51
|
The Impact of Radiological Response to Peptide Receptor Radionuclide Therapy on Overall Survival in Patients With Metastatic Midgut Neuroendocrine Tumors. Clin Nucl Med 2017; 42:e135-e141. [PMID: 27922860 DOI: 10.1097/rlu.0000000000001457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
52
|
Sandblom V, Ståhl I, Olofsson Bagge R, Forssell-Aronsson E. Evaluation of two intraoperative gamma detectors for assessment of 177Lu activity concentration in vivo. EJNMMI Phys 2017; 4:3. [PMID: 28070731 PMCID: PMC5222763 DOI: 10.1186/s40658-016-0168-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 12/06/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Patients with somatostatin receptor-expressing neuroendocrine tumours can be treated with intravenously administered 177Lu-octreotate. Few patients are cured with the present protocol due to the current dose limitation of normal organs at risk, such as the kidneys. By locally administering 177Lu-octreotate to the liver for the purpose of treating liver metastases, a substantially reduced absorbed dose to organs at risk could be achieved. The development of such a technique requires the capability of measuring the 177Lu activity concentration in tissues in vivo. The aim of this study was to evaluate different performance parameters of two commercially available intraoperative gamma detectors in order to investigate whether intraoperative gamma detector measurements could be used to determine 177Lu activity concentration in vivo. RESULTS Measurements were made using different sources containing 177Lu. Response linearity, sensitivity, spatial resolution and its depth dependence, organ thickness dependence of the measured count rate and tumour detectability were assessed for two intraoperative gamma detectors. The two detectors (a scintillation and a semiconductor detector) showed differences in technical performance. For example, the sensitivity was higher for the scintillation detector, while the spatial resolution was better for the semiconductor detector. Regarding organ thickness dependence and tumour detectability, similar results were obtained for both detectors, and even relatively small simulated tumours of low tumour-to-background activity concentration ratios could be detected. CONCLUSIONS Acceptable results were obtained for both detectors, although the semiconductor detector proved more advantageous for our purpose. The measurements demonstrated factors that must be corrected for, such as organ thickness or dead-time effects. Altogether, intraoperative gamma detector measurements could be used to determine 177Lu activity concentration in vivo.
Collapse
Affiliation(s)
- Viktor Sandblom
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, SE-413 45, Gothenburg, Sweden.
| | - Ingun Ståhl
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, SE-413 45, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, SE-413 45, Gothenburg, Sweden
| | - Eva Forssell-Aronsson
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, SE-413 45, Gothenburg, Sweden
| |
Collapse
|
53
|
Luna Pais H, Alho I, Vendrell I, Mansinho A, Costa L. Radionuclides in oncology clinical practice – review of the literature. Dalton Trans 2017; 46:14475-14487. [DOI: 10.1039/c7dt01929g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Radionuclide therapy is a promising type of targeted therapy for cancer and its use is becoming more common in several types of malignant tumors.
Collapse
Affiliation(s)
- Helena Luna Pais
- Medical Oncology Department
- Hospital de Santa Maria
- 1649-035 Lisbon
- Portugal
| | - Irina Alho
- Instituto de Medicina Molecular
- Faculdade de Medicina
- Universidade de Lisboa
- 1649-035 Lisbon
- Portugal
| | - Inês Vendrell
- Medical Oncology Department
- Hospital de Santa Maria
- 1649-035 Lisbon
- Portugal
| | - André Mansinho
- Medical Oncology Department
- Hospital de Santa Maria
- 1649-035 Lisbon
- Portugal
| | - Luís Costa
- Medical Oncology Department
- Hospital de Santa Maria
- 1649-035 Lisbon
- Portugal
- Instituto de Medicina Molecular
| |
Collapse
|
54
|
Kaifi JT, Kayser G, Ruf J, Passlick B. The Diagnosis and Treatment of Bronchopulmonary Carcinoid. DEUTSCHES ARZTEBLATT INTERNATIONAL 2016. [PMID: 26214234 DOI: 10.3238/arztebl.2015.0479] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The spectrum of primary neuroendocrine tumors of the lungs ranges from typical carcinoid tumors, which are relatively benign, to highly aggressive small-cell carcinoma. In this review, we summarize the treatment of bronchopulmonary carcinoid, a disease with an incidence of 0.5 per 100,000 persons per year in Western countries. METHOD We selectively searched the PubMed database for scientific evidence on the treatment of bronchopulmonary carcinoid, considering only articles published up to February 2015. We also performed a survival analysis of 84 patients with this disease who underwent interdisciplinary treatment at the University of Freiburg Medical Center. RESULTS Carcinoid tumors account for less than 1% of all lung tumors. They manifest themselves clinically with cough (35%), hemoptysis (25%), and/or bronchial obstruction (40%), depending on their location, size, and pattern of growth. 30% of patients are asymptomatic, and less than 1% have hormone-associated symptoms. Typical and atypical carcinoid tumors are distinguished on a histological basis; the histologic differential diagnosis also includes large-cell neuroendocrine tumors and small-cell carcinoma of the lung. 80% of patients who undergo resection of typical carcinoid tumors survive at least 10 years. Atypical carcinoid tumors recur more commonly than typical ones. If the mediastinal lymph nodes are involved, adjuvant treatment should be considered. CONCLUSION Because of their rarity, the treatment of bronchopulmonary carcinoid tumors presents an interdisciplinary challenge. Surgical resection, the treatment of choice for local carcinoid tumors, generally leads to long-term survival. The existing registers should be made more comprehensive so that the treatment of this disease can be better in the future.
Collapse
|
55
|
Björklund P, Pacak K, Crona J. Precision medicine in pheochromocytoma and paraganglioma: current and future concepts. J Intern Med 2016; 280:559-573. [PMID: 27165774 PMCID: PMC7441825 DOI: 10.1111/joim.12507] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pheochromocytoma and paraganglioma (PPGL) are rare diseases but are also amongst the most characterized tumour types. Hence, patients with PPGL have greatly benefited from precision medicine for more than two decades. According to current molecular biology and genetics-based taxonomy, PPGL can be divided into three different clusters characterized by: Krebs cycle reprogramming with oncometabolite accumulation or depletion (group 1a); activation of the (pseudo)hypoxia signalling pathway with increased tumour cell proliferation, invasiveness and migration (group 1b); and aberrant kinase signalling causing a pro-mitogenic and anti-apoptotic state (group 2). Categorization into these clusters is highly dependent on mutation subtypes. At least 12 different syndromes with distinct genetic causes, phenotypes and outcomes have been described. Genetic screening tests have a documented benefit, as different PPGL syndromes require specific approaches for optimal diagnosis and localization of various syndrome-related tumours. Genotype-tailored treatment options, follow-up and preventive care are being investigated. Future new developments in precision medicine for PPGL will mainly focus on further identification of driver mechanisms behind both disease initiation and malignant progression. Identification of novel druggable targets and prospective validation of treatment options are eagerly awaited. To achieve these goals, we predict that collaborative large-scale studies will be needed: Pheochromocytoma may provide an example for developing precision medicine in orphan diseases that could ultimately aid in similar efforts for other rare conditions.
Collapse
Affiliation(s)
- P Björklund
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - K Pacak
- Section on Medical Neuroendocrinology, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - J Crona
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
56
|
Embolotherapy for Neuroendocrine Tumor Liver Metastases: Prognostic Factors for Hepatic Progression-Free Survival and Overall Survival. Cardiovasc Intervent Radiol 2016; 40:69-80. [PMID: 27738818 DOI: 10.1007/s00270-016-1478-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/27/2016] [Indexed: 12/27/2022]
Abstract
PURPOSE The purpose of the study was to evaluate prognostic factors for survival outcomes following embolotherapy for neuroendocrine tumor (NET) liver metastases. MATERIALS AND METHODS This was a multicenter retrospective study of 155 patients (60 years mean age, 57 % male) with NET liver metastases from pancreas (n = 71), gut (n = 68), lung (n = 8), or other/unknown (n = 8) primary sites treated with conventional transarterial chemoembolization (TACE, n = 50), transarterial radioembolization (TARE, n = 64), or transarterial embolization (TAE, n = 41) between 2004 and 2015. Patient-, tumor-, and treatment-related factors were evaluated for prognostic effect on hepatic progression-free survival (HPFS) and overall survival (OS) using unadjusted and propensity score-weighted univariate and multivariate Cox proportional hazards models. RESULTS Median HPFS and OS were 18.5 and 125.1 months for G1 (n = 75), 12.2 and 33.9 months for G2 (n = 60), and 4.9 and 9.3 months for G3 tumors (n = 20), respectively (p < 0.05). Tumor burden >50 % hepatic volume demonstrated 5.5- and 26.8-month shorter median HPFS and OS, respectively, versus burden ≤50 % (p < 0.05). There were no significant differences in HPFS or OS between gut or pancreas primaries. In multivariate HPFS analysis, there were no significant differences among embolotherapy modalities. In multivariate OS analysis, TARE had a higher hazard ratio than TACE (unadjusted Cox model: HR 2.1, p = 0.02; propensity score adjusted model: HR 1.8, p = 0.11), while TAE did not differ significantly from TACE. CONCLUSION Higher tumor grade and tumor burden prognosticated shorter HPFS and OS. TARE had a higher hazard ratio for OS than TACE. There were no significant differences in HPFS among embolotherapy modalities.
Collapse
|
57
|
Stoelben E, Yordanova A, Gossmann A, Hammer-Hellmig M, Brossart P, Mayer K, Essler M. Karzinoide der Lunge. BEST PRACTICE ONKOLOGIE 2016; 11:22-28. [DOI: 10.1007/s11654-016-0586-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
58
|
Abstract
Neuroendocrine tumors are increasingly diagnosed, either incidentally as part of screening processes, or for symptoms, which have commonly been mistaken for other disorders initially. The diagnostic workup to characterize tumor behaviour and prognosis focuses on histologic, anatomic, and functional imaging assessments. Several therapeutic options exist for patients ranging from curative and debulking surgery through to liver-directed therapies and systemic treatments. Multimodal therapies are often required over the patient's disease history. The management paradigm can be complex but should be focused on curative resections and then on controlling symptoms and limiting disease progression. There are several new systemic therapies that have completed phase 3 studies with new compounds being studied in phase 2. Genetic and epigenetic markers may lead to a new era of personalised therapy in the future.
Collapse
Affiliation(s)
- Ron Basuroy
- Neuroendocrine Tumour Unit, Institute of Liver Studies, Kings College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Raj Srirajaskanthan
- Neuroendocrine Tumour Unit, Institute of Liver Studies, Kings College Hospital, Denmark Hill, London SE5 9RS, UK
| | - John K Ramage
- Neuroendocrine Tumour Unit, Institute of Liver Studies, Kings College Hospital, Denmark Hill, London SE5 9RS, UK.
| |
Collapse
|
59
|
177Lu-DKFZ-PSMA-617 therapy in metastatic castration resistant prostate cancer: safety, efficacy, and quality of life assessment. Eur J Nucl Med Mol Imaging 2016; 44:81-91. [PMID: 27506431 DOI: 10.1007/s00259-016-3481-7] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/31/2016] [Indexed: 02/07/2023]
Abstract
PURPOSE The purpose of this study was to evaluate the efficacy and safety of a novel theranostic agent, 177Lu-DKFZ-PSMA-617 therapy in metastatic castration resistant prostate cancer (mCRPC). METHODS Thirty-one mCRPC patients with progressive disease despite second-line hormonal therapy and/or docetaxel chemotherapy were recruited for the study. All patients underwent diagnostic68Ga-PSMA-HBED-CCPET/CT, prior to inclusion for therapy. Included patients then underwent quarterly 177Lu-DKFZ-PSMA-617 therapy. Hematological, kidney function, liver function tests, and serum PSA levels were recorded before and after therapy at 2 weeks, 4 weeks, and 3 month intervals. Biochemical response was assessed with trend in serum PSA levels. Metabolic response was assessed by PERCIST 1 criteria. Clinical response was assessed by visual analogue score (VASmax) analgesic score (AS), Karanofsky performance status (KPS), and ttoxicity and response criteria of the Eastern Cooperative Oncology Group (ECOG) criteria. RESULTS The mean age of patients was 65.93 ± 9.77 years (range: 38-81 years). The mean activity administered in the 31 patients was 5069 ± 1845 MBq ranging from one to four cycles. There was a decline in the mean serum PSA levels from the baseline (baseline: 275 ng/mL, post 1st cycle therapy: 141.75 ng/mL). Based on biochemical response criteria 2/31, 20/31, 3/31, and 6/31 had complete response (CR), partial response(PR), stable disease (SD), and progressive disease (PD), respectively. Metabolic response revealed 2/6 patients with CR, and the remaining 3/6 patients with PR and 1/6 patients with SD. The mean VASmax score decreased from 7.5 to 3. The mean analgesic score decreased from 2.5 to 1.8 after therapy. The mean KPS score improved from 50.32 to 65.42 after therapies. The mean ECOG performance status improved from 2.54 to 1.78 after therapy. Two patients experienced grade I and grade II hemoglobin toxicity each. None of the patients experienced nephrotoxicity or hepatotoxicity. CONCLUSION 177Lu-DKFZ-PSMA-617 radionuclide therapy is a safe and effective approach in the treatment of mCRPC patients.
Collapse
|
60
|
Rogowski W, Wachuła E, Lewczuk A, Buscombe JR, Seklecka N, Sankowski A, Ćwikła JB. Long-term efficacy of (90)Y-DOTATATE in patients with nonresectable pancreatic and small bowel neuroendocrine neoplasms. Future Oncol 2016; 12:1877-85. [PMID: 27156864 DOI: 10.2217/fon-2016-0031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
AIM To determine the efficacy of (90)Y [DOTA(0), D-Phe(1), Tyr(3)]-octreotate (DOTATATE) in 67 patients with pancreatic and small bowel neuroendocrine tumors (NETs). PATIENTS & METHODS The primary efficacy end point was overall survival (OS) and secondary end points were progression-free survival (PFS) and tumor response. RESULTS Median PFS in pancreatic and small bowel NETs was 25 and 28 months, respectively, and median OS was 42 and 38.5 months, respectively. No intergroup differences in median OS (p = 0.945) or PFS (p = 0.174) were found, also after adjustment for tumor origin, secretory status and grade, and patient's gender. CONCLUSION (90)Y-DOTATATE may have similar efficacy in pancreatic and small bowel NETs. Better WHO performance status at baseline seems to be associated with more favorable outcomes.
Collapse
Affiliation(s)
- Wojciech Rogowski
- Clinical Department of Chemotherapy, Hospital Ministry of the Interior & Administration & Warmia & Mazury Oncology Centre, Olsztyn, Poland
| | - Ewa Wachuła
- Clinical Department of Chemotherapy, Hospital Ministry of the Interior & Administration & Warmia & Mazury Oncology Centre, Olsztyn, Poland
| | - Anna Lewczuk
- Department of Endocrinology, Medical University of Gdansk, Gdansk, Poland
| | - John R Buscombe
- Department of Nuclear Medicine & PET, Addenbrooke's Hospital, Cambridge, UK
| | - Nina Seklecka
- Department of Radiology & Diagnostic Imaging, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Artur Sankowski
- Department of Radiology & Diagnostic Imaging, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Jarosław B Ćwikła
- Faculty of Medical Science, University of Varmia & Masuria, Olsztyn, Poland
| |
Collapse
|
61
|
Martin RF, Feinendegen LE. The quest to exploit the Auger effect in cancer radiotherapy - a reflective review. Int J Radiat Biol 2016; 92:617-632. [PMID: 26926313 DOI: 10.3109/09553002.2015.1136854] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
To identify the emergence of the recognition of the potential of the Auger effect for clinical application, and after tracing the salient milestones towards that goal, to evaluate the status quo and future prospects. It was not until 40 years after the discovery of Auger electrons, that the availability of radioactive DNA precursors enabled the biological power, and the clinical potential, of the Auger effect to be appreciated. Important milestones on the path to clinical translation have been identified and reached, but hurdles remain. Nevertheless the potential is still evident, and there is reasonable optimism that the goal of clinical translation is achievable.
Collapse
Affiliation(s)
- Roger F Martin
- a Molecular Radiation Biology Laboratory , Peter MacCallum Cancer Centre.,b The Sir Peter MacCallum Department of Oncology , The University of Melbourne.,c School of Chemistry and Bio-21 Molecular Science and Biotechnology Institute , University of Melbourne , Melbourne , Australia
| | - Ludwig E Feinendegen
- d Heinrich-Heine-University Düsseldorf , Germany.,e Brookhaven National Laboratory , Upton , NY , USA
| |
Collapse
|
62
|
Pusceddu S, Lo Russo G, Macerelli M, Proto C, Vitali M, Signorelli D, Ganzinelli M, Scanagatta P, Duranti L, Trama A, Buzzoni R, Pelosi G, Pastorino U, de Braud F, Garassino MC. Diagnosis and management of typical and atypical lung carcinoids. Crit Rev Oncol Hematol 2016; 100:167-76. [PMID: 26917456 DOI: 10.1016/j.critrevonc.2016.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 02/15/2016] [Indexed: 01/08/2023] Open
Abstract
An estimated 20% to 30% of all neuroendocrine tumours originate in the bronchial tree and lungs. According to the 2015 World Health Organization categorization, these tumours are separated into four subtypes characterized by increasing biological aggressiveness: typical carcinoid, atypical carcinoid, large-cell neuroendocrine carcinoma and small-cell carcinoma. Although typical and atypical lung carcinoids account for less than 1-5% of all pulmonary malignancies, the incidence of these neoplasms has risen significantly in recent decades. Surgery is the treatment of choice for loco-regional disease but for advanced lung carcinoids there is no recognized standard of care and successful management requires a multidisciplinary approach. The aim of this review is to provide a useful guide for the clinical management of lung carcinoids.
Collapse
Affiliation(s)
- Sara Pusceddu
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Giuseppe Lo Russo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Marianna Macerelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Claudia Proto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Milena Vitali
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Diego Signorelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Monica Ganzinelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Paolo Scanagatta
- Division of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Leonardo Duranti
- Division of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Annalisa Trama
- Department of Preventive and Predictive Medicine, Fondazione IRCSS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Roberto Buzzoni
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Giuseppe Pelosi
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Ugo Pastorino
- Division of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Marina Chiara Garassino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy.
| |
Collapse
|
63
|
Song KB, Kim SC, Kim JH, Seo DW, Hong SM, Park KM, Hwang DW, Lee JH, Lee YJ. Prognostic Value of Somatostatin Receptor Subtypes in Pancreatic Neuroendocrine Tumors. Pancreas 2016; 45:187-192. [PMID: 26474434 DOI: 10.1097/mpa.0000000000000493] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Studies on the expression of somatostatin receptor (SSTR) subtypes in pancreatic neuroendocrine tumors (PNETs) are rare. The aim of this study was to determine the expression of the SSTR subtypes via immunohistochemistry analyses and assess the correlation between SSTR subtype expression and prognosis. METHODS We examined 199 patients with PNET who underwent surgical resection between January 1995 and December 2010 at the Asan Medical Center. For all cases, medical records, including demographic data, clinical symptoms, radiological findings, postoperative treatment outcomes, and expression of SSTR subtypes, were carefully reviewed. RESULTS In total, 162 (81.4%) PNETs expressed more than 1 SSTR subtype. Functioning PNET expressed significantly more SSTR subtypes, compared to nonfunctioning PNET. The SSTR2(+) and SSTR5(+) groups had better prognosis than the SSTR2(-) (P = 0.009) and SSTR5(-) groups (P = 0.03), respectively. In the grade 2 PNET of 2010 World Health Organization classification, the SSTR(+) group had better prognosis than SSTR(-) group. CONCLUSIONS The expression of SSTR 2 and 5 were related with good prognosis of PNET. In World Health Organization grade 2 PNET, the SSTR(+) group had better prognosis than SSTR(-) group. The SSTR expression(+) by immunohistochemistry might be related with good prognosis of the patients with surgically resected PNET.
Collapse
Affiliation(s)
- Ki Byung Song
- From the *Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, †Department of Pathology, and ‡Division of Gastroenterology, Department of Internal Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Li Y, Ma L, Gaddam V, Gallazzi F, Hennkens HM, Harmata M, Lewis MR, Deakyne CA, Jurisson SS. Synthesis, Characterization, and In Vitro Evaluation of New (99m)Tc/Re(V)-Cyclized Octreotide Analogues: An Experimental and Computational Approach. Inorg Chem 2016; 55:1124-33. [PMID: 26789775 DOI: 10.1021/acs.inorgchem.5b02306] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Radiolabeled proteolytic degradation-resistant somatostatin analogues have been of long-standing interest as cancer imaging and radiotherapy agents for targeting somatostatin receptor-positive tumors. Our interest in developing (186)Re- and (188)Re-based therapeutic radiopharmaceuticals led to investigation of a new Re(V)-cyclized octreotide analogue, Re(V)-cyclized, thiolated-DPhe(1)-Cys(2)-Tyr(3)-DTrp(4)-Lys(5)-Thr(6)-Cys(7)-Thr(OH)(8) (Re-SDPhe-TATE) using both experimental and quantum chemical methods. The metal is directly coordinated to SDPhe-TATE through cyclization of the peptide around the [ReO](3+) core. Upon complexation, four isomers were observed; the isolated/semi-isolated isomers exhibited different somatostatin receptor (sstr) binding affinities, 0.13 to 1.5 μM, in rat pancreatic tumor cells. Two-dimensional NMR experiments and electronic structure calculations were employed to elucidate the structural differences among the different isomers. According to NMR studies, the metal is coordinated to three thiolates and the backbone amide of Cys(2) in isomers 1 and 4, whereas the metal is coordinated to three thiolates and the backbone amide of Tyr(3) in isomer 2. Quantum chemical methods clarified the stereochemistry of Re-SDPhe-TATE and the possible peptide arrangements around the [ReO](3+) core. The re-cyclization reaction was translated to the (99m)Tc radiotracer level with four isomers observed on complexation with comparable HPLC retention times as the Re-SDPhe-TATE isomers. About 85% total (99m)Tc labeling yield was achieved by ligand exchange from (99m)Tc-glucoheptonate at 60 °C for an hour. About 100% and 51% of (99m)Tc(V)-cyclized SDPhe-TATE remained intact in phosphate buffered saline and 1 mM cysteine solution under physiological conditions at 6 h, respectively.
Collapse
Affiliation(s)
| | - Lixin Ma
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65201, United States
| | | | | | - Heather M Hennkens
- University of Missouri Research Reactor Center , Columbia, Missouri 65211, United States
| | | | - Michael R Lewis
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65201, United States
| | | | | |
Collapse
|
65
|
Abstract
Targeted therapy is gaining prominence in the management of different cancers. Given different mechanism of action compared with traditional chemoradiotherapy, selection of patients for targeted therapy and monitoring response to these agents is difficult with conventional imaging. Various new PET radiopharmaceuticals have been evaluated for molecular imaging of these targets to achieve specific patient selection and response monitoring. These PET/computed tomography (CT) agents target the cell surface receptors, hormone receptors, receptor tyrosine kinases, or angiogenesis components. This article reviews the established and potential role of PET/CT with new radiopharmaceuticals for guiding targeted therapy.
Collapse
Affiliation(s)
- Punit Sharma
- Department of Nuclear Medicine and PET/CT, Eastern Diagnostics India Ltd, 13C Mirza Ghalib Street, Kolkata 700016, India; Diagnostic Nuclear Medicine Division, Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Rakesh Kumar
- Diagnostic Nuclear Medicine Division, Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| | - Abass Alavi
- Division of Nuclear Medicine, Department of Radiology, University of Pennsylvania School of Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
66
|
A systematic evaluation of the potential of PCTA-NCS ligand as a bifunctional chelating agent for design of 177Lu radiopharmaceuticals. J Radioanal Nucl Chem 2015. [DOI: 10.1007/s10967-015-4281-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
67
|
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: 36] [Impact Index Per Article: 3.6] [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
|
68
|
Beaino W, Nedrow JR, Anderson CJ. Evaluation of (68)Ga- and (177)Lu-DOTA-PEG4-LLP2A for VLA-4-Targeted PET Imaging and Treatment of Metastatic Melanoma. Mol Pharm 2015; 12:1929-38. [PMID: 25919487 PMCID: PMC5167571 DOI: 10.1021/mp5006917] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Malignant melanoma is a highly aggressive cancer, and the incidence of this disease is increasing worldwide at an alarming rate. Despite advances in the treatment of melanoma, patients with metastatic disease still have a poor prognosis and low survival rate. New strategies, including targeted radiotherapy, would provide options for patients who become resistant to therapies such as BRAF inhibitors. Very late antigen-4 (VLA-4) is expressed on melanoma tumor cells in higher levels in more aggressive and metastatic disease and may provide an ideal target for drug delivery and targeted radiotherapy. In this study, we evaluated (177)Lu- and (68)Ga-labeled DOTA-PEG4-LLP2A as a VLA-4-targeted radiotherapeutic with a companion PET agent for diagnosis and monitoring metastatic melanoma treatment. DOTA-PEG4-LLP2A was synthesized by solid-phase synthesis. The affinity of (177)Lu- and (68)Ga-labeled DOTA-PEG4-LLP2A to VLA-4 was determined in B16F10 melanoma cells by saturation binding and competitive binding assays, respectively. Biodistribution of the LLP2A conjugates was determined in C57BL/6 mice bearing B16F10 subcutaneous tumors, while PET/CT imaging was performed in subcutaneous and metastatic models. (177)Lu-DOTA-PEG4-LLP2A showed high affinity to VLA-4 with a Kd of 4.1 ± 1.5 nM and demonstrated significant accumulation in the B16F10 melanoma tumor after 4 h (31.5 ± 7.8%ID/g). The tumor/blood ratio of (177)Lu-DOTA-PEG4-LLP2A was highest at 24 h (185 ± 26). PET imaging of metastatic melanoma with (68)Ga-DOTA-PEG4-LLP2A showed high uptake in sites of metastases and correlated with bioluminescence imaging of the tumors. These data demonstrate that (177)Lu-DOTA-PEG4-LLP2A has potential as a targeted therapeutic for treating melanoma as well as other VLA-4-expressing tumors. In addition, (68)Ga-DOTA-PEG4-LLP2A is a readily translatable companion PET tracer for imaging of metastatic melanoma.
Collapse
Affiliation(s)
- Wissam Beaino
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| | - Jessie R. Nedrow
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| | - Carolyn J. Anderson
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| |
Collapse
|
69
|
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
|
70
|
Tan TH, Hussein Z, Saad FFA, Shuaib IL. Diagnostic Performance of (68)Ga-DOTATATE PET/CT, (18)F-FDG PET/CT and (131)I-MIBG Scintigraphy in Mapping Metastatic Pheochromocytoma and Paraganglioma. Nucl Med Mol Imaging 2015; 49:143-51. [PMID: 26085860 DOI: 10.1007/s13139-015-0331-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 02/27/2015] [Accepted: 03/02/2015] [Indexed: 12/15/2022] Open
Abstract
PURPOSE To evaluate the diagnostic performance of (68)Ga-DOTATATE (18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography (PET)/computed tomography (CT), (18)F-FDG PET/CT and (131)I-MIBG scintigraphy in the mapping of metastatic pheochromocytoma and paraganglioma. MATERIALS AND METHODS Seventeen patients (male = 8, female = 9; age range, 13-68 years) with clinically proven or suspicious metastatic pheochromocytoma or paraganglioma were included in this prospective study. Twelve patients underwent all three modalities, whereas five patients underwent (68)Ga-DOTATATE and (131)I-MIBG without (18)F-FDG. A composite reference standard derived from anatomical and functional imaging findings, along with histopathological information, was used to validate the findings. Results were analysed on a per-patient and on per-lesion basis. Sensitivity and accuracy were assessed using McNemar's test. RESULTS On a per-patient basis, 14/17 patients were detected in (68)Ga-DOTATATE, 7/17 patients in (131)I-MIBG, and 10/12 patients in (18)F-FDG. The sensitivity and accuracy of (68)Ga-DOTATATE, (131)I-MIBG and (18)F-FDG were (93.3 %, 94.1 %), (46.7 %, 52.9 %) and (90.9 %, 91.7 %) respectively. On a per-lesion basis, an overall of 472 positive lesions were detected; of which 432/472 were identified by (68)Ga-DOTATATE, 74/472 by (131)I-MIBG, and 154/300 (patient, n = 12) by (18)F-FDG. The sensitivity and accuracy of (68)Ga-DOTATATE, (131)I-MIBG and (18)F-FDG were (91.5 %, 92.6 % p < 0.0001), (15.7 %, 26.0 % p < 0.0001) and (51.3 %, 57.8 % p < 0.0001) respectively. Discordant lesions were demonstrated on (68)Ga-DOTATATE, (131)I-MIBG and (18)F-FDG. CONCLUSIONS Ga-DOTATATE PET/CT shows high diagnostic accuracy than (131)I-MIBG scintigraphy and (18)F-FDG PET/ CT in mapping metastatic pheochromocytoma and paraganglioma.
Collapse
Affiliation(s)
- Teik Hin Tan
- Nuclear Medicine Department, National Cancer Institute, No 4, Jalan P7, Presint 7, 62550 Putrajaya, Malaysia
| | - Zanariah Hussein
- Department of Endocrine, Hospital Putrajaya, Presint 7, 62250 Putrajaya, Wilayah Persekutuan Malaysia
| | | | - Ibrahim Lutfi Shuaib
- Department of Radiology, Advanced Medical and Dental Institute, University Sains Malaysia, Bertam, 13200 Kepala Batas, Pulau Pinang Malaysia
| |
Collapse
|
71
|
Schüler E, Österlund A, Forssell-Aronsson E. The amount of injected 177Lu-octreotate strongly influences biodistribution and dosimetry in C57BL/6N mice. Acta Oncol 2015; 55:68-76. [PMID: 25813472 DOI: 10.3109/0284186x.2015.1027001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND (177)Lu-octreotate therapy has proven to give favorable results after treatment of patients with neuroendocrine tumors. Much focus has been on the binding and uptake of (177)Lu-octreotate in tumor tissue, but biodistribution properties in normal tissues is still not fully understood, and the effect of receptor saturation may be important. The aim of this study was to investigate the influence of the amount of (177)Lu-octreotate on the biodistribution of (177)Lu-octreotate in normal tissues in mice. MATERIAL AND METHODS C57BL/6N female mice were intravenously injected with 0.1-150 MBq (177)Lu-octreotate (0.039 μg peptide/MBq). The mice were killed 0.25 h to 14 days after injection by cardiac puncture under anesthesia. Activity concentration was determined in blood, bone marrow, kidneys, liver, lungs, pancreas, and spleen, and mean absorbed doses were calculated. RESULTS The activity concentration varied with time and amount of injected activity. At 4-8 h after injection, a local maximum in activity concentration was found for liver, lungs, pancreas, and spleen. With the exception for the lower injected activities (0.1-1 MBq), the overall highest uptake was found in the kidneys (%IA/g). Large variations were found and the activity concentration in kidneys was 11-23%IA/g at 4 h, and 0.22-1.9%IA/g at 7 days after injection. Furthermore, a clear reduction in activity concentration with increased injected activity was observed for lungs, pancreas and spleen. CONCLUSION The activity concentration in all tissues investigated was strongly influenced by the amount of (177)Lu-octreotate injected. Large differences in mean absorbed dose per unit injected activity were found between low (0.1-1 MBq, 0.0039-0.039 μg) and moderate amounts (5-45 MBq, 0.2-1.8 μg). Furthermore, the results clearly showed the need for better ways to estimate absorbed dose to bone marrow other than methods based on a single blood sample analysis. Since the absorbed dose to critical organs will limit the amount of (177)Lu-octreotate administered, these findings must be taken into consideration when optimizing this type of therapy.
Collapse
Affiliation(s)
- Emil Schüler
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Andreas Österlund
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Eva Forssell-Aronsson
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
72
|
Wolin EM. Treatment options for advanced pancreatic neuroendocrine tumors: what is on the horizon? INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2015. [DOI: 10.2217/ije.14.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract: Treatment options for patients with advanced or metastatic pancreatic tumors (pNET) have expanded greatly in recent years. These treatments include resection, ablation or embolization of liver metastases and systemic therapy with everolimus or sunitinib and cytotoxic agents. New investigational approaches include the use of inhibitors of multiple downstream effectors in the PI3K/Akt/mTOR pathway, novel antiangiogenics, somatostatin analogues, new tyrosine kinase inhibitors and peptide receptor radionuclide therapy. The treatment horizon for pNET patients may offer improved duration of tumor control and survival and more effective symptom control.
Collapse
|
73
|
Dash A, Chakraborty S, Pillai MRA, Knapp FFR. Peptide receptor radionuclide therapy: an overview. Cancer Biother Radiopharm 2015; 30:47-71. [PMID: 25710506 DOI: 10.1089/cbr.2014.1741] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is a site-directed targeted therapeutic strategy that specifically uses radiolabeled peptides as biological targeting vectors designed to deliver cytotoxic levels of radiation dose to cancer cells, which overexpress specific receptors. Interest in PRRT has steadily grown because of the advantages of targeting cellular receptors in vivo with high sensitivity as well as specificity and treatment at the molecular level. Recent advances in molecular biology have not only stimulated advances in PRRT in a sustainable manner but have also pushed the field significantly forward to several unexplored possibilities. Recent decades have witnessed unprecedented endeavors for developing radiolabeled receptor-binding somatostatin analogs for the treatment of neuroendocrine tumors, which have played an important role in the evolution of PRRT and paved the way for the development of other receptor-targeting peptides. Several peptides targeting a variety of receptors have been identified, demonstrating their potential to catalyze breakthroughs in PRRT. In this review, the authors discuss several of these peptides and their analogs with regard to their applications and potential in radionuclide therapy. The advancement in the availability of combinatorial peptide libraries for peptide designing and screening provides the capability of regulating immunogenicity and chemical manipulability. Moreover, the availability of a wide range of bifunctional chelating agents opens up the scope of convenient radiolabeling. For these reasons, it would be possible to envision a future where the scope of PRRT can be tailored for patient-specific application. While PRRT lies at the interface between many disciplines, this technology is inextricably linked to the availability of the therapeutic radionuclides of required quality and activity levels and hence their production is also reviewed.
Collapse
Affiliation(s)
- Ashutosh Dash
- 1 Isotope Production and Applications Division, Bhabha Atomic Research Centre , Mumbai, India
| | | | | | | |
Collapse
|
74
|
Kratochwil C, Stefanova M, Mavriopoulou E, Holland-Letz T, Dimitrakopoulou-Strauss A, Afshar-Oromieh A, Mier W, Haberkorn U, Giesel FL. SUV of [68Ga]DOTATOC-PET/CT Predicts Response Probability of PRRT in Neuroendocrine Tumors. Mol Imaging Biol 2014; 17:313-8. [DOI: 10.1007/s11307-014-0795-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
75
|
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
|
76
|
Ashton CE, Jeans SP, Valle JW. Combination therapy of 177Lu and 90Y Dotatate for treatment of neuroendocrine cancer. Nucl Med Commun 2014; 35:585-7. [DOI: 10.1097/mnm.0000000000000094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
77
|
Delpassand ES, Samarghandi A, Zamanian S, Wolin EM, Hamiditabar M, Espenan GD, Erion JL, O'Dorisio TM, Kvols LK, Simon J, Wolfangel R, Camp A, Krenning EP, Mojtahedi A. Peptide receptor radionuclide therapy with 177Lu-DOTATATE for patients with somatostatin receptor-expressing neuroendocrine tumors: the first US phase 2 experience. Pancreas 2014; 43:518-25. [PMID: 24632546 DOI: 10.1097/mpa.0000000000000113] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Peptide receptor radionuclide therapy with radiolabeled somatostatin analogs is a novel method of treatment in patients with metastatic neuroendocrine tumors (NETs). For the first time in the United States, we present preliminary results of the treatment with Lutetium (177)(Lu) DOTATATE in patients with progressive NETs. METHODS Thirty-seven patients with grade 1 and grade 2 disseminated and progressive gastroenteropancreatic NET were enrolled in a nonrandomized, phase 2 clinical trial. Repeated cycles of 200 mCi (7.4 GBq; ±10%) were administered up to the cumulative dose of 800 mCi (29.6 GBq; ±10%). RESULTS Among 32 evaluable patients, partial response and minimal response to treatment were seen in 28% and 3%, respectively, and stable disease was seen in 41% of patients. A total of 28% had progressive disease. A response to treatment was significantly associated with lower burden of disease in the liver. No significant acute or delayed hematologic or kidney toxicity was observed. An impressive improvement of performance status and quality of life were seen after Lu-DOTATATE therapy. CONCLUSIONS Treatment with multiple cycles of (177)Lu-DOTATATE peptide receptor radionuclide therapy is well tolerated. This treatment results in control of the disease in most patients, whereas systemic toxicities are limited and reversible. Quality of life is also improved.
Collapse
Affiliation(s)
- Ebrahim S Delpassand
- From the *Excel Diagnostics and Nuclear Oncology Center, Houston, TX; †RadioMedix, Inc, Houston, TX; ‡Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA; §Physics Services Inc, Metairie, LA; ∥BioSynthema Inc, St Louis, MO; ¶Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Health Care, Iowa City, IA; #Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL; **IsoTherapeutics Group, Angleton, TX; ††Certus International, Inc, St Louis, MO; ‡‡Iso-Tex Diagnostics, Inc, Friendswood, TX; and §§Department of Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Synthesis, Radiolabelling and In Vitro Characterization of the Gallium-68-, Yttrium-90- and Lutetium-177-Labelled PSMA Ligand, CHX-A''-DTPA-DUPA-Pep. Pharmaceuticals (Basel) 2014; 7:517-29. [PMID: 24787458 PMCID: PMC4035767 DOI: 10.3390/ph7050517] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 03/27/2014] [Accepted: 04/14/2014] [Indexed: 01/19/2023] Open
Abstract
Since prostate-specific membrane antigen (PSMA) has been identified as a diagnostic target for prostate cancer, many urea-based small PSMA-targeting molecules were developed. First, the clinical application of these Ga-68 labelled compounds in positron emission tomography (PET) showed their diagnostic potential. Besides, the therapy of prostate cancer is a demanding field, and the use of radiometals with PSMA bearing ligands is a valid approach. In this work, we describe the synthesis of a new PSMA ligand, CHX-A''-DTPA-DUPA-Pep, the subsequent labelling with Ga-68, Lu-177 and Y-90 and the first in vitro characterization. In cell investigations with PSMA-positive LNCaP C4-2 cells, KD values of ≤14.67 ± 1.95 nM were determined, indicating high biological activities towards PSMA. Radiosyntheses with Ga-68, Lu-177 and Y-90 were developed under mild reaction conditions (room temperature, moderate pH of 5.5 and 7.4, respectively) and resulted in nearly quantitative radiochemical yields within 5 min.
Collapse
|
79
|
Hörsch D, Schmid KW, Anlauf M, Darwiche K, Denecke T, Baum RP, Spitzweg C, Grohé C, Presselt N, Stremmel C, Heigener DF, Serke M, Kegel T, Pavel M, Waller CF, Deppermann KM, Arnold R, Huber RM, Weber MM, Hoffmann H. Neuroendocrine tumors of the bronchopulmonary system (typical and atypical carcinoid tumors): current strategies in diagnosis and treatment. Conclusions of an expert meeting February 2011 in Weimar, Germany. Oncol Res Treat 2014; 37:266-76. [PMID: 24853787 DOI: 10.1159/000362430] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 02/24/2014] [Indexed: 11/19/2022]
Abstract
Neuroendocrine tumors (NETs; syn. carcinoid tumors) are highly or moderately differentiated neoplasms. They comprise a large variety of rare and heterogeneous tumors with an estimated incidence of 3-5/100,000/year. They can arise in virtually every internal organ, but mainly occur in the gastroenteropancreatic and bronchopulmonary systems. Around 25% of the NETs are localized in the bronchopulmonary system. Approximately 2% of all lung tumors are NETs. According to the World Health Organization (WHO) classification of lung tumors, bronchopulmonary NETs are subdivided into typical carcinoids (TCs) and atypical carcinoids (ACs). The parameter with the highest impact on NET behavior and prognosis is the histological classification and staging according to the tumor/node/metastasis (TNM) system. The diagnosis of NETs is established by histological examination and the immunohistochemical detection of general neuroendocrine markers, such as chromogranin A (CgA) and synaptophysin. Serum markers and the use of functional imaging techniques are important additive tools to establish the diagnosis of a NET. The only curative option for lung NETs is complete surgical resection. Beyond that, the currently available interdisciplinary therapeutic options are local ablation, biotherapy (somatostatin analogues), or chemotherapy. New therapeutic options such as peptide receptor radionuclide therapy (PRRT) and molecularly targeted therapies achieve promising results and are under further evaluation. This report is a consensus summary of the interdisciplinary symposium 'Neuroendocrine Tumors of the Lung and of the Gastroenteropancreatic System (GEP NET) - Expert Dialogue' held on February 25-26, 2011 in Weimar, Germany. At this conference, a panel of 23 German experts shared their knowledge and exchanged their thoughts about research, diagnosis, and clinical management of NETs, whereby special attention was paid to NETs of the respiratory tract.
Collapse
Affiliation(s)
- Dieter Hörsch
- Klinik für Innere Medizin, Gastroenterologie und Endokrinologie, Zentralklinik Bad Berka, Bad Berka, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Bol K, Haeck JC, Groen HC, Niessen WJ, Bernsen MR, de Jong M, Veenland JF. Can DCE-MRI explain the heterogeneity in radiopeptide uptake imaged by SPECT in a pancreatic neuroendocrine tumor model? PLoS One 2013; 8:e77076. [PMID: 24116203 PMCID: PMC3792933 DOI: 10.1371/journal.pone.0077076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 09/05/2013] [Indexed: 11/19/2022] Open
Abstract
Although efficient delivery and distribution of treatment agents over the whole tumor is essential for successful tumor treatment, the distribution of most of these agents cannot be visualized. However, with single-photon emission computed tomography (SPECT), both delivery and uptake of radiolabeled peptides can be visualized in a neuroendocrine tumor model overexpressing somatostatin receptors. A heterogeneous peptide uptake is often observed in these tumors. We hypothesized that peptide distribution in the tumor is spatially related to tumor perfusion, vessel density and permeability, as imaged and quantified by DCE-MRI in a neuroendocrine tumor model. Four subcutaneous CA20948 tumor-bearing Lewis rats were injected with the somatostatin-analog (111)In-DTPA-Octreotide (50 MBq). SPECT-CT and MRI scans were acquired and MRI was spatially registered to SPECT-CT. DCE-MRI was analyzed using semi-quantitative and quantitative methods. Correlation between SPECT and DCE-MRI was investigated with 1) Spearman's rank correlation coefficient; 2) SPECT uptake values grouped into deciles with corresponding median DCE-MRI parametric values and vice versa; and 3) linear regression analysis for median parameter values in combined datasets. In all tumors, areas with low peptide uptake correlated with low perfusion/density/ /permeability for all DCE-MRI-derived parameters. Combining all datasets, highest linear regression was found between peptide uptake and semi-quantitative parameters (R(2)>0.7). The average correlation coefficient between SPECT and DCE-MRI-derived parameters ranged from 0.52-0.56 (p<0.05) for parameters primarily associated with exchange between blood and extracellular extravascular space. For these parameters a linear relation with peptide uptake was observed. In conclusion, the 'exchange-related' DCE-MRI-derived parameters seemed to predict peptide uptake better than the 'contrast amount- related' parameters. Consequently, fast and efficient diffusion through the vessel wall into tissue is an important factor for peptide delivery. DCE-MRI helps to elucidate the relation between vascular characteristics, peptide delivery and treatment efficacy, and may form a basis to predict targeting efficiency.
Collapse
Affiliation(s)
- Karin Bol
- Biomedical Imaging Group Rotterdam, Departments of Radiology and Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- * E-mail:
| | - Joost C. Haeck
- Biomedical Imaging Group Rotterdam, Departments of Radiology and Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Harald C. Groen
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wiro J. Niessen
- Biomedical Imaging Group Rotterdam, Departments of Radiology and Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Imaging Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Monique R. Bernsen
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marion de Jong
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jifke F. Veenland
- Biomedical Imaging Group Rotterdam, Departments of Radiology and Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
81
|
Sabet A, Haslerud T, Pape UF, Sabet A, Ahmadzadehfar H, Grünwald F, Guhlke S, Biersack HJ, Ezziddin S. Outcome and toxicity of salvage therapy with 177Lu-octreotate in patients with metastatic gastroenteropancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2013; 41:205-10. [DOI: 10.1007/s00259-013-2547-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/14/2013] [Indexed: 11/30/2022]
|
82
|
Öksüz MÖ, Winter L, Pfannenberg C, Reischl G, Müssig K, Bares R, Dittmann H. Peptide receptor radionuclide therapy of neuroendocrine tumors with (90)Y-DOTATOC: is treatment response predictable by pre-therapeutic uptake of (68)Ga-DOTATOC? Diagn Interv Imaging 2013; 95:289-300. [PMID: 24034971 DOI: 10.1016/j.diii.2013.07.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PURPOSE PET with (68)Ga-DOTATOC allows for imaging and quantitative assessment of somatostatin receptor expression in neuroendocrine tumors (NET). The aim of this retrospective study was to analyze whether pre-therapeutic (68)Ga-DOTATOC PET/CT is able to predict response to Peptide Receptor Radionuclide Therapy (PRRT). PATIENTS AND METHODS Forty patients with advanced stage NET were treated with a fixed dose of (90)Y-DOTATOC (5550 or 3700MBq). Prior to PRRT, each patient received (68)Ga-DOTATOC PET/CT. Treatment results were evaluated after 3months by CT, tumor marker levels and clinical course and correlated with (68)Ga-DOTATOC uptake (SUVmax) and the assumed uptake of (90)Y-DOTATOC in tumor manifestations (MBq/g). ROC analysis and pairwise comparison of area under the curve (AUC) were performed with pre-treatment uptake of (68)Ga-DOTATOC, assumed uptake of (90)Y-DOTATOC and treatment activity alone and in relation to body weight as continuous variables, and response/no response as classification variable. RESULTS According to conventional criteria (tumor shrinkage, decrease of tumor markers, improved or stable clinical condition), 20 patients were classified as responders, 16 as non-responders and in four patients findings were equivocal. Using a SUV more than 17.9 as cut-off for favorable outcome, PET was able to predict treatment response of all responders and 15 out of 16 non-responders. All four patients with equivocal findings showed SUV less than or equal to 17.9 and soon experienced tumor progression. The assumed uptake of (90)Y-DOTATOC in tumor manifestations using a cut-off more than 1.26MBq/g as predictor of response was able to correctly classify 19 out of 20 responders, and 14 out of 16 non-responders. In all patients with equivocal findings, the assumed uptake of (90)Y-DOTATOC was below 1.26MBq/g. CONCLUSION Pre-therapeutic (68)Ga-DOTATOC tumor uptake as well as assumed uptake of (90)Y-DOTATOC are strongly associated with the results of subsequent PRRT. The defined cut-off values should be confirmed by prospective studies and may then provide the rationale for individual dosing and selecting patients with high likelihood of favorable treatment outcome.
Collapse
Affiliation(s)
- M Ö Öksüz
- Département d'Imagerie Médicale, Hôpital neuchâtelois, Maladière 45, 2000 Neuchâtel, Switzerland; Department of Nuclear Medicine, Tübingen University Hospital, Otfried-Müller-Strasse 14, 72076 Tübingen, Germany.
| | - L Winter
- Department of Radiology and Nuclear Medicine, Basel University Hospital, Petersgraben 4, 4031 Basel, Switzerland
| | - C Pfannenberg
- Department of Radiology, Tübingen University Hospital, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - G Reischl
- Department of Radiopharmacy, Tübingen University Hospital, Röntgenweg 15, 72076 Tübingen, Germany
| | - K Müssig
- Deutsches Diabetes-Zentrum (DDZ), Leibniz-Zentrum für Diabetes-Forschung an der Heinrich-Heine-Universität Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - R Bares
- Department of Nuclear Medicine, Tübingen University Hospital, Otfried-Müller-Strasse 14, 72076 Tübingen, Germany
| | - H Dittmann
- Department of Nuclear Medicine, Tübingen University Hospital, Otfried-Müller-Strasse 14, 72076 Tübingen, Germany
| |
Collapse
|
83
|
Somatostatin receptor-based molecular imaging and therapy for neuroendocrine tumors. BIOMED RESEARCH INTERNATIONAL 2013; 2013:102819. [PMID: 24106690 PMCID: PMC3784148 DOI: 10.1155/2013/102819] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 08/08/2013] [Indexed: 12/25/2022]
Abstract
Neuroendocrine tumors (NETs) are tumors originated from neuroendocrine cells in the body. The localization and the detection of the extent of NETs are important for diagnosis and treatment, which should be individualized according to the tumor type, burden, and symptoms. Molecular imaging of NETs with high sensitivity and specificity is achieved by nuclear medicine method using single photon-emitting and positron-emitting radiopharmaceuticals. Somatostatin receptor imaging (SRI) using SPECT or PET as a whole-body imaging technique has become a crucial part of the management of NETs. The radiotherapy with somatostatin analogues labeled with therapeutic beta emitters, such as lutetium-177 or yttrium-90, has been proved to be an option of therapy for patients with unresectable and metastasized NETs. Molecular imaging can deliver an important message to improve the outcome for patients with NETs by earlier diagnosis, better choice of the therapeutic method, and evaluation of the therapeutic response.
Collapse
|
84
|
Dong C, Zhao H, Yang S, Shi J, Huang J, Cui L, Zhong L, Jin X, Li F, Liu Z, Jia B, Wang F. 99mTc-Labeled Dimeric Octreotide Peptide: A Radiotracer with High Tumor Uptake for Single-Photon Emission Computed Tomography Imaging of Somatostatin Receptor Subtype 2-Positive Tumors. Mol Pharm 2013; 10:2925-33. [DOI: 10.1021/mp400040z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Chengyan Dong
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Department of Radiation
Medicine,
School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Huiyun Zhao
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Medical and Healthy Analytical
Center, Peking University, Beijing 100191,
China
| | - Sujuan Yang
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Department of Radiation
Medicine,
School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jiyun Shi
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Medical and Healthy Analytical
Center, Peking University, Beijing 100191,
China
| | - Jinming Huang
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Department of Radiation
Medicine,
School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Liyang Cui
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Department of Radiation
Medicine,
School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Lijun Zhong
- Medical and Healthy Analytical
Center, Peking University, Beijing 100191,
China
| | - Xiaona Jin
- Department of Nuclear
Medicine, Peking Union Medical College Hospital, Beijing 100857,
China
| | - Fang Li
- Department of Nuclear
Medicine, Peking Union Medical College Hospital, Beijing 100857,
China
| | - Zhaofei Liu
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Department of Radiation
Medicine,
School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Bing Jia
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Department of Radiation
Medicine,
School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Fan Wang
- Medical Isotopes Research
Center, Peking University, Beijing 100191,
China
- Department of Radiation
Medicine,
School of Basic Medical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
85
|
Kulkarni HR, Prasad V, Schuchardt C, Baum RP. Is there a correlation between peptide receptor radionuclide therapy-associated hematological toxicity and spleen dose? Recent Results Cancer Res 2013; 194:561-6. [PMID: 22918783 DOI: 10.1007/978-3-642-27994-2_33] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
AIM The spleen receives a high mean absorbed radiation dose during peptide receptor radionuclide therapy (PRRNT). The aim of this study is to correlate the radiation dose to spleen with the effect on blood cell count after PRRNT. METHODS Fifty-three neuroendocrine tumor (NET) patients were treated with 3.8-8.5 GBq (177)Lu-DOTATATE or (177)Lu-DOTATOC. Dosimetry was performed according to MIRD scheme. Eleven NET patients who had undergone splenectomy before PRRNT and who received 4.7-7.6 GBq (177)Lu-DOTATATE or (177)Lu-DOTATOC were selected as controls. RBC, WBC (total and differential), and platelet counts before and after each cycle of PRRNT were documented. RESULTS The median dose to the spleen in the study group was 6.34 Gy (2.32-20.06 Gy). There was no significant difference in the posttherapy changes in the blood cell counts (RBC, WBC, or platelets) between the study group and the control group. Mild hematological toxicity was found in 7 of the 53 (13.2%) patients in the study group and in 1 out of the 11 patients (9.1%) in the control group. However, there was no correlation between the incidence or grade of hematological toxicity and the dose to the spleen. CONCLUSION This study demonstrates for the first time that hematological toxicity after PRRNT is not related to the radiation dose to the spleen.
Collapse
Affiliation(s)
- Harshad R Kulkarni
- Department of Nuclear Medicine/Center for PET/CT, ENETS Center of Excellence Zentralklinik Bad Berka, Bad Berka, Germany
| | | | | | | |
Collapse
|
86
|
Atkinson H, England JA, Rafferty A, Jesudason V, Bedford K, Karsai L, Atkin SL. Somatostatin receptor expression in thyroid disease. Int J Exp Pathol 2013; 94:226-9. [PMID: 23672766 PMCID: PMC3664968 DOI: 10.1111/iep.12024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 03/05/2013] [Indexed: 11/30/2022] Open
Abstract
Somatostatin analogues are commercially available and used for the management of acromegaly and neuroendocrine tumours, but the expression of the receptors as a target in thyroid disease has not been explored. To assess somatostatin (SST) and somatostatin receptor (SSTR1-5) expression in both normal and thyroid disorders, as a potential target for somatostatin analogue therapy, 67 thyroid tissue specimens were reviewed: 12 differentiated thyroid carcinomas, 14 follicular adenomas, 17 multinodular goitres, 14 Graves disease, 10 Hashimotos thyroiditis specimens and five normal thyroids. Tissue was immunostained for SST and SSTR1-5. Positivity and the degree of positivity were recorded by double-blinded observers. Somatostatin receptor expression was highly expressed in normal tissue for SSTR1, 3, 4 and 5 (5 of 5, 4 of 5, 4 of 5 and 5 of 5 respectively) whilst SST and SSTR 2a and b were not expressed at all. The commonest receptor expressed for all pathological subtypes grouped together was SSTR2b (63 specimens). The commonest receptors expressed in differentiated thyroid cancer were SSTR5 (11 of 12 specimens) and SSTR2b (10 of 12 specimens). The commonest receptor expressed in benign disease was SSTR2b (53 of 55 specimens). SSTR5 was significantly under-expressed in Graves disease (P < 0.05). This study illustrates that SSTR 1, 3, 4 and 5 are highly expressed in normal, benign and malignant thyroid tissue. SSTR 2a and 2b appear absent in normal tissue and present in benign and malignant thyroid tissue (P < 0.02). This suggests that focussed SSTR2 treatment may be a potential therapeutic target.
Collapse
Affiliation(s)
- Helen Atkinson
- Department of Otolaryngology Head and Neck Surgery, Castle Hill Hospital, East Yorks, UK.
| | | | | | | | | | | | | |
Collapse
|
87
|
Klimstra DS. Pathology reporting of neuroendocrine tumors: essential elements for accurate diagnosis, classification, and staging. Semin Oncol 2013; 40:23-36. [PMID: 23391110 DOI: 10.1053/j.seminoncol.2012.11.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Much recent debate has focused on the optimal classification of epithelial neuroendocrine tumors (NETs). Multiple different systems of terminology, grading, and staging have been proposed, and some systems combine elements of grade and stage into a single prognostic classification. Recently, national and international consensus groups have attempted to standardize the classification of NETs, especially for those arising in the gastrointestinal tract and pancreas. Furthermore, the recognition that common classification criteria (such as proliferative rate) span multiple different systems allows the basic data necessary to predict outcome and tailor therapy to be included in pathology reports, even though a single uniform system of terminology may remain elusive. Formal tumor-node-metastasis (TNM)-based staging systems also have been developed recently, and advances in the treatment of some NETs (pancreatic in particular) are pointing towards the need to assess therapeutic biomarkers in routine practice. This review will present the most widely used systems for classifying, grading, and staging NETs and will summarize the recommendations for the data to be included in standard pathology reports of these uncommon tumors.
Collapse
Affiliation(s)
- David S Klimstra
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
88
|
Gayana S, Mittal BR, Bhattacharya A, Radotra BD, Gupta AK. 68Ga-DOTATATE PET/CT Imaging in Carotid Body Tumor. Clin Nucl Med 2013; 38:e191-3. [DOI: 10.1097/rlu.0b013e318279b71e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
89
|
Macedo D, Amaral T, Fernandes I, Sousa AR, Costa AL, Távora I, Quintela A, Cortes P, Costa L. The Treatment of Liver Metastases in Patients with Neuroendocrine Tumors in 2012. ISRN HEPATOLOGY 2013; 2013:702167. [PMID: 27335831 PMCID: PMC4890909 DOI: 10.1155/2013/702167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/08/2013] [Indexed: 02/07/2023]
Abstract
Neuroendocrine tumors (NETs) comprise a heterogeneous group of tumors that form a distinct entity. Approximately 75-80% of patients present with liver metastases at the time of their diagnosis, and 20%-25% will develop these lesions in the course of their disease. The presence of secondary deposits in the liver significantly increases the morbidity and mortality in these patients. The only potentially curative treatment is the surgical resection of the primary tumor and hepatic lesions. However, only 10% of patients presents under ideal conditions for that approach. Several techniques aimed at localized liver lesions have been applied also with interesting results in terms of survival and symptom control. The same has been demonstrated with new systemic therapies (target therapies). However, these are still under study, in order to define their true role in the management of these patients. This paper intends to address, in a general way, the various treatment options in patients with liver metastases from neuroendocrine tumors.
Collapse
Affiliation(s)
- Daniela Macedo
- Oncology Division, Hospital de Santa Maria, Lisbon, Portugal
| | - Teresa Amaral
- Oncology Division, Hospital de Santa Maria, Lisbon, Portugal
| | - Isabel Fernandes
- Oncology Division, Hospital de Santa Maria, Lisbon, Portugal
- Instituto de Medicina Molecular, University of Lisbon, Portugal
| | - Ana Rita Sousa
- Oncology Division, Hospital de Santa Maria, Lisbon, Portugal
| | - Ana Lúcia Costa
- Oncology Division, Hospital de Santa Maria, Lisbon, Portugal
| | - Isabel Távora
- Radiology Division, Hospital de Santa Maria, Lisbon AV Professor Egas Moniz, 1649-039 Lisbon, Portugal
| | | | - Paulo Cortes
- Oncology Division, Hospital de Santa Maria, Lisbon, Portugal
| | - Luís Costa
- Oncology Division, Hospital de Santa Maria, Lisbon, Portugal
- Instituto de Medicina Molecular, University of Lisbon, Portugal
| |
Collapse
|
90
|
Semiautomated labelling and fractionation of yttrium-90 and lutetium-177 somatostatin analogues using disposable syringes and vials. Nucl Med Commun 2012; 33:1144-52. [DOI: 10.1097/mnm.0b013e328358131f] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
91
|
Rustagi T, Rai M, Bauer F. Non-functional Pancreatic Neuroendocrine Tumor as an Incidentaloma—A Case Report and Review of Literature. J Gastrointest Cancer 2012; 44:336-42. [DOI: 10.1007/s12029-012-9445-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
92
|
Myelodysplastic Syndrome with t(1;7) Associated with Marked Dysmegakarypoiesis & Severe Thrombocytopenia: A Case Report and Review of the Literature. Case Rep Hematol 2012; 2012:167653. [PMID: 22937318 PMCID: PMC3420701 DOI: 10.1155/2012/167653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 02/01/2012] [Indexed: 11/17/2022] Open
Abstract
We present the case of a 70-year-old woman who had a bone marrow examination performed to investigate marked thrombocytopenia in the context of a recent history of metastatic glucagonoma. Surprisingly this identified marked dysmegakaryopoiesis and fulfilled diagnostic criteria for refractory cytopenia with multilineage dysplasia, with a relatively uncommon associated cytogenetic lesion t(1;7). We present the case and review the literature of this cytogenetic lesion.
Collapse
|
93
|
Fani M, Maecke HR, Okarvi SM. Radiolabeled peptides: valuable tools for the detection and treatment of cancer. Am J Cancer Res 2012; 2:481-501. [PMID: 22737187 PMCID: PMC3364555 DOI: 10.7150/thno.4024] [Citation(s) in RCA: 234] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/31/2012] [Indexed: 12/17/2022] Open
Abstract
Human cancer cells overexpress many peptide receptors as molecular targets. Radiolabeled peptides that bind with high affinity and specificity to the receptors on tumor cells hold great potential for both diagnostic imaging and targeted radionuclide therapy. The advantage of solid-phase peptide synthesis, the availability of different chelating agents and prosthetic groups and bioconjugation techniques permit the facile preparation of a wide variety of peptide-based targeting molecules with diverse biological and tumor targeting properties. Some of these peptides, including somatostatin, bombesin, vasoactive intestinal peptide, gastrin, neurotensin, exendin and RGD are currently under investigation. It is anticipated that in the near future many of these peptides may find applications in nuclear oncology. This article presents recent developments in the field of small peptides, and their applications in the diagnosis and treatment of cancer.
Collapse
|
94
|
Li J, Chen F, Cona MM, Feng Y, Himmelreich U, Oyen R, Verbruggen A, Ni Y. A review on various targeted anticancer therapies. Target Oncol 2012; 7:69-85. [PMID: 22350489 DOI: 10.1007/s11523-012-0212-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 01/30/2012] [Indexed: 12/11/2022]
Abstract
Translational oncology aims to translate laboratory research into new anticancer therapies. Contrary to conventional surgery, chemotherapy, and radiotherapy, targeted anticancer therapy (TAT) refers to systemic administration of drugs with particular mechanisms that specifically act on well-defined targets or biologic pathways that, when activated or inactivated, may cause regression or destruction of the malignant process, meanwhile with minimized adverse effects on healthy tissues. In this article, we intend to first give a brief review on various known TAT approaches that are deemed promising for clinical applications in the current trend of personalized medicine, and then we will introduce our newly developed approach namely small molecular sequential dual targeting theragnostic strategy as a generalized class of TAT for the management of most solid malignancies, which, after optimization, is expected to help improve overall cancer treatability and curability.
Collapse
Affiliation(s)
- Junjie Li
- Section of Radiology, Department of Diagnostic Sciences, Faculty of Medicine, University of Leuven, Herestraat 49, BE-3000, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Velikyan I, Xu H, Nair M, Hall H. Robust labeling and comparative preclinical characterization of DOTA-TOC and DOTA-TATE. Nucl Med Biol 2012; 39:628-39. [PMID: 22336375 DOI: 10.1016/j.nucmedbio.2011.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 12/09/2011] [Accepted: 12/14/2011] [Indexed: 01/15/2023]
Abstract
OBJECTIVES Various radionuclide-labeled somatostatin analogues are used currently for diagnosis and therapy of neuroendocrine tumors. In particular, [68Ga]Ga-DOTA-TOC is commonly used for diagnosis, while [177Lu]Lu-DOTA-TATE is used for therapy. With the development of theranostics and personalized medicine where the imaging diagnosis is tailored to the subsequent radiotherapy, it is of paramount importance to investigate the relevance of the ligand exchange. The aim of this study was to compare binding capacity of [67/68Ga]Ga-DOTA-TOC ([67/68Ga]Ga-N-(4,7,10-(tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetyl-D-Phe-c[Cys-D-Tyr-Trp-Lys-Thr-Cys]-Thr(ol)) and [67/68Ga]Ga-DOTA-TATE ([67/68Ga]Ga-N-(4,7,10-(tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetyl-D-Phe-c[Cys-D-Tyr-Trp-Lys-Thr-Cys]-Thr) in vitro in monkey brain cryosections and in vivo in the rat, where, in contrast to transfected cell lines, there is a heterogeneous distribution of somatostatin receptor (SSTR) subtypes. The influence of various production methods of [68Ga]Ga-DOTA-TOC and [68Ga]Ga-DOTA-TATE on the biological performance of the tracers was also studied. MATERIAL AND METHODS [67Ga]Ga-DOTA-TOC, [68Ga]Ga-DOTA-TOC, [67Ga]Ga-DOTA-TATE and [68Ga]Ga-DOTA-TATE were synthesized including preconcentration and purification of the generator eluate. The binding of the radioligands was assessed in vitro using autoradiography on cryosections of Rhesus monkey brains and in vivo/ex vivo using organ distribution studies in rats. RESULTS AND DISCUSSION The tracer production method was improved in terms of higher robustness, simplification and good manufacturing practice (GMP) relevance. The synthesis variation did not influence the biological performance of the tracers. There was no statistically significant difference observed in the binding of [67/68Ga]Ga-DOTA-TOC and [67/68Ga]Ga-DOTA-TATE either in brain cortex in vitro or in rat biodistribution and uptake in SSTR-positive tissues such as pancreas, adrenals and pituitary. The uptake in these organs was precluded by the excess of octreotide (Sandostatin). The 10-fold higher affinity to SSTR2 of DOTA-TATE as compared to DOTA-TOC known from studies in transfected cells was reflected in a slightly more intense binding of [67/68Ga]Ga-DOTA-TATE than of [67/68Ga]Ga-DOTA-TOC in the monkey brain sections in vitro, but not in vivo in the rat. CONCLUSION A robust 68Ga-labeling method was introduced. The difference in the uptake of [67/68Ga]Ga-DOTA-TOC and [67/68Ga]Ga-DOTA-TATE in SSTR2-positive organs was not statistically significant either in vitro in tissue studies or in vivo/ex vivo in rat experiments. The results indicate that the more complex environment in vitro and in vivo diminishes the difference observed in transfected cell line binding.
Collapse
Affiliation(s)
- Irina Velikyan
- Department of Radiology, Oncology and Radiation Sciences, Uppsala University, PET Center, Uppsala University Hospital, Uppsala, SE-75185 Uppsala, Sweden.
| | | | | | | |
Collapse
|
96
|
Dasanu CA, Majumder S, Gopal S, Stoica-Mustafa E, Trikudanathan G. Emerging therapeutic options for advanced enteropancreatic neuroendocrine tumors. Expert Opin Pharmacother 2012; 13:461-71. [PMID: 22292707 DOI: 10.1517/14656566.2012.656089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Several chemotherapy agents and combinations have proven effective in the therapy of advanced enteropancreatic neuroendocrine tumors (EP-NETs). However, their toxicity can be significant. Recent understanding of the molecular mechanisms of these tumors, especially the central role of tumor angiogenesis, has led to the identification of new therapeutic targets and agents directed at the molecular level. AREAS COVERED This paper gives a comprehensive evaluation of the existing therapeutic armamentarium for EP-NETs. Narrated in a historical perspective, this review analyzes the available information on traditional chemotherapy agents, interferon-α and somatostatin analogs, as well as newer therapies and experimental agents. EXPERT OPINION Despite recent advances, a curative approach for metastatic EP-NETs is yet to be discovered. To date, sunitinib and everolimus have been shown to impact progression-free survival only in pancreatic NETs, and the duration of this benefit has not yet been established. Further research is necessary to determine whether a combination of these drugs, either together or with other therapies, may yield superior outcomes. Moreover, sequential use of these agents should be explored in an attempt to improve survival. Efficacy of a variety of experimental agents is also being tested in clinical trials.
Collapse
Affiliation(s)
- Constantin A Dasanu
- Department of Hematology-Oncology, St. Francis Hospital and Medical Center, Hartford, CT 06105, USA.
| | | | | | | | | |
Collapse
|
97
|
Das T, Chakraborty S, Kallur KG, Venkatesh M, Banerjee S. Preparation of patient doses of (177)Lu-DOTA-TATE using indigenously produced (177)Lu: the Indian experience. Cancer Biother Radiopharm 2012; 26:395-400. [PMID: 21728843 DOI: 10.1089/cbr.2010.0881] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE (177)Lu (T(1/2) = 6.73 days, E(β(max)) = 0.497 MeV, E(γ) = 113 KeV [6.4%] and 208 KeV [11%])-labeled DOTA-TATE, a somatostatin analog, is presently being considered a promising agent for the treatment of patients suffering from inoperable neuroendocrine tumors, which overexpress somatostatin receptors. The objective of the present work was to develop an optimized protocol for the preparation of therapeutic dose of (177)Lu-DOTA-TATE with as high as achievable specific activity at the time of its administration, taking into account the variable specific activity of (177)Lu available during the preparation of the agent. METHODS (177)Lu labeling of DOTA-TATE was carried out using a precalculated amount of DOTA-TATE based on the available specific activity of (177)Lu at the time of preparation, keeping a minimum molar ratio of [DOTA-TATE]:[Lu] = 4:1, so that (177)Lu-DOTA-TATE could be obtained with highest possible specific activity without compromising its radiochemical purity and stability. RESULTS One hundred (100) batches of (177)Lu-DOTA-TATE were prepared following this protocol till date at five different nuclear medicine centers of India, with a radiochemical purity of 98.25% ± 1.1% and specific activity of 32.74-65.49 GBq/μmol (885-1770 mCi/μmol). Till date, 250 patient doses of (177)Lu-DOTA-TATE have been dispensed and administered in 150 patients suffering from various types of neuroendocrine-originated tumors. CONCLUSIONS The developed method ensures that patient doses of (177)Lu-DOTA-TATE could be prepared with highest possible specific activity depending upon the available specific activity of (177)Lu at the hospital radio-pharmacy.
Collapse
Affiliation(s)
- Tapas Das
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | | | | | | | | |
Collapse
|
98
|
A multimodal approach to the management of neuroendocrine tumour liver metastases. Int J Hepatol 2012; 2012:819193. [PMID: 22518323 PMCID: PMC3296190 DOI: 10.1155/2012/819193] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 12/02/2011] [Indexed: 12/13/2022] Open
Abstract
Neuroendocrine tumours (NETs) are often indolent malignancies that commonly present with metastatic disease in the liver. Surgical, locoregional, and systemic treatment modalities are reviewed. A multidisciplinary approach to patient care is suggested to ensure all therapeutic options explored.
Collapse
|
99
|
Schmid HA, Lambertini C, van Vugt HH, Barzaghi-Rinaudo P, Schäfer J, Hillenbrand R, Sailer AW, Kaufmann M, Nuciforo P. Monoclonal antibodies against the human somatostatin receptor subtypes 1-5: development and immunohistochemical application in neuroendocrine tumors. Neuroendocrinology 2012; 95:232-47. [PMID: 22156600 DOI: 10.1159/000330616] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 06/25/2011] [Indexed: 01/26/2023]
Abstract
BACKGROUND Activation of somatostatin receptors (sstr1-5) by somatostatin and its analogues exerts an inhibitory effect on hormone secretion and provides the basis for the treatment of a range of endocrine diseases such as acromegaly, Cushing's disease and neuroendocrine tumors (NET). The lack of well-characterized commercially available sstr subtype-specific antibodies prevents routine identification of the sstr expression profile in patients. METHODS We generated and characterized new mouse monoclonal antibodies (mAbs) targeting the five human sstr subtypes using ELISA and immunohistochemistry, and tested their suitability in formalin-fixed and paraffin-embedded (FFPE) human tissues and archival samples of normal pancreatic tissue and NET. RESULTS All mAbs were highly specific with no cross-reactivity. The sstr1-5 immunoreactivity in gastrointestinal NET (n=67) was correlated with clinicopathologic data. With the exception of sstr3, NET were highly positive for all receptor subtypes (42, 63, 6, 32 and 65% of tumors were positive for sstr1, sstr2a, sstr3, sstr4 and sstr5, respectively). sstr1, sstr2a and sstr5 were present at the plasma membrane and in the cytoplasm of tumor cells, whereas sstr3 and sstr4 were almost exclusively cytoplasmic. Immunoreactivity of sstr1, sstr2a and sstr4 tended to decrease as tumor aggressiveness increased. sstr5 showed an opposite pattern, with higher staining in well-differentiated carcinomas compared with well-differentiated tumors. sstr5 immunoreactivity was correlated with the presence of metastases and angioinvasion, suggesting a possible association with more aggressive behavior. CONCLUSION Determination of the sstr1-5 by immunohistochemistry using subtype-specific mAbs is feasible in FFPE tissue and may provide a tool for routine clinical practice.
Collapse
Affiliation(s)
- Herbert A Schmid
- Novartis Institutes for BioMedical Research, Basel, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Oddstig J, Bernhardt P, Lizana H, Nilsson O, Ahlman H, Kölby L, Forssell-Aronsson E. Inhomogeneous activity distribution of 177Lu-DOTA0-Tyr3-octreotate and effects on somatostatin receptor expression in human carcinoid GOT1 tumors in nude mice. Tumour Biol 2011; 33:229-39. [PMID: 22108870 DOI: 10.1007/s13277-011-0268-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 11/07/2011] [Indexed: 10/15/2022] Open
Abstract
The aim of this study was to investigate the activity distribution in neouroendocrine tumors after diagnostic, or therapeutic, amounts of [(177)Lu-DOTA(0)-Tyr(3)]-octreotate and to investigate how the activity distribution influences the absorbed dose. Furthermore, the activity distribution of a second administration of radiolabeled octreotate was studied. Nude mice with subcutaneously grown human midgut carcinoid (GOT1) were injected intravenously with different amounts of (177)Lu-octreotate. At different time points thereafter (4 h to 13 days), a second injection of [(111)In-DOTA(0)-Tyr(3)]-octreotate was given to estimate the somatostatin receptor (sstr) expression. The activity distribution in the tumors was then determined. Monte Carlo simulations with PENELOPE were performed for dosimetry. Fifty-one out of 58 investigated tumors showed a lower activity concentration in the peripheral part than in the central part of the tumor. The amount of activity injected, or time after administration, did neither influence the relative activity nor the sstr distribution in the tumor. After an initial down-regulation (at 4-24 h), there was an up-regulation of sstr (1.5-2 times, at 7-14 days). Monte Carlo simulations demonstrated an inhomogeneous absorbed dose distribution in the tumor using (177)Lu, with twice as high absorbed dose centrally than peripherally. The high activity concentration centrally and the up-regulation of sstr demonstrated will facilitate fractionated therapy using radiolabeled somatostatin analogues if similar results will be obtained also in patients. The inhomogeneous activity distribution in the tumor has to be taken into account when the absorbed dose distribution in tumor is calculated.
Collapse
Affiliation(s)
- Jenny Oddstig
- Department of Radiation Physics, Lundberg Laboratory for Cancer Research, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden.
| | | | | | | | | | | | | |
Collapse
|