151
|
Aerts A, Eberlein U, Holm S, Hustinx R, Konijnenberg M, Strigari L, van Leeuwen FWB, Glatting G, Lassmann M. EANM position paper on the role of radiobiology in nuclear medicine. Eur J Nucl Med Mol Imaging 2021; 48:3365-3377. [PMID: 33912987 PMCID: PMC8440244 DOI: 10.1007/s00259-021-05345-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022]
Abstract
With an increasing variety of radiopharmaceuticals for diagnostic or therapeutic nuclear medicine as valuable diagnostic or treatment option, radiobiology plays an important role in supporting optimizations. This comprises particularly safety and efficacy of radionuclide therapies, specifically tailored to each patient. As absorbed dose rates and absorbed dose distributions in space and time are very different between external irradiation and systemic radionuclide exposure, distinct radiation-induced biological responses are expected in nuclear medicine, which need to be explored. This calls for a dedicated nuclear medicine radiobiology. Radiobiology findings and absorbed dose measurements will enable an improved estimation and prediction of efficacy and adverse effects. Moreover, a better understanding on the fundamental biological mechanisms underlying tumor and normal tissue responses will help to identify predictive and prognostic biomarkers as well as biomarkers for treatment follow-up. In addition, radiobiology can form the basis for the development of radiosensitizing strategies and radioprotectant agents. Thus, EANM believes that, beyond in vitro and preclinical evaluations, radiobiology will bring important added value to clinical studies and to clinical teams. Therefore, EANM strongly supports active collaboration between radiochemists, radiopharmacists, radiobiologists, medical physicists, and physicians to foster research toward precision nuclear medicine.
Collapse
Affiliation(s)
- An Aerts
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Uta Eberlein
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany.
| | - Sören Holm
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University Hospital Copenhagen, Copenhagen, Denmark
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, GIGA-CRC in vivo Imaging, University of Liège, Liège, Belgium
| | - Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Lidia Strigari
- Medical Physics Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerhard Glatting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Michael Lassmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
152
|
Prasad V, Prasad S, Lehnert W, Brenner W, Kai H, Bronzel M, Kluge A. Effect of Peptide Dose on Radiation Dosimetry for Peptide Receptor Radionuclide Therapy with 177Lu-DOTATOC: A Pilot Study. Indian J Nucl Med 2021; 36:412-421. [PMID: 35125759 PMCID: PMC8771073 DOI: 10.4103/ijnm.ijnm_15_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 11/14/2022] Open
Abstract
Background: Optimal peptide concentration in treatment with 177Lu-DOTATOC/DOTATATE is a matter of debate. Most of the studies with peptide receptor radionuclide therapy mention peptide dose ranging between 100 and 250 μg. The aim of this is to identify possible differences in radiation-absorbed doses (D/Gy) to tumor and kidney as a function of the peptide mass dose in order to identify the most suitable peptide dose for treatment. The therapeutic index (Dtumor/Dkidneys) was assessed as a key parameter for the treatment response. Materials and Methods: Five patients with metastasized Grade 1 to Grade 2 neuroendocrine tumor were analyzed in this study. Patients (n = 4) received two cycles of treatment with intravenously injected 177Lu-DOTATOC containing peptide mass doses of 200 μg and 90 μg, alternatively; one patient was treated with 90 μg peptide mass in both the therapy cycles. Whole-body (head to mid-thigh) three-dimensional single-photon emission computerized tomography (3D SPECT)/CT images were acquired at 1, 4, 24, 48, and 72 h following the injection of 177Lu-DOTATOC. Attenuation correction for 3D SPECT images was performed using CT data acquired and fused with the SPECT data (SPECT/CT). Results: Overall, 28 target lesions (liver n = 17, lung n = 4, lymph nodes n = 1, and bone n = 2) were analyzed after 1st and 2nd therapy cycles. Tumor normalized absorbed doses varied by a factor of 74 between 0.35 and 26 mGy/MBq. Averaged over all patients, a higher normalized mean tumor dose (10.51 mGy/MBq) was achieved for a peptide dose of 200 μg compared to 90 μg (4.58 mGy/MBq). Kidneys doses varied by a factor of up to 4 between patients (0.25–1.0 mGy/MBq) (independent of dose cycle and peptide dose) and by a factor of up to 2 between dose cycles. The mean kidney dose was 13.7% higher for the 90 μg peptide dose compared to 200 μg. Given the higher tumor dose, the mean therapeutic index of a 200 μg mass dose was considerably higher (16.95), compared to a 90 μg mass dose (9.63). This coincided with the observation, that lesion volume reduction was more pronounced after an initial treatment with a 200 μg mass dose. Biologically effective dose was only 5. 1%–19.3% higher than the absorbed dose for individual dose cycles. Conclusions: Higher peptide dose of 200 μg appears to be more suitable than 90 μg in terms of tumor dose, kidney dose, and therapeutic index for treatment with 177Lu-DOTATOC.
Collapse
Affiliation(s)
- Vikas Prasad
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Nuclear Medicine, University Hospital Ulm, Ulm, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
| | - Sonal Prasad
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin Experimental Radionuclide Imaging Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Wencke Lehnert
- ABX - CRO Advanced Pharmaceutical Services, Dresden, Germany.,Department of Nuclear Medicine, University Hospital Hamburg Eppendorf, Berlin, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,Berlin Experimental Radionuclide Imaging Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Huang Kai
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Bronzel
- ABX - CRO Advanced Pharmaceutical Services, Dresden, Germany
| | - Andreas Kluge
- ABX - CRO Advanced Pharmaceutical Services, Dresden, Germany
| |
Collapse
|
153
|
Gear J, McGowan D, Rojas B, Craig AJ, Smith AL, Scott CJ, Scuffam J, Aldridge M, Tipping J. The internal dosimetry user group position statement on molecular radiotherapy. Br J Radiol 2021; 94:20210547. [PMID: 34433005 PMCID: PMC9328072 DOI: 10.1259/bjr.20210547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Internal Dosimetry User Group (IDUG) is an independent, non-profit group of medical professionals dedicated to the promotion of dosimetry in molecular radiotherapy (www.IDUG.org.uk). The Ionising Radiation (Medical Exposure) Regulations 2017, IR(ME)R, stipulate a requirement for optimisation and verification of molecular radiotherapy treatments, ensuring doses to non-target organs are as low as reasonably practicable. For many molecular radiotherapy treatments currently undertaken within the UK, this requirement is not being fully met. The growth of this field is such that we risk digressing further from IR(ME)R compliance potentially delivering suboptimal therapies that are not in the best interest of our patients. For this purpose, IDUG proposes ten points of action to aid in the successful implementation of this legislation. We urge stakeholders to support these proposals and ensure national provision is sufficient to meet the criteria necessary for compliance, and for the future advancement of molecular radiotherapy within the UK.
Collapse
Affiliation(s)
- Jonathan Gear
- The Internal Dosimetry User Group, England, United Kingdom.,The Joint Department of Physics, The Royal Marsden NHS Foundation Trust & Institute of Cancer Research, Sutton, United Kingdom
| | - Daniel McGowan
- The Internal Dosimetry User Group, England, United Kingdom.,Radiation Physics and Protection, Oxford University Hospitals NHS Foundation Trust, England, United Kingdom
| | - Bruno Rojas
- The Internal Dosimetry User Group, England, United Kingdom.,The Joint Department of Physics, The Royal Marsden NHS Foundation Trust & Institute of Cancer Research, Sutton, United Kingdom
| | - Allison J Craig
- The Internal Dosimetry User Group, England, United Kingdom.,The Joint Department of Physics, The Royal Marsden NHS Foundation Trust & Institute of Cancer Research, Sutton, United Kingdom
| | - April-Louise Smith
- The Internal Dosimetry User Group, England, United Kingdom.,Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Catherine J Scott
- The Internal Dosimetry User Group, England, United Kingdom.,Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - James Scuffam
- The Internal Dosimetry User Group, England, United Kingdom.,Nuclear Medicine Physics, The Royal Surrey NHS Foundation Trust, England, United Kingdom
| | - Matthew Aldridge
- The Internal Dosimetry User Group, England, United Kingdom.,Maidstone and Royal Tunbridge Wells NHS Trust, England, United Kingdom
| | - Jill Tipping
- The Internal Dosimetry User Group, England, United Kingdom.,Nuclear Medicine, The Christie NHS Foundation Trust, England, United Kingdom
| |
Collapse
|
154
|
Chiesa C, Strigari L, Pacilio M, Richetta E, Cannatà V, Stasi M, Marzola MC, Schillaci O, Bagni O, Maccauro M. Dosimetric optimization of nuclear medicine therapy based on the Council Directive 2013/59/EURATOM and the Italian law N. 101/2020. Position paper and recommendations by the Italian National Associations of Medical Physics (AIFM) and Nuclear Medicine (AIMN). Phys Med 2021; 89:317-326. [PMID: 34583307 DOI: 10.1016/j.ejmp.2021.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/27/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022] Open
Abstract
This recommendation by the Italian Associations of Nuclear Medicine (AIMN) and Medical Physics (AIFM) focuses on the dosimetric optimization of Nuclear Medicine Therapy (NMT) as clearly requested by the article 56 of the EURATOM Directive 2013/59 and its consequent implementation in article 158 in the Italian Law n. 101/2020. However, this statement must deal with scientific and methodological limits that still exist and, above all, with the currently available limited resources. This paper addresses these specific issues. It distinguishes among many possible kinds of NMT. For each type, dosimetric optimization is recommended or considered optional, according to the general criteria adopted in any human choice, i.e. a check of technical feasibility first, followed by a cost/benefit argument. The classification of therapies as standardized or non-standardized is presented. This is based on the complexity of the type of pathology, on the variability of the treatment outcome, and on the risks involved. According to the present document, which was officially delivered to Italian Health Ministry as necessary interpretation of the law, a therapeutic team can, in science and consciousness, overcome the indications of posology, to optimize and tailoring a treatment with dosimetry, on the basis of published national or international data or guidelines, without need of an Ethics Committee approval. Data collected in this way will provide additional evidence about optimal dosimetric reference values. As conclusion, a formal appeal is made to the European and National regulatory agencies for pharmaceuticals to obtain the official acknowledgment of this principle.
Collapse
Affiliation(s)
- Carlo Chiesa
- Nuclear Medicine, Foundation IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | - Lidia Strigari
- Director of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Massimiliano Pacilio
- Director of Medical Physics, Azienda Ospedaliero-Universitaria Policlinico Umberto I, Rome, Italy
| | - Elisa Richetta
- Medical Physics, Azienda Ospedaliera Ordine Mauriziano, Turin, Italy
| | - Vittorio Cannatà
- Director of Medical Physics Unit, Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Michele Stasi
- Medical Physics, Azienda Ospedaliera Ordine Mauriziano, Turin, Italy
| | - Maria Cristina Marzola
- Department of Nuclear Medicine PET/CT Centre, S. Maria della Misericordia Hospital, Rovigo, Italy
| | - Orazio Schillaci
- Dean of University Tor Vergata, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Oreste Bagni
- Director of Nuclear Medicine, S. Maria Goretti Hospital, Latina, Italy
| | - Marco Maccauro
- Nuclear Medicine, Foundation IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
155
|
Brosch-Lenz J, Yousefirizi F, Zukotynski K, Beauregard JM, Gaudet V, Saboury B, Rahmim A, Uribe C. Role of Artificial Intelligence in Theranostics:: Toward Routine Personalized Radiopharmaceutical Therapies. PET Clin 2021; 16:627-641. [PMID: 34537133 DOI: 10.1016/j.cpet.2021.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We highlight emerging uses of artificial intelligence (AI) in the field of theranostics, focusing on its significant potential to enable routine and reliable personalization of radiopharmaceutical therapies (RPTs). Personalized RPTs require patient-specific dosimetry calculations accompanying therapy. Additionally we discuss the potential to exploit biological information from diagnostic and therapeutic molecular images to derive biomarkers for absorbed dose and outcome prediction; toward personalization of therapies. We try to motivate the nuclear medicine community to expand and align efforts into making routine and reliable personalization of RPTs a reality.
Collapse
Affiliation(s)
- Julia Brosch-Lenz
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada
| | - Fereshteh Yousefirizi
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada
| | - Katherine Zukotynski
- Department of Medicine and Radiology, McMaster University, 1200 Main Street West, Hamilton, Ontario L9G 4X5, Canada
| | - Jean-Mathieu Beauregard
- Department of Radiology and Nuclear Medicine, Cancer Research Centre, Université Laval, 2325 Rue de l'Université, Québec City, Quebec G1V 0A6, Canada; Department of Medical Imaging, Research Center (Oncology Axis), CHU de Québec - Université Laval, 2325 Rue de l'Université, Québec City, Quebec G1V 0A6, Canada
| | - Vincent Gaudet
- Department of Electrical and Computer Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Babak Saboury
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA; Department of Computer Science and Electrical Engineering, University of Maryland Baltimore County, Baltimore, MD, USA; Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada; Department of Radiology, University of British Columbia, 11th Floor, 2775 Laurel St, Vancouver, British Columbia V5Z 1M9, Canada; Department of Physics, University of British Columbia, 325 - 6224 Agricultural Road, Vancouver, British Columbia V6T 1Z1, Canada
| | - Carlos Uribe
- Department of Radiology, University of British Columbia, 11th Floor, 2775 Laurel St, Vancouver, British Columbia V5Z 1M9, Canada; Department of Functional Imaging, BC Cancer, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada.
| |
Collapse
|
156
|
Uccelli L, Boschi A, Cittanti C, Martini P, Panareo S, Tonini E, Nieri A, Urso L, Caracciolo M, Lodi L, Carnevale A, Giganti M, Bartolomei M. 90Y/ 177Lu-DOTATOC: From Preclinical Studies to Application in Humans. Pharmaceutics 2021; 13:1463. [PMID: 34575538 PMCID: PMC8469896 DOI: 10.3390/pharmaceutics13091463] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/31/2021] [Accepted: 09/10/2021] [Indexed: 12/30/2022] Open
Abstract
The PRRT (Peptide Receptor Radionuclide Therapy) is a promising modality treatment for patients with inoperable or metastatic neuroendocrine tumors (NETs). Progression-free survival (PFS) and overall survival (OS) of these patients are favorably comparable with standard therapies. The protagonist in this type of therapy is a somatostatin-modified peptide fragment ([Tyr3] octreotide), equipped with a specific chelating system (DOTA) capable of creating a stable bond with β-emitting radionuclides, such as yttrium-90 and lutetium-177. In this review, covering twenty five years of literature, we describe the characteristics and performances of the two most used therapeutic radiopharmaceuticals for the NETs radio-treatment: [90Y]Y-DOTATOC and [177Lu]Lu-DOTATOC taking this opportunity to retrace the most significant results that have determined their success, promoting them from preclinical studies to application in humans.
Collapse
Affiliation(s)
- Licia Uccelli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Petra Martini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
| | - Stefano Panareo
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Eugenia Tonini
- Medical Physics Unit, University Hospital, 44124 Ferrara, Italy;
| | - Alberto Nieri
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Luca Urso
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Matteo Caracciolo
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Luca Lodi
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Aldo Carnevale
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Melchiore Giganti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| |
Collapse
|
157
|
Artigas C, Mileva M, Flamen P, Karfis I. Targeted radionuclide therapy: an emerging field in solid tumours. Curr Opin Oncol 2021; 33:493-499. [PMID: 34183491 DOI: 10.1097/cco.0000000000000762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Targeted radionuclide therapy (TRNT) is characterized by systemic administration of radiolabelled drugs, targeting specific molecular alterations expressed on the tumour cells. Small molecules, labelled with β- or α- emitting radioisotopes, are used to deliver radiation directly to the tumour sites. Pretreatment imaging to visualize whole body biodistribution of the target, using the same drugs labelled with positron or γ-emitting radionuclides, completes the concept of theranostic. This review will briefly summarize the current clinical research findings and applications of TRNT in solid tumours, mostly focusing on neuroendocrine and prostate neoplasms. RECENT FINDINGS Peptide receptor radionuclide therapy is a major component in the management of gastroentropancreatic neuroendocrine tumours, with favourable safety profile, quality-of-life improvement and survival benefit. On the NETTER-1 study, it proved to be more effective than high-dose long-acting-release octreotide, leading to its regulatory approval. Prostate-specific membrane antigen (PSMA) is an excellent target for TRNT in prostate cancer. 177Lu-PSMA radioligand therapy demonstrated higher response rates in patients with metastatic castration resistant prostate cancer, when compared with second-line chemotherapy. New developments, including targeting of fibroblast activation proteins overexpressed in the tumour stroma, show promising preliminary results in the theranostic setting. SUMMARY Recent research has demonstrated and consolidated the use of TRNT against well established targets in neuroendocrine tumours and prostate cancer. The identification of new promising molecular targets for TRNT, will further expand the theranostic applications of radionuclides in the field of nuclear medicine.
Collapse
Affiliation(s)
- Carlos Artigas
- Department of Nuclear Medicine, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | |
Collapse
|
158
|
Peptide Receptor Radionuclide Therapy and Primary Brain Tumors: An Overview. Pharmaceuticals (Basel) 2021; 14:ph14090872. [PMID: 34577572 PMCID: PMC8470698 DOI: 10.3390/ph14090872] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 08/19/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023] Open
Abstract
Primary brain tumors (PBTs) are some of the most difficult types of cancer to treat, and despite advancements in surgery, chemotherapy and radiotherapy, new strategies for the treatment of PBTs are needed, especially for those with poor prognosis such as inoperable/difficult-to-reach lesions or relapsing disease. In regard to the last point, malignant primary brain tumors remain some of the most lethal types of cancer. Nuclear medicine may provide exciting new weapons and significant contributions in the treatment of PBTs. In this review, we performed literature research in order to highlight the possible role of peptide receptor radionuclide therapy (PRRT) in the treatment of PBTs with radiolabeled molecules that bind with high-affinity transmembrane receptors such as somatostatin receptors (SSTRs), neurokinin type-1 receptor and prostate-specific membrane antigen (PSMA). These receptors are overexpressed in some cancer types such as gliomas, meningiomas, pituitary tumors and medulloblastomas. A comprehensive overview of possible applications in this field will be shown, providing knowledge about benefits, feasibility, developments and limitations of PRRT in this type of tumor, also revealing new advantages in the management of the disease.
Collapse
|
159
|
Chicheportiche A, Sason M, Godefroy J, Krausz Y, Zidan M, Oleinikov K, Meirovitz A, Gross DJ, Grozinsky-Glasberg S, Ben-Haim S. Simple model for estimation of absorbed dose by organs and tumors after PRRT from a single SPECT/CT study. EJNMMI Phys 2021; 8:63. [PMID: 34436698 PMCID: PMC8390741 DOI: 10.1186/s40658-021-00409-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 08/10/2021] [Indexed: 12/25/2022] Open
Abstract
Background Following each cycle of peptide receptor radionuclide therapy (PRRT), absorbed doses by tumors and normal organs are typically calculated from three quantitative single-photon emission computed tomography (SPECT)/computed tomography (CT) studies acquired at t1 = 24 h, t2 = 96 h, t3 = 168 h after the first cycle of treatment and from a single study at t1 after the subsequent cycles. In the present study, we have assessed the feasibility of a single SPECT/CT study after each PRRT cycle using a trained multiple linear regression (MLR) model for absorbed dose calculation and have evaluated its impact on patient management. Quantitative [177Lu]-DOTA-TATE SPECT/CT data after PRRT of seventy-two consecutive metastatic neuroendocrine tumors patients were retrospectively evaluated. A set of 40 consecutive studies was used to train the MLR model. The two independent variables of the model included the time of imaging after administration of the treatment and the radiopharmaceutical activity concentration in a given organ/tumor. The dependent variable was the dose absorbed by the organ/tumor obtained with the standard protocol. For bone marrow dosimetry, the independent variables included the time of imaging, and the blood and remainder of the body activity concentration. The model was evaluated in 32 consecutive patients. Absorbed doses were assessed for kidneys, bone marrow, liver, spleen and tumor sites. Results There was no difference in management decisions, whether PRRT can be safely continued or not because unsafe absorbed dose to risk organs between the standard and the MLR model-based protocol using a single SPECT/CT study performed at t3 = 168 h after the first cycle and at t1 = 24 h after the subsequent cycles. Cumulative absorbed doses were obtained with mean relative differences of − 0.5% ± 5.4%, 1.6% ± 15.1%, − 6.2% ± 7.3%, − 5.5% ± 5.8% and 2.9% ± 12.7% for kidneys, bone marrow, liver, spleen and tumors, respectively (Pearson’s r correlation coefficient 0.99, 0.91, 0.99, 0.99 and 0.97, respectively). Conclusion Dosimetry calculations using a MLR model with a single SPECT/CT study are in good agreement with the standard protocol, while avoiding the use of dosimetry software and enabling improved patient comfort and reduced scanner and staff time. Supplementary Information The online version contains supplementary material available at 10.1186/s40658-021-00409-z.
Collapse
Affiliation(s)
- Alexandre Chicheportiche
- Department of Nuclear Medicine & Biophysics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel.
| | - Moshe Sason
- Department of Nuclear Medicine & Biophysics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Jeremy Godefroy
- Department of Nuclear Medicine & Biophysics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Yodphat Krausz
- Department of Nuclear Medicine & Biophysics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Mahmoud Zidan
- Department of Nuclear Medicine & Biophysics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Kira Oleinikov
- Neuroendocrine Tumor Unit, ENETS Center of Excellence, Endocrinology and Metabolism Department, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Amichay Meirovitz
- Oncology Department and Radiation Therapy Unit, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - David J Gross
- Neuroendocrine Tumor Unit, ENETS Center of Excellence, Endocrinology and Metabolism Department, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Simona Grozinsky-Glasberg
- Neuroendocrine Tumor Unit, ENETS Center of Excellence, Endocrinology and Metabolism Department, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Simona Ben-Haim
- Department of Nuclear Medicine & Biophysics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, 91120, Jerusalem, Israel.,Institute of Nuclear Medicine, University College London Hospitals, London, UK
| |
Collapse
|
160
|
Schreuder N, de Romijn I, Jager PL, Kosterink JGW, van Puijenbroek EP. Safe use of radiopharmaceuticals in patients with chronic kidney disease: a systematic review. EJNMMI Radiopharm Chem 2021; 6:27. [PMID: 34417933 PMCID: PMC8380202 DOI: 10.1186/s41181-021-00145-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) may need to have their radiopharmaceutical dosage adjusted to prevent adverse effects and poor outcomes, but there are few recommendations on radiopharmaceutical dosing for this group of patients. The aim of this study is to provide an overview of the available information on radiopharmaceutical dose recommendations for patients with CKD. METHODS We performed a systematic literature review according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. We conducted a literature search in the MEDLINE (PubMed) and Embase databases and screened potentially relevant studies using inclusion and exclusion criteria. We independently assessed the included observational studies' methodologies and extracted relevant data. RESULTS Of the 5795 studies first identified, 34 were included in this systematic review. These studies described three radiopharmaceuticals: [131I]sodium iodine, [18F]fludeoxyglucose, and [131I]iobenguane. Twenty-nine studies (85.3%) reported data on patients with CKD stage 5, while only three studies mentioned CKD patients in other stages (8.8%). CONCLUSION We found no consistent recommendations for radiopharmaceutical dosing in patients with CKD. Although some studies do mention dosing difficulties in patients with CKD, information is available for only a few radiopharmaceuticals, and recommendations are sometimes contradictory. Further research on radiopharmaceutical dosing in patients with CKD is needed to determine whether these patients require specific dosing, especially for therapeutic radiopharmaceuticals where a non-optimised dose may lead to an increased risk of toxicity for non-targeted organs. Including patients with CKD in studies and providing specific information about dosing in these patients should be a priority for the radiopharmaceutical community.
Collapse
Affiliation(s)
- Nanno Schreuder
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands.
- GE Healthcare Radiopharmacy Zwolle, Zwolle, The Netherlands.
| | - Iris de Romijn
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Pieter L Jager
- Department of Nuclear Medicine, Isala Hospital, Zwolle, The Netherlands
| | - Jos G W Kosterink
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eugène P van Puijenbroek
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| |
Collapse
|
161
|
Chakraborty A, Mitra A, Tawate M, Sahoo S, Lad S, Rakshit S, Gaikwad S, Basu S, Shimpi H, Banerjee S. Therapeutic Multidose Preparation of a Ready-to-Use 177Lu-PSMA-617 Using Carrier Added Lutetium-177 in a Hospital Radiopharmacy and Its Clinical Efficacy. Cancer Biother Radiopharm 2021; 36:682-692. [PMID: 34402687 DOI: 10.1089/cbr.2020.4261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Introduction: [177Lu]Lu-prostate-specific membrane antigen (PSMA)-617 has emerged as a promising radiopharmaceutical for targeting PSMA in metastatic castrate-resistant prostate carcinoma (mCRPC). We have optimized the radiolabeling protocol for a multidose formulation (27-28.8 GBq equivalent to 6-7 patient-doses) of [177Lu]Lu-PSMA-617 using [177Lu]Lu3+ produced via 176Lu(n,γ)177Lu route with moderate specific activity (0.66-0.81 GBq/μg). Methods: [177Lu]Lu-PSMA-617 was synthesized using moderate specific activity [177Lu]LuCl3 (0.74 GBq/μg) with PSMA-617 having metal-to-ligand molar ratio ∼1: 2.5 in CH3COONH4 buffer (0.1 M) containing gentisic acid at pH 4.0-4.5. Human prostate carcinoma cell line LNCaP cell (high PSMA expression) was used for in vitro cell-binding studies and generating tumor xenograft models in nude mice for tissue biodistribution studies. Several batches of the present formulation have been clinically administered in mCRPC patients (single patient dose: 4.44-5.55 GBq per cycle). Results: In this study we report a consistent and reproducible protocol for multidose formulations of [177Lu]Lu-PSMA-617 for adopting in a hospital radiopharmacy setting. Although the radiochemical yield of [177Lu]Lu-PSMA-617 was found to be 97.30% ± 1.03%, the radiochemical purity was 98.24% ± 0.50% (n = 19). In vitro and serum stability of [177Lu]Lu-PSMA-617 was retained up to 72 and 120 h after radiolabeling and upon storage at -20°C with a radioactive concentration between 0.37 and 0.74 GBq/mL upon using stabilizer concentration as low as 43-48 μg/mCi. Preclinical cell-binding studies of [177Lu]Lu-PSMA-617 revealed specific binding with LNCaP cells of 17.4% ± 2.4%. The uptake in LnCaP xenografted tumor (nude mice) was 7.5 ± 2.6% ID/g for ∼1.5-2.0 cm3 tumor volume at 24-h post-injection. Post-therapy (24 h) SPECT image of mCRPC patients with prior orchidectomy and various hormone therapy showed specific localization of [177Lu]Lu-PSMA-617 in the tumor region. Conclusions: Formulation of a ready-to-use multidose formulation of [177Lu]Lu-PSMA-617 was successfully achieved and the procedure was optimized for routine preparation at a hospital radiopharmacy set-up. High degree of localization of [177Lu]Lu-PSMA-617 in post-therapy SPECT scan and the post-therapeutic response confirms its therapeutic efficacy. Clinical Trials.gov ID: RPC/51/Minutes/Final dated 16th October, 2019.
Collapse
Affiliation(s)
- Avik Chakraborty
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India.,Department of Atomic Energy, Homi Bhabha National Institute, Mumbai, India
| | - Arpit Mitra
- Medical Cyclotron Facility, Radiation Medicine Centre, Board of Radiation and Isotope Technology, Mumbai, India
| | - Megha Tawate
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
| | - Sudip Sahoo
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
| | - Sangita Lad
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
| | - Sutapa Rakshit
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
| | - Sujay Gaikwad
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India.,Department of Atomic Energy, Homi Bhabha National Institute, Mumbai, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India.,Department of Atomic Energy, Homi Bhabha National Institute, Mumbai, India
| | - Hemant Shimpi
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India
| | - Sharmila Banerjee
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Mumbai, India.,Department of Atomic Energy, Homi Bhabha National Institute, Mumbai, India.,Medical Cyclotron Facility, Radiation Medicine Centre, Board of Radiation and Isotope Technology, Mumbai, India
| |
Collapse
|
162
|
Abstract
Peptide receptor radionuclide therapy (PRRT) is an effective form of treatment of patients with metastatic neuroendocrine tumors, delivering modest objective tumor response rates but notable survival and symptomatic benefits. The first PRRT approved by the US Food and Drug Administration was lutetium 177-DOTATATE and is for use in adults with somatostatin receptor-positive gastroenteropancreatic neuroendocrine tumors. The treatment paradigm typically leads to significant improvement in symptomology coupled with an extended period of progression-free survival. Side effects are limited, with a small fraction of individuals experiencing clinically significant long-term renal or hematologic toxicity.
Collapse
|
163
|
Kristiansson A, Örbom A, Vilhelmsson Timmermand O, Ahlstedt J, Strand SE, Åkerström B. Kidney Protection with the Radical Scavenger α 1-Microglobulin (A1M) during Peptide Receptor Radionuclide and Radioligand Therapy. Antioxidants (Basel) 2021; 10:antiox10081271. [PMID: 34439519 PMCID: PMC8389303 DOI: 10.3390/antiox10081271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 02/07/2023] Open
Abstract
α1-Microglobulin (A1M) is an antioxidant found in all vertebrates, including humans. It has enzymatic reductase activity and can scavenge radicals and bind free heme groups. Infused recombinant A1M accumulates in the kidneys and has therefore been successful in protecting kidney injuries in different animal models. In this review, we focus on A1M as a radioprotector of the kidneys during peptide receptor radionuclide/radioligand therapy (PRRT/RLT). Patients with, e.g., neuroendocrine tumors or castration resistant prostate cancer can be treated by administration of radiolabeled small molecules which target and therefore enable the irradiation and killing of cancer cells through specific receptor interaction. The treatment is not curative, and kidney toxicity has been reported as a side effect since the small, radiolabeled substances are retained and excreted through the kidneys. In recent studies, A1M was shown to have radioprotective effects on cell cultures as well as having a similar biodistribution as the somatostatin analogue peptide 177Lu-DOTATATE after intravenous infusion in mice. Therefore, several animal studies were conducted to investigate the in vivo radioprotective potential of A1M towards kidneys. The results of these studies demonstrated that A1M co-infusion yielded protection against kidney toxicity and improved overall survival in mouse models. Moreover, two different mouse studies reported that A1M did not interfere with tumor treatment itself. Here, we give an overview of radionuclide therapy, the A1M physiology and the results from the radioprotector studies of the protein.
Collapse
Affiliation(s)
- Amanda Kristiansson
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
- Correspondence:
| | - Anders Örbom
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
| | - Oskar Vilhelmsson Timmermand
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
| | - Jonas Ahlstedt
- Department of Clinical Sciences Lund, CIPA, Lund University, 221 84 Lund, Sweden;
| | - Sven-Erik Strand
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
- Department of Clinical Sciences Lund, Medical Radiation Physics, Lund University, 221 00 Lund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, 221 84 Lund, Sweden;
| |
Collapse
|
164
|
Abstract
The landscape of nuclear oncology is rapidly changing. The advent of molecular radionuclide theranostics, multidisciplinary tumor board decision making, artificial intelligence and radiomics interpretation of diagnostic imaging, evolution of pharmacogenomics prediction of tumor response, and regulatory requirements for prospective individual dosimetry are just some of the elements which are broadening the essence of physician responsibility. The burgeoning knowledge base essential for mastering the emergent technologies, and their profound effect on moral philosophic aspects of provision of cancer care, are challenging. The new relationship of the theranostic nuclear physician with respect to shared care of the individual patient, particularly with regard to transparency, accountability, and responsibility for targeted radionuclide diagnosis and therapy of cancer, will be explored in this update.
Collapse
Affiliation(s)
- J Harvey Turner
- Department of Nuclear Medicine, The University of Western Australia, Fiona Stanley Fremantle Hospitals, Murdoch, Australia
| |
Collapse
|
165
|
Demirkol MO, Özkan A, Uçar B, Wester HJ, Ferhanoğlu B. Extramedullary Relapsed Multiple Myeloma Treatment With 177Lu-Labeled CXCR4 Endoradiotherapy and Dosimetric Results. Clin Nucl Med 2021; 46:656-658. [PMID: 34034308 DOI: 10.1097/rlu.0000000000003705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT We created our first national clinical protocol of 177Lu-CXCR4 therapy for patient who have failed to respond to current therapy options. We also calculated the kidney, liver, and tumor dosimetry. The kidney's mean absorbed dose was calculated to be 0.45 Gy/GBq, the calculated radiation absorbed dose of the liver was 0.63 Gy/GBq, and the radiation absorbed doses of the tumors vary between 9.2 and 82 Gy/GBq. 177Lu-CXCR4 therapy produced a promising clinical response in our patient in acceptable radiation dose limits as a treatment option in heavily pretreated patients with advanced multiple myeloma.
Collapse
Affiliation(s)
| | | | - Burcu Uçar
- Nuclear Medicine and Molecular Imaging Department, VKF American Hospital, Istanbul, Turkey
| | - Hans-Jürgen Wester
- Chair for Pharmaceutical Radiochemistry, Technische Universität München, Garching, Germany
| | | |
Collapse
|
166
|
Vonken EJPA, Bruijnen RCG, Snijders TJ, Seute T, Lam MGEH, de Keizer B, Braat AJAT. Intra-arterial administration boosts 177Lu-HA-DOTATATE accumulation in salvage meningioma patients. J Nucl Med 2021; 63:406-409. [PMID: 34301783 DOI: 10.2967/jnumed.121.262491] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/14/2021] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION: Intravenous 177Lu-(HA-)DOTATATE has shown promising results for the treatment of surgery- and radiotherapy-refractory meningiomas. We aimed to investigate the added value of intra-arterial administration. METHODS: Patients underwent at least one intravenous 177Lu-HA-DOTATATE treatment first and subsequent intra-arterial cycles. In(tra)-patient comparison was based on post-treatment 177Lu-HA-DOTATATE imaging 24 hours post-injection. Technical success rates and adverse events were recorded. RESULTS: Four patients provided informed consent. Technical success rate was 100% and no angiography related or unexpected adverse events occurred. Intra-patient comparison showed an increased target lesion accumulation on both planar imaging (mean +220%) and SPECT/CT (mean +398%) after intra-arterial administration compared to intravenous. No unexpected adverse events during follow-up occurred. CONCLUSION: Intra-arterial PRRT significantly increases tracer accumulation, and is a safe and promising improvement for salvage meningioma patients. Future prospective studies on intra-arterial PRRT are needed to determine gain on efficacy and survival.
Collapse
|
167
|
Kreppel B, Gonzalez-Carmona MA, Feldmann G, Küppers J, Moon ES, Marinova M, Bundschuh RA, Kristiansen G, Essler M, Roesch F, Gaertner FC. Fibroblast activation protein inhibitor (FAPi) positive tumour fraction on PET/CT correlates with Ki-67 in liver metastases of neuroendocrine tumours. Nuklearmedizin 2021; 60:344-354. [PMID: 34256394 DOI: 10.1055/a-1521-8604] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Gallium-68-labelled inhibitors of the fibroblast activation protein (FAPi) enable positron emission tomography/computed tomography (PET/CT) imaging of fibroblast activation. We evaluated if [68Ga]Ga-DATA5 m.SA.FAPi PET/CT is related to Ki-67 as a marker of tumour aggressiveness in patients with liver metastases of NET. METHODS Thirteen patients with liver metastases of a histologically confirmed NET who underwent PET/CT with [68Ga]Ga-DATA5 m.SA.FAPi, [18F]FDG and [68Ga]Ga-DOTA-TOC were retrospectively analyzed. PET-positive liver tumour volumes were segmented for calculation of volume, SUVmax and PET-positive tumour fraction (TF). PET parameters were correlated with Ki-67. RESULTS FDGSUVmax correlated positively (rho = 0.543, p < 0.05) and DOTATOCSUVmax correlated negatively (rho = -0.618, p < 0.05) with Ki-67, the correlation coefficients were in the moderate range. There was no significant correlation between FAPiSUVmax and Ki-67 (rho = 0.382, p > 0.05). FAPiTF correlated positively (rho = 0.770, p < 0.01) and DOTATOCTF correlated negatively (rho = -0.828, p < 0.01) with Ki-67, both significantly with high correlation coefficients. FDGTF also correlated significantly with Ki-67, with a moderate correlation coefficient (rho = 0.524, p < 0.05). The ratio FAPiVOL:DOTATOCVOL showed a significant and strong correlation with Ki-67 (rho = 0.808, p < 0.01). CONCLUSION The ratio FAPiVOL:DOTATOCVOL might serve as a clinical parameter for the assessment of dedifferentiation and aggressiveness of liver metastases in patients with NET. [68Ga]Ga-DATA5 m.SA.FAPi might hold potential for identification of high-risk patients. Further studies are warranted to evaluate its prognostic significance in comparison to [18F]FDG in patients with NET.
Collapse
Affiliation(s)
- Barbara Kreppel
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | | | - Georg Feldmann
- Department of Internal Medicine III, University Hospital Bonn, Bonn, Germany
| | - Jim Küppers
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Euy Sung Moon
- Department of Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Milka Marinova
- Department of Radiology, University Hospital Bonn, Bonn, Germany
| | | | | | - Markus Essler
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Frank Roesch
- Department of Chemistry, Johannes Gutenberg University, Mainz, Germany
| | | |
Collapse
|
168
|
Gomes Marin JF, Nunes RF, Coutinho AM, Zaniboni EC, Costa LB, Barbosa FG, Queiroz MA, Cerri GG, Buchpiguel CA. Theranostics in Nuclear Medicine: Emerging and Re-emerging Integrated Imaging and Therapies in the Era of Precision Oncology. Radiographics 2021; 40:1715-1740. [PMID: 33001789 DOI: 10.1148/rg.2020200021] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Theranostics refers to the pairing of diagnostic biomarkers with therapeutic agents that share a specific target in diseased cells or tissues. Nuclear medicine, particularly with regard to applications in oncology, is currently one of the greatest components of the theranostic concept in clinical and research scenarios. Theranostics in nuclear medicine, or nuclear theranostics, refers to the use of radioactive compounds to image biologic phenomena by means of expression of specific disease targets such as cell surface receptors or membrane transporters, and then to use specifically designed agents to deliver ionizing radiation to the tissues that express these targets. The nuclear theranostic approach has sparked increasing interest and gained importance in parallel to the growth in molecular imaging and personalized medicine, helping to provide customized management for various diseases; improving patient selection, prediction of response and toxicity, and determination of prognosis; and avoiding futile and costly diagnostic examinations and treatment of many diseases. The authors provide an overview of theranostic approaches in nuclear medicine, starting with a review of the main concepts and unique features of nuclear theranostics and aided by a retrospective discussion of the progress of theranostic agents since early applications, with illustrative cases emphasizing the imaging features. Advanced concepts regarding the role of fluorine 18-fluorodeoxyglucose PET in theranostics, as well as developments in and future directions of theranostics, are discussed. ©RSNA, 2020 See discussion on this article by Greenspan and Jadvar.
Collapse
Affiliation(s)
- José Flávio Gomes Marin
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Rafael F Nunes
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Artur M Coutinho
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Elaine C Zaniboni
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Larissa B Costa
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Felipe G Barbosa
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Marcelo A Queiroz
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Giovanni G Cerri
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Carlos A Buchpiguel
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| |
Collapse
|
169
|
The Dependence of Renal 68Ga[Ga]-DOTATOC Uptake on Kidney Function and Its Relevance for Peptide Receptor Radionuclide Therapy with 177Lu[Lu]-DOTATOC. Diagnostics (Basel) 2021; 11:diagnostics11071216. [PMID: 34359299 PMCID: PMC8307408 DOI: 10.3390/diagnostics11071216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022] Open
Abstract
Background: In addition to its SSTR-specific binding in tumors and healthy tissues, DOTATOC analogues accumulate in kidney parenchyma. Renal tracer uptake might be a surrogate of kidney function or dysfunction. This study aimed to evaluate if kidney function can be estimated from 68Ga[Ga]-DOTATOC uptake in PET/CT and its impact on the nephrotoxicity of 177Lu[Lu]-DOTATOC PRRT. Methods: Two cohorts of patients (A: 128 diagnostic patients; B: 32 PRRT patients) were evaluated retrospectively. SUV values of the kidneys, physiologically SSTR-expressing organs and in background compartments were assessed. Kidney function was calculated as eGFR by CKD-EPI creatinine equation. Pearson’s correlation coefficients and treatment-induced changes of uptake and kidney function were assessed and compared. Results: Kidney function and renal DOTATOC uptake showed a significant inverse correlation (R2 = 0.037; p = 0.029). Evaluated models of PET/CT measurements were not able to predict kidney function sufficiently. The uptake of other organs did not depend on eGFR. While the renal uptake increased after PRRT (p < 0.001), the kidney function did not change significantly (p = 0.382). Neither low pre-therapeutic eGFR nor high pre-therapeutic kidney uptake were risk factors of PRRT-induced deterioration in kidney function. Conclusion: The relevance of kidney function for renal 68Ga[Ga]-DOTATOC uptake is limited. The nephrotoxicity of 177Lu[Lu]-DOTATOC PRRT might be low and cannot be reliably predicted by pre-therapeutic measurements.
Collapse
|
170
|
Abstract
Neuroendocrine tumors (NET) are a heterogeneous group of neoplasms, arising from cells of the endocrine system, with various clinical behaviors. Although these neoplasms are considered rare, a significant increase in the incidence and detectability of NET has been noted in many epidemiological studies in recent years. Among the various therapeutic options, peptide receptor radionuclide therapy (PRRT), using somatostatine has been shown to be highly effective and a well-tolerated therapy, improving survival parameters. The current use of radionuclides for PRRT is β-emitters. Due to hypoxia cancer tissue could be resistant for β-emitters. Quite long penetration range had a significant impact on side effects. α-particles with higher energy and shorter penetration range in comparison to β-particles, have distinct advantages for use in targeted therapy. The clinical experience with somatostatine based targeted α therapy (TAT) in NET showed very promising results even in patienicts refractory to treatment with β-emitters. This article summarizes current developments in preclinical and clinical investigation on TAT in NET.
Collapse
Affiliation(s)
| | - Leszek Królicki
- Nuclear Medicine Department, Medical University of Warsaw, Poland
| |
Collapse
|
171
|
Ga-68 DOTATATE PET/CT and F-18 FDG PET/CT in the evaluation of low and intermediate versus high-grade neuroendocrine tumors. Nucl Med Commun 2021; 41:1060-1065. [PMID: 32732600 DOI: 10.1097/mnm.0000000000001255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We investigated the role of Ga-68 DOTATATE PET/CT in comparison to F-18 FDG PET/CT in patients with low and intermediate versus high-grade neuroendocrine tumors (NETs). METHODS We identified 81 patients who underwent Ga-68 DOTATATE PET/CT at our institution between May 2017 and December 2018 and met inclusion criteria of biopsy-proven NET with known Ki-67 index, histologic grade, or differentiation. Patients were divided into two groups. Control group included Ki-67 ≤20%, grade 1 or 2, or well-differentiated tumors. Experimental group included Ki-67 >20%, grade 3, or poorly-differentiated tumors. RESULTS Mean age was 57 years, with 36 males and 45 females. Most common primary sites were small bowel, pancreas, and lung. Most common distant metastatic sites were liver and bone. In the control group (n = 67), median Ki-67 was 4% (range 1-30%). 55/67 (82.1%) DOTATATE and 6/11 (54.5%) FDG scans were positive (P = 0.04). Positive scans showed >10 lesions in 25/55 (45.5%) DOTATATE and 1/6 (16.7%) FDG scans (P = 0.18). 40/55 (72.7%) positive DOTATATE and 3/6 (50%) FDG scans showed distant disease (P = 0.25). In the experimental group (n = 14), median Ki-67 was 68% (range 25-95%). All 14 DOTATATE and all nine FDG scans were positive. Positive scans showed >10 lesions in 4/14 (28.6%) DOTATATE and 5/9 (55.6%) FDG scans (P = 0.20). 10/14 (71.4%) positive DOTATATE and 7/9 (77.8%) FDG scans showed distant disease (P = 0.74). CONCLUSION All patients with high grade, poorly-differentiated NETs had positive DOTATATE PET/CTs. In these patients, DOTATATE PET/CT did not significantly differ from FDG PET/CT in identifying >10 lesions or distant disease.
Collapse
|
172
|
Emerging Preclinical and Clinical Applications of Theranostics for Nononcological Disorders. PET Clin 2021; 16:429-440. [PMID: 34053586 DOI: 10.1016/j.cpet.2021.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Studies in nuclear medicine have shed light on molecular imaging and therapeutic approaches for oncological and nononcological conditions. Using the same radiopharmaceuticals for diagnosis and therapeutics of malignancies, the theranostics approach, has improved clinical management of patients. Theranostic approaches for nononcological conditions are recognized as emerging topics of research. This review focuses on preclinical and clinical studies of nononcological disorders that include theranostic strategies. Theranostic approaches are demonstrated as possible in the clinical management of infections and inflammations. There is an emerging need for randomized trials to specify the factors affecting validity and efficacy of theranostic approaches in nononcological diseases.
Collapse
|
173
|
Sekikawa Y, Funada K, Akamatsu G, Himuro K, Takahashi A, Baba S, Sasaki M. Monte Carlo simulation of the acquisition conditions for 177Lu molecular imaging of hepatic tumors. Ann Nucl Med 2021; 35:823-833. [PMID: 34057655 DOI: 10.1007/s12149-021-01620-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/21/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To examine the impact of acquisition time on Lutetium-177 (177Lu) single-photon emission computed tomography (SPECT) images using Monte Carlo simulation. METHODS A gamma camera simulation based on the Monte Carlo method was performed to produce SPECT images. The phantom was modeled on a NEMA IEC BODY phantom including six spheres as tumors. After the administration of 7.4 GBq of 177Lu, radioactivity concentrations of the tumor/liver at 6, 24, and 72 h after administration were set to 1.85/0.201, 2.12/0.156, and 1.95/0.117 MBq/mL, respectively. In addition, the radioactivity concentrations of the tumor at 72 h after administration varied by 1/2, 1/4, and 1/8 when comparison was made. Acquisition times examined were 1.2, 1.5, 2, 3, 6, and 12 min. To assess the impact of collimators, SPECT data acquired at 72 h after the administration using six collimators of low-energy high-resolution (LEHR), extended low-energy general-purpose (ELEGP), medium-energy, and general-purpose (MEGP-1, MEGP-2, and MEGP-3) and high-energy general-purpose (HEGP) were examined. After prefiltering using a Butterworth filter, projection images were reconstructed using ordered subset expectation maximization. The detected photons were classified into direct rays, scattered rays, penetrating rays, and characteristic X-rays from lead. The image quality was evaluated through visual assessment, and physical assessment of contrast recovery coefficient (CRC) and contrast-to-noise ratio (CNR). In this study, the CNR threshold for detectability was assumed to be 5.0. RESULTS To compare collimators, the highest sensitivity was observed with ELEGP, followed by LEHR and MEGP-1. The highest ratio of direct ray was also observed in ELEGP followed by MEGP-1. In comparison of the radioactivity concentration ratios of tumor/liver, CRC and CNR were significantly decreased with smaller radioactivity concentration ratios. This effect was greater with larger spheres. According to the visual assessment, the acquisition time of 6, 6, and 3 min or longer was required using ELEGP collimator at 6, 24, and 72 h after administration, respectively. Physical assessment based on CNR and CRC also suggested that 6, 6, and 3 min or longer acquisition time was necessary at 6, 24, and 72 h after administration. CONCLUSION 177Lu-SPECT images generated via the Monte Carlo simulation suggested that the recommended acquisition time was 6 min or longer at 6 and 24 h and 3 min or longer at 72 h after administration.
Collapse
Affiliation(s)
- Yuya Sekikawa
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Department of Radiological Technology, Faculty of Fukuoka Medical Technology, Teikyo University, 6-22 Misakimachi, Omuta, Fukuoka, 836-8505, Japan
| | - Keita Funada
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Go Akamatsu
- National Institutes for Quantum and Radiological Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Kazuhiko Himuro
- Division of Radiology, Department of Medical Technology, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akihiko Takahashi
- Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Shingo Baba
- Division of Radiology, Department of Medical Technology, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masayuki Sasaki
- Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
174
|
Peptide receptor radionuclide therapy for GEP-NET: consolidated knowledge and innovative applications. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00443-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
175
|
Mix M, Renaud T, Kind F, Nemer U, Yousetzadeh-Nowsha E, Moalosi TCG, Omrane MA, Meyer PT, Ruf J. Kidney doses in 177Lu-based radioligand therapy in prostate cancer: Is dose estimation based on reduced dosimetry measurements feasible? J Nucl Med 2021; 63:253-258. [PMID: 34088773 DOI: 10.2967/jnumed.121.262245] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
The radiation dose to the kidneys should be monitored in prostate cancer patients treated with radioligand therapy (RLT) targeting the prostate-specific membrane antigen (PSMA). We analyzed whether pretherapeutic kidney function is predictive of subsequent kidney dose and to what extend the cumulative kidney dose after multiple therapy cycles at the end of treatment can be predicted from a dosimetry based on the first cycle. Methods: Data of 59 patients treated with at least 2 cycles of 177Lu-PSMA-617 (PSMA-RLT) were analyzed. Treatment (median: 6 GBq/cycle) was performed at 6-8 week intervals, accompanied by voxel-based 3D-dosimetry (measured kidney dose) with SPECT/CT on each of days 0-3 and once during days 6-9. Pretherapeutic kidney function (eGFR, MAG3-clearance) was correlated to the kidney doses. Cumulative kidney doses at the end of treatment were compared to a dose estimation based on the population-based mean kidney dose, individual first cycle kidney dose and mean kidney doses of cycles 1, 3 and 5 per administered activity. Results: A total of 176 PSMA-RLT cycles were performed with a median of 3 cycles per patient. The average kidney dose per administered activity of all 176 cycles was 0.67 ± 0.24 Gy/GBq (range 0.21 - 1.60). MAG3-clearance and eGFR were no reliable predictors of subsequent absorbed kidney dose and showed only small effect sizes (R2 = 0.080 and 0.014, P = 0.039 and 0.375). All simplified estimations of cumulative kidney dose correlated significantly (P < 0.001) with measured kidney doses: Estimations based on the individual first-cycle dose were more accurate than the use of the population-based average kidney dose (R2 = 0.853 vs. R2 = 0.560). Dose estimation was best when the doses of cycles 3 and 5 were included as well (R2 = 0.960). Conclusion: Pretherapeutic renal function was not predictive for subsequent kidney dose during therapy. Extrapolation of individual data from dosimetry of the first cycle was highly predictive for the cumulative kidney dose at the end of treatment. This is further improved by the integration of dose information from every other cycle. In any case, because of a high interindividual variance, an individual dosimetry is advisable.
Collapse
Affiliation(s)
- Michael Mix
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tobias Renaud
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Felix Kind
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Ursula Nemer
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Elham Yousetzadeh-Nowsha
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tumelo C G Moalosi
- Department of Medical Imaging and Clinical Oncology, Nuclear Medicine Division, Faculty of Medicine and Health Science, Stellenbosch University, South Africa
| | - Mohamed Aymen Omrane
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
176
|
Basu S, Parghane R, Ranade R, Thapa P, Ramaswamy A, Ostwal V, Sirohi B, Panda D, Shrikhande SV. Peptide Receptor Radionuclide Therapy in the Management of Neuroendocrine Tumors (Neoplasms): Fundamentals and Salient Clinical Practice Points for Medical Oncologists. Indian J Med Paediatr Oncol 2021. [DOI: 10.4103/ijmpo.ijmpo_161_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
AbstractThis editorial commentary is an expert summary of “Peptide Receptor Radionuclide Therapy (PRRT),” encompassing the essential fundamentals and salient clinical practice points, deliberated and designed in a point-wise manner with theme-based subheadings. Emphasis has been laid on the topics of practical relevance to the referring oncologists with relevant finer points where necessary. A part of the presented overview has been generated from the authors' own practical experience of more than 3500 successful therapies delivered over the last 9 years at a large tertiary care PRRT setting by the joint efforts of Radiation Medicine Centre (RMC), Bhabha Atomic Research Centre (BARC), and Gastrointestinal services of Tata Memorial Hospital (TMH) at the TMH-RMC premises. While the technical indigenization is beyond the scope of this treatise, we must mention here that India had been one of the frontrunners in this treatment modality, and the PRRT services in this country were developed purely as an indigenous effort right from the production of the radionuclide (177-Lutetium) at the reactor and radiolabeling and production of the radiopharmaceutical (177Lu-DOTATATE) by the radiopharmaceutical scientists at the BARC and RMC; such an endeavor allowed this very specialized therapy to be delivered at a very affordable cost in our setting which could be viewed as a major societal contribution of the atomic energy research in this country.
Collapse
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai, Maharashtra
- Homi Bhabha National Institute, DAE’s University, Mumbai, Maharashtra
| | - Rahul Parghane
- Radiation Medicine Centre Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai, Maharashtra
- Homi Bhabha National Institute, DAE’s University, Mumbai, Maharashtra
| | - Rohit Ranade
- Radiation Medicine Centre Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai, Maharashtra
- Homi Bhabha National Institute, DAE’s University, Mumbai, Maharashtra
| | - Pradeep Thapa
- Radiation Medicine Centre Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai, Maharashtra
- Homi Bhabha National Institute, DAE’s University, Mumbai, Maharashtra
| | - Anant Ramaswamy
- Homi Bhabha National Institute, DAE’s University, Mumbai, Maharashtra
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra
| | - Vikas Ostwal
- Homi Bhabha National Institute, DAE’s University, Mumbai, Maharashtra
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra
| | - Bhawna Sirohi
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra
- Department of Medical Oncology, Max Institute of Cancer Care, New Delhi, India
| | - Dipanjan Panda
- Department of Medical Oncology, Indraprastha Apollo Hospital, New Delhi, India
| | - Shailesh V Shrikhande
- Department of Surgical Oncology, Gastrointestinal and Hepato-Pancreato-Biliary Service, Tata Memorial Hospital, Mumbai, Maharashtra
| |
Collapse
|
177
|
Chen KS, Lawhn-Heath C, Behr S, Juarez R, Whitman J, Paciorek A, Nakakura EK, Fidelman N, Feng MUS, Bergsland EK, Anwar M. Outcomes after high-dose radiation in the management of neuroendocrine neoplasms. PLoS One 2021; 16:e0252574. [PMID: 34077464 PMCID: PMC8171937 DOI: 10.1371/journal.pone.0252574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/18/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Neuroendocrine neoplasms (NENs) comprise a rare and heterogenous group of cancers, for which the role of radiation therapy continues to evolve. The purpose of this study is to analyze oncologic outcomes after the use of high-dose radiation in management of NENs at a tertiary hospital. MATERIALS AND METHODS We performed a retrospective review of patients who received high-dose radiation with intent to cure or provide durable local control (defined as biologically effective dose (BED) ≥40, α/β = 10) for a localized or metastatic NEN from 2006 to 2019. Evaluation of disease status after radiation was performed according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria when possible. Patients were grouped by differentiation (well-differentiated (WD) or poorly-differentiated (PD)) and stage (localized/locally advanced disease (L) or metastatic (M)) in analysis of probabilities of progression after radiation. RESULTS 45 patients completed a radiation course with BED ≥40 for a NEN (median BED 72). With a median follow-up of 24 months after radiation, the 2-year actuarial rates of local relapse-free survival, new metastasis-free survival, progression-free survival, and overall survival after radiation were 98%, 45%, 41%, and 69%, respectively. 25 patients (56%) developed new metastases after completion of radiation, including 33% (n = 3) of patients with WD-L disease, 44% (n = 8) of WD-M, 77% (n = 10) of PD-L, and 80% (n = 4) of PD-M, with progressively shorter median times to progression (26, 9, 8, and 3 months, respectively; p = 0.093). Of the 25 patients evaluable by RECIST, 68% (n = 17) achieved either a complete or partial best response in the irradiated lesion. CONCLUSIONS These data suggest that focal, high-dose radiation has a role in the management of selected patients with NENs. Local failure is rare in patients with both well-differentiated and poorly-differentiated disease, although the predominant pattern of failure remains development of new metastases.
Collapse
Affiliation(s)
- Katherine S. Chen
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, United States of America
| | - Courtney Lawhn-Heath
- Department of Radiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Spencer Behr
- Department of Radiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Roxanna Juarez
- Department of Radiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Julia Whitman
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, United States of America
| | - Alan Paciorek
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
| | - Eric K. Nakakura
- Department of Surgery, Division of Surgical Oncology, University of California San Francisco, San Francisco, CA, United States of America
| | - Nicholas Fidelman
- Department of Radiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Mary Uan-Sian Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, United States of America
| | - Emily K. Bergsland
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA, United States of America
| | - Mekhail Anwar
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
178
|
Maharaj M, Heslop L, Govender T, Korowlay N, Singh A, Choudhary P, Sathekge M. The Outcome and Safety of Re-challenge Lutetium-177 PSMA ( 177Lu-PSMA) Therapy with Low-Dose Docetaxel as a Radiosensitizer-a Promising Combination in Metastatic Castrate-Resistant Prostate Cancer (mCRPC): a Case Report. Nucl Med Mol Imaging 2021; 55:136-140. [PMID: 34093893 PMCID: PMC8139998 DOI: 10.1007/s13139-021-00696-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 01/08/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA)-directed radioligand therapy (PSMA-RLT) with lutetium-177 (177Lu-PSMA) has been used in metastatic castrate-resistant prostate cancer (mCRPC), and retrospective data have shown this therapy to be favourably safe with attractive clinical responses. Re-challenge 177Lu-PSMA therapy in early responders has been shown to be safe and effective. We report the use of low-dose Taxol-based chemotherapy (modified dose 25 mg/m2 weekly × 6 weeks) as a radiosensitizer with re-challenge 177Lu-PSMA therapy (4 cycles). In a period of 3 years, the patient underwent a total of 8 cycles of 177Lu-PSMA with a cumulative dose of 51.8 GBq. All therapies were uneventful and well tolerated. There was a good response to re-challenge 177Lu-PSMA therapy and low-dose docetaxel (Taxol-177Lu-PSMA) with no recorded tumour resistance.
Collapse
Affiliation(s)
- Masha Maharaj
- Department of Nuclear Medicine, Imaging and Therapy Centres of Excellence, Durban, KwaZulu Natal South Africa
| | - Lucille Heslop
- Durban Oncology Centre, West Ridge, Durban, South Africa
| | - Trisha Govender
- Department of Nuclear Medicine, Imaging and Therapy Centres of Excellence, Durban, KwaZulu Natal South Africa
| | - Nisaar Korowlay
- Division of Nuclear Medicine, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa
| | - Aviral Singh
- Theranostics Center for Molecular Radiotherapy and Molecular Imaging (PET/CT) ENETS Center of Excellence, Zentralklinik Bad Berka GmbH, Bad Berka, Germany
| | - Partha Choudhary
- Department of Nuclear Medicine, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| |
Collapse
|
179
|
Ebbers SC, Barentsz MW, de Keizer B, Krijger GC, Lam MGEH, Braat AJAT. A Rapid and Safe Infusion Protocol for 177Lu Peptide Receptor Radionuclide Therapy. J Nucl Med 2021; 62:816-822. [PMID: 33246981 DOI: 10.2967/jnumed.120.252494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/29/2020] [Indexed: 11/16/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) with 177Lu-labeled somatostatin analogs in patients with somatostatin receptor-expressing tumors is often performed using administration protocols prescribing a 30-min infusion time. The most often used method of infusion is the gravity method, by which the complete dose is effectively administered exponentially. However, there is no evidence to explicitly support an infusion time of 30 min. This study aims to investigate the safety of an infusion time of less than 5 min. Methods: A cohort study was performed, examining the biochemical and clinical toxicity after PRRT when using a fast-infusion protocol with a maximum infusion time of 5 min. Data on patient characteristics, laboratory tests, follow-up visits, and pre- and posttreatment imaging using 68Ga-DOTATOC PET/CT from patients treated with PRRT at the University Medical Center Utrecht (UMC Utrecht) were collected. All patients receiving PRRT using the fast-infusion protocol were included. If no laboratory or clinical follow-up was available, patients were excluded. In addition, a laboratory experiment was performed, simulating the standard-infusion protocol using the gravity method. Results: Thirty-one patients, treated using the fast-infusion protocol, were included. Clinical toxicity mainly consisted of grade 1/2 fatigue (87.1%) and grade 1 nausea or vomiting (67.7%) during follow-up. No acute or long-term clinical toxicity possibly related to the fast-infusion protocol was reported. Grade 3/4 hematologic toxicity occurred after PRRT in 1 patient (3.2%). No grade 3/4 renal toxicity occurred. The laboratory experiment showed that when using the gravity method for infusion, half of the activity is infused after 3.5 min, and 95% is infused within 15 min. Conclusion: A faster infusion of PRRT using an infusion time of less than 5 min is safe and feasible in clinical practice.
Collapse
Affiliation(s)
- Sander C Ebbers
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands; and
| | - Maarten W Barentsz
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bart de Keizer
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands; and
| | - Gerard C Krijger
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands; and
| | - Marnix G E H Lam
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands; and
| | - Arthur J A T Braat
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands; and
| |
Collapse
|
180
|
Kertels O, Breun M, Hänscheid H, Kircher M, Hartrampf PE, Schirbel A, Monoranu CM, Ernestus RI, Buck AK, Löhr M, Matthies C, Lapa C. Peptide Receptor Radionuclide Therapy in Patients With Neurofibromatosis Type 2: Initial Experience. Clin Nucl Med 2021; 46:e312-e316. [PMID: 33826573 DOI: 10.1097/rlu.0000000000003627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Neurofibromatosis type 2 (NF2) is a genetic disorder that is associated with multiple tumors of the nervous system, and approximately one half of patients present with meningiomas. For patients with multifocal disease, somatostatin receptor-targeted peptide receptor radionuclide therapy (PRRT) might be a suitable systemic treatment option. PATIENTS AND METHODS Between March 2015 and August 2017, 11 NF2 patients (7 females and 4 males; mean age, 39 ± 12 years) with multifocal, progressive meningiomas underwent a median of 4 cycles of PRRT (range, 2-6 cycles). Acute and chronic adverse events were recorded according to National Institutes of Health's Common Toxicity Criteria (CTC) version 5.0. Follow-up MRIs (every 3 to 6 months), using the Response Assessment in Neuro-Oncology response criteria for meningiomas, were used to assess treatment responses. RESULTS Peptide receptor radionuclide therapy was well tolerated in all patients without any relevant acute adverse effects. Transient hematologic toxicity (CTC grade 3) was observed in 2 subjects. Somatostatin receptor-directed radiopeptide therapy resulted in radiological disease stabilization in 6 of 11 patients. Median progression-free survival was 12 months (range, 1-55 months), and overall survival was 37 months (range, 5-61 months). CONCLUSIONS Based on our retrospective pilot data, PRRT is feasible and well-tolerated in NF2 patients. It might offer a suitable treatment option in subjects with multiple, recurrent, or treatment-refractory meningiomas.
Collapse
Affiliation(s)
| | | | | | | | | | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg
| | - Camelia-Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg
| | | | | | | |
Collapse
|
181
|
Abstract
Several studies have demonstrated the effectiveness of somatostatin receptor (SSTR)-targeted imaging for diagnosis, staging, evaluating the possibility of treatment with cold somatostatin analogs, as well peptide receptor radionuclide therapy (PRRT), and evaluation of treatment response. PET with 68Ga-labeled somatostatin analogs provides excellent sensitivity and specificity for diagnosing and staging neuroendocrine tumors (NETs). Metabolic imaging with PET with fludeoxyglucose 18F/computed tomography (CT) complements the molecular imaging with 68Ga-SSTR PET/CT toward a personalized therapy in NET patients. The documented response rate of PRRT in NET summing up complete response, partial response, minor response, and stable disease is 70% to 80%.
Collapse
Affiliation(s)
- Margarida Rodrigues
- Department of Nuclear Medicine, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria.
| | - Hanna Svirydenka
- Department of Nuclear Medicine, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| |
Collapse
|
182
|
Widjaja L, Werner RA, Ross TL, Bengel FM, Derlin T. Comparison of pretherapeutic osseous tumor volume and standard hematology for prediction of hematotoxicity after PSMA-targeted radioligand therapy. Eur J Nucl Med Mol Imaging 2021; 48:4077-4088. [PMID: 34041564 PMCID: PMC8484194 DOI: 10.1007/s00259-021-05412-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Purpose Hematotoxicity is a potentially dose-limiting adverse event in patients with metastasized castration-resistant prostate cancer (mCRPC) undergoing prostate-specific membrane antigen (PSMA)-directed radioligand therapy (RLT). We aimed to identify clinical or PSMA-targeted imaging-derived parameters to predict hematological adverse events at early and late stages in the treatment course. Methods In 67 patients with mCRPC scheduled for 177Lu-PSMA-617 RLT, pretherapeutic osseous tumor volume (TV) from 68Ga-PSMA-11 PET/CT and laboratory values were assessed. We then tested the predictive capability of these parameters for early and late hematotoxicity (according to CTCAE vers. 5.0) after one cycle of RLT and in a subgroup of 32/67 (47.8%) patients after four cycles of RLT. Results After one cycle, 10/67 (14.9%) patients developed leukocytopenia (lymphocytopenia, 39/67 [58.2%]; thrombocytopenia, 17/67 [25.4%]). A cut-off of 5.6 × 103/mm3 for baseline leukocytes was defined by receiver operating characteristics (ROC) and separated between patients with and without leukocytopenia (P < 0.001). Baseline leukocyte count emerged as a stronger predictive factor in multivariate analysis (hazard ratio [HR], 33.94, P = 0.001) relative to osseous TV (HR, 14.24, P = 0.01). After four cycles, 4/32 (12.5%) developed leukocytopenia and the pretherapeutic leukocyte cut-off (HR, 9.97, P = 0.082) tended to predict leukocytopenia better than TV (HR, 8.37, P = 0.109). In addition, a cut-off of 1.33 × 103/mm3 for baseline lymphocytes separated between patients with and without lymphocytopenia (P < 0.001), which was corroborated in multivariate analysis (HR, 21.39, P < 0.001 vs. TV, HR, 4.57, P = 0.03). After four cycles, 19/32 (59.4%) developed lymphocytopenia and the pretherapeutic cut-off for lymphocytes (HR, 46.76, P = 0.007) also demonstrated superior predictive performance for late lymphocytopenia (TV, HR, 5.15, P = 0.167). Moreover, a cut-off of 206 × 103/mm3 for baseline platelets separated between patients with and without thrombocytopenia (P < 0.001) and also demonstrated superior predictive capability in multivariate analysis (HR, 115.02, P < 0.001 vs.TV, HR, 12.75, P = 0.025). After four cycles, 9/32 (28.1%) developed thrombocytopenia and the pretherapeutic cut-off for platelets (HR, 5.44, P = 0.048) was also superior for the occurrence of late thrombocytopenia (TV, HR, 1.44, P = 0.7). Conclusions Pretherapeutic leukocyte, lymphocyte, and platelet levels themselves are strong predictors for early and late hematotoxicity under PSMA-directed RLT, and are better suited than PET-based osseous TV for this purpose.
Collapse
Affiliation(s)
- Liam Widjaja
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
183
|
van Andel L, Aalbersberg EA, Geluk-Jonker MM, Stokkel MPM, Beijnen JH, Hendrikx JJMA. The development and validation of a high performance liquid chromatography method to determine the radiochemical purity of [ 177Lu]Lu-HA-DOTA-TATE in pharmaceutical preparations. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1171:122605. [PMID: 33740691 DOI: 10.1016/j.jchromb.2021.122605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/02/2021] [Accepted: 02/16/2021] [Indexed: 10/22/2022]
Abstract
Lutetium-177 [177Lu] tetra-azacyclododecanetetra-acetic acid [DOTA]-(Tyr3)-octreotate [TATE] ([177Lu]Lu-DOTA-TATE) is a radiopeptide used for peptide receptor radionuclide therapy in patients with neuroendocrine tumours (NETs). This radiopeptide is made by labelling the ligand octreotate with Lutetium-177 using the linker DOTA. After labelling, and before clinical application quality control of the radiopeptide is needed and the radiochemical purity is assessed. Acceptance limits for radiochemical purity should be within 90-110% of the label claim for radiopharmaceuticals for diagnostic use and within 95-105% of the label claim for radiopharmaceuticals for therapeutic use. Moreover, the amount of unlabelled [177Lu]LuCl3 cannot exceed 2% of the radioactive dose. Since no monograph is available for [177Lu]Lu-DOTA-TATE in the European Pharmacopeia (Ph Eur), this article describes the development and validation of a high-performance liquid chromatography (HPLC) method with ultraviolet (UV) detection and radiodetection. A Waters Acquity Arc UHPLC system equipped with a Waters 2998 photodiode array (PDA) detector was used coupled to a Berthold Lb 514 Flowstar detector equipped with a BGO-X gamma measuring cell. A reversed phase Symmetry Shield C18 column (4.6 mm × 250 mm, 5 µm) was used for chromatographic separation. A flow of 1.5 mL/min was maintained during analysis, using 0.1% TFA in water as mobile phase A and 0.1% TFA in ACN as mobile phase B. The retention time was around 1.7 min and 13.5 min for [177Lu]LuCl3 and [177Lu]Lu-HA-DOTA-TATE, respectively. Stock solutions of [177Lu]LuCl3 were made by serial dilution and were injected to test for linearity, accuracy and precision, carry over and signal-to-noise ratio. A [177Lu]Lu-HA-DOTA-TATE sample was prepared and injected to determine the carry over. The results showed that the method is linear over a range of 0.300-130 MBq/mL, which covers the range for clinical samples, provided that the clinical sample is diluted ten times before analysis. The LLOQ can be measured accurately even after dilution, with a signal-to-noise ratio of at least 5. In short, the method is accurate, precise and sensitive and can be implemented as part of the quality control of [177Lu]Lu-HA-DOTA-TATE.
Collapse
Affiliation(s)
- L van Andel
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - E A Aalbersberg
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - M M Geluk-Jonker
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - M P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - J H Beijnen
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht, the Netherlands
| | - J J M A Hendrikx
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands.
| |
Collapse
|
184
|
Lu X, Lu C, Yang Y, Shi X, Wang H, Yang N, Yang K, Zhang X. Current Status and Trends in Peptide Receptor Radionuclide Therapy in the Past 20 Years (2000-2019): A Bibliometric Study. Front Pharmacol 2021; 12:624534. [PMID: 33986664 PMCID: PMC8111084 DOI: 10.3389/fphar.2021.624534] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/15/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Peptide receptor radionuclide therapy (PRRT) is an emerging therapeutic option for the treatment of neuroendocrine tumors (NETs), and the number of publications in this field has been increasing in recent years. The aim of the present study was to present the research status and summarize the key topics through bibliometric analysis of published PRRT literature. Methods: A literature search for PRRT research from 2000 to 2019 was conducted using the Science Citation Index Expanded of Web of Science Core Collection (limited to SCIE) on August 4, 2020. The VOSviewer, R-bibliometrix, and CiteSpace software were used to conduct the bibliometric analysis. Results: From 2000 to 2019, a total of 681 publications (523 articles and 158 reviews) were retrieved. Annual publication outputs grew from three to 111 records. Germany had the largest number of publications, making the largest contribution to the field (n = 151, 22.17%). Active cooperation between countries/regions was observed. Kwekkeboom from the Erasmus Medical Center is perhaps a key researcher in the field of PRRT. The European Journal of Nuclear Medicine and Molecular Imaging and Journal of Nuclear Medicine ranked first for productive (n = 84, 12.33%) and co-cited (n = 3,438) journals, respectively. Important topics mainly included matters related to the efficacy of PRRT (e.g., 90Y-dotatoc and 177Lu-dotatate), the long-term adverse effects of PRRT (e.g., hematologic and renal toxicities), standardization of NETs and PRRT in practice, the development of medical imaging techniques, and the individual dose optimization of PRRT. Conclusion: Using bibliometric analysis, we gained deep insight into the global status and trends of studies investigating PRRT for the first time. The PRRT field is undergoing a period of rapid development, and our study provides a valuable reference for clinical researchers and practitioners.
Collapse
Affiliation(s)
- Xiaojing Lu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cuncun Lu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongjie Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangfen Shi
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haibo Wang
- School of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Nan Yang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Kehu Yang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
185
|
Roll W, Weckesser M, Seifert R, Bodei L, Rahbar K. Imaging and liquid biopsy in the prediction and evaluation of response to PRRT in neuroendocrine tumors: implications for patient management. Eur J Nucl Med Mol Imaging 2021; 48:4016-4027. [PMID: 33903926 PMCID: PMC8484222 DOI: 10.1007/s00259-021-05359-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/05/2021] [Indexed: 12/16/2022]
Abstract
Purpose The aim of this narrative review is to give an overview on current and emerging imaging methods and liquid biopsy for prediction and evaluation of response to PRRT. Current limitations and new perspectives, including artificial intelligence, are discussed. Methods A literature review of PubMed/Medline was performed with representative keywords. The search included articles published online through August 31, 2020. All searches were restricted to English language manuscripts. Results Peptide radio receptor therapy (PRRT) is a prospectively evaluated and approved therapy option in neuroendocrine tumors (NETs). Different ligands targeting the somatostatin receptor (SSTR) are used as theranostic pairs for imaging NET and for PRRT. Response assessment in prospective trials often relies on the morphological RECIST 1.1 criteria, based on lesion size in CT or MRI. The role of SSTR-PET and quantitative uptake parameters and volumetric data is still not defined. Monoanalyte tumor marker chromogranin A has a limited value for response assessment after PRRT. New emerging liquid biopsy techniques are offering prediction of response to PRRT and prognostic value. Conclusions New response criteria for NET patients undergoing PRRT will comprise multiparametric hybrid imaging and blood-based multianalyte markers. This represents tumor biology and heterogeneity.
Collapse
Affiliation(s)
- Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany
| | - Matthias Weckesser
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany.,Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Lisa Bodei
- Department of Nuclear Medicine, Radiology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany. .,West German Cancer Center, Muenster and Essen, Essen, Germany.
| |
Collapse
|
186
|
Radiological and Clinical Efficacy of Intra-Arterial 90Y-DOTATATE in Patients with Unresectable, Progressive, Liver Dominant Neuroendocrine Neoplasms. J Clin Med 2021; 10:jcm10081794. [PMID: 33924160 PMCID: PMC8074370 DOI: 10.3390/jcm10081794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/08/2021] [Accepted: 04/16/2021] [Indexed: 01/14/2023] Open
Abstract
This study was performed to determine if intra-arterial (i.a.) administration of 90Y DOTATATE can provide an effective and safe alternative to the accepted standard for i.v. of peptide receptor radionuclide therapy (PRRT) in liver-dominant metastases of gastrointestinal pancreatic neuroendocrine neoplasm (GEP-NEN). A single site, prospective, preliminary case series study included 39 patients with histologically proven liver-dominant NEN. PRRT in the form of 1.15GBq 90Y DOTATATE was given selectively into the liver via radiological catheterization of the hepatic artery, up to four times. The endpoint was radiological response (RECIST). Secondary endpoints assessed clinical well-being post-treatment, progression-free survival (PFS), overall survival (OS), and toxicity. Partial response (PR) was noted in 13% of subjects six weeks post-therapy, increasing to 24% at six months and dropping to 13% at 36 months. Disease progression (DP) was not seen at six weeks, was 5% at six months, and 47% at 36 months. Clinical response based on PS seen in 74% of patients at six weeks, 69% at six months, and 39% at 36 months had PFS and OS, respectively, of 22.7 months and 38.2 months. There was no difference in OS/PFS between those with RECIST PR and SD. One patient had significant toxicity (3%). Use of i.a. PRRT appears to be safe and effective in treating patients with liver-dominant NEN. In addition, the best OS (51 vs. 22 months) was seen when i.a. was used as an upfront treatment of bulky GEP-NEN liver metastases and not after i.v. 90Y DOTATATE. The use of i.a. 90Y DOTATATE PRRT appears to be safe and effective in treating patients with liver-dominant NEN.
Collapse
|
187
|
Simultaneous Visualization of 161Tb- and 177Lu-Labeled Somatostatin Analogues Using Dual-Isotope SPECT Imaging. Pharmaceutics 2021; 13:pharmaceutics13040536. [PMID: 33921467 PMCID: PMC8070648 DOI: 10.3390/pharmaceutics13040536] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/29/2021] [Accepted: 04/07/2021] [Indexed: 11/17/2022] Open
Abstract
The decay of terbium-161 results in the emission of β¯-particles as well as conversion and Auger electrons, which makes terbium-161 interesting for therapeutic purposes. The aim of this study was to use dual-isotope SPECT imaging in order to demonstrate visually that terbium-161 and lutetium-177 are interchangeable without compromising the pharmacokinetic profile of the radiopharmaceutical. The 161Tb- and 177Lu-labeled somatostatin (SST) analogues DOTATOC (agonist) and DOTA-LM3 (antagonist) were tested in vitro to demonstrate equal properties regarding distribution coefficients and cell uptake into SST receptor-positive AR42J tumor cells. The radiopeptides were further investigated in AR42J tumor-bearing nude mice using the method of dual-isotope (terbium-161/lutetium-177) SPECT/CT imaging to enable the visualization of their distribution profiles in the same animal. Equal pharmacokinetic profiles were demonstrated for either of the two peptides, irrespective of whether it was labeled with terbium-161 or lutetium-177. Moreover, the visualization of the sub-organ distribution confirmed similar behavior of 161Tb- and 177Lu-labeled SST analogues. The data were verified in quantitative biodistribution studies using either type of peptide labeled with terbium-161 or lutetium-177. While the radionuclide did not have an impact on the organ distribution, this study confirmed previous data of a considerably higher tumor uptake of radiolabeled DOTA-LM3 as compared to the radiolabeled DOTATOC.
Collapse
|
188
|
Toyama Y, Werner RA, Ruiz-Bedoya CA, Ordonez AA, Takase K, Lapa C, Jain SK, Pomper MG, Rowe SP, Higuchi T. Current and future perspectives on functional molecular imaging in nephro-urology: theranostics on the horizon. Theranostics 2021; 11:6105-6119. [PMID: 33897902 PMCID: PMC8058716 DOI: 10.7150/thno.58682] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/21/2021] [Indexed: 01/01/2023] Open
Abstract
In recent years, a paradigm shift from single-photon-emitting radionuclide radiotracers toward positron-emission tomography (PET) radiotracers has occurred in nuclear oncology. Although PET-based molecular imaging of the kidneys is still in its infancy, such a trend has emerged in the field of functional renal radionuclide imaging. Potentially allowing for precise and thorough evaluation of renal radiotracer urodynamics, PET radionuclide imaging has numerous advantages including precise anatomical co-registration with CT images and dynamic three-dimensional imaging capability. In addition, relative to scintigraphic approaches, PET can allow for significantly reduced scan time enabling high-throughput in a busy PET practice and further reduces radiation exposure, which may have a clinical impact in pediatric populations. In recent years, multiple renal PET radiotracers labeled with 11C, 68Ga, and 18F have been utilized in clinical studies. Beyond providing a precise non-invasive read-out of renal function, such radiotracers may also be used to assess renal inflammation. This manuscript will provide an overview of renal molecular PET imaging and will highlight the transformation of conventional scintigraphy of the kidneys toward novel, high-resolution PET imaging for assessing renal function. In addition, future applications will be introduced, e.g. by transferring the concept of molecular image-guided diagnostics and therapy (theranostics) to the field of nephrology.
Collapse
Affiliation(s)
- Yoshitaka Toyama
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Department of Diagnostic Radiology, Tohoku University, Sendai, Japan
| | - Rudolf A. Werner
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Wuerzburg, Wuerzburg Germany
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camilo A. Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alvaro A. Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kei Takase
- Department of Diagnostic Radiology, Tohoku University, Sendai, Japan
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Sanjay K. Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin G. Pomper
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven P. Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Wuerzburg, Wuerzburg Germany
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
189
|
Lodge MA, Solnes LB, Chaudhry MA, Wahl RL. Prospective Within-Patient Assessment of the Impact of an Unlabeled Octreotide Pre-dose on the Biodistribution and Tumor Uptake of 68Ga DOTATOC as Assessed by Dynamic Whole-body PET in Patients with Neuroendocrine Tumors: Implications for Diagnosis and Therapy. Mol Imaging Biol 2021; 23:766-774. [PMID: 33829361 DOI: 10.1007/s11307-021-01600-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Gastroenteropancreatic neuroendocrine tumors (GEP NETs) are often associated with high expression of somatostatin receptors (SSTRs) which allows for PET/CT imaging with radiolabeled somatostatin analogs such as 68Ga-DOTATOC. The interplay between 68Ga-DOTATOC and the synthetic somatostatin analogs commonly used to manage patient symptoms may lead to competition between the labelled and unlabeled peptides for receptor binding sites and current product labelling recommends patients be taken off somatostatin analogs before imaging. In this study, we prospectively investigated in human patients the effect of a pre-dose of octreotide, a short-acting somatostatin analog, on the distribution of 68Ga-DOTATOC in GEP NETs and normal organs. PROCEDURE Research participants with GEP NETs were studied on two occasions using dynamic whole-body 68Ga-DOTATOC PET/CT. The two imaging studies were performed within 21 days of each other, using an identical acquisition protocol except for the administration of 50 μg of short-acting octreotide (pre-dose) immediately before the second PET/CT. Paired t-tests were used to compare tracer uptake with and without octreotide, for tumor and various normal organs. RESULTS Seven participants with a mean age of 53 ± 10 years were studied. Octreotide pre-dosing decreased radiotracer uptake in the normal liver and spleen by 25 % (p = 0.04) and 47 % (p = 0.05) respectively but did not significantly change uptake in tumor (p = 0.53), red marrow (p = 0.12), kidneys (p =0.57), or pituitary gland (p = 0.27). CONCLUSIONS Our data indicate SSTR imaging can be improved with a pre-dose of unlabeled octreotide given just prior to injection of the radiotracer. These data suggest there may be no need to discontinue somatostatin analog therapy prior to PET/CT with 68Ga-DOTATOC, allowing for a simpler, less disruptive patient protocol. This approach warrants further study in a variety of settings.
Collapse
Affiliation(s)
- Martin A Lodge
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, 21287, USA.
| | - Lilja B Solnes
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Muhammad A Chaudhry
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Richard L Wahl
- The Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
190
|
Tylski P, Pina-Jomir G, Bournaud-Salinas C, Jalade P. Tissue dose estimation after extravasation of 177Lu-DOTATATE. EJNMMI Phys 2021; 8:33. [PMID: 33788043 PMCID: PMC8012450 DOI: 10.1186/s40658-021-00378-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 03/16/2021] [Indexed: 12/31/2022] Open
Abstract
Background Extravasation of radiopharmaceuticals used for vectorized internal radiotherapy can lead to severe tissue damage (van der Pol et al., Eur J Nucl Med Mol Imaging 44:1234–1243, 2017). Clinical management of these extravasations requires the preliminary estimation of the dose distribution in the extravasation area. Data are scarce regarding the dose estimation in the literature. This work presents a methodology for estimating the dose distribution after an extravasation occurred in September 2017, in the arm of a patient during a 7.4-GBq infusion of Lutathera ® (AAA). Methods A local quantification procedure initially developed for renal dosimetry was used. A calibration factor was determined and verified by phantom study. Extravasation volume of interest and its variation in time were determined using 4 whole body (WB) planar acquisitions performed at 2 h (T2h), 5 h (T5h), 20 h (T20h), and 26 h (T26h) after the beginning of the infusion and three SPECT/CT thoracic acquisitions at T5h, T20h, and T26h. For better estimation of initial extravasation volume, 3 volumes were defined on SPECT images using a 3D activity threshold. Cumulated activities and associated absorbed doses (D1, D2, D3) were calculated in the 3 volumes using the MIRD formalism. Results Volumes estimated using 3D threshold were V1 = 1000 mL, V2 =400 mL, and V3 =180 mL. Cumulated activities were evaluated using a monoexponential fit on activities calculated on SPECT images. Estimated local absorbed doses in V1, V2, and V3 were D1 = 2.3 Gy, D2 = 4.1 Gy, and D3 = 6.8 Gy. Evolution in time of local activity in the extravasation area was consistent with an effective local half-life (Teff) of 2.3 h. Conclusions Rapid local dose estimation was permitted thanks to knowledge of the calibration factor determined previous to accidental extravasation. Lutathera® lymphatic drainage was quick in the arm (Teff = 2.3h). Estimated doses were in the lower range of deterministic effects and far under soft tissue necrosis threshold. Thus, no surgical rinse was proposed. The patient did not show any clinical consequence of the extravasation.
Collapse
Affiliation(s)
- Perrine Tylski
- Service de Physique Médicale et Radioprotection, Hospices Civils de Lyon, Lyon, France.
| | - Géraldine Pina-Jomir
- Service de Médecine Nucléaire, Groupement Hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Claire Bournaud-Salinas
- Service de Médecine Nucléaire, Groupement Hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Patrice Jalade
- Service de Physique Médicale et Radioprotection, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
191
|
Derlin T, Bogdanova N, Ohlendorf F, Ramachandran D, Werner RA, Ross TL, Christiansen H, Bengel FM, Henkenberens C. Assessment of γ-H2AX and 53BP1 Foci in Peripheral Blood Lymphocytes to Predict Subclinical Hematotoxicity and Response in Somatostatin Receptor-Targeted Radionuclide Therapy for Advanced Gastroenteropancreatic Neuroendocrine Tumors. Cancers (Basel) 2021; 13:cancers13071516. [PMID: 33806081 PMCID: PMC8036952 DOI: 10.3390/cancers13071516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/16/2021] [Accepted: 03/20/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND We aimed to characterize γ-H2AX and 53BP1 foci formation in patients receiving somatostatin receptor-targeted radioligand therapy, and explored its role for predicting treatment-related hematotoxicity, and treatment response. METHODS A prospective analysis of double-strand break (DSB) markers was performed in 21 patients with advanced gastroenteropancreatic neuroendocrine tumors. γ-H2AX and 53BP1 foci formation were evaluated in peripheral blood lymphocytes (PBLs) at baseline, +1 h and +24 h after administration of 7.4 GBq (177Lu)Lu-DOTA-TATE. Hematotoxicity was evaluated using standard hematology. Therapy response was assessed using (68Ga)Ga-DOTA-TATE PET/CT before enrollment and after 2 cycles of PRRT according to the volumetric modification of RECIST 1.1. RESULTS DSB marker kinetics were heterogeneous among patients. Subclinical hematotoxicity was associated with γ-H2AX and 53BP1 foci formation (e.g., change in platelet count vs change in γ-H2AX+ cells between baseline and +1 h (r = -0.6080; p = 0.0045). Patients showing early development of new metastases had less γ-H2AX (p = 0.0125) and less 53BP1 foci per cell at +1 h (p = 0.0289), and demonstrated a distinct kinetic pattern with an absence of DSB marker decrease at +24 h (γ-H2AX: p = 0.0025; 53BP1: p = 0.0008). CONCLUSIONS Assessment of γ-H2AX and 53BP1 foci formation in PBLs of patients receiving radioligand therapy may hold promise for predicting subclinical hematotoxicity and early treatment response.
Collapse
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.O.); (R.A.W.); (T.L.R.); (F.M.B.)
- Correspondence: ; Tel.: +49-(0)5115322579; Fax: +49-(0)5115323761
| | - Natalia Bogdanova
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany; (N.B.); (H.C.); (C.H.)
| | - Fiona Ohlendorf
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.O.); (R.A.W.); (T.L.R.); (F.M.B.)
| | - Dhanya Ramachandran
- Department of Radiation Oncology, and Gynaecology Research Unit, Hannover Medical School, 30625 Hannover, Germany;
| | - Rudolf A. Werner
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.O.); (R.A.W.); (T.L.R.); (F.M.B.)
| | - Tobias L. Ross
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.O.); (R.A.W.); (T.L.R.); (F.M.B.)
| | - Hans Christiansen
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany; (N.B.); (H.C.); (C.H.)
| | - Frank M. Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.O.); (R.A.W.); (T.L.R.); (F.M.B.)
| | - Christoph Henkenberens
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany; (N.B.); (H.C.); (C.H.)
| |
Collapse
|
192
|
Nephrotoxicity/renal failure after therapy with 90Yttrium- and 177Lutetium-radiolabeled somatostatin analogs in different types of neuroendocrine tumors: a systematic review. Nucl Med Commun 2021; 41:601-617. [PMID: 32404645 DOI: 10.1097/mnm.0000000000001198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVE Data regarding the nephrotoxicity of the peptide receptor radionuclide therapy (PRRT) with Yttrium- and Lutetium-radiolabeled somatostatin analogs (RSA) are inconclusive. We aimed to evaluate the short- and long-term nephrotoxicity following PRRT usage in patients with all types of neuroendocrine tumors (NETs). METHODS A systematic review of observational studies reporting data about nephrotoxicity after treatment with Yttrium and Lutetium RSA was performed. Data on serum creatinine, creatinine clearance, glomerular filtration rate (GFR) and need for renal replacement therapy were compiled. We included patients with progressive, inoperable symptomatic G1, G2 and G3 different types of NETs. After searching in three electronic databases PubMed, Scopus and the Cochrane Library, from 1 January 1978 to November 2018, data were extracted and summarized using a random-effects model. RESULTS The final analysis included 34 studies, comprising 5386 participants, enrolling patients with G1, G2, G3 NETs and a follow-up from 12 up to 191 months. Compared with renal function before treatment, measured/estimated glomerular filtration rate (m/eGFR) values changed after PRRT, with a mean annual decrease following PRRT between 2 and 4 mL/min/1.73 m suggesting different grades of nephrotoxicity after PRRT. When compared, Y-RSA and the Y-RSA-Lu-RSA combination are associated with a higher m/eGFR decline compared to Lu-RSA alone. CONCLUSIONS PRRT can be followed by potentially serious long-term nephrotoxicity, despite kidney protection. The use of the quantified renal function combined with a long follow-up period and personalized dosimetry-based PRRT can reduce nephrotoxicity, in order to use the whole PRRT potential in the management of NETs.
Collapse
|
193
|
Ramonaheng K, van Staden JA, du Raan H. The effect of calibration factors and recovery coefficients on 177Lu SPECT activity quantification accuracy: a Monte Carlo study. EJNMMI Phys 2021; 8:27. [PMID: 33738605 PMCID: PMC7973313 DOI: 10.1186/s40658-021-00365-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Different gamma camera calibration factor (CF) geometries have been proposed to convert SPECT data into units of activity concentration. However, no consensus has been reached on a standardised geometry. The CF is dependent on the selected geometry and is further affected by partial volume effects. This study investigated the effect of two CF geometries and their corresponding recovery coefficients (RCs) on the quantification accuracy of 177Lu SPECT images using Monte Carlo simulations. METHODS The CF geometries investigated were (i) a radioactive-sphere surrounded by non-radioactive water (sphere-CF) and (ii) a cylindrical phantom uniformly filled with radioactive water (cylinder-CF). Recovery coefficients were obtained using the sphere-CF and cylinder-CF, yielding the sphere-RC and cylinder-RC values, respectively, for partial volume correction (PVC). The quantification accuracy was evaluated using four different-sized spheres (15.6-65.4 ml) and a kidney model with known activity concentrations inside a cylindrical, torso and patient phantom. Images were reconstructed with the 3D OS-EM algorithm incorporating attenuation, scatter and detector-response corrections. Segmentation was performed using the physical size and a small cylindrical volume inside the cylinder for the sphere-CF and cylinder-CF, respectively. RESULTS The sphere quantification error (without PVC) was better for the sphere-CF (≤ - 5.54%) compared to the cylinder-CF (≤ - 20.90%), attributed to the similar geometry of the quantified and CF spheres. Partial volume correction yielded comparable results for the sphere-CF-RC (≤ 3.47%) and cylinder-CF-RC (≤ 3.53%). The accuracy of the kidney quantification was poorer (≤ 22.34%) for the sphere-CF without PVC compared to the cylinder-CF (≤ 2.44%). With PVC, the kidney quantification results improved and compared well for the sphere-CF-RC (≤ 3.50%) and the cylinder-CF-RC (≤ 3.45%). CONCLUSION The study demonstrated that upon careful selection of CF-RC combinations, comparable quantification errors (≤ 3.53%) were obtained between the sphere-CF-RC and cylinder-CF-RC, when all corrections were applied.
Collapse
Affiliation(s)
- Keamogetswe Ramonaheng
- Department of Medical Physics, Faculty of Health Sciences, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa.
| | - Johannes A van Staden
- Department of Medical Physics, Faculty of Health Sciences, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa
| | - Hanlie du Raan
- Department of Medical Physics, Faculty of Health Sciences, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa
| |
Collapse
|
194
|
Prognostic and Theranostic Applications of Positron Emission Tomography for a Personalized Approach to Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2021; 22:ijms22063036. [PMID: 33809749 PMCID: PMC8002334 DOI: 10.3390/ijms22063036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 01/25/2023] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) represents a condition of progressive disease in spite of androgen deprivation therapy (ADT), with a broad spectrum of manifestations ranging from no symptoms to severe debilitation due to bone or visceral metastatization. The management of mCRPC has been profoundly modified by introducing novel therapeutic tools such as antiandrogen drugs (i.e., abiraterone acetate and enzalutamide), immunotherapy through sipuleucel-T, and targeted alpha therapy (TAT). This variety of approaches calls for unmet need of biomarkers suitable for patients’ pre-treatment selection and prognostic stratification. In this scenario, imaging with positron emission computed tomography (PET/CT) presents great and still unexplored potential to detect specific molecular and metabolic signatures, some of whom, such as the prostate specific membrane antigen (PSMA), can also be exploited as therapeutic targets, thus combining diagnosis and therapy in the so-called “theranostic” approach. In this review, we performed a web-based and desktop literature research to investigate the prognostic and theranostic potential of several PET imaging probes, such as 18F-FDG, 18F-choline and 68Ga-PSMA-11, also covering the emerging tracers still in a pre-clinical phase (e.g., PARP-inhibitors’ analogs and the radioligands binding to gastrin releasing peptide receptors/GRPR), highlighting their potential for defining personalized care pathways in mCRPC.
Collapse
|
195
|
Minczeles NS, Hofland J, de Herder WW, Brabander T. Strategies Towards Improving Clinical Outcomes of Peptide Receptor Radionuclide Therapy. Curr Oncol Rep 2021; 23:46. [PMID: 33721105 PMCID: PMC7960621 DOI: 10.1007/s11912-021-01037-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Peptide receptor radionuclide therapy (PRRT) with [177Lu-DOTA0,Tyr3] octreotate is an effective and safe second- or third-line treatment option for patients with low-grade advanced gastroenteropancreatic (GEP) neuroendocrine neoplasms (NEN). In this review, we will focus on possible extensions of the current use of PRRT and on new approaches which could further improve its treatment efficacy and safety. RECENT FINDINGS Promising results were published regarding PRRT in other NENs, including lung NENs or high-grade NENs, and applying PRRT as neoadjuvant or salvage therapy. Furthermore, a diversity of strategic approaches, including dosimetry, somatostatin receptor antagonists, somatostatin receptor upregulation, radiosensitization, different radionuclides, albumin binding, alternative renal protection, and liver-directed therapy in combination with PRRT, have the potential to improve the outcome of PRRT. Also, novel biomarkers are presented that could predict response to PRRT. Multiple preclinical and early clinical studies have shown encouraging potential to advance the clinical outcome of PRRT in NEN patients. However, at this moment, most of these strategies have not yet reached the clinical setting of randomized phase III trials.
Collapse
Affiliation(s)
- N S Minczeles
- Department of Internal Medicine, Section of Endocrinology, Erasmus MC and Erasmus MC Cancer Center, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- ENETS Center of Excellence Rotterdam, Rotterdam, The Netherlands
| | - J Hofland
- Department of Internal Medicine, Section of Endocrinology, Erasmus MC and Erasmus MC Cancer Center, Rotterdam, The Netherlands
- ENETS Center of Excellence Rotterdam, Rotterdam, The Netherlands
| | - W W de Herder
- Department of Internal Medicine, Section of Endocrinology, Erasmus MC and Erasmus MC Cancer Center, Rotterdam, The Netherlands
- ENETS Center of Excellence Rotterdam, Rotterdam, The Netherlands
| | - T Brabander
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
- ENETS Center of Excellence Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
196
|
Predictive and Prognostic Impact of Blood-Based Inflammatory Biomarkers in Patients with Gastroenteropancreatic Neuroendocrine Tumors Commencing Peptide Receptor Radionuclide Therapy. Diagnostics (Basel) 2021; 11:diagnostics11030504. [PMID: 33809226 PMCID: PMC8000284 DOI: 10.3390/diagnostics11030504] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/03/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment inflammation contributes to the proliferation and survival of malignant cells, angiogenesis, metastasis, subversion of adaptive immunity, and reduced treatment response. We aimed to evaluate the early predictive and prognostic significance of markers of systemic inflammation in patients receiving somatostatin-receptor targeted peptide receptor radionuclide therapy (PRRT). This retrospective observational cohort study included 33 patients with advanced gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs) treated with PRRT. Pretreatment blood-based inflammatory biomarkers, e.g., C-reactive protein levels (CRP), white blood cell count (WBC), and absolute neutrophil count (ANC), were documented and inflammation indexes, e.g., neutrophil-lymphocyte ratio (NLR) and Platelet × CRP multiplier (PCM), were calculated. Tumor burden was determined using [68Ga]Ga-DOTA-TATE PET/CT before enrollment and every 2 cycles thereafter until progression. Therapy response was assessed using RECIST 1.1, including its volumetric modification. Inflammatory biomarkers and inflammatory indexes demonstrated marked heterogeneity among patients, and were significantly higher in non-responders (e.g., CRP (p < 0.001), ANC (p = 0.002), and PCM (p < 0.001)). Change in whole-body tumor burden after two cycles of PRRT was significantly associated with CRP (p = 0.0157) and NLR (p = 0.0040) in multivariate regression analysis. A cut-off of 2.5 mg/L for CRP (AUC = 0.84, p = 0.001) revealed a significant outcome difference between patients with adversely high vs. low CRP (median PFS 508 days vs. not yet reached (HR = 4.52; 95% CI, 1.27 to 16.18; p = 0.02)). Tumor-driven systemic inflammatory networks may be associated with treatment response, change in tumor burden, and prognosis in patients with GEP-NETs receiving PRRT.
Collapse
|
197
|
Peptide Receptor Radionuclide Therapy of Pulmonary Neuroendocrine Neoplasms: a Single-Centre Experience. Nucl Med Mol Imaging 2021; 55:38-45. [PMID: 33643488 DOI: 10.1007/s13139-020-00679-y] [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: 07/21/2020] [Revised: 11/25/2020] [Accepted: 12/07/2020] [Indexed: 10/22/2022] Open
Abstract
Purpose Peptide receptor radionuclide therapy represents a therapeutic option for neuroendocrine neoplasms; to date, experiences with peptide receptor radionuclide therapy of pulmonary neuroendocrine neoplasms are still limited. We report our experience with peptide receptor radionuclide therapy of pulmonary neuroendocrine neoplasm patients. Materials and Methods Clinical records of 14 pulmonary neuroendocrine neoplasm patients (7 female and 7 male) who received at least 2 cycles of peptide receptor radionuclide therapy were retrospectively reviewed. Tumoural uptake of somatostatin analogues at pre-treatment imaging was graded as 2 to 3 in all patients. RECIST criteria were used to evaluate response. Results No treated patient had significant toxicity. Partial response was found in 3 (21.4%) patients, stable disease in 7 (50%), and progressive disease in 4 (28.6%). A statistically significant difference between disease state at enrolment and after peptide receptor radionuclide therapy was found. Conclusions Our data furtherly support peptide receptor radionuclide therapy as a safe and effective treatment of patients affected by pulmonary neuroendocrine neoplasms allowing disease control in about 71% of patients without showing significant toxicity. Other studies are needed to confirm our results.
Collapse
|
198
|
Zacho MD, Iversen P, Villadsen GE, Baunwall SMD, Arveschoug AK, Grønbaek H, Dam G. Clinical efficacy of first and second series of peptide receptor radionuclide therapy in patients with neuroendocrine neoplasm: a cohort study. Scand J Gastroenterol 2021; 56:289-297. [PMID: 33470864 DOI: 10.1080/00365521.2021.1872095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Peptide receptor radionuclide therapy (PRRT) is an established treatment for metastatic neuroendocrine neoplasms (NEN). However, only limited data exists for the effect of multiple series of PRRT. The aim of this study was to investigate PFS and OS inNEN patients treated with multiple series of PRRT conforming to the ENETS treatment protocol. METHODS We included all patients with gastrointestinal (GI), pancreatic and bronchopulmonary (BP) NEN treated with PRRT from 2008 to 2018. We used Kaplan-Meier estimation to evaluate PFS and OS with subgroup analysis of primary tumor, Ki67-index, type of radioisotope and number of PRRT series. RESULTS 133 patients (female/male 61/72) were included, median age 70 (interquartile range 64-76) years. GI-NEN comprised 62%, pancreatic 23% and BP 11%. Median Ki67-index was 5%. After first PRRTG1- and G2-tumors had PFS of 25 and 22 months, compared to 11 months in G3-NENs (p < .05) and PFS was longer in G1/G2 GI-NENs than BP-NEN (30vs. 12 months, p < .05). After retreatment with a second series of PRRT, the overall PFS (G1-G3) was 19 months, with G1- and G2-tumors having the highest PFS of 19 and 22 months, respectively. Overall, the GI and BP tumors had an OS of 54 and 51 months. CONCLUSIONS PRRT is an effective therapy with long-term PFS and OS, especially in G1 and G2 NENs, and with better prognosis in GI-NEN compared with BP-NENs. OS and PFS was shorter after the second series of PRRT compared with the first, however results were still encouraging.
Collapse
Affiliation(s)
- M D Zacho
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - P Iversen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - G E Villadsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - S M D Baunwall
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - A K Arveschoug
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - H Grønbaek
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - G Dam
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
199
|
Loharkar S, Basu S. Case work-up and monitoring of systemic radionuclide therapies: A proposed 3-sheet excel format with integrated graph for implementation in a busy treatment set-up. World J Nucl Med 2021; 19:447-451. [PMID: 33623523 PMCID: PMC7875039 DOI: 10.4103/wjnm.wjnm_98_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 11/04/2022] Open
Affiliation(s)
- Sarvesh Loharkar
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
200
|
Bıçakçı N, Elli M. 18Fluorine-fluorodeoxyglucose PET/CT Imaging in Childhood Malignancies. Mol Imaging Radionucl Ther 2021; 30:18-27. [PMID: 33586403 PMCID: PMC7885281 DOI: 10.4274/mirt.galenos.2020.64436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objectives: The aim of the study was to evaluate the utility of 18fluorine-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) in the diagnosis, staging, restaging, and treatment response of childhood malignancies. Methods: This study included 52 patients (32 boys, 20 girls) who were referred to our clinic between November 2008 and December 2018 with the diagnosis of malignancy. The patients were evaluated retrospectively. Median age of the patients was 13 years (range 2-17). 18F-FDG was given to the patients intravenously, and time of flight with PET/16 slice CT was performed 1 hour thereafter. The lowest dose was 2 mCi (74 MBq) and the highest dose was 10 mCi (370 MBq). Fasting blood sugars of all patients were found below 200 mg/dL (11.1 mmol/L). Results: 18F-FDG PET/CT was performed to evaluate the response to treatment in 38 of 52 children, staging in 11 patients (staging and evaluation of the response to treatment in nine of them), restaging in 2 patients, restaging, and evaluation of the response to treatment in 1 patient. 18F-FDG PET/CT examination was reported as normal in 13 patients (5 girls, 8 boys). The pathological 18F-FDG uptake was detected in 39 patients (14 girls, 25 boys), which indicated metastasis and/or recurrence of the primary disease. Total number of deaths was 30 (13 girls, 17 boys). Conclusion: 18F-FDG PET/CT has a significant role for staging, restaging, treatment response, and detection of metastatic disease but it is limited for the early diagnosis of childhood cancers
Collapse
Affiliation(s)
- Nilüfer Bıçakçı
- University of Health Sciences Turkey, Samsun Training and Research Hospital, Clinic of Nuclear Medicine, Samsun, Turkey
| | - Murat Elli
- İstanbul Medipol University Faculty of Medicine, Department of Pediatric Oncology, İstanbul, Turkey
| |
Collapse
|