1
|
Han SA, Ryu J, Song SW, Kim JS, Ryu JS, Oh M. 68Ga-DOATATOC Brain PET/CT Imaging in a case of Dural Metastasis from Synovial Sarcoma. Nucl Med Mol Imaging 2024; 58:310-316. [PMID: 39036463 PMCID: PMC11255146 DOI: 10.1007/s13139-024-00854-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 07/23/2024] Open
Abstract
We present the case of a 70-year-old male patient who underwent a gallium-68 (68Ga)-DOTATOC brain positron emission tomography (PET)/computed tomography (CT) for the assessment of a tumorous lesion on the dura. The patient had previously undergone below-knee amputation due to a mass of synovial sarcoma on the left foot and completed adjuvant chemotherapy approximately 3 months ago. Subsequently, a well-demarcated papillary solid mass located on the dura was surgically excised. Pathological examination confirmed that the dural metastasis originated from synovial sarcoma and post-operative magnetic resonance imaging (MRI) revealed no residual tumor. We conducted a 68Ga-DOTATOC brain PET/CT suspecting a meningioma given the presence of a dural mass. The result showed lower uptake (maximum standardized uptake [SUVmax 4.9]) than the pituitary gland (SUVmax 9.3). Thus, we successfully conducted a differential diagnosis of metastasis from the preexisting malignancy rather than the meningioma. 68Ga-DOTATOC PET/CT is a valuable tool for the differential diagnosis of meningioma. However, metastasis should also be considered, especially in patients with a history of malignancy and lesions showing mild 68Ga-DOTATOC uptake.
Collapse
Affiliation(s)
- Shin Ae Han
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Jeongryul Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Sang Woo Song
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae-Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Jin-Sook Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| |
Collapse
|
2
|
Boursier C, Zaragori T, Bros M, Bordonne M, Melki S, Taillandier L, Blonski M, Roch V, Marie PY, Karcher G, Imbert L, Verger A. Semi-automated segmentation methods of SSTR PET for dosimetry prediction in refractory meningioma patients treated by SSTR-targeted peptide receptor radionuclide therapy. Eur Radiol 2023; 33:7089-7098. [PMID: 37148355 DOI: 10.1007/s00330-023-09697-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/10/2023] [Accepted: 03/12/2023] [Indexed: 05/08/2023]
Abstract
OBJECTIVES Tumor dosimetry with somatostatin receptor-targeted peptide receptor radionuclide therapy (SSTR-targeted PRRT) by 177Lu-DOTATATE may contribute to improved treatment monitoring of refractory meningioma. Accurate dosimetry requires reliable and reproducible pretherapeutic PET tumor segmentation which is not currently available. This study aims to propose semi-automated segmentation methods to determine metabolic tumor volume with pretherapeutic 68Ga-DOTATOC PET and evaluate SUVmean-derived values as predictive factors for tumor-absorbed dose. METHODS Thirty-nine meningioma lesions from twenty patients were analyzed. The ground truth PET and SPECT volumes (VolGT-PET and VolGT-SPECT) were computed from manual segmentations by five experienced nuclear physicians. SUV-related indexes were extracted from VolGT-PET and the semi-automated PET volumes providing the best Dice index with VolGT-PET (Volopt) across several methods: SUV absolute-value (2.3)-threshold, adaptative methods (Jentzen, Otsu, Contrast-based method), advanced gradient-based technique, and multiple relative thresholds (% of tumor SUVmax, hypophysis SUVmean, and meninges SUVpeak) with optimal threshold optimized. Tumor-absorbed doses were obtained from the VolGT-SPECT, corrected for partial volume effect, performed on a 360° whole-body CZT-camera at 24, 96, and 168 h after administration of 177Lu-DOTATATE. RESULTS Volopt was obtained from 1.7-fold meninges SUVpeak (Dice index 0.85 ± 0.07). SUVmean and total lesion uptake (SUVmeanxlesion volume) showed better correlations with tumor-absorbed doses than SUVmax when determined with the VolGT (respective Pearson correlation coefficients of 0.78, 0.67, and 0.56) or Volopt (0.64, 0.66, and 0.56). CONCLUSION Accurate definition of pretherapeutic PET volumes is justified since SUVmean-derived values provide the best tumor-absorbed dose predictions in refractory meningioma patients treated by 177Lu-DOTATATE. This study provides a semi-automated segmentation method of pretherapeutic 68Ga-DOTATOC PET volumes to achieve good reproducibility between physicians. CLINICAL RELEVANCE STATEMENT SUVmean-derived values from pretherapeutic 68Ga-DOTATOC PET are predictive of tumor-absorbed doses in refractory meningiomas treated by 177Lu-DOTATATE, justifying to accurately define pretherapeutic PET volumes. This study provides a semi-automated segmentation of 68Ga-DOTATOC PET images easily applicable in routine. KEY POINTS • SUVmean-derived values from pretherapeutic 68Ga-DOTATOC PET images provide the best predictive factors of tumor-absorbed doses related to 177Lu-DOTATATE PRRT in refractory meningioma. • A 1.7-fold meninges SUVpeak segmentation method used to determine metabolic tumor volume on pretherapeutic 68Ga-DOTATOC PET images of refractory meningioma treated by 177Lu-DOTATATE is as efficient as the currently routine manual segmentation method and limits inter- and intra-observer variabilities. • This semi-automated method for segmentation of refractory meningioma is easily applicable to routine practice and transferrable across PET centers.
Collapse
Affiliation(s)
- Caroline Boursier
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France.
- Université de Lorraine, IADI, INSERM U1254, F-54000, Nancy, France.
- Nancyclotep Imaging Platform, F-54000, Nancy, France.
| | | | - Marie Bros
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
| | - Manon Bordonne
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
| | - Saifeddine Melki
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
| | - Luc Taillandier
- Department of Neuro-Oncology, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
- Centre de Recherche en Automatique de Nancy CRAN, UMR 7039, Université de Lorraine, CNRS, F-54000, Nancy, France
| | - Marie Blonski
- Department of Neuro-Oncology, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
- Centre de Recherche en Automatique de Nancy CRAN, UMR 7039, Université de Lorraine, CNRS, F-54000, Nancy, France
| | - Veronique Roch
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
- Nancyclotep Imaging Platform, F-54000, Nancy, France
| | - Pierre-Yves Marie
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
- Université de Lorraine, IADI, INSERM U1254, F-54000, Nancy, France
- Nancyclotep Imaging Platform, F-54000, Nancy, France
| | - Gilles Karcher
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
- Nancyclotep Imaging Platform, F-54000, Nancy, France
| | - Laëtitia Imbert
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
- Université de Lorraine, IADI, INSERM U1254, F-54000, Nancy, France
- Nancyclotep Imaging Platform, F-54000, Nancy, France
| | - Antoine Verger
- Department of Nuclear Medicine, Université de Lorraine, CHRU Nancy, F-54000, Nancy, France
- Université de Lorraine, IADI, INSERM U1254, F-54000, Nancy, France
- Nancyclotep Imaging Platform, F-54000, Nancy, France
| |
Collapse
|
3
|
Iglseder S, Iglseder A, Beliveau V, Heugenhauser J, Gizewski ER, Kerschbaumer J, Stockhammer G, Uprimny C, Virgolini I, Dudas J, Nevinny-Stickel M, Nowosielski M, Scherfler C. Somatostatin receptor subtype expression and radiomics from DWI-MRI represent SUV of [68Ga]Ga-DOTATOC PET in patients with meningioma. J Neurooncol 2023; 164:711-720. [PMID: 37707754 PMCID: PMC10589159 DOI: 10.1007/s11060-023-04414-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/03/2023] [Indexed: 09/15/2023]
Abstract
OBJECTIVE This retrospective study aimed to analyse the correlation between somatostatin receptor subtypes (SSTR 1-5) and maximum standardized uptake value (SUVmax) in meningioma patients using Gallium-68 DOTA-D-Phe1-Tyr3-octreotide Positron Emission Tomography ([68Ga]Ga-DOTATOC PET). Secondly, we developed a radiomic model based on apparent diffusion coefficient (ADC) maps derived from diffusion weighted magnetic resonance images (DWI MRI) to reproduce SUVmax. METHOD The study included 51 patients who underwent MRI and [68Ga]Ga-DOTATOC PET before meningioma surgery. SUVmax values were quantified from PET images and tumour areas were segmented on post-contrast T1-weighted MRI and mapped to ADC maps. A total of 1940 radiomic features were extracted from the tumour area on each ADC map. A random forest regression model was trained to predict SUVmax and the model's performance was evaluated using repeated nested cross-validation. The expression of SSTR subtypes was quantified in 18 surgical specimens and compared to SUVmax values. RESULTS The random forest regression model successfully predicted SUVmax values with a significant correlation observed in all 100 repeats (p < 0.05). The mean Pearson's r was 0.42 ± 0.07 SD, and the root mean square error (RMSE) was 28.46 ± 0.16. SSTR subtypes 2A, 2B, and 5 showed significant correlations with SUVmax values (p < 0.001, R2 = 0.669; p = 0.001, R2 = 0.393; and p = 0.012, R2 = 0.235, respectively). CONCLUSION SSTR subtypes 2A, 2B, and 5 correlated significantly with SUVmax in meningioma patients. The developed radiomic model based on ADC maps effectively reproduces SUVmax using [68Ga]Ga-DOTATOC PET.
Collapse
Affiliation(s)
- Sarah Iglseder
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Anna Iglseder
- Department of Geodesy and Geoinformation, Technical University Vienna, Vienna, Austria
| | - Vincent Beliveau
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
- Neuroimaging Research Core Facility, Innsbruck Medical University, Innsbruck, Austria
| | | | - Elke R Gizewski
- Neuroimaging Research Core Facility, Innsbruck Medical University, Innsbruck, Austria
- Department of Neuroradiology, Innsbruck Medical University, Innsbruck, Austria
| | | | | | - Christian Uprimny
- Department of Nuclear Medicine, Innsbruck Medical University, Innsbruck, Austria
| | - Irene Virgolini
- Department of Nuclear Medicine, Innsbruck Medical University, Innsbruck, Austria
| | - Jozsef Dudas
- Department of Otorhinolaryngology, Innsbruck Medical University, Innsbruck, Austria
| | - Meinhard Nevinny-Stickel
- Department of Therapeutic Radiology and Oncology, Innsbruck Medical University, Innsbruck, Austria
| | - Martha Nowosielski
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria.
| | - Christoph Scherfler
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
- Department of Neuroradiology, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
4
|
Akhavanallaf A, Peterson AB, Fitzpatrick K, Roseland M, Wong KK, El-Naqa I, Zaidi H, Dewaraja YK. The predictive value of pretherapy [ 68Ga]Ga-DOTA-TATE PET and biomarkers in [ 177Lu]Lu-PRRT tumor dosimetry. Eur J Nucl Med Mol Imaging 2023; 50:2984-2996. [PMID: 37171633 PMCID: PMC10981963 DOI: 10.1007/s00259-023-06252-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023]
Abstract
PURPOSE Metastatic neuroendocrine tumors (NETs) overexpressing type 2 somatostatin receptors are the target for peptide receptor radionuclide therapy (PRRT) through the theragnostic pair of 68Ga/177Lu-DOTATATE. The main purpose of this study was to develop machine learning models to predict therapeutic tumor dose using pre therapy 68Ga -PET and clinicopathological biomarkers. METHODS We retrospectively analyzed 90 segmented metastatic NETs from 25 patients (M14/F11, age 63.7 ± 9.5, range 38-76) treated by 177Lu-DOTATATE at our institute. Patients underwent both pretherapy [68Ga]Ga-DOTA-TATE PET/CT and four timepoints SPECT/CT at ~ 4, 24, 96, and 168 h post-177Lu-DOTATATE infusion. Tumors were segmented by a radiologist on baseline CT or MRI and transferred to co-registered PET/CT and SPECT/CT, and normal organs were segmented by deep learning-based method on CT of the PET and SPECT. The SUV metrics and tumor-to-normal tissue SUV ratios (SUV_TNRs) were calculated from 68Ga -PET at the contour-level. Posttherapy dosimetry was performed based on the co-registration of SPECT/CTs to generate time-integrated-activity, followed by an in-house Monte Carlo-based absorbed dose estimation. The correlation between delivered 177Lu Tumor absorbed dose and PET-derived metrics along with baseline clinicopathological biomarkers (such as Creatinine, Chromogranin A and prior therapies) were evaluated. Multiple interpretable machine-learning algorithms were developed to predict tumor dose using these pretherapy information. Model performance on a nested tenfold cross-validation was evaluated in terms of coefficient of determination (R2), mean-absolute-error (MAE), and mean-relative-absolute-error (MRAE). RESULTS SUVmean showed a significant correlation (q-value < 0.05) with absorbed dose (Spearman ρ = 0.64), followed by TLSUVmean (SUVmean of total-lesion-burden) and SUVpeak (ρ = 0.45 and 0.41, respectively). The predictive value of PET-SUVmean in estimation of posttherapy absorbed dose was stronger compared to PET-SUVpeak, and SUV_TNRs in terms of univariate analysis (R2 = 0.28 vs. R2 ≤ 0.12). An optimal trivariate random forest model composed of SUVmean, TLSUVmean, and total liver SUVmean (normal and tumoral liver) provided the best performance in tumor dose prediction with R2 = 0.64, MAE = 0.73 Gy/GBq, and MRAE = 0.2. CONCLUSION Our preliminary results demonstrate the feasibility of using baseline PET images for prediction of absorbed dose prior to 177Lu-PRRT. Machine learning models combining multiple PET-based metrics performed better than using a single SUV value and using other investigated clinicopathological biomarkers. Developing such quantitative models forms the groundwork for the role of 68Ga -PET not only for the implementation of personalized treatment planning but also for patient stratification in the era of precision medicine.
Collapse
Affiliation(s)
- Azadeh Akhavanallaf
- Department of Radiology, University of Michigan, 1301 Catherine, 2276 Medical Science I/5610, Ann Arbor, MI, 48109, USA.
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland.
| | - Avery B Peterson
- Department of Radiology, University of Michigan, 1301 Catherine, 2276 Medical Science I/5610, Ann Arbor, MI, 48109, USA
| | - Kellen Fitzpatrick
- Department of Radiology, University of Michigan, 1301 Catherine, 2276 Medical Science I/5610, Ann Arbor, MI, 48109, USA
| | - Molly Roseland
- Department of Radiology, University of Michigan, 1301 Catherine, 2276 Medical Science I/5610, Ann Arbor, MI, 48109, USA
| | - Ka Kit Wong
- Department of Radiology, University of Michigan, 1301 Catherine, 2276 Medical Science I/5610, Ann Arbor, MI, 48109, USA
| | - Issam El-Naqa
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, USA
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
- Geneva University Neurocenter, Geneva University, CH-1205, Geneva, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, DK-500, Odense, Denmark
| | - Yuni K Dewaraja
- Department of Radiology, University of Michigan, 1301 Catherine, 2276 Medical Science I/5610, Ann Arbor, MI, 48109, USA
| |
Collapse
|
5
|
Peterson AB, Wang C, Wong KK, Frey KA, Muzik O, Schipper MJ, Dewaraja YK. 177Lu-DOTATATE Theranostics: Predicting Renal Dosimetry From Pretherapy 68Ga-DOTATATE PET and Clinical Biomarkers. Clin Nucl Med 2023; 48:393-399. [PMID: 37010563 PMCID: PMC10353839 DOI: 10.1097/rlu.0000000000004599] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
PURPOSE Pretreatment predictions of absorbed doses can be especially valuable for patient selection and dosimetry-guided individualization of radiopharmaceutical therapy. Our goal was to build regression models using pretherapy 68Ga-DOTATATE PET uptake data and other baseline clinical factors/biomarkers to predict renal absorbed dose delivered by 177Lu-DOTATATE peptide receptor radionuclide therapy (177Lu-PRRT) for neuroendocrine tumors. We explore the combination of biomarkers and 68Ga PET uptake metrics, hypothesizing that they will improve predictive power over univariable regression. PATIENTS AND METHODS Pretherapy 68Ga-DOTATATE PET/CTs were analyzed for 25 patients (50 kidneys) who also underwent quantitative 177Lu SPECT/CT imaging at approximately 4, 24, 96, and 168 hours after cycle 1 of 177Lu-PRRT. Kidneys were contoured on the CT of the PET/CT and SPECT/CT using validated deep learning-based tools. Dosimetry was performed by coupling the multi-time point SPECT/CT images with an in-house Monte Carlo code. Pretherapy renal PET SUV metrics, activity concentration per injected activity (Bq/mL/MBq), and other baseline clinical factors/biomarkers were investigated as predictors of the 177Lu SPECT/CT-derived mean absorbed dose per injected activity to the kidneys using univariable and bivariable models. Leave-one-out cross-validation (LOOCV) was used to estimate model performance using root mean squared error and absolute percent error in predicted renal absorbed dose including mean absolute percent error (MAPE) and associated standard deviation (SD). RESULTS The median therapy-delivered renal dose was 0.5 Gy/GBq (range, 0.2-1.0 Gy/GBq). In LOOCV of univariable models, PET uptake (Bq/mL/MBq) performs best with MAPE of 18.0% (SD = 13.3%), and estimated glomerular filtration rate (eGFR) gives an MAPE of 28.5% (SD = 19.2%). Bivariable regression with both PET uptake and eGFR gives LOOCV MAPE of 17.3% (SD = 11.8%), indicating minimal improvement over univariable models. CONCLUSIONS Pretherapy 68Ga-DOTATATE PET renal uptake can be used to predict post-177Lu-PRRT SPECT-derived mean absorbed dose to the kidneys with accuracy within 18%, on average. Compared with PET uptake alone, including eGFR in the same model to account for patient-specific kinetics did not improve predictive power. Following further validation of these preliminary findings in an independent cohort, predictions using renal PET uptake can be used in the clinic for patient selection and individualization of treatment before initiating the first cycle of PRRT.
Collapse
Affiliation(s)
- Avery B. Peterson
- Department of Radiology, University of Michigan, Ann Arbor
- Department of Radiation Oncology, Wayne State University, Detroit
| | - Chang Wang
- Department of Biostatistics, University of Michigan, Ann Arbor
| | - Ka Kit Wong
- Department of Radiology, University of Michigan, Ann Arbor
| | - Kirk A. Frey
- Department of Radiology, University of Michigan, Ann Arbor
| | - Otto Muzik
- Department of Pediatrics, Wayne State University, Detroit, MI
| | | | | |
Collapse
|
6
|
Stenvall A, Gustafsson J, Larsson E, Roth D, Sundlöv A, Jönsson L, Hindorf C, Ohlsson T, Sjögreen Gleisner K. Relationships between uptake of [ 68Ga]Ga-DOTA-TATE and absorbed dose in [ 177Lu]Lu-DOTA-TATE therapy. EJNMMI Res 2022; 12:75. [PMID: 36534192 PMCID: PMC9763525 DOI: 10.1186/s13550-022-00947-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Somatostatin receptor 68Ga PET imaging is standard for evaluation of a patient's suitability for 177Lu peptide receptor radionuclide therapy of neuroendocrine tumours (NETs). The 68Ga PET serves to ensure sufficient somatostatin receptor expression, commonly evaluated qualitatively. The aim of this study is to investigate the quantitative relationships between uptake in 68Ga PET and absorbed doses in 177Lu therapy. METHOD Eighteen patients underwent [68Ga]Ga-DOTA-TATE PET imaging within 20 weeks prior to their first cycle of [177Lu]Lu-DOTA-TATE. Absorbed doses for therapy were estimated for tumours, kidney, spleen, and normal liver parenchyma using a hybrid SPECT/CT-planar method. Gallium-68 activity concentrations were retrieved from PET images and also used to calculate SUVs and normalized SUVs, using blood and tissue for normalization. The 68Ga activity concentrations per injected activity, SUVs, and normalized SUVs were compared with 177Lu activity concentrations 1 d post-injection and 177Lu absorbed doses. For tumours, for which there was a variable number per patient, both inter- and intra-patient correlations were analysed. Furthermore, the prediction of 177Lu tumour absorbed doses based on a combination of tumour-specific 68Ga activity concentrations and group-based estimates of the effective half-lives for grade 1 and 2 NETs was explored. RESULTS For normal organs, only spleen showed a significant correlation between the 68Ga activity concentration and 177Lu absorbed dose (r = 0.6). For tumours, significant, but moderate, correlations were obtained, with respect to both inter-patient (r = 0.7) and intra-patient (r = 0.45) analyses. The correlations to absorbed doses did not improve when using 68Ga SUVs or normalized SUVs. The relationship between activity uptakes for 68Ga PET and 177Lu SPECT was stronger, with correlation coefficients r = 0.8 for both inter- and intra-patient analyses. The 177Lu absorbed dose to tumour could be predicted from the 68Ga activity concentrations with a 95% coverage interval of - 65% to 248%. CONCLUSIONS On a group level, a high uptake of [68Ga]Ga-DOTA-TATE is associated with high absorbed doses at 177Lu-DOTA-TATE therapy, but the relationship has a limited potential with respect to individual absorbed dose planning. Using SUV or SUV normalized to reference tissues do not improve correlations compared with using activity concentration per injected activity.
Collapse
Affiliation(s)
- Anna Stenvall
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Johan Gustafsson
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden
| | - Erik Larsson
- grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Daniel Roth
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden
| | - Anna Sundlöv
- grid.4514.40000 0001 0930 2361Division of Oncology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Lena Jönsson
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Cecilia Hindorf
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Ohlsson
- grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | |
Collapse
|
7
|
Xue S, Gafita A, Dong C, Zhao Y, Tetteh G, Menze BH, Ziegler S, Weber W, Afshar-Oromieh A, Rominger A, Eiber M, Shi K. Application of machine learning to pretherapeutically estimate dosimetry in men with advanced prostate cancer treated with 177Lu-PSMA I&T therapy. Eur J Nucl Med Mol Imaging 2022; 49:4064-4072. [PMID: 35771265 PMCID: PMC9525373 DOI: 10.1007/s00259-022-05883-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/16/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE Although treatment planning and individualized dose application for emerging prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) are generally recommended, it is still difficult to implement in practice at the moment. In this study, we aimed to prove the concept of pretherapeutic prediction of dosimetry based on imaging and laboratory measurements before the RLT treatment. METHODS Twenty-three patients with metastatic castration-resistant prostate cancer (mCRPC) treated with 177Lu-PSMA I&T RLT were included retrospectively. They had available pre-therapy 68 Ga-PSMA-HEBD-CC PET/CT and at least 3 planar and 1 SPECT/CT imaging for dosimetry. Overall, 43 cycles of 177Lu-PSMA I&T RLT were applied. Organ-based standard uptake values (SUVs) were obtained from pre-therapy PET/CT scans. Patient dosimetry was calculated for the kidney, liver, spleen, and salivary glands using Hermes Hybrid Dosimetry 4.0 from the planar and SPECT/CT images. Machine learning methods were explored for dose prediction from organ SUVs and laboratory measurements. The uncertainty of these dose predictions was compared with the population-based dosimetry estimates. Mean absolute percentage error (MAPE) was used to assess the prediction uncertainty of estimated dosimetry. RESULTS An optimal machine learning method achieved a dosimetry prediction MAPE of 15.8 ± 13.2% for the kidney, 29.6% ± 13.7% for the liver, 23.8% ± 13.1% for the salivary glands, and 32.1 ± 31.4% for the spleen. In contrast, the prediction based on literature population mean has significantly larger MAPE (p < 0.01), 25.5 ± 17.3% for the kidney, 139.1% ± 111.5% for the liver, 67.0 ± 58.3% for the salivary glands, and 54.1 ± 215.3% for the spleen. CONCLUSION The preliminary results confirmed the feasibility of pretherapeutic estimation of treatment dosimetry and its added value to empirical population-based estimation. The exploration of dose prediction may support the implementation of treatment planning for RLT.
Collapse
Affiliation(s)
- Song Xue
- Dept. Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andrei Gafita
- Dept. Nuclear Medicine, Technical University of Munich, Munich, Germany
- Dept. Molecular & Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Chao Dong
- Dept. Electrical Engineering, Technical University of Munich, Munich, Germany
| | - Yu Zhao
- Dept. Informatics, Technical University of Munich, Munich, Germany
| | - Giles Tetteh
- Dept. Informatics, Technical University of Munich, Munich, Germany
| | - Bjoern H Menze
- Dept. Informatics, Technical University of Munich, Munich, Germany
| | - Sibylle Ziegler
- Dept. Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Dept. Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Ali Afshar-Oromieh
- Dept. Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Dept. Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matthias Eiber
- Dept. Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Kuangyu Shi
- Dept. Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Dept. Informatics, Technical University of Munich, Munich, Germany.
| |
Collapse
|
8
|
Wong KK, Frey KA, Niedbala J, Kaza RK, Worden FP, Fitzpatrick KJ, Dewaraja YK. Differences in tumor-to-normal organ SUV ratios measured with 68 Ga-DOTATATE PET compared with 177 Lu-DOTATATE SPECT in patients with neuroendocrine tumors. Nucl Med Commun 2022; 43:892-900. [PMID: 35703269 PMCID: PMC9288505 DOI: 10.1097/mnm.0000000000001592] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Our goal is to quantitatively compare radiotracer biodistributions within tumors and major normal organs on pretherapy 68 Ga-DOTATATE PET to post-therapy 177 Lu-DOTATATE single-photon emission computed tomography (SPECT) in patients receiving peptide receptor radionuclide therapy (PRRT). METHODS PET/CT at ~ 60 min postinjection of Ga-68 DOTATATE and research 177 Lu-SPECT/CT imaging ~ at 4 h (SPECT1) and ~ 24 h (SPECT2) post-cycle#1 were available. Manual contours of lesions on baseline CT or MRI were applied to co-registered SPECT/CT and PET/CT followed by deep learning-based CT auto-segmentation of organs. Tumor-to-normal organ ratios (TNR) were calculated from standardized uptake values (SUV) mean and SUV peak for tumor, and SUV mean for non-tumoral liver (nliver), spleen and kidney. RESULTS There were 90 lesons in 24 patients with progressive metastatic neuroendocrine tumor. The correlation between PET and SPECT SUV TNRs were poor/moderate: PET versus SPECT1 R 2 = 0.19, 0.21, 0.29; PET versus SPECT2 R 2 = 0.06, 0.16, 0.33 for TNR nliver ,TNR spleen ,TNR kidney , respectively. Across all patients, the average value of the TNR measured on PET was significantly lower than on SPECT at both time points ( P < 0.001). Using SUV mean for tumor, average TNR values and 95% confidence intervals (CI) were PET: TNR nliver = 3.5 [CI: 3.0-3.9], TNR spleen = 1.3 [CI, 1.2-1.5], TNR kidney = 1.7 [CI: 1.6-1.9]; SPECT1: TNR nliver = 10 [CI: 8.2-11.7], TNR spleen = 2.9 [CI: 2.5-3.4], TNR kidney = 2.8 [CI: 2.3-3.3]; SPECT2: TNR nliver = 16.9 [CI: 14-19.9], TNR spleen = 3.6 [CI: 3-4.2], TNR kidney = 3.6 [CI: 3.0-4.2]. Comparison of PET and SPECT results in a sphere phantom study demonstrated that these differences are not attributed to imaging modality. CONCLUSIONS Differences in TNR exist for the theranostic pair, with significantly higher SUV TNR on 177 Lu SPECT compared with 68 Ga PET. We postulate this phenomenon is due to temporal differences in DOTATATE uptake and internalization in tumor as compared to normal organs.
Collapse
Affiliation(s)
- Ka Kit Wong
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Kirk A. Frey
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Jeremy Niedbala
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Ravi K. Kaza
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Francis P. Worden
- Department of Endocrine Oncology, University of Michigan, Ann Arbor, Michigan 48109
| | - Kellen J. Fitzpatrick
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Yuni K. Dewaraja
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
9
|
Fodi CK, Schittenhelm J, Honegger J, Castaneda-Vega SG, Behling F. The Current Role of Peptide Receptor Radionuclide Therapy in Meningiomas. J Clin Med 2022; 11:jcm11092364. [PMID: 35566491 PMCID: PMC9104797 DOI: 10.3390/jcm11092364] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/14/2022] [Accepted: 04/20/2022] [Indexed: 02/06/2023] Open
Abstract
Meningiomas are the most common primary intracranial tumors. The majority of patients can be cured by surgery, or tumor growth can be stabilized by radiation. However, the management of recurrent and more aggressive tumors remains difficult because no established alternative treatment options exist. Therefore, innovative therapeutic approaches are needed. Studies have shown that meningiomas express somatostatin receptors. It is well known from treating neuroendocrine tumors that peptide radioreceptor therapy that targets somatostatin receptors can be effective. As yet, this therapy has been used for treating meningiomas only within individual curative trials. However, small case series and studies have demonstrated stabilization of the disease. Therefore, we see potential for optimizing this therapeutic option through the development of new substances and specific adaptations to the different meningioma subtypes. The current review provides an overview of this topic.
Collapse
Affiliation(s)
- Christina-Katharina Fodi
- Department of Neurosurgery and Neurotechnology, University Hospital Tübingen, Eberhard-Karls University, 72076 Tübingen, Germany; (C.-K.F.); (J.H.)
- Center for CNS Tumors, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Eberhard-Karls-University, 72076 Tübingen, Germany;
| | - Jens Schittenhelm
- Center for CNS Tumors, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Eberhard-Karls-University, 72076 Tübingen, Germany;
- Department of Neuropathology, University Hospital Tübingen, Eberhard-Karls University, 72076 Tübingen, Germany
| | - Jürgen Honegger
- Department of Neurosurgery and Neurotechnology, University Hospital Tübingen, Eberhard-Karls University, 72076 Tübingen, Germany; (C.-K.F.); (J.H.)
- Center for CNS Tumors, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Eberhard-Karls-University, 72076 Tübingen, Germany;
| | - Salvador Guillermo Castaneda-Vega
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tübingen, Eberhard-Karls University, 72076 Tübingen, Germany;
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University, 72076 Tübingen, Germany
| | - Felix Behling
- Department of Neurosurgery and Neurotechnology, University Hospital Tübingen, Eberhard-Karls University, 72076 Tübingen, Germany; (C.-K.F.); (J.H.)
- Center for CNS Tumors, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Eberhard-Karls-University, 72076 Tübingen, Germany;
- Correspondence: ; Tel.: +49-707129-80235; Fax: +49-707129-4549
| |
Collapse
|
10
|
Miller C, Rousseau J, Ramogida CF, Celler A, Rahmim A, Uribe CF. Implications of physics, chemistry and biology for dosimetry calculations using theranostic pairs. Theranostics 2022; 12:232-259. [PMID: 34987643 PMCID: PMC8690938 DOI: 10.7150/thno.62851] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/18/2021] [Indexed: 12/15/2022] Open
Abstract
Theranostics is an emerging paradigm that combines imaging and therapy in order to personalize patient treatment. In nuclear medicine, this is achieved by using radiopharmaceuticals that target identical molecular targets for both imaging (using emitted gamma rays) and radiopharmaceutical therapy (using emitted beta, alpha or Auger-electron particles) for the treatment of various diseases, such as cancer. If the therapeutic radiopharmaceutical cannot be imaged quantitatively, a “theranostic pair” imaging surrogate can be used to predict the absorbed radiation doses from the therapeutic radiopharmaceutical. However, theranostic dosimetry assumes that the pharmacokinetics and biodistributions of both radiopharmaceuticals in the pair are identical or very similar, an assumption that still requires further validation for many theranostic pairs. In this review, we consider both same-element and different-element theranostic pairs and attempt to determine if factors exist which may cause inaccurate dose extrapolations in theranostic dosimetry, either intrinsic (e.g. chemical differences) or extrinsic (e.g. injecting different amounts of each radiopharmaceutical) to the radiopharmaceuticals. We discuss the basis behind theranostic dosimetry and present common theranostic pairs and their therapeutic applications in oncology. We investigate general factors that could create alterations in the behavior of the radiopharmaceuticals or the quantitative accuracy of imaging them. Finally, we attempt to determine if there is evidence showing some specific pairs as suitable for theranostic dosimetry. We show that there are a variety of intrinsic and extrinsic factors which can significantly alter the behavior among pairs of radiopharmaceuticals, even if they belong to the same chemical element. More research is needed to determine the impact of these factors on theranostic dosimetry estimates and on patient outcomes, and how to correctly account for them.
Collapse
|
11
|
Abstract
This article summarizes the role of PET imaging for detection, characterization, and theranostic/therapy planning for neuroendocrine tumors. Topics in this article span overall imaging accuracy with mostly 68Ga-DOTA-peptide imaging as well as basic principles of individualized dosimetry. There is also some discussion around further specialized approaches in dosimetry in theranostics. In addition, an overview of the literature on functional imaging in neuroendocrine tumors and the current understanding of imaging-derived clinical outcome prediction are presented.
Collapse
Affiliation(s)
- Rebecca K S Wong
- Radiation Medicine Program, Princess Margaret Cancer Center, University Health Network, 610 University Ave, Toronto, ON M5G 2M9, Canada; Department of Radiation Oncology Temerty Faculty of Medicine, University of Toronto, 149 College Street, Suite 504, Toronto, ON M5T 1P5, Canada
| | - Ur Metser
- Joint Department Medical Imaging, University Health Network, Mount Sinai Hospital & Women's College Hospital, University of Toronto, 610 University Ave, Suite 3-920, Toronto, ON M5G 2M9, Canada; Department of Medical Imaging, University of Toronto, 263 McCaul Street, 4th Floor, Toronto, ON M5T 1W7, Canada
| | - Patrick Veit-Haibach
- Joint Department Medical Imaging, University Health Network, Mount Sinai Hospital & Women's College Hospital, University of Toronto, 610 University Ave, Suite 3-920, Toronto, ON M5G 2M9, Canada; Department of Medical Imaging, University of Toronto, 263 McCaul Street, 4th Floor, Toronto, ON M5T 1W7, Canada.
| |
Collapse
|
12
|
Differences in the expression of SSTR1-5 in meningiomas and its therapeutic potential. Neurosurg Rev 2021; 45:467-478. [PMID: 33899156 PMCID: PMC8827401 DOI: 10.1007/s10143-021-01552-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/03/2021] [Accepted: 04/15/2021] [Indexed: 12/05/2022]
Abstract
Beyond microsurgical resection and radiation therapy, there are currently no established treatment alternatives for meningioma patients. In selected cases, peptide radio receptor therapy (PRRT) can be implemented. For this purpose, a radionuclide is bound to a substance targeting specific receptors in meningiomas. One of them is somatostatin receptor 2, which can be found in most meningiomas. However, other somatostatin receptors (SSTR) exist, but their expressions have only been described in small case series. In this study, we analyzed the expression of SSTR1, 2A, 3, 4, and 5 in a large cohort of meningiomas in order to enable further refinement of this innovative treatment option. Overall, 726 tumor samples were processed into tissue microarrays and stained for SSTR1, 2A, 3, 4, and 5 immunohistochemically. Microscopic evaluation was done with an established semiquantitative score regarding percentual quantification and staining intensity, and results were correlated with clinical data. There was a significant lower rate of SSTR1 expression in meningiomas of male patients. Older age was associated with higher expression of SSTR1, 2A, and 5 and lower scores for SSTR3 and 4. Tumors treated with radiotherapy before resection showed lower rates of SSTR1 and 5 expression, while recurrent meningiomas had lower SSTR1 scores. Tumor tissue from patients suffering from neurofibromatosis type 2 had lower expression scores for SSTR1, 2, and 5. For SSTR3 and 4, NF2 patients showed higher scores than sporadic tumors. Spinal meningiomas had higher scores for SSTR1, 4, and 5 compared tumor location of the skull base and convexity/falx. Overall, higher WHO grade was associated with lower SSTR scores. While all SSTRs were expressed, there are marked differences of SSTR expression between meningioma subgroups. This has the potential to drive the development of more selective PRRT substances with higher treatment efficacy.
Collapse
|
13
|
Mirian C, Duun-Henriksen AK, Maier A, Pedersen MM, Jensen LR, Bashir A, Graillon T, Hrachova M, Bota D, van Essen M, Spanjol P, Kreis C, Law I, Broholm H, Poulsgaard L, Fugleholm K, Ziebell M, Munch T, Walter MA, Mathiesen T. Somatostatin Receptor-Targeted Radiopeptide Therapy in Treatment-Refractory Meningioma: Individual Patient Data Meta-analysis. J Nucl Med 2021; 62:507-513. [PMID: 32859705 DOI: 10.2967/jnumed.120.249607] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/29/2020] [Indexed: 11/16/2022] Open
Abstract
Somatostatin receptor (SSTR)-targeted peptide receptor radionuclide therapy (PRRT) represents a promising approach for treatment-refractory meningiomas. Methods: We performed an individual patient data meta-analysis, including all published data on meningioma patients treated with SSTR-targeted PRRT. The main outcomes were toxicity, response to treatment, progression-free survival (PFS), and overall survival (OS). We applied the Kaplan-Meier method to estimate survival probabilities and report incidence rates per 100 person-years. We applied Cox proportional hazards models to determine the effect of covariates. Results: We screened 537 papers and identified 6 eligible cohort studies. We included a total of 111 patients who had treatment-refractory meningioma and received SSTR-targeted PRRT. Disease control was achieved in 63% of patients. The 6-mo PFS rates were 94%, 48%, and 0% for World Health Organization grades I, II, and III, respectively. The risk of disease progression decreased by 13% per 1,000-MBq increase in the total applied activity. The 1-y OS rates were 88%, 71%, and 52% for World Health Organization grades I, II, and III, respectively. The risk of death decreased by 17% per 1,000-MBq increase in the total applied activity. The main side effects comprised transient hematotoxicity, such as anemia in 22% of patients, leukopenia in 13%, lymphocytopenia in 24%, and thrombocytopenia in 17%. Conclusion: To our knowledge, this individual patient data meta-analysis represents the most comprehensive analysis of the benefits of and adverse events associated with SSTR-targeted PRRT for treatment-refractory meningioma. The treatment was well tolerated, achieved disease control in most cases, and showed promising results regarding PFS and OS.
Collapse
Affiliation(s)
- Christian Mirian
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Nuclear Medicine, University Hospital of Geneva, Geneva, Switzerland
| | | | - Andrea Maier
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Maria Møller Pedersen
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lasse Rehné Jensen
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Asma Bashir
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas Graillon
- APHM, Department of Neurosurgery, La Timone Hospital, Marseille, France
| | - Maya Hrachova
- Department of Neurology, UC Irvine Medical Center, Irvine, California
| | - Daniela Bota
- Department of Neurology, UC Irvine Medical Center, Irvine, California
- Department of Neurosurgery, UC Irvine Medical Center, Irvine, California
| | - Martjin van Essen
- Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Petar Spanjol
- Department of Nuclear Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Christian Kreis
- Department of Nuclear Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Copenhagen, Denmark
| | - Helle Broholm
- Department of Neuropathology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lars Poulsgaard
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kåre Fugleholm
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Morten Ziebell
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Tina Munch
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark; and
| | - Martin A Walter
- Department of Nuclear Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Tiit Mathiesen
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
14
|
Galldiks N, Langen KJ, Albert NL, Chamberlain M, Soffietti R, Kim MM, Law I, Le Rhun E, Chang S, Schwarting J, Combs SE, Preusser M, Forsyth P, Pope W, Weller M, Tonn JC. PET imaging in patients with brain metastasis-report of the RANO/PET group. Neuro Oncol 2020; 21:585-595. [PMID: 30615138 DOI: 10.1093/neuonc/noz003] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/11/2018] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Brain metastases (BM) from extracranial cancer are associated with significant morbidity and mortality. Effective local treatment options are stereotactic radiotherapy, including radiosurgery or fractionated external beam radiotherapy, and surgical resection. The use of systemic treatment for intracranial disease control also is improving. BM diagnosis, treatment planning, and follow-up is most often based on contrast-enhanced magnetic resonance imaging (MRI). However, anatomic imaging modalities including standard MRI have limitations in accurately characterizing posttherapeutic reactive changes and treatment response. Molecular imaging techniques such as positron emission tomography (PET) characterize specific metabolic and cellular features of metastases, potentially providing clinically relevant information supplementing anatomic MRI. Here, the Response Assessment in Neuro-Oncology working group provides recommendations for the use of PET imaging in the clinical management of patients with BM based on evidence from studies validated by histology and/or clinical outcome.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University Hospital Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine 3, 4, Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology, Universities of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine 3, 4, Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany
| | - Marc Chamberlain
- Departments of Neurology and Neurological Surgery, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Denmark
| | - Emilie Le Rhun
- Department of Neurosurgery, University Hospital Lille, Lille, France
| | - Susan Chang
- Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Julian Schwarting
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Technical University Munich, Munich, Germany
| | - Matthias Preusser
- Department of Medicine I and Comprehensive Cancer Centre CNS Tumours Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Forsyth
- Moffitt Cancer Center, University of South Florida, Tampa, Florida, USA
| | - Whitney Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California , USA
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jörg C Tonn
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Germany
| |
Collapse
|
15
|
Brastianos PK, Galanis E, Butowski N, Chan JW, Dunn IF, Goldbrunner R, Herold-Mende C, Ippen FM, Mawrin C, McDermott MW, Sloan A, Snyder J, Tabatabai G, Tatagiba M, Tonn JC, Wen PY, Aldape K, Nassiri F, Zadeh G, Jenkinson MD, Raleigh DR. Advances in multidisciplinary therapy for meningiomas. Neuro Oncol 2020; 21:i18-i31. [PMID: 30649489 DOI: 10.1093/neuonc/noy136] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Surgery has long been established as the first-line treatment for the majority of symptomatic and enlarging meningiomas, and evidence for its success is derived from retrospective case series. Despite surgical resection, a subset of meningiomas display aggressive behavior with early recurrences that are difficult to treat. The decision to radically resect meningiomas and involved structures is balanced against the risk for neurological injury in patients. Radiation therapy has largely been used as a complementary and safe therapeutic strategy in meningiomas with evidence primarily stemming from retrospective, single-institution reports. Two of the first cooperative group studies (RTOG 0539 and EORTC 22042) evaluating the outcomes of adjuvant radiation therapy in higher-risk meningiomas have shown promising preliminary results. Historically, systemic therapy has resulted in disappointing results in meningiomas. However, several clinical trials are under way evaluating the efficacy of chemotherapies, such as trabectedin, and novel molecular agents targeting Smoothened, AKT1, and focal adhesion kinase in patients with recurrent meningiomas.
Collapse
Affiliation(s)
- Priscilla K Brastianos
- Divisions of Hematology/Oncology & Neuro-Oncology, Departments of Medicine & Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evanthia Galanis
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Jason W Chan
- Department of Radiation Oncology, University of California, San Francisco, California, USA
| | - Ian F Dunn
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Roland Goldbrunner
- Department of General Neurosurgery, University Hospital Cologne, Cologne, Germany
| | | | - Franziska M Ippen
- Divisions of Hematology/Oncology & Neuro-Oncology, Departments of Medicine & Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Christian Mawrin
- Institute of Neuropathology, Otto-von-Guericke University, Magdeburg, Germany
| | - Michael W McDermott
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Andrew Sloan
- Department of Neurological Surgery, University Hospital-Case Medical Center, Cleveland, Ohio, USA
| | - James Snyder
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Ghazaleh Tabatabai
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research & Centre for CNS Tumors, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
| | - Marcos Tatagiba
- Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | - Joerg C Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Farshad Nassiri
- MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Division of Neurosurgery, University Health Network, University of Toronto, Ontario, Canada.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Michael D Jenkinson
- Department of Neurosurgery & Institute of Translational Medicine, The Walton Centre NHS Foundation Trust & University of Liverpool, Lower Lane, Liverpool, Merseyside, UK
| | - David R Raleigh
- Department of Neurological Surgery, University of California, San Francisco, California, USA.,Department of Radiation Oncology, University of California, San Francisco, California, USA
| | | |
Collapse
|
16
|
Huang RY, Bi WL, Griffith B, Kaufmann TJ, la Fougère C, Schmidt NO, Tonn JC, Vogelbaum MA, Wen PY, Aldape K, Nassiri F, Zadeh G, Dunn IF. Imaging and diagnostic advances for intracranial meningiomas. Neuro Oncol 2020; 21:i44-i61. [PMID: 30649491 DOI: 10.1093/neuonc/noy143] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The archetypal imaging characteristics of meningiomas are among the most stereotypic of all central nervous system (CNS) tumors. In the era of plain film and ventriculography, imaging was only performed if a mass was suspected, and their results were more suggestive than definitive. Following more than a century of technological development, we can now rely on imaging to non-invasively diagnose meningioma with great confidence and precisely delineate the locations of these tumors relative to their surrounding structures to inform treatment planning. Asymptomatic meningiomas may be identified and their growth monitored over time; moreover, imaging routinely serves as an essential tool to survey tumor burden at various stages during the course of treatment, thereby providing guidance on their effectiveness or the need for further intervention. Modern radiological techniques are expanding the power of imaging from tumor detection and monitoring to include extraction of biologic information from advanced analysis of radiological parameters. These contemporary approaches have led to promising attempts to predict tumor grade and, in turn, contribute prognostic data. In this supplement article, we review important current and future aspects of imaging in the diagnosis and management of meningioma, including conventional and advanced imaging techniques using CT, MRI, and nuclear medicine.
Collapse
Affiliation(s)
- Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Wenya Linda Bi
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brent Griffith
- Department of Radiology, Henry Ford Health System, Detroit, Michigan, USA
| | - Timothy J Kaufmann
- Department of Radiology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tubingen, Tubingen, Germany
| | - Nils Ole Schmidt
- Department of Neurosurgery, University Medical Center, Hamburg-Eppendorf, Germany
| | - Jöerg C Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael A Vogelbaum
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kenneth Aldape
- Department of Laboratory Pathology, National Cancer Institute, National Institute of Health, Bethesda, Maryland, USA.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Farshad Nassiri
- Division of Neurosurgery, University Health Network, University of Toronto, Ontario, Canada.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Division of Neurosurgery, University Health Network, University of Toronto, Ontario, Canada.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Ian F Dunn
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
17
|
Castillo Seoane D, de Saint-Hubert M, Crabbe M, Struelens L, Koole M. Targeted alpha therapy: a critical review of translational dosimetry research with emphasis on actinium-225. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:265-277. [PMID: 32441067 DOI: 10.23736/s1824-4785.20.03266-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This review provides a general overview of the current achievements and challenges in translational dosimetry for targeted alpha therapy (TAT). The concept of targeted radionuclide therapy (TRNT) is described with an overview of its clinical applicability and the added value of TAT is discussed. For TAT, we focused on actinium-225 (225Ac) as an example for alpha particle emitting radionuclides and their features, such as limited range within tissue and high linear energy transfer, which make alpha particle emissions more effective in targeted killing of tumour cells compared to beta radiation. Starting with the state-of-the-art dosimetry for TRNT and TAT, we then describe the challenges that still need to be met in order to move to a personalized dosimetry approach for TAT. Specifically for 225Ac, we discuss the recoiled daughter effect which may provoke significant damage to healthy tissue or organs and should be considered. Next, a broad overview is given of the pre-clinical research on 225Ac-TAT with an extensive description of tools which are only available in a pre-clinical setting and their added value. In addition, we review the preclinical biodistribution and dosimetry studies that have been performed on TAT-agents and more specifically of 225Ac and its multiple progeny, and describe their potential role to better characterize the pharmacokinetic (PK) profile of TAT-agents and to optimize the use of theranostic approaches for dosimetry. Finally, we discuss the support pre-clinical studies may provide in understanding dose-effect relationships, linking radiation dose quantities to biological endpoints and even moving away from macro- to microdosimetry. As such, the translation of pre-clinical findings may provide valuable information and new approaches for improved clinical dosimetry, thus paving the way to personalized TAT.
Collapse
Affiliation(s)
- Dayana Castillo Seoane
- Unit of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit Leuven (KUL), Leuven, Belgium - .,Research Unit in Dosimetric Applications, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium -
| | - Marijke de Saint-Hubert
- Research Unit in Dosimetric Applications, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| | - Melissa Crabbe
- Research Unit in Dosimetric Applications, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| | - Lara Struelens
- Research Unit in Dosimetric Applications, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| | - Michel Koole
- Unit of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit Leuven (KUL), Leuven, Belgium
| |
Collapse
|
18
|
Pharmacokinetic analysis of [ 68Ga]Ga-DOTA-TOC PET in meningiomas for assessment of in vivo somatostatin receptor subtype 2. Eur J Nucl Med Mol Imaging 2020; 47:2577-2588. [PMID: 32170347 DOI: 10.1007/s00259-020-04759-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/04/2020] [Indexed: 01/10/2023]
Abstract
PURPOSE DOTA-D-Phe1-Tyr3-octreotide with gallium-68 ([68Ga]Ga-DOTA-TOC) is one of the PET tracers that forms the basis for peptide receptor radionuclide therapy based on somatostatin receptor subtype 2 (SSTR2) expression in meningiomas. Yet, the quantitative relationship between [68Ga]Ga-DOTA-TOC accumulation and SSTR2 is unknown. We conducted a correlative analysis of a range of [68Ga]Ga-DOTA-TOC PET metric(s) as imaging surrogate(s) of the receptor binding in meningiomas by correlating the PET results with SSTR2 expression from surgical specimens. We additionally investigated possible influences of secondary biological factors such as vascularization, inflammation and proliferation. METHODS Fifteen patients with MRI-presumed or recurrent meningiomas underwent a 60-min dynamic [68Ga]Ga-DOTA-TOC PET/CT before surgery. The PET data comprised maximum and mean standardized uptake values (SUVmax, SUVmean) with and without normalization to reference regions, and quantitative measurements derived from kinetic modelling using a reversible two-tissue compartment model with the fractional blood volume (VB). Expressions of SSTR2 and proliferation (Ki-67, phosphohistone-H3, proliferating cell nuclear antigen) were determined by immunohistochemistry and/or quantitative polymerase chain reaction (qPCR), while biomarkers of vascularization (vascular endothelial growth factor A (VEGFA), endothelial marker CD34) and inflammation (cytokine interleukin-18, microglia/macrophage-specific marker CD68) by qPCR. RESULTS Histopathology revealed 12 World Health Organization (WHO) grade I and three WHO grade II meningiomas showing no link to SSTR2. The majority of [68Ga]Ga-DOTA-TOC PET metrics showed significant associations with SSTR2 protein, while all PET metrics were positively correlated with SSTR2 mRNA with the best results for mean tumour-to-blood ratio (TBRmean) (r = 0.757, P = 0.001) and SUVmean (r = 0.714, P = 0.003). Significant positive correlations were also found between [68Ga]Ga-DOTA-TOC PET metrics, and VEGFA and VB. SSTR2 mRNA was moderately correlated with VEGFA (r = 0.539, P = 0.038). Neither [68Ga]Ga-DOTA-TOC PET metrics nor SSTR2 were correlated with proliferation or inflammation. CONCLUSION [68Ga]Ga-DOTA-TOC accumulation in meningiomas is associated with SSTR2 binding and vascularization with TBRmean being the best PET metric for assessing SSTR2.
Collapse
|
19
|
Pfister D, Drude N, Mottaghy F, Behrendt F, Verburg F. PSA levels, PSA doubling time, Gleason score and prior therapy cannot predict measured uptake of [68Ga]PSMA-HBED-CC lesion uptake in recurrent/metastatic prostate cancer. Nuklearmedizin 2018; 56:225-232. [DOI: 10.3413/nukmed-0917-17-07] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
SummaryAim: To assess whether clinical prostate cancer (PCA) related factors and therapy status can predict the degree of tracer uptake on [68Ga]PSMA-HBED-CC PET/CT.Materials & methods: We retrospectively studied 124 patients with recurrent an/or metastatic PCA who underwent [68Ga]PSMAHBED-CC PET/CT. The maximum standardized uptake value (SUVmax) was determined in the prostate bed as well as in three size categories (≤ 5 mm, > 5–15 mm, > 15 mm) in pelvic lymph node, extrapelvic lymph node, bone and visceral metastases.Results: Significant positive correlations between lesion size and SUVmax were found in pelvic lymph node metastases > 5 -≤15 mm (Spearmans rho = 0.502, p = 0.002) as well as in extrapelvic lymph node metastases5 mm (rho = 0.314, p = 0.033) and > 5 ≤-15 mm (rho = 0.614, p < 0.001). SUVmax tended to be higher in the largest diameter category in each anatomic station than in the middle and lower categories. We were unable to find evidence for a relationship between SUVmax and PSA, PSAdt, Gleason score, androgen deprivation therapy, radiation therapy or chemotherapy status.Conclusion: Measured tracer uptake in [68Ga]PSMA-HBED-CC PET/CT in patients with recurrent/metastasized prostate cancer is significantly influenced by lesion size as a result of partial volume effects in the very small lesions. Clinical indicators of aggressive prostate cancer behaviour such as PSA levels, PSA doubling time or the Gleason score of the primary tumour, as well as the androgen deprivation therapy, radiation therapy or chemotherapy status are not related to measured tracer uptake.
Collapse
|
20
|
Hardiansyah D, Guo W, Attarwala AA, Kletting P, Mottaghy FM, Glatting G. Treatment planning in PRRT based on simulated PET data and a PBPK model. Nuklearmedizin 2018; 56:23-30. [DOI: 10.3413/nukmed-0819-16-04] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 11/07/2016] [Indexed: 11/20/2022]
Abstract
SummaryAim: To investigate the accuracy of treatment planning in peptide-receptor radionuclide therapy (PRRT) based on simulated PET data (using a PET noise model) and a physiologically based pharmacokinetic (PBPK) model. Methods: The parameters of a PBPK model were fitted to the biokinetic data of 15 patients. True mathematical phantoms of patients (MPPs) were the PBPK model with the fitted parameters. PET measurements after bolus injection of 150 MBq 68Ga-DOTATATE were simulated for the true MPPs. PET noise with typical noise levels was added to the data (i.e. c=0.3 [low], 3, 30 and 300 [high]). Organ activity data in the kidneys, tumour, liver and spleen were simulated at 0.5, 1 and 4 h p.i. PBPK model parameters were fitted to the simulated noisy PET data to derive the PET-predicted MPPs. Therapy was simulated assuming an infusion of 3.3 GBq of 90Y-DOTATATE over 30 min. Time-integrated activity coefficients (TIACs) of simulated therapy in tumour, kidneys, liver, spleen and remainder were calculated from both, true MPPs (true TIACs) and predicted MPPs (predicted TIACs). Variability v between true TIACs and predicted TIACs were calculated and analysed. Variability< 10 % was considered to be an accurate prediction. Results: For all noise level, variabilities for the kidneys, liver, and spleen showed an accurate prediction for TIACs, e.g. c=300: vkidney=(5 ± 2)%, vliver=(5 ± 2)%, vspleen=(4 ± 2)%. However, tumour TIAC predictions were not accurate for all noise levels, e.g. c=0.3: vtumour=(8 ± 5)%. Conclusion: PET based treatment planning with kidneys as the dose limiting organ seems possible for all reported noise levels using an adequate PBPK model and previous knowledge about the individual patient.
Collapse
|
21
|
Yamaguchi A, Hanaoka H, Higuchi T, Tsushima Y. Radiolabeled (4-Fluoro-3-Iodobenzyl)Guanidine Improves Imaging and Targeted Radionuclide Therapy of Norepinephrine Transporter–Expressing Tumors. J Nucl Med 2017; 59:815-821. [DOI: 10.2967/jnumed.117.201525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022] Open
|
22
|
Eberlein U, Cremonesi M, Lassmann M. Individualized Dosimetry for Theranostics: Necessary, Nice to Have, or Counterproductive? J Nucl Med 2017; 58:97S-103S. [PMID: 28864620 DOI: 10.2967/jnumed.116.186841] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/10/2017] [Indexed: 11/16/2022] Open
Abstract
In 2005, the term theragnostics (theranostics) was introduced for describing the use of imaging for therapy planning in radiation oncology. In nuclear medicine, this expression describes the use of tracers for predicting the absorbed doses in molecular radiotherapy and, thus, the safety and efficacy of a treatment. At present, the most successful groups of isotopes for this purpose are 123I/124I/131I, 68Ga/177Lu, and 111In/86Y/90Y. The purpose of this review is to summarize available data on the dosimetry and dose-response relationships of several theranostic compounds, with a special focus on radioiodine therapy for differentiated thyroid cancer and peptide receptor radionuclide therapy. These are treatment modalities for which dose-response relationships for healthy tissues and tumors have been demonstrated. In addition, available data demonstrate that posttherapeutic dosimetry after a first treatment cycle predicts the absorbed doses in further cycles. Both examples show the applicability of the concept of theranostics in molecular radiotherapies. Nevertheless, unanswered questions need to be addressed in clinical trials incorporating dosimetry-related concepts for determining the amount of therapeutic activity to be administered.
Collapse
Affiliation(s)
- Uta Eberlein
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Germany; and
| | - Marta Cremonesi
- Radiation Research Unit, Istituto Europeo di Oncologia, Milano, Italy
| | - Michael Lassmann
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Germany; and
| |
Collapse
|
23
|
Hardiansyah D, Attarwala AA, Kletting P, Mottaghy FM, Glatting G. Prediction of time-integrated activity coefficients in PRRT using simulated dynamic PET and a pharmacokinetic model. Phys Med 2017; 42:298-304. [PMID: 28739143 DOI: 10.1016/j.ejmp.2017.06.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 10/19/2022] Open
Abstract
PURPOSE To investigate the accuracy of predicted time-integrated activity coefficients (TIACs) in peptide-receptor radionuclide therapy (PRRT) using simulated dynamic PET data and a physiologically based pharmacokinetic (PBPK) model. METHODS PBPK parameters were estimated using biokinetic data of 15 patients after injection of (152±15)MBq of 111In-DTPAOC (total peptide amount (5.78±0.25)nmol). True mathematical phantoms of patients (MPPs) were the PBPK model with the estimated parameters. Dynamic PET measurements were simulated as being done after bolus injection of 150MBq 68Ga-DOTATATE using the true MPPs. Dynamic PET scans around 35min p.i. (P1), 4h p.i. (P2) and the combination of P1 and P2 (P3) were simulated. Each measurement was simulated with four frames of 5min each and 2 bed positions. PBPK parameters were fitted to the PET data to derive the PET-predicted MPPs. Therapy was simulated assuming an infusion of 5.1GBq of 90Y-DOTATATE over 30min in both true and PET-predicted MPPs. TIACs of simulated therapy were calculated, true MPPs (true TIACs) and predicted MPPs (predicted TIACs) followed by the calculation of variabilities v. RESULTS For P1 and P2 the population variabilities of kidneys, liver and spleen were acceptable (v<10%). For the tumours and the remainders, the values were large (up to 25%). For P3, population variabilities for all organs including the remainder further improved, except that of the tumour (v>10%). CONCLUSION Treatment planning of PRRT based on dynamic PET data seems possible for the kidneys, liver and spleen using a PBPK model and patient specific information.
Collapse
Affiliation(s)
- Deni Hardiansyah
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Electrical Engineering, Universitas Padjadjaran, Bandung, Indonesia
| | - Ali Asgar Attarwala
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Kletting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Felix M Mottaghy
- Klinik für Nuklearmedizin, University Hospital, RWTH Aachen University, Aachen, Germany; Department of Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Gerhard Glatting
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| |
Collapse
|
24
|
Hardiansyah D, Guo W, Kletting P, Mottaghy FM, Glatting G. Time-integrated activity coefficient estimation for radionuclide therapy using PET and a pharmacokinetic model: A simulation study on the effect of sampling schedule and noise. Med Phys 2017; 43:5145. [PMID: 27587044 DOI: 10.1118/1.4961012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE The aim of this study was to investigate the accuracy of PET-based treatment planning for predicting the time-integrated activity coefficients (TIACs). METHODS The parameters of a physiologically based pharmacokinetic (PBPK) model were fitted to the biokinetic data of 15 patients to derive assumed true parameters and were used to construct true mathematical patient phantoms (MPPs). Biokinetics of 150 MBq (68)Ga-DOTATATE-PET was simulated with different noise levels [fractional standard deviation (FSD) 10%, 1%, 0.1%, and 0.01%], and seven combinations of measurements at 30 min, 1 h, and 4 h p.i. PBPK model parameters were fitted to the simulated noisy PET data using population-based Bayesian parameters to construct predicted MPPs. Therapy simulations were performed as 30 min infusion of (90)Y-DOTATATE of 3.3 GBq in both true and predicted MPPs. Prediction accuracy was then calculated as relative variability vorgan between TIACs from both MPPs. RESULTS Large variability values of one time-point protocols [e.g., FSD = 1%, 240 min p.i., vkidneys = (9 ± 6)%, and vtumor = (27 ± 26)%] show inaccurate prediction. Accurate TIAC prediction of the kidneys was obtained for the case of two measurements (1 and 4 h p.i.), e.g., FSD = 1%, vkidneys = (7 ± 3)%, and vtumor = (22 ± 10)%, or three measurements, e.g., FSD = 1%, vkidneys = (7 ± 3)%, and vtumor = (22 ± 9)%. CONCLUSIONS (68)Ga-DOTATATE-PET measurements could possibly be used to predict the TIACs of (90)Y-DOTATATE when using a PBPK model and population-based Bayesian parameters. The two time-point measurement at 1 and 4 h p.i. with a noise up to FSD = 1% allows an accurate prediction of the TIACs in kidneys.
Collapse
Affiliation(s)
- Deni Hardiansyah
- Medical Radiation Physics/Radiation Protection, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, Mannheim 68167, Germany and Department of Radiation Oncology, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Wei Guo
- Medical Radiation Physics/Radiation Protection, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Peter Kletting
- Department of Nuclear Medicine, Ulm University, Ulm 89081, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital, RWTH Aachen University, Aachen 52074, Germany and Department of Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht 6229, The Netherlands
| | - Gerhard Glatting
- Medical Radiation Physics/Radiation Protection, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, Mannheim 68167, Germany
| |
Collapse
|
25
|
Abstract
This article provides an overview of the key considerations for the development and application of molecular imaging agents for brain tumors and the major classes of PET tracers that have been used for imaging brain tumors in humans. The mechanisms of uptake, biological implications, primary applications, and limitations of PET tracers in neuro-oncology are reviewed. The available data indicate that several of these classes of tracers, including radiolabeled amino acids, have imaging properties superior to those of (18)F-fluorodeoxyglucose, and can complement contrast-enhanced magnetic resonance imaging in the evaluation of brain tumors.
Collapse
|
26
|
Seystahl K, Stoecklein V, Schüller U, Rushing E, Nicolas G, Schäfer N, Ilhan H, Pangalu A, Weller M, Tonn JC, Sommerauer M, Albert NL. Somatostatin receptor-targeted radionuclide therapy for progressive meningioma: benefit linked to 68Ga-DOTATATE/-TOC uptake. Neuro Oncol 2016; 18:1538-1547. [PMID: 27106404 DOI: 10.1093/neuonc/now060] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 03/16/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The prognosis of patients with progressive meningioma after failure of surgery and radiotherapy is poor. METHODS We retrospectively evaluated the safety and efficacy of somatostatin-receptor (SSTR)-targeted radionuclide therapy (177Lu-DOTATATE [n = 16], 90Y-DOTATOC [n = 3], or both [n = 1]) in patients with progressive, treatment-refractory meningiomas (5 World Health Organization [WHO] grade I, 7 WHO grade II, 8 WHO grade III) and in part multifocal disease (17 of 20 patients). RESULTS SSTR radionuclide treatment (median of 3 treatment cycles, median administered dose/cycle 7400 MBq) led to a disease stabilization in 10 of 20 patients for a median time of 17 months. Stratification according to WHO grade showed a median progression-free survival (PFS) of 32.2 months for grade I tumors, 7.2 for grade II, and 2.1 for grade III. PFS at 6 months was 100% for grade I, 57% for grade II, and 0% for grade III. Median overall survival was 17.2 months in WHO grade III patients and not reached for WHO I and II at a median follow-up of 20 months. In the analysis of single meningioma lesions, maximal and mean standardized uptake values in pretherapeutic 68Ga-DOTATOC/-TATE PET/CT were significantly higher in those lesions with radiographic stability after 6 months. In line with this, high expression of SSTR via immunohistochemistry was associated with PFS >6 months. CONCLUSIONS SSTR-targeted radionuclide treatment has activity in a subset of patients with meningioma. Expression of SSTR via immunohistochemistry or radionuclide uptake might serve as a predictive biomarker for outcome to facilitate individualized treatment optimization in patients with uni- and multifocal meningiomas.
Collapse
Affiliation(s)
- Katharina Seystahl
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Veit Stoecklein
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Ulrich Schüller
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Elisabeth Rushing
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Guillaume Nicolas
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Niklaus Schäfer
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Harun Ilhan
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Athina Pangalu
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Michael Weller
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Jörg-Christian Tonn
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Michael Sommerauer
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Nathalie L Albert
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| |
Collapse
|
27
|
Abstract
One early application of PET/MRI in clinical practice may be the imaging of head and neck cancers. This is because the morphologic imaging modalities, CT and MR, are recognized as similarly effective tools in cross-sectional oncological imaging of the head and neck. The addition of PET with FDG is believed to enhance the accuracy of both modalities to a similar degree. However, there are a few specific scenarios in head and neck cancer imaging where MR is thought to provide an edge over CT, including perineural spread of tumors and the infiltration of important anatomical landmarks, such as the prevertebral fascia and great vessel walls. Here, hybrid PET/MR might provide higher diagnostic certainty than PET/CT or a separate acquisition of PET/CT and MR. Another advantage of MR is the availability of several functional techniques. Although some of them might enhance the imaging of head and neck cancer with PET/MR, other functional techniques actually might prove dispensable in the presence of PET. In this overview, we discuss current trends and potential clinical applications of PET/MR in the imaging of head and neck cancers, including clinical protocols. We also discuss potential benefits of implementing functional MR techniques into hybrid PET/MRI of head and neck cancers.
Collapse
Affiliation(s)
- Marcelo A Queiroz
- Research and Education Institute, Hospital Sirio-Libanes, Sao Paulo, Brazil; Department of Radiology, Cancer Institute, Hospital das Clinicas/University of Sao Paulo, Sao Paulo, Brazil
| | - Martin W Huellner
- Research and Education Institute, Hospital Sirio-Libanes, Sao Paulo, Brazil; Department of Medical Radiology, Divisions of Nuclear Medicine and Neuroradiology,University Hospital Zurich/University of Zurich, Zurich, Switzerland.
| |
Collapse
|
28
|
Sommerauer M, Burkhardt JK, Frontzek K, Rushing E, Buck A, Krayenbuehl N, Weller M, Schaefer N, Kuhn FP. 68Gallium-DOTATATE PET in meningioma: A reliable predictor of tumor growth rate? Neuro Oncol 2016; 18:1021-7. [PMID: 26865086 DOI: 10.1093/neuonc/now001] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/03/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND DOTATATE-based radionuclides have added new options in the diagnosis and treatment of meningiomas; however, a reliable predictor of tumor growth has still not been established. METHODS We analyzed 64 meningiomas imaged with (68)Ga-DOTATATE PET. Tumor growth rates were calculated by volumetric analysis of sequential MRI scans. Maximums of standardized uptake values (SUVmax) were correlated with tumor growth and covariates. RESULTS World Health Organization (WHO) grades I and II meningiomas showed a correlation of SUVmax and tumor growth rate (meningiomas limited to the intracranial compartment: r = 0.757, P < .001, and transosseous growing meningiomas: r = 0.819, P = .024). SUVmax was significantly higher and the slope of the linear regression significantly steeper in transosseous compared with intracranial meningiomas (both P < .001). The association remained significant in multivariate analysis, and the prediction of tumor growth rate was independent of WHO grade. Anaplastic meningiomas showed no significant correlation of SUVmax and tumor growth. CONCLUSIONS (68)Ga-DOTATATE PET is a reliable predictor of tumor growth in WHO grades I and II meningiomas and provides additional information to conventional cross-sectional imaging modalities. Hence, (68)Ga-DOTATATE PET can assist in selecting the time point for treatment initiation. Furthermore, meningiomas with fast tumor growth and transosseous expansion elicit the highest DOTATATE binding; therefore, they might be especially suited for DOTATATE-based therapy.
Collapse
Affiliation(s)
- Michael Sommerauer
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| | - Jan-Karl Burkhardt
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| | - Karl Frontzek
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| | - Elisabeth Rushing
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| | - Niklaus Krayenbuehl
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| | - Michael Weller
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| | - Niklaus Schaefer
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| | - Felix P Kuhn
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (M.S., A.B., N.S., F.P.K.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.S., M.W.); Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland (J.-K.B., N.K.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (K.F., E.R.); Department of Medical Oncology, University Hospital Zurich, Zurich, Switzerland (N.S.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (F.P.K.)
| |
Collapse
|
29
|
Hardiansyah D, Maass C, Attarwala AA, Müller B, Kletting P, Mottaghy FM, Glatting G. The role of patient-based treatment planning in peptide receptor radionuclide therapy. Eur J Nucl Med Mol Imaging 2015; 43:871-880. [PMID: 26577941 DOI: 10.1007/s00259-015-3248-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/30/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Accurate treatment planning is recommended in peptide-receptor radionuclide therapy (PRRT) to minimize the toxicity to organs at risk while maximizing tumor cell sterilization. The aim of this study was to quantify the effect of different degrees of individualization on the prediction accuracy of individual therapeutic biodistributions in patients with neuroendocrine tumors (NETs). METHODS A recently developed physiologically based pharmacokinetic (PBPK) model was fitted to the biokinetic data of 15 patients with NETs after pre-therapeutic injection of (111)In-DTPAOC. Mathematical phantom patients (MPP) were defined using the assumed true (true MPP), mean (MPP 1A) and median (MPP 1B) parameter values of the patient group. Alterations of the degree of individualization were introduced to both mean and median patients by including patient-specific information as a priori knowledge: physical parameters and hematocrit (MPP 2A/2B). Successively, measurable individual biokinetic parameters were added: tumor volume V tu (MPP 3A/3B), glomerular filtration rate GFR (MPP 4A/4B), and tumor perfusion f tu (MPP 5A/5B). Furthermore, parameters of MPP 5A/5B and a simulated (68)Ga-DOTATATE PET measurement 60 min p.i. were used together with the population values used as Bayesian parameters (MPP 6A/6B). Therapeutic biodistributions were simulated assuming an infusion of (90)Y-DOTATATE (3.3 GBq) over 30 min to all MPPs. Time-integrated activity coefficients were predicted for all MPPs and compared to the true MPPs for each patient in tumor, kidneys, spleen, liver, remainder, and whole body to obtain the relative differences RD. RESULTS The large RD values of MPP 1A [RDtumor = (625 ± 1266)%, RDkidneys = (11 ± 38)%], and MPP 1B [RDtumor = (197 ± 505)%, RDkidneys = (11 ± 39)%] demonstrate that individual treatment planning is needed due to large physiological differences between patients. Although addition of individual patient parameters reduced the deviations considerably [MPP 5A: RDtumor = (-2 ± 27)% and RDkidneys = (16 ± 43)%; MPP 5B: RDtumor = (2 ± 28)% and RDkidneys = (7 ± 40)%] errors were still large. For the kidneys, prediction accuracy was considerably improved by including the PET measurement [MPP 6A/MPP 6B: RDtumor = (-2 ± 22)% and RDkidneys = (-0.1 ± 0.5)%]. CONCLUSION Individualized treatment planning is needed in the investigated patient group. The use of a PBPK model and the inclusion of patient specific data, e.g., weight, tumor volume, and glomerular filtration rate, do not suffice to predict the therapeutic biodistribution. Integrating all available a priori information in the PBPK model and using additionally PET data measured at one time point for tumor, kidneys, spleen, and liver could possibly be sufficient to perform an individualized treatment planning.
Collapse
Affiliation(s)
- Deni Hardiansyah
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian Maass
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ali Asgar Attarwala
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Berthold Müller
- Klinik für Nuklearmedizin, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Peter Kletting
- Klinik für Nuklearmedizin, Universität Ulm, Ulm, Germany
| | - Felix M Mottaghy
- Klinik für Nuklearmedizin, University Hospital, RWTH Aachen University, Aachen, Germany.,Department of Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Gerhard Glatting
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Brain tumors differ in histology, biology, prognosis and treatment options. Although structural magnetic resonance is still the gold standard for morphological tumor characterization, molecular imaging has gained an increasing importance in assessment of tumor activity and malignancy. RECENT FINDINGS Amino acid PET is frequently used for surgery and biopsy planning as well as therapy monitoring in suspected primary brain tumors as well as metastatic lesions, whereas 18F-fluorodeoxyglucose (18F-FDG) remains the tracer of choice for evaluation of patients with primary central nervous system lymphoma. Application of somatostatin receptor ligands has improved tumor delineation in skull base meningioma and concurrently opened up new treatment possibilities in recurrent or surgically not assessable tumors.Recent development focuses on the implementation of hybrid PET/MRI as well as on the development of new tracers targeting tumor hypoxia, enzymes involved in neoplastic metabolic pathways and the combination of PET tracers with therapeutic agents. SUMMARY Implementation of molecular imaging in the clinical routine continues to improve management in patients with brain tumors. However, more prospective large sample studies are needed to validate the additional informative value of PET.
Collapse
|
31
|
Kiviniemi A, Gardberg M, Frantzén J, Pesola M, Vuorinen V, Parkkola R, Tolvanen T, Suilamo S, Johansson J, Luoto P, Kemppainen J, Roivainen A, Minn H. Somatostatin receptor subtype 2 in high-grade gliomas: PET/CT with (68)Ga-DOTA-peptides, correlation to prognostic markers, and implications for targeted radiotherapy. EJNMMI Res 2015; 5:25. [PMID: 25977882 PMCID: PMC4420768 DOI: 10.1186/s13550-015-0106-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 04/14/2015] [Indexed: 01/20/2023] Open
Abstract
Background High-grade gliomas (HGGs) express somatostatin receptors (SSTR), rendering them candidates for peptide receptor radionuclide therapy (PRRT). Our purpose was to evaluate the potential of 68Ga-DOTA-1-Nal3-octreotide (68Ga-DOTANOC) or 68Ga-DOTA-Tyr3-octreotide (68Ga-DOTATOC) to target SSTR subtype 2 (SSTR2) in HGGs, and to study the association between SSTR2 expression and established biomarkers. Methods Twenty-seven patients (mean age 52 years) with primary or recurrent HGG prospectively underwent 68Ga-DOTA-peptide positron emission tomography/computed tomography (PET/CT) before resection. Maximum standardized uptake values (SUVmax) and receptor binding potential (BP) were calculated on PET/CT and disruption of blood–brain barrier (BBB) from contrast-enhanced T1-weighted magnetic resonance imaging (MRI-T1-Gad). Tumor volume concordance between PET and MRI-T1-Gad was assessed by Dice similarity coefficient (DC) and correlation by Spearman’s rank. Immunohistochemically determined SSTR2 status was compared to receptor imaging findings, prognostic biomarkers, and survival with Kruskal-Wallis, Pearson chi-square, and multivariate Cox regression, respectively. Results All 19 HGGs with disrupted BBB demonstrated tracer uptake. Tumor SUVmax (2.25 ± 1.33) correlated with MRI-T1-Gad (r = 0.713, P = 0.001) although DC 0.41 ± 0.19 suggested limited concordance. SSTR2 immunohistochemistry was regarded as positive in nine HGGs (32%) but no correlation with SUVmax or BP was found. By contrast, SSTR2 expression was associated with IDH1 mutation (P = 0.007), oligodendroglioma component (P = 0.010), lower grade (P = 0.005), absence of EGFR amplification (P = 0.021), and longer progression-free survival (HR 0.161, CI 0.037 to 0.704, P = 0.015). Conclusions In HGGs, uptake of 68Ga-DOTA-peptides is associated with disrupted BBB and cannot be predicted by SSTR2 immunohistochemistry. Thus, PET/CT shows limited value to detect HGGs suitable for PRRT. However, high SSTR2 expression portends favorable outcome along with established biomarkers such as IDH1 mutation. Trial registration ClinicalTrials.gov NCT01460706
Collapse
Affiliation(s)
- Aida Kiviniemi
- Turku PET Centre, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, 20521 Turku, Finland ; Department of Radiology, Medical Imaging Centre of Southwest Finland, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Maria Gardberg
- Department of Pathology, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Janek Frantzén
- Department of Neurosurgery, Turku University Hospital, Hämeentie 11, 20521 Turku, Finland
| | - Marko Pesola
- Turku PET Centre, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Ville Vuorinen
- Department of Neurosurgery, Turku University Hospital, Hämeentie 11, 20521 Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, Medical Imaging Centre of Southwest Finland, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Tuula Tolvanen
- Turku PET Centre, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Sami Suilamo
- Department of Oncology and Radiotherapy, Turku University Hospital, Hämeentie 11, 20521 Turku, Finland
| | - Jarkko Johansson
- Turku PET Centre, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Pauliina Luoto
- Turku PET Centre, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Jukka Kemppainen
- Turku PET Centre, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, 20521 Turku, Finland ; Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | - Heikki Minn
- Department of Oncology and Radiotherapy, Turku University Hospital, Hämeentie 11, 20521 Turku, Finland
| |
Collapse
|
32
|
Lapa C, Linsenmann T, Lückerath K, Samnick S, Herrmann K, Stoffer C, Ernestus RI, Buck AK, Löhr M, Monoranu CM. Tumor-associated macrophages in glioblastoma multiforme-a suitable target for somatostatin receptor-based imaging and therapy? PLoS One 2015; 10:e0122269. [PMID: 25807228 PMCID: PMC4373835 DOI: 10.1371/journal.pone.0122269] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 02/11/2015] [Indexed: 12/22/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. Tumor-associated macrophages (TAM) have been shown to promote malignant growth and to correlate with poor prognosis. [1,4,7,10-tetraazacyclododecane-NN′,N″,N′″-tetraacetic acid]-d-Phe1,Tyr3-octreotate (DOTATATE) labeled with Gallium-68 selectively binds to somatostatin receptor 2A (SSTR2A) which is specifically expressed and up-regulated in activated macrophages. On the other hand, the role of SSTR2A expression on the cell surface of glioma cells has not been fully elucidated yet. The aim of this study was to non-invasively assess SSTR2A expression of both glioma cells as well as macrophages in GBM. Methods 15 samples of patient-derived GBM were stained immunohistochemically for macrophage infiltration (CD68), proliferative activity (Ki67) as well as expression of SSTR2A. Anti-CD45 staining was performed to distinguish between resident microglia and tumor-infiltrating macrophages. In a subcohort, positron emission tomography (PET) imaging using 68Ga-DOTATATE was performed and the semiquantitatively evaluated tracer uptake was compared to the results of immunohistochemistry. Results The amount of microglia/macrophages ranged from <10% to >50% in the tumor samples with the vast majority being resident microglial cells. A strong SSTR2A immunostaining was observed in endothelial cells of proliferating vessels, in neurons and neuropile. Only faint immunostaining was identified on isolated microglial and tumor cells. Somatostatin receptor imaging revealed areas of increased tracer accumulation in every patient. However, retention of the tracer did not correlate with immunohistochemical staining patterns. Conclusion SSTR2A seems not to be overexpressed in GBM samples tested, neither on the cell surface of resident microglia or infiltrating macrophages, nor on the surface of tumor cells. These data suggest that somatostatin receptor directed imaging and treatment strategies are less promising in GBM.
Collapse
Affiliation(s)
- Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- * E-mail:
| | - Thomas Linsenmann
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Carolin Stoffer
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Camelia-Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
33
|
Maclean J, Aldridge M, Bomanji J, Short S, Fersht N. Peptide receptor radionuclide therapy for aggressive atypical pituitary adenoma/carcinoma: variable clinical response in preliminary evaluation. Pituitary 2014; 17:530-8. [PMID: 24323313 DOI: 10.1007/s11102-013-0540-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE There are limited treatment options for progressive atypical pituitary adenomas and carcinomas. Peptide receptor radionuclide therapy that targets somatostatin receptors has recently been proposed as a potential treatment option. The theoretical rationale for efficacy is elegant but evaluation of outcomes in the first patients treated for this indication is required to assess whether further study is warranted. METHODS We performed a case review of the three pituitary patients we have treated with (177)Lutetium DOTATATE in our institution (two atypical adenomas, one carcinoma) and dosimetric analysis of the radiation uptake in one patient. RESULTS Treatment was well tolerated. One patient with slowly progressive pituitary carcinoma has stable disease 40 months after completing the planned 4 cycles of treatment. Two patients with rapidly progressive atypical adenomas terminated treatment early due to continued disease progression. Dosimetric evaluation revealed inhomogenous uptake across the tumour (1.3-11.9 Gy with one cycle). CONCLUSION We have found mixed results in our first 3 patients with stable disease achieved only in the patient with the more slowly progressive tumour. As only a limited number of centres offer Peptide receptor radionuclide therapy, a formal study with prospective data collection may be feasible and if carried out should include dosimetric evaluation of absorbed dose.
Collapse
Affiliation(s)
- Jillian Maclean
- University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK,
| | | | | | | | | |
Collapse
|
34
|
Somatostatin receptor-based PET/CT of intracranial tumors: a potential area of application for 68 Ga-DOTA peptides? AJR Am J Roentgenol 2014; 201:1340-7. [PMID: 24896203 DOI: 10.2214/ajr.13.10987] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Similar to neuroendocrine tumors (NETs) at other sites, a wide array of intracranial tumors also express somatostatin receptors (SSTRs). This expression can be exploited for both imaging and therapy. The introduction of (68)Ga-labeled tetraazacyclododecanetetraacetic acid (DOTA)-peptide PET/CT has given new dimension to SSTR-based imaging because of its improved sensitivity and excellent spatial resolution. CONCLUSION However, in contrast to gastropancreatic and bronchopulmonary NETs, limited literature is available regarding the use of (68)Ga-DOTA-peptide PET/CT in intracranial tumors. Here, we briefly review the available literature and highlight the potential role that (68)Ga-DOTA-peptide PET/CT can play in the management of intracranial tumors.
Collapse
|
35
|
Sollini M, Erba PA, Fraternali A, Casali M, Di Paolo ML, Froio A, Frasoldati A, Versari A. PET and PET/CT with 68gallium-labeled somatostatin analogues in Non GEP-NETs Tumors. ScientificWorldJournal 2014; 2014:194123. [PMID: 24693229 PMCID: PMC3947736 DOI: 10.1155/2014/194123] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 10/30/2013] [Indexed: 12/27/2022] Open
Abstract
Somatostatin (SST) is a 28-amino-acid cyclic neuropeptide mainly secreted by neurons and endocrine cells. A major interest for SST receptors (SSTR) as target for in vivo diagnostic and therapeutic purposes was born since a series of stable synthetic SST-analouges PET became available, being the native somatostatin non feasible for clinical use due to the very low metabolic stability. The rationale for the employment of SST-analogues to image cancer is both based on the expression of SSTR by tumor and on the high affinity of these compounds for SSTR. The primary indication of SST-analogues imaging is for neuroendocrine tumors (NETs), which usually express a high density of SSTR, so they can be effectively targeted and visualized with radiolabeled SST-analogues in vivo. Particularly, SST-analogues imaging has been widely employed in gastroenteropancreatic (GEP) NETs. Nevertheless, a variety of tumors other than NETs expresses SSTR thus SST-analogues imaging can also be used in these tumors, particularly if treatment with radiolabeled therapeutic SST-analouges PET is being considered. The aim of this paper is to provide a concise overview of the role of positron emission tomography/computed tomography (PET/CT) with (68)Ga-radiolabeled SST-analouges PET in tumors other than GEP-NETs.
Collapse
Affiliation(s)
- Martina Sollini
- Nuclear Medicine Unit, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Paola Anna Erba
- Regional Center of Nuclear Medicine, University of Pisa, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, 56125 Pisa, Italy
| | - Alessandro Fraternali
- Nuclear Medicine Unit, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Massimiliano Casali
- Nuclear Medicine Unit, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Maria Liberata Di Paolo
- Nuclear Medicine Unit, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Armando Froio
- Nuclear Medicine Unit, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Andrea Frasoldati
- Endocrinology Unit, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Annibale Versari
- Nuclear Medicine Unit, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, 42123 Reggio Emilia, Italy
| |
Collapse
|
36
|
Verburg FA, Heinzel A, Hänscheid H, Mottaghy FM, Luster M, Giovanella L. Nothing new under the nuclear sun: towards 80 years of theranostics in nuclear medicine. Eur J Nucl Med Mol Imaging 2013; 41:199-201. [DOI: 10.1007/s00259-013-2609-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 10/26/2022]
|
37
|
Walker RC, Smith GT, Liu E, Moore B, Clanton J, Stabin M. Measured human dosimetry of 68Ga-DOTATATE. J Nucl Med 2013; 54:855-60. [PMID: 23516312 DOI: 10.2967/jnumed.112.114165] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Measured human dosimetry of the (68)Ga-labeled synthetic somatostatin analog (68)Ga-DOTATATE has not been reported in the peer-reviewed literature. (68)Ga-DOTATATE is an investigational PET/CT imaging agent that binds with high affinity to somatostatin receptor subtype 2, found on many human cancers, most classically neuroendocrine tumors but also others. Reporting of measured dosimetry of (68)Ga-DOTATATE could be useful for investigations for diagnosis, staging, and restaging of somatostatin receptor-expressing tumors. METHODS We performed measured dosimetry with (68)Ga-DOTATATE PET/CT scanning in 6 volunteer human subjects as part of an Institutional Review Board-approved biodistribution investigation of the use of this radiopharmaceutical for possible future use in the diagnosis of indeterminate lung nodules or lung cancer. Five subjects were imaged at 3 time points, and 1 subject was imaged at 2 time points. Dosimetry was then measured for the whole body and for specific organs. RESULTS There were no observed adverse events to the radiopharmaceutical in the immediate or delayed time frames, with a follow-up of 1 y. One patient had stage IV non-small cell lung cancer and remains alive but with disease progressing on treatment. For the other 5 patients, it was ultimately proven that they had benign nodules. The measured dosimetry shows that the critical organ with (68)Ga-DOTATATE is the spleen, followed by the uroepithelium of the bladder, the kidneys, and the liver, in that order. Organ-specific and whole-body dosimetries for (68)Ga-DOTATATE were similar to but often slightly greater than those for (68)Ga-DOTATOC or (68)Ga-DOTANOC but less than those for (111)In-diethylenetriaminepentaacetic acid-octreotide. CONCLUSION No toxicity was observed in our 6 patients, and no adverse events occurred. The measured human dosimetry of (68)Ga-DOTATATE is similar to that of other (68)Ga-labeled somatostatin receptor analogs.
Collapse
Affiliation(s)
- Ronald C Walker
- Medical Imaging Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37232-2675, USA.
| | | | | | | | | | | |
Collapse
|