DTPA-octreotide), (90)Y-[DOTA(0),Tyr(3)]-octreotide ((90)Y-DOTATOC) and (177)Lu-[DOTA(0),Tyr(3),Thr(8)]-octreotide ((177)Lu-DOTATATE). Following the Medical Internal Radiation Dose (MIRD) Committee formalism, dosimetric analysis demonstrates large interpatient variability in tumour and organ uptake, with kidneys and bone marrow being the critical organs. The results are dependent on the image acquisition and processing protocol, as well as the dosimetric imaging radiopharmaceutical.
Collapse
Affiliation(s)
- M T Chalkia
- University General Hospital of Athens "Attikon", 1, Rimini Street, Chaidari, 12462, Athens, Greece
| | | | | | | | | | | |
Collapse
|
477
|
Czepczyński R, Matysiak-Grześ M, Gryczyńska M, Bączyk M, Wyszomirska A, Stajgis M, Ruchała M. Peptide receptor radionuclide therapy of differentiated thyroid cancer: efficacy and toxicity. Arch Immunol Ther Exp (Warsz) 2014; 63:147-54. [PMID: 25403743 PMCID: PMC4359293 DOI: 10.1007/s00005-014-0318-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 08/01/2014] [Indexed: 11/24/2022]
Abstract
In rare cases of differentiated thyroid carcinoma (DTC), radioiodine treatment is no longer effective due to cell dedifferentiation. Targeting somatostatin receptors in DTC cells by radiolabelled somatostatin analogues could provide an alternative therapy option. The aim of this study was to evaluate safety and efficacy of peptide receptor radionuclide therapy (PRRT) in patients with advanced, non-iodine avid DTC. Eleven patients aged 47–81 years (median: 65 years) with a history of several courses of radioiodine therapy, increasing thyroglobulin (Tg) and negative whole body scan, were qualified to the study. After confirming receptor expression by somatostatin receptor scintigraphy, PRRT with yttrium-90 labelled analogue was initiated. Fractionated treatment protocol was used with four doses of 90Y-DOTA-TOC in 12-week intervals. Activity of each dose was 3.7 GBq (100 mCi). Of 11 patients, 5 died before receiving the fourth course of PRRT. In the remaining six patients, morphological response, evaluated 3 months after the last course using RECIST criteria showed partial remission (PR) in one patient, stable disease (SD) in two patients and progressive disease (PD) in three patients. Biochemical response based on Tg measurements before and after PRRT showed PR in one patient, SD in four patients and PD in one patient. Median survival was 21 months from the first course of PRRT. Only minor and transient hematological toxicity was observed in some patients. We conclude that PRRT is generally well-tolerated and may be a valuable option for some patients with radioiodine-refractory DTC.
Collapse
Affiliation(s)
- Rafał Czepczyński
- Department of Endocrinology, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznań, Poland,
| | | | | | | | | | | | | |
Collapse
|
478
|
Dalmo J, Westberg E, Barregard L, Svedbom L, Johansson M, Törnqvist M, Forssell-Aronsson E. Evaluation of retinol binding protein 4 and carbamoylated haemoglobin as potential renal toxicity biomarkers in adult mice treated with (177)Lu-octreotate. EJNMMI Res 2014; 4:59. [PMID: 26116120 PMCID: PMC4452688 DOI: 10.1186/s13550-014-0059-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 10/13/2014] [Indexed: 12/25/2022] Open
Abstract
Background The kidneys are regarded as one of the main dose-limiting organs in the treatment of neuroendocrine tumours with 177Lu-[DOTA0, Tyr3]-octreotate (177Lu-octreotate), despite the successful use of kidney uptake blocking agents such as lysine and arginine. To avoid renal toxicity but still give each patient as high amount of 177Lu-octreotate as possible, there is a need for methods/biomarkers that indicate renal injury in an early stage of the treatment. The aim of this study was to investigate the potential of using urinary retinol binding protein 4 (RBP4) and carbamoylated haemoglobin (Hb) in blood as biomarkers of nephrotoxic effects on adult mice after 177Lu-octreotate treatment. Methods Adult BALB/c nude mice were injected with 60 MBq or 120 MBq of 177Lu-octreotate or with saline (control). Urine was collected before injection and concentrations of urinary RBP4 and creatinine were determined 14 to 90 days after injection Blood samples were collected after 90 days, and carbamoylated N-terminal valine in Hb, formed from urea, was measured as valine hydantoin (VH) after detachment from Hb. Results The RBP4 values increased with administered activity and time. For the 60 and 120 MBq groups, statistically significantly higher RBP4 levels (p <0.05) were found at day 60 and 90 compared to baseline, also at day 30 for 120 MBq group. For VH, the mean values were similar for the 60 MBq and control groups, while a small increase was observed for the 120 MBq group; but there were no statistically significant differences between any of the groups (p >0.05). No morphological changes in the kidney tissue were found. Conclusions Urinary RBP4 is a promising new biomarker for radiation-induced renal toxicity. For the conditions used in this experiment, carbamoylated Hb (from urea) measured as VH may not be a sufficiently sensitive biomarker to be used for renal toxicity. Trial registration ID 326-2008
Collapse
Affiliation(s)
- Johanna Dalmo
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Centre, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, SE-413 45, Sweden,
| | | | | | | | | | | | | |
Collapse
|
479
|
Lim JC, Cho EH, Kim JJ, Choi SM, Lee SY, Nam SS, Park UJ, Park SH. Preclinical pharmacokinetic, biodistribution, imaging and therapeutic efficacy of (177)Lu-Labeled glycated bombesin analogue for gastrin-releasing peptide receptor-positive prostate tumor targeting. Nucl Med Biol 2014; 42:234-41. [PMID: 25498002 DOI: 10.1016/j.nucmedbio.2014.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 10/24/2022]
Abstract
UNLABELLED The gastrin-releasing peptide receptor (GRPR) has been shown to be overexpressed in many human tumors, including prostate, colon, gastric, breast, pancreatic, and small cell lung cancers. Because bombesin (BBS) binds to GRPR with high affinity, BBS derivatives have been labeled with various radionuclides and have been demonstrated to be successful candidates for peptide receptor radiotherapy (PRRT). The present study describes the in vitro and in vivo preclinical characteristics of (177)Lu-DOTA-Lys(glucose)-4 aminobenzoic acid-BBS7-14 ((177)Lu-DOTA-gluBBN) to prepare radiolabeled candidates for the treatment of GRPR-expressing prostate tumors. METHODS (177)Lu-DOTA-gluBBN was prepared as previously published [1]. Human prostate PC-3 tumor cells were used to determine the binding (Kd) retention and efflux of (177)Lu-DOTA-gluBBN. Pharmacokinetic, imaging, and radiotherapy studies were performed in PC-3 xenografted mice. RESULTS The Kd value of (177)Lu-DOTA-gluBBN was 0.63 nM, with a maximum binding capacity (Bmax) of 669.7 fmol/10(6) cells (4.04×10(5) GRPR/cell). During a 2-hr incubation, 90.1±0.4% of the cell-associated radio-peptide was internalized, and 56.3±7.1% of the internalized radio-peptide was externalized in vitro. High amounts of the radio-peptide were rapidly accumulated in a PC-3 tumor in vivo, and the % ID/g of the tumor was 12.42±2.15 1 hr p.i. The radio-peptide was quickly cleared from the blood, yielding tumor-to-blood ratios of 39.22±17.36 at 1 hr p.i. and 330.67±131.23 at 24hr p.i. In addition, (177)Lu-DOTA-gluBBN was clearly visualized in PC-3 tumors 1 hr p.i. and significantly inhibited the tumor growth (P<0.05). Treatment-related toxicity in the pancreas and kidneys was not observed, except for slight glomerulopathy. CONCLUSIONS The pharmacokinetic, imaging, and therapy studies suggest that this (177)Lu-DOTA-gluBBN has promising characteristics for application in nuclear medicine, namely, for the diagnosis and treatment of GRPR-overexpressing prostate tumors.
Collapse
Affiliation(s)
- Jae Cheong Lim
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 305-353, Republic of Korea; Bio-therapy Human Resources Center, Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea.
| | - Eun Ha Cho
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 305-353, Republic of Korea
| | - Jin Joo Kim
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 305-353, Republic of Korea
| | - Sang Mu Choi
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 305-353, Republic of Korea
| | - So Young Lee
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 305-353, Republic of Korea
| | - Sung Soo Nam
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 305-353, Republic of Korea
| | - Ul Jae Park
- Radioisotope Research Division, Department of Research Reactor Utilization, Korea Atomic Energy Research Institute, Daejeon 305-353, Republic of Korea
| | - Soo Hyun Park
- Bio-therapy Human Resources Center, Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea.
| |
Collapse
|
480
|
Prasad V, Brenner W, Modlin IM. How smart is peptide receptor radionuclide therapy of neuroendocrine tumors especially in the salvage setting? The clinician's perspective. Eur J Nucl Med Mol Imaging 2014; 41:202-4. [PMID: 24196915 DOI: 10.1007/s00259-013-2593-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Vikas Prasad
- Department of Nuclear Medicine, Campus Virchow-Klinikum, Charité University Hospital, Berlin, Germany,
| | | | | |
Collapse
|
481
|
Wong KK, Fig LM, Youssef E, Ferretti A, Rubello D, Gross MD. Endocrine scintigraphy with hybrid SPECT/CT. Endocr Rev 2014; 35:717-46. [PMID: 24977318 DOI: 10.1210/er.2013-1030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nuclear medicine imaging of endocrine disorders takes advantage of unique cellular properties of endocrine organs and tissues that can be depicted by targeted radiopharmaceuticals. Detailed functional maps of biodistributions of radiopharmaceutical uptake can be displayed in three-dimensional tomographic formats, using single photon emission computed tomography (CT) that can now be directly combined with simultaneously acquired cross-sectional anatomic maps derived from CT. The integration of function depicted by scintigraphy and anatomy with CT has synergistically improved the efficacy of nuclear medicine imaging across a broad spectrum of clinical applications, which include some of the oldest imaging studies of endocrine dysfunction.
Collapse
Affiliation(s)
- Ka Kit Wong
- Nuclear Medicine/Radiology Department (K.K.W., E.Y., M.D.G.), University of Michigan Hospital, Ann Arbor, Michigan 48109; Nuclear Medicine Service (K.K.W., L.M.F., M.D.G.), Department of Veterans Affairs Health System, Ann Arbor, Michigan 48105; and Department of Nuclear Medicine (A.F., D.R.), Radiology, Medical Physics, Santa Maria della Misericordia Hospital, 45100 Rovigo, Italy
| | | | | | | | | | | |
Collapse
|
482
|
Gene expression accurately distinguishes liver metastases of small bowel and pancreas neuroendocrine tumors. Clin Exp Metastasis 2014; 31:935-44. [PMID: 25241033 DOI: 10.1007/s10585-014-9681-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/12/2014] [Indexed: 12/31/2022]
Abstract
Small bowel (SBNETs) and pancreatic neuroendocrine tumors (PNETs) often present with liver metastases. Although liver biopsy establishes a neuroendocrine diagnosis, the primary tumor site is frequently unknown without exploratory surgery. Gene expression differences in metastases may distinguish primary SBNETs and PNETs. This study sought to determine expression differences of four genes in neuroendocrine metastases and to create a gene expression algorithm to distinguish the primary site. Nodal and liver metastases from SBNETs and PNETs (n = 136) were collected at surgery under an Institutional Review Board-approved protocol. Quantitative PCR measured expression of bombesin-like receptor-3, opioid receptor kappa-1, oxytocin receptor, and secretin receptor in metastases. Logistic regression models defined an algorithm predicting the primary tumor site. Models were developed on a training set of 21 nodal metastases and performance was validated on an independent set of nodal and liver metastases. Expression of all four genes was significantly different in SBNET compared to PNET metastases. The optimal model employed expression of bombesin-like receptor-3 and opioid receptor kappa-1. When these genes did not amplify, the algorithm used oxytocin receptor and secretin receptor expression, which allowed classification of all 136 metastases with 94.1 % accuracy. In the independent liver metastasis validation set, 52/56 (92.9 %) were correctly classified. Positive predictive values were 92.5 % for SBNETs and 93.8 % for PNETs. This validated algorithm accurately distinguishes SBNET and PNET metastases based on their expression of four genes. High accuracy in liver metastases demonstrates applicability to the clinical setting. Studies assessing this algorithm's utility in prospective clinical decision-making are warranted.
Collapse
|
483
|
Basu S, Ostwal V, Ranade R, Panda D. Supportive Measures and Finer Practice Points in 177Lu-DOTATATE PRRT for NET: Aiming for Optimal Disease Management. J Nucl Med 2014; 55:1916-7. [DOI: 10.2967/jnumed.114.146282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
484
|
Lapa C, Werner RA, Bluemel C, Lückerath K, Schirbel A, Strate A, Buck AK, Herrmann K. Influence of the amount of co-infused amino acids on post-therapeutic potassium levels in peptide receptor radionuclide therapy. EJNMMI Res 2014; 4:46. [PMID: 25977877 PMCID: PMC4412194 DOI: 10.1186/s13550-014-0046-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/13/2014] [Indexed: 11/22/2022] Open
Abstract
Background Peptide receptor radionuclide therapy (PRRT) is routinely used for advanced or metastasized neuroendocrine tumours (NET). To prevent nephrotoxicity, positively charged amino acids (AA) are co-infused. The aim of this study was to correlate the risk for therapy-related hyperkalaemia with the total amount of AA infused. Methods Twenty-two patients undergoing PRRT with standard activities of 177Lu-DOTATATE/-TOC were monitored during two following treatment cycles with co-infusion of 75 and 50 g of AA (l-arginine and l-lysine), respectively. Mean serum levels of potassium and other parameters (glomerular filtration rate [GFR], creatinine, blood urea nitrogen [BUN], phosphate, chloride, lactate dehydrogenase) prior to, 4 h and 24 h after AA infusion were compared. Results Self-limiting hyperkalaemia (>5.0 mmol/l) resolving after 24 h occurred in 91% (20/22) of patients in both protocols. Potassium levels, BUN, creatinine, GFR, phosphate, chloride and LDH showed a similar range at 4 h after co-infusion of 75 or 50 g of AA, respectively (p?>?0.05). Only GFR and creatinine levels at 24 h varied significantly between the two co-infusion protocols (p?<?0.05). Conclusions Hyperkalaemia is a frequent side effect of AA infusion in PRRT. Varying the dose of co-infused amino acids did not impact on the incidence and severity of hyperkalaemia.
Collapse
Affiliation(s)
- Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Christina Bluemel
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Alexander Strate
- Institute of Clinical Chemistry, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg 97080, Germany
| |
Collapse
|
485
|
68Gallium- and 90Yttrium-/ 177Lutetium: "theranostic twins" for diagnosis and treatment of NETs. Ann Nucl Med 2014; 29:1-7. [PMID: 25139472 PMCID: PMC4306729 DOI: 10.1007/s12149-014-0898-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 08/07/2014] [Indexed: 12/16/2022]
Abstract
Abundant expression of somatostatin receptors (SSTR) is frequently identified in differentiated neuroendocrine tumors and may serve as potential target for diagnostic imaging and treatment. This article discusses the “theranostic approach” of SSTR-targeting compounds including an overview of its role for diagnosis, staging and restaging, discussing its way to being established in clinical routine, and giving an outlook about further potentially relevant developments.
Collapse
|
486
|
Spampatti M, Vlotides G, Spöttl G, Maurer J, Göke B, Auernhammer CJ. Aspirin inhibits cell viability and mTOR downstream signaling in gastroenteropancreatic and bronchopulmonary neuroendocrine tumor cells. World J Gastroenterol 2014; 20:10038-10049. [PMID: 25110431 PMCID: PMC4123333 DOI: 10.3748/wjg.v20.i29.10038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of aspirin on neuroendocrine tumor (NET) cell growth and signaling in vitro.
METHODS: Human pancreatic BON1, bronchopulmonary NCI-H727 and midgut GOT1 neuroendocrine tumor cells were treated with different concentrations of aspirin (from 0.001 to 5 mmol/L), and the resulting effects on metabolic activity/cell proliferation were measured using cell proliferation assays and SYBR-DNA-labeling after 72, 144 and 216 h of incubation. The effects of aspirin on the expression and phosphorylation of several critical proteins that are involved in the most common intracellular growth factor signaling pathways (especially Akt protein kinase B) and mammalian target of rapamycin (mTOR) were determined by Western blot analyses. Propidium iodide staining and flow cytometry were used to evaluate changes in cell cycle distribution and apoptosis. Statistical analysis was performed using a 2-tailed Student’s t-test to evaluate the proliferation assays and cell cycle analyses. The results are expressed as the mean ± SD of 3 or 4 independently performed experiments. Statistical significance was set at P < 0.05.
RESULTS: Treatment with aspirin suppressed the viability/proliferation of BON1, NCI-H727 and GOT1 cells in a time- and dose-dependent manner. Significant effects were observed at starting doses of 0.5-1 mmol/L and peaked at 5 mmol/L. For instance, after treatment with 1 mmol/L aspirin for 144 h, the viability of pancreatic BON1 cells decreased to 66% ± 13% (P < 0.05), the viability of bronchopulmonary NCI-H727 cells decreased to 53% ± 8% (P < 0.01) and the viability of midgut GOT1 cells decreased to 89% ± 6% (P < 0.01). These effects were associated with a decreased entry into the S phase, the induction of the cyclin-dependent kinase inhibitor p21 and reduced expression of cyclin-dependent kinase 4 and cyclin D3. Aspirin suppressed mTOR downstream signaling, evidenced by the reduced phosphorylation of the mTOR substrates 4E binding protein 1, serine/threonine kinase P70S6K and S6 ribosomal protein and inhibited glycogen synthase kinase 3 activity. We observed the (compensatory) activation of tuberous sclerosis 2, the serine/threonine specific protein kinase AKT and extracellular signal-regulated kinases.
CONCLUSION: Aspirin demonstrates promising anticancer properties for NETs in vitro. Further preclinical and clinical studies are needed.
Collapse
|
487
|
Chan LWC, Ngo CHC, Wang F, Zhao MY, Zhao M, Law HKW, Wong SCC, Yung BYM. Disease-specific target gene expression profiling of molecular imaging probes: database development and clinical validation. Mol Imaging 2014; 13. [PMID: 25022454 DOI: 10.2310/7290.2014.00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Molecular imaging probes can target abnormal gene expression patterns in patients and allow early diagnosis of disease. For selecting a suitable imaging probe, the current Molecular Imaging and Contrast Agent Database (MICAD) provides descriptive and qualitative information on imaging probe characteristics and properties. However, MICAD does not support linkage with the expression profiles of target genes. The proposed Disease-specific Imaging Probe Profiling (DIPP) database quantitatively archives and presents the gene expression profiles of targets across different diseases, anatomic regions, and subcellular locations, providing an objective reference for selecting imaging probes. The DIPP database was validated with a clinical positron emission tomography (PET) study on lung cancer and an in vitro study on neuroendocrine cancer. The retrieved records show that choline kinase beta and glucose transporters were positively and significantly associated with lung cancer among the targets of 11C-choline and [18F]fluoro-2-deoxy-2-d-glucose (FDG), respectively. Their significant overexpressions corresponded to the findings that the uptake rate of FDG increased with tumor size but that of 11C-choline remained constant. Validated with the in vitro study, the expression profiles of disease-associated targets can indicate the eligibility of patients for clinical trials of the treatment probe. A Web search tool of the DIPP database is available at http://www.polyu.edu.hk/bmi/dipp/.
Collapse
|
488
|
Kraeber-Bodéré F, Barbet J. Challenges in nuclear medicine: innovative theranostic tools for personalized medicine. Front Med (Lausanne) 2014; 1:16. [PMID: 25705627 PMCID: PMC4335403 DOI: 10.3389/fmed.2014.00016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/04/2014] [Indexed: 12/17/2022] Open
Affiliation(s)
- Françoise Kraeber-Bodéré
- Nuclear Medicine, University Hospital-ICO, CRCNA, INSERM U892, CNRS UMR 6299, Nantes and GIP Arronax , Saint-Herblain , France
| | - Jacques Barbet
- Nuclear Medicine, University Hospital-ICO, CRCNA, INSERM U892, CNRS UMR 6299, Nantes and GIP Arronax , Saint-Herblain , France
| |
Collapse
|
489
|
Bodei L, Cremonesi M, Kidd M, Grana CM, Severi S, Modlin IM, Paganelli G. Peptide receptor radionuclide therapy for advanced neuroendocrine tumors. Thorac Surg Clin 2014; 24:333-49. [PMID: 25065935 DOI: 10.1016/j.thorsurg.2014.04.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) consists of the systemic administration of a synthetic peptide, labeled with a suitable β-emitting radionuclide, able to irradiate tumors and their metastases via internalization through a specific receptor (usually somatostatin S2), over-expressed on the cell membrane. After almost 2 decades of experience, PRRT, with either (90)Y-octreotide or (177)Lu-octreotate, has established itself to be an efficient and effective therapeutic modality. As a treatment, it is relatively safe up to the known thresholds of absorbed and bio-effective isotope dosages and the renal and hematological toxicity profiles are acceptable if adequate protective measures are undertaken.
Collapse
Affiliation(s)
- Lisa Bodei
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, Milan 20141, Italy.
| | - Marta Cremonesi
- Division of Health Physics, European Institute of Oncology, via Ripamonti 435, Milan 20141, Italy
| | - Mark Kidd
- Department of Surgery, Yale School of Medicine, 310 Cedar Street, New Haven, CT 06520, USA
| | - Chiara M Grana
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, Milan 20141, Italy
| | - Stefano Severi
- Radiometabolic Unit, Department of Nuclear Medicine, IRST-IRCCS, Via Maroncelli 40, Meldola 47014, Italy
| | - Irvin M Modlin
- Department of Surgery, Yale School of Medicine, 310 Cedar Street, New Haven, CT 06520, USA; Clifton Life Sciences, Branford, CT 06405, USA
| | - Giovanni Paganelli
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, Milan 20141, Italy; Radiometabolic Unit, Department of Nuclear Medicine, IRST-IRCCS, Via Maroncelli 40, Meldola 47014, Italy
| |
Collapse
|
490
|
Jois B, Asopa R, Basu S. Somatostatin Receptor Imaging in Non–131I-Avid Metastatic Differentiated Thyroid Carcinoma for Determining the Feasibility of Peptide Receptor Radionuclide Therapy With 177Lu-DOTATATE. Clin Nucl Med 2014; 39:505-10. [DOI: 10.1097/rlu.0000000000000429] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
491
|
Basu S, Sirohi B, Shrikhande SV. Dual tracer imaging approach in assessing tumor biology and heterogeneity in neuroendocrine tumors: its correlation with tumor proliferation index and possible multifaceted implications for personalized clinical management decisions, with focus on PRRT. Eur J Nucl Med Mol Imaging 2014; 41:1492-6. [PMID: 24863431 DOI: 10.1007/s00259-014-2805-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/05/2014] [Indexed: 12/28/2022]
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Jerbai Wadia Road, Parel, Mumbai, 400 012, India,
| | | | | |
Collapse
|
492
|
Kong G, Thompson M, Collins M, Herschtal A, Hofman MS, Johnston V, Eu P, Michael M, Hicks RJ. Assessment of predictors of response and long-term survival of patients with neuroendocrine tumour treated with peptide receptor chemoradionuclide therapy (PRCRT). Eur J Nucl Med Mol Imaging 2014; 41:1831-44. [PMID: 24844348 PMCID: PMC4159597 DOI: 10.1007/s00259-014-2788-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 04/15/2014] [Indexed: 01/08/2023]
Abstract
PURPOSE To review the response and outcomes of (177)Lu-DOTA-octreotate chemoradionuclide therapy (LuTate PRCRT) in patients with neuroendocrine tumour (NET) expressing high levels of somatostatin receptors with uncontrolled symptoms or disease progression. METHODS A total of 68 patients (39 men; 17 - 76 years of age) who had completed an induction course of at least three cycles of LuTate PRCRT between January 2006 and June 2010 were reviewed. Ten patients were treated for uncontrolled symptoms and 58 had disease progression despite conventional treatment. The majority had four induction LuTate cycles (median treatment duration 5 months and cumulative activity 31 GBq), and 63 patients had concomitant 5-FU radiosensitizing infusional chemotherapy. Factors predicting overall survival were assessed using the log-rank test and Cox proportional hazards regression. RESULTS Of those treated for uncontrolled symptoms, 70 % received benefit maintained for at least 6 months after treatment. Among patients with progressive disease 68 % showed stabilization or regression on CT, 67 % on molecular imaging and 56 % biochemically up to 12 months after treatment; 32 patients died. Overall survival rates at 2 and 5 year were 72.1 % and 52.1 %, respectively. Median overall survival was not estimable at a median follow-up of 60 months (range 5 - 86 months). Nonpancreatic primary sites, dominant liver metastases, lesion size <5 cm and the use of 5-FU chemotherapy were statistically significantly associated with objective response. A disseminated pattern and a high disease burden (whole-body retention index) were associated with an increased risk of death. Objective biochemical, molecular imaging and CT responses were all associated with longer overall survival. CONCLUSION A high proportion of patients with progressive NET or uncontrolled symptoms received therapeutic benefit from LuTate with concomitant 5-FU chemotherapy. The achievement of objective biochemical, molecular or CT responses within 12 months was associated with improved overall survival. Patients with a primary pancreatic site and larger lesions (>5 cm) appeared to have lower objective response rates and may need a more aggressive treatment approach.
Collapse
Affiliation(s)
- G Kong
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
493
|
Minutoli F, Amato E, Sindoni A, Cardile D, Conti A, Herberg A, Baldari S. Peptide receptor radionuclide therapy in patients with inoperable meningiomas: our experience and review of the literature. Cancer Biother Radiopharm 2014; 29:193-9. [PMID: 24811687 DOI: 10.1089/cbr.2013.1599] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE Few studies describe peptide receptor radionuclide therapy (PRRT) using (90)Y- or (177)Lu-labeled peptides in patients with recurrent meningiomas. No clinical data about (111)In-Pentetreotide in such patients are available. We report on (111)In-Pentetreotide therapy in patients with inoperable meningiomas and review the literature about PRRT of meningiomas. METHODS We reviewed clinical records of 8 patients with meningioma/meningiomatosis showing high (111)In-Pentetreotide uptake on pretherapy scintigraphy who were treated with at least one cycle of (111)In-Pentetreotide. In 2 patients, a cocktail of (111)In-Pentetreotide and beta-emitting radiolabeled peptides had been administered. RESULTS No patient experienced acute toxicity, neurological or renal function impairment. Mild transient bone marrow toxicity was observed in 4 patients. Objective partial response was observed in 2 patients, stable disease in 5 and disease progression in one. There were no statistically significant correlations between objective response and patient age, tumor WHO grade, baseline Karnofsky performance score, (111)In-Pentetreotide tumoral uptake grade, tumor/nontumor ratio, disease state at baseline, and cumulative dose. CONCLUSIONS In consideration of its efficacy and the lack of significant toxicity, PRRT of meningiomas using (111)In-Pentetreotide could be proposed even nowadays when the use of (177)Lu- or (90)Y-labeled peptides seems unsafe, namely in patients with renal impairment/toxicity.
Collapse
Affiliation(s)
- Fabio Minutoli
- 1 Department of Biomedical Sciences and Morphological and Functional Images, University of Messina , Messina, Italy
| | | | | | | | | | | | | |
Collapse
|
494
|
de Herder WW. GEP-NETS update: functional localisation and scintigraphy in neuroendocrine tumours of the gastrointestinal tract and pancreas (GEP-NETs). Eur J Endocrinol 2014; 170:R173-83. [PMID: 24723670 DOI: 10.1530/eje-14-0077] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For patients with neuroendocrine tumours (NETs) of the gastrointestinal tract and pancreas (GEP) (GEP-NETs), excellent care should ideally be provided by a multidisciplinary team of skilled health care professionals. In these patients, a combination of nuclear medicine imaging and conventional radiological imaging techniques is usually mandatory for primary tumour visualisation, tumour staging and evaluation of treatment. In specific cases, as in patients with occult insulinomas, sampling procedures can provide a clue as to where to localise the insulin-hypersecreting pancreatic NETs. Recent developments in these fields have led to an increase in the detection rate of primary GEP-NETs and their metastatic deposits. Radiopharmaceuticals targeted at specific tumour cell properties and processes can be used to provide sensitive and specific whole-body imaging. Functional imaging also allows for patient selection for receptor-based therapies and prediction of the efficacy of such therapies. Positron emission tomography/computed tomography (CT) and single-photon emission CT/CT are used to map functional images with anatomical localisations. As a result, tumour imaging and tumour follow-up strategies can be optimised for every individual GEP-NET patient. In some cases, functional imaging might give indications with regard to future tumour behaviour and prognosis.
Collapse
Affiliation(s)
- Wouter W de Herder
- Section of Endocrinology, Department of Internal Medicine, Erasmus MC, 's Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| |
Collapse
|
495
|
Sherman SK, Maxwell JE, O'Dorisio MS, O'Dorisio TM, Howe JR. Pancreastatin predicts survival in neuroendocrine tumors. Ann Surg Oncol 2014; 21:2971-80. [PMID: 24752611 DOI: 10.1245/s10434-014-3728-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Serum neurokinin A, chromogranin A, serotonin, and pancreastatin reflect tumor burden in neuroendocrine tumors. We sought to determine whether their levels correlate with survival in surgically managed small bowel (SBNETs) and pancreatic neuroendocrine tumors (PNETs). METHODS Clinical data were collected with Institutional Review Board approval for patients undergoing surgery at one center. Progression-free (PFS) and overall (OS) survival were from the time of surgery. Event times were estimated by the Kaplan-Meier method. Preoperative and postoperative laboratory values were tested for correlation with outcomes. A multivariate Cox model adjusted for confounders. RESULTS Included were 98 SBNETs and 78 PNETs. Median follow-up was 3.8 years; 62 % had metastatic disease. SBNETs had lower median PFS than PNETs (2.0 vs. 5.6 years; p < 0.01). Median OS was 10.5 years for PNETs and was not reached for SBNETs. Preoperative neurokinin A did not correlate with PFS or OS. Preoperative serotonin correlated with PFS but not OS. Higher levels of preoperative chromogranin A and pancreastatin showed significant correlation with worse PFS and OS (p < 0.05). After multivariate adjustment for confounders, preoperative and postoperative pancreastatin remained independently predictive of worse PFS and OS (p < 0.05). Whether pancreastatin normalized postoperatively further discriminated outcomes. Median PFS was 1.7 years in patients with elevated preoperative pancreastatin versus 6.5 years in patients with normal levels (p < 0.001). CONCLUSIONS Higher pancreastatin levels are significantly associated with worse PFS and OS in SBNETs and PNETs. This effect is independent of age, primary tumor site, and presence of nodal or metastatic disease. Pancreastatin provides valuable prognostic information and identifies surgical patients at high risk of recurrence who could benefit most from novel therapies.
Collapse
Affiliation(s)
- Scott K Sherman
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | | | | | | |
Collapse
|
496
|
Versari A, Sollini M, Frasoldati A, Fraternali A, Filice A, Froio A, Asti M, Fioroni F, Cremonini N, Putzer D, Erba PA. Differentiated thyroid cancer: a new perspective with radiolabeled somatostatin analogues for imaging and treatment of patients. Thyroid 2014; 24:715-26. [PMID: 24102584 DOI: 10.1089/thy.2013.0225] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND The expression of somatostatin receptors (SSTR) in thyroid cells may offer the possibility to identify metastatic lesions and to select patients for peptide receptor radionuclide therapy (PRRT). We investigated (68)Ga-DOTATOC positron emission tomography/computed tomography (PET/CT) to select patients with progressive differentiated thyroid cancer (DTC) for PRRT as well as treatment response and toxicity in treated patients. METHODS We enrolled 41 patients with progressive radioiodine-negative DTC (24 women and 17 men; mean age=54.3 years, median=59 years, range=19-78 years). In all patients, [(18)F]FDG-PET/CT was performed to determine recurrent disease with enhanced glucose metabolism, and (68)Ga-DOTATOC PET/CT was used to identify SSTR expression. Dosimetric evaluation was performed with (111)In-DOTATOC scintigraphy. Eleven patients were treated with PRRT receiving a fractionated injection of 1.5-3.7 GBq (90)Y-DOTATOC/administration. Serial (68)Ga-DOTATOC PET/CT scans were performed in all treated patients to evaluate treatment response. Parameters provided by (68)Ga-DOTATOC PET/CT were analyzed as potential therapeutic predictors to differentiate responding from nonresponding. In all treated patients, adverse events and toxicity were recorded. RESULTS (68)Ga-DOTATOC PET/CT were positive in 24/41 of radioiodine-negative DTC patients. Based on the high expression of SSTR detected by (68)Ga-DOTATOC PET/CT, 13 patients were suitable for PRRT. Two out of 13 patients were not treated due to the lack of fulfillment of other study inclusion criteria. PRRT induced disease control in 7/11 patients (two partial response and five stabilization) with a duration of response of 3.5-11.5 months. Objective response was associated with symptoms relief. Functional volume (FV) over time obtained by PET/CT was the only parameter demonstrating a significant difference between lesions responding and nonresponding to PRRT (p=0.001). Main PRRT adverse events were nausea, asthenia, and transient hematologic toxicity. One patient experienced permanent renal toxicity. CONCLUSIONS In our series, SSTR imaging provided positive results in more than half of the cases with radioiodine-negative DTC, and about one third of patients were eligible for PRRT. (68)Ga-DOTATOC PET/CT seems a reliable tool both for patient selection and evaluation of treatment response. In our experience, FV determination over time seems to represent a reliable parameter to determine tumor response to PRRT, although further investigations are needed to better define its role.
Collapse
Affiliation(s)
- Annibale Versari
- 1 Nuclear Medicine, Arcispedale Santa Maria Nuova, Clinical Cancer Research Institute (IRCCS) , Reggio Emilia, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
497
|
177Lu-DOTATATE therapy in patients with neuroendocrine tumours: 5 years’ experience from a tertiary cancer care centre in India. Eur J Nucl Med Mol Imaging 2014; 41:1319-26. [DOI: 10.1007/s00259-014-2710-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/20/2014] [Indexed: 01/08/2023]
|
498
|
Herbertson RA, Tebbutt NC, Lee FT, Gill S, Chappell B, Cavicchiolo T, Saunder T, O'Keefe GJ, Poon A, Lee ST, Murphy R, Hopkins W, Scott FE, Scott AM. Targeted chemoradiation in metastatic colorectal cancer: a phase I trial of 131I-huA33 with concurrent capecitabine. J Nucl Med 2014; 55:534-9. [PMID: 24556590 DOI: 10.2967/jnumed.113.132761] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED huA33 is a humanized antibody that targets the A33 antigen, which is highly expressed in intestinal epithelium and more than 95% of human colon cancers but not other normal tissues. Previous studies have shown huA33 can target and be retained in a metastatic tumor for 6 wk but eliminated from normal colonocytes within days. This phase I study used radiolabeled huA33 in combination with capecitabine to target chemoradiation to metastatic colorectal cancer. The primary objective was safety and tolerability of the combination of capecitabine and (131)I-huA33. Pharmacokinetics, biodistribution, immunogenicity, and tumor response were also assessed. METHODS Eligibility included measurable metastatic colorectal cancer, adequate hematologic and biochemical function, and informed consent. An outpatient scout (131)I-huA33 dose was followed by a single-therapy infusion 1 wk later, when capecitabine was commenced. Dose escalation occurred over 5 dose levels. Patients were evaluated weekly, with tumor response assessment at the end of the 12-wk trial. Tumor targeting was assessed using a γ camera and SPECT imaging. RESULTS Nineteen eligible patients were enrolled. The most frequently observed toxicity included myelosuppression, gastrointestinal symptoms, and asymptomatic hyperbilirubinemia. Biodistribution analysis demonstrated excellent tumor targeting of the known tumor sites, expected transient bowel uptake, but no other normal tissue uptake. (131)I-huA33 demonstrated a mean terminal half-life and serum clearance suited to radioimmunotherapy (T1/2β, 100.24 ± 20.92 h, and clearance, 36.72 ± 8.01 mL/h). The mean total tumor dose was 13.8 ± 7.6 Gy (range, 5.1-26.9 Gy). One patient had a partial response, and 10 patients had stable disease. CONCLUSION (131)I-huA33 achieves specific targeting of radiotherapy to colorectal cancer metastases and can be safely combined with chemotherapy, providing an opportunity to deliver chemoradiation specifically to metastatic disease in colorectal cancer patients.
Collapse
|
499
|
Sherman SK, Maxwell JE, Carr JC, Wang D, O'Dorisio MS, O'Dorisio TM, Howe JR. GIPR expression in gastric and duodenal neuroendocrine tumors. J Surg Res 2014; 190:587-93. [PMID: 24565507 DOI: 10.1016/j.jss.2014.01.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/16/2014] [Accepted: 01/24/2014] [Indexed: 01/14/2023]
Abstract
BACKGROUND Compounds targeting somatostatin-receptor-type-2 (SSTR2) are useful for small bowel neuroendocrine tumor (SBNET) and pancreatic neuroendocrine tumor (PNET) imaging and treatment. We recently characterized expression of 13 cell surface receptor genes in SBNETs and PNETs, identifying three drug targets (GIPR, OXTR, and OPRK1). This study set out to characterize expression of this gene panel in the less common neuroendocrine tumors of the stomach and duodenum (gastric and duodenal neuroendocrine tumors [GDNETs]). METHODS Primary tumors and adjacent normal tissue were collected at surgery, RNA was extracted, and expression of 13 target genes was determined by quantitative polymerase chain reaction. Expression was normalized to GAPDH and POLR2A internal control genes. Expression relative to normal tissue (ddCT) and absolute expression (dCT) were calculated. Wilcoxon tests compared median expression with false discovery rate correction for multiple comparisons. RESULTS Gene expression was similar in two gastric and seven duodenal tumors, and these were analyzed together. Like SBNETs (n = 63) and PNETs (n = 51), GDNETs showed significant overexpression compared with normal tissue of BRS3, GIPR, GRM1, GPR113, OPRK1, and SSTR2 (P < 0.05 for all). Of these, SSTR2 had the highest absolute expression in GDNETs (median dCT 4.0). Absolute expression of BRS3, GRM1, GPR113, and OPRK1 was significantly lower than SSTR2 in GDNETs (P < 0.05 for all), whereas expression of GIPR was similar to SSTR2 (median 4.3, P = 0.4). CONCLUSIONS As in SBNETs and PNETs, GIPR shows absolute expression close to SSTR2 but has greater overexpression relative to normal tissue (21.1 versus 3.5-fold overexpression). We conclude that GIPR could provide an improved signal-to-noise ratio for imaging versus SSTR2 and represents a promising novel therapeutic target in GDNETs.
Collapse
Affiliation(s)
- Scott K Sherman
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jessica E Maxwell
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jennifer C Carr
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Donghong Wang
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - M Sue O'Dorisio
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Thomas M O'Dorisio
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - James R Howe
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa.
| |
Collapse
|
500
|
Sherman SK, Carr JC, Wang D, O'Dorisio MS, O'Dorisio TM, Howe JR. Gastric inhibitory polypeptide receptor (GIPR) is a promising target for imaging and therapy in neuroendocrine tumors. Surgery 2014; 154:1206-13; discussion 1214. [PMID: 24238043 DOI: 10.1016/j.surg.2013.04.052] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/25/2013] [Indexed: 01/18/2023]
Abstract
BACKGROUND Ligands binding the somatostatin receptor type 2 (SSTR2) are useful for imaging and treatment of neuroendocrine tumors (NETs), but not all tumors express high levels of these receptors. The aim of this study was to evaluate gene expression of new therapeutic targets in NETs relative to SSTR2. METHODS RNA was extracted from 103 primary small bowel and pancreatic NETs, matched normal tissue, and 123 metastases. Expression of 12 candidate genes was measured by quantitative polymerase chain reaction normalized to internal controls; candidate gene expression was compared with SSTR2. RESULTS Relative to normal tissue, primary NET expression of SSTR2, GPR98, BRS3, GIPR, GRM1, and OPRK1 were increased by 3, 8, 13, 13, 17, and 20-fold, respectively. Similar changes were found in metastases. Although most candidate genes showed lesser absolute expressions than SSTR2, absolute GIPR expression was closest to SSTR2 (mean dCT 3.6 vs. 2.7, P = .01). Absolute OPRK1 and OXTR expression varied greatly by primary tumor type and was close to SSTR2 in small bowel NETs but not pancreatic NETs. CONCLUSION Compared with the current treatment standard SSTR2, GIPR has only somewhat lesser absolute gene expression in tumor tissue but much lesser expression in normal tissue, making it a promising new target for NET imaging and therapy.
Collapse
Affiliation(s)
- Scott K Sherman
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA
| | | | | | | | | | | |
Collapse
|