73 results on '"Mark Konijnenberg"'
Search Results
2. Overcoming nephrotoxicity in peptide receptor radionuclide therapy using [177Lu]Lu-DOTA-TATE for the treatment of neuroendocrine tumours
- Author
-
Lorain Geenen, Sarah Baatout, Julie Nonnekens, Marion de Jong, An Aerts, and Mark Konijnenberg
- Subjects
EXPRESSION ,Cancer Research ,Receptor complex ,Kidney ,LU-177-DOTATATE ,GEP-NETs ,Nephrotoxicity ,chemistry.chemical_compound ,SDG 3 - Good Health and Well-being ,Medicine and Health Sciences ,medicine ,Somatostatin receptor 2 ,Radiology, Nuclear Medicine and imaging ,DOTA-TATE ,Science & Technology ,business.industry ,Radiology, Nuclear Medicine & Medical Imaging ,RADIOLABELED PEPTIDES ,DOSIMETRY ,[Lu-177]Lu-DOTA-TATE ,Renal protection ,HEMATOLOGICAL TOXICITY ,SOMATOSTATIN ANALOGS ,medicine.anatomical_structure ,Cell killing ,chemistry ,Radionuclide therapy ,KIDNEYS ,Cancer research ,RADIATION ,Molecular Medicine ,PRRT ,Bone marrow ,RENAL UPTAKE ,business ,Life Sciences & Biomedicine - Abstract
Peptide receptor radionuclide therapy (PRRT) is used for the treatment of patients with unresectable or metastasized somatostatin receptor type 2 (SSTR2)-expressing gastroenteropancreatic neuroendocrine tumours (GEP-NETs). The radiolabelled somatostatin analogue [177Lu]Lu-DOTA-TATE delivers its radiation dose to SSTR2-overexpressing tumour cells, resulting in selective cell killing during radioactive decay. While tumour control can be achieved in many patients, complete remissions remain rare, causing the majority of patients to relapse after a certain period of time. This raises the question whether the currently fixed treatment regime (4 × 7.4 GBq) leaves room for dose escalation as a means of improving therapy efficacy. The kidneys have shown to play an important role in defining a patient's tolerability to PRRT. As a consequence of the proximal tubular reabsorption of [177Lu]Lu-DOTA-TATE, via the endocytic megalin/cubilin receptor complex, the radionuclides are retained in the renal interstitium. This results in extended retention of radioactivity in the kidneys, generating a risk for the development of radiation nephropathy. In addition, a decreased kidney function has shown to be associated with a prolonged circulation of [177Lu]Lu-DOTA-TATE, causing increased irradiation to the bone marrow. This can on its turn lead to myelosuppression and haematological toxicity, owing to the marked radio sensitivity of the rapidly proliferating cells in the bone marrow. In contrast to external beam radiotherapy (EBRT), the exact absorbed dose limits for these critical organs (kidneys and bone marrow) in PRRT with [177Lu]Lu-DOTA-TATE are still unclear. Better insights into these uncertainties, can help in optimizing PRRT to reach its maximum therapeutic potential, while avoiding severe adverse events, like nephropathy and hematologic toxicities. In this review we focus on the nephrotoxic effects of PRRT with [177Lu]Lu-DOTA-TATE for the treatment of GEP-NETs. If the absorbed dose to the kidneys can be lowered, higher activities can be administered, enlarging the therapeutic window for PRRT. Therefore, we evaluated the renal protective potential of current and promising future strategies and discuss the importance of (renal) dosimetry in PRRT. ispartof: NUCLEAR MEDICINE AND BIOLOGY vol:102 pages:1-11 ispartof: location:United States status: published
- Published
- 2021
- Full Text
- View/download PDF
3. An EANM position paper on advancing radiobiology for shaping the future of nuclear medicine
- Author
-
Jean-Pierre, Pouget, Mark, Konijnenberg, Uta, Eberlein, Gerhard, Glatting, Pablo Minguez, Gabina, Ken, Herrmann, Søren, Holm, Lidia, Strigari, Fijs W B, van Leeuwen, and Michael, Lassmann
- Published
- 2022
4. Albutate-1, a Novel Long-Circulating Radiotracer Targeting the Somatostatin Receptor Subtype 2
- Author
-
Sandra van Tiel, Theodosia Maina, Berthold Nock, Mark Konijnenberg, Erik de Blois, Yann Seimbille, Monique Bernsen, and Marion de Jong
- Subjects
DOTA-TATE ,Biodistribution ,chemistry.chemical_compound ,Therapeutic index ,medicine.anatomical_structure ,Chemistry ,In vivo ,Somatostatin receptor ,medicine ,DOTA ,Somatostatin receptor 2 ,Bone marrow ,Pharmacology - Abstract
Currently, radiolabeled DOTA-[Tyr3]-octreotate (DOTA-TATE) is most commonly used in the clinic to image and treat neuroendocrine tumors. To further improve tumor uptake, and thus treatment, the amount of radiotracer that can accumulate in the tumor might be increased by prolonging the blood circulation time of the radiotracer. To achieve this, we designed Albutate-1, with both DOTA and an albumin-binding domain coupled to TATE via a suitable linker. The aim of this study was to determine the characteristics of the novel radiotracer Albutate-1. A competition binding assay was performed using [111In]In-DOTA-TATE on fresh-frozen SSTR2+ tumor sections. In vitro cell-uptake and internalization of [111In]In-Albutate-1 and [111In]In-DOTA-TATE were determined. The stability of [177Lu]Lu-Albutate-1 was tested. A biodistribution study was performed to provide tumor and organ uptake of [177Lu]Lu-Albutate-1. The biodistribution data was used to determine time-activity curves and the radiation dose for each organ and the tumor. The in vitro IC50 value of Albutate-1 was 1.2 nM. A higher amount of the tracer was found in the intracellular fraction than in the membrane fraction ([111In]In-Albutate-1 14.0 vs 1.9% of the added amount; [111In]In-DOTA-TATE 11.0 vs 1.5% of the added amount). After radiolabeling [111In] In-Albutate-1 was stable up to 3 days (93.1-88.9%) in labeling solution and very stable in mouse serum (90-94%) for at least 24 h. In vivo, [177Lu]Lu-Albutate-1 was cleared slowly from the circulation (1 h p.i. 58%ID/g, 168h p.i. 2%ID/g). The addition of an albumin-binding domain to DOTA-TATE extended the blood circulation to T1/2= 27.5h. The tumor absorbed dose was raised to 1455 mGy/MBq. Bone marrow, the dose-limiting organ in the mouse spine, unfortunately, received 765 mGy/MBq. All other organs also received a high radiation dose, reducing the therapeutic index.
- Published
- 2021
- Full Text
- View/download PDF
5. Phase I study to assess safety, biodistribution and radiation dosimetry for (89)Zr-girentuximab in patients with renal cell carcinoma
- Author
-
Daphne Lobeek, Mark Rijpkema, Luis David Jiménez-Franco, Egbert Oosterwijk, Andreas Kluge, Robin I. J. Merkx, Mark Konijnenberg, and Peter F.A. Mulders
- Subjects
Biodistribution ,Vascular damage Radboud Institute for Health Sciences [Radboudumc 16] ,Girentuximab ,Effective dose (radiation) ,Renal cell carcinoma ,Positron Emission Tomography Computed Tomography ,Urological cancers Radboud Institute for Molecular Life Sciences [Radboudumc 15] ,medicine ,Humans ,Dosimetry ,Tissue Distribution ,Radiology, Nuclear Medicine and imaging ,Radiometry ,Adverse effect ,Carcinoma, Renal Cell ,business.industry ,Antibodies, Monoclonal ,Zirconium-89 ,General Medicine ,medicine.disease ,RCC ,Kidney Neoplasms ,Phase i study ,Clear cell renal cell carcinoma ,Organ-based dosimetry ,Positron-Emission Tomography ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,Original Article ,Nuclear medicine ,business ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,medicine.drug - Abstract
Purpose In this phase I study, we evaluated the safety, biodistribution and dosimetry of [89Zr]Zr-DFO-girentuximab (89Zr-girentuximab) PET/CT imaging in patients with suspicion of clear cell renal cell carcinoma (ccRCC). Methods Ten eligible patients received an intravenous administration of 37 MBq (± 10%) of 89Zr-girentuximab at mass doses of 5 mg or 10 mg. Safety was evaluated according to the NCI CTCAE (version 4.03). Biodistribution and normal organ dosimetry was performed based on PET/CT images acquired at 0.5, 4, 24, 72 and 168 h post-administration. Additionally, tumour dosimetry was performed in patients with confirmed ccRCC and visible tumour uptake on PET/CT imaging. Results 89Zr-girentuximab was administered in ten patients as per protocol. No treatment-related adverse events ≥ grade 3 were reported. 89Zr-girentuximab imaging allowed successful differentiation between ccRCC and non-ccRCC lesions in all patients, as confirmed with histological data. Dosimetry analysis using OLINDA/EXM 2.1 showed that the organs receiving the highest doses (mean ± SD) were the liver (1.86 ± 0.40 mGy/MBq), the kidneys (1.50 ± 0.22 mGy/MBq) and the heart wall (1.45 ± 0.19 mGy/MBq), with a mean whole body effective dose of 0.57 ± 0.08 mSv/MBq. Tumour dosimetry was performed in the 6 patients with histologically confirmed ccRCC resulting in a median tumour-absorbed dose of 4.03 mGy/MBq (range 1.90–11.6 mGy/MBq). Conclusions This study demonstrates that 89Zr-girentuximab is safe and well tolerated for the administered activities and mass doses and allows quantitative assessment of 89Zr-girentuximab PET/CT imaging in patients with suspicion of ccRCC. Trial registration NCT03556046—14th of June, 2018
- Published
- 2021
6. Effect of epigenetic treatment on SST
- Author
-
Julie, Refardt, Maria J, Klomp, Peter M, van Koetsveld, Fadime, Dogan, Mark, Konijnenberg, Tessa, Brabander, Richard A, Feelders, Wouter W, de Herder, Leo J, Hofland, and Johannes, Hofland
- Subjects
Neuroendocrine Tumors ,Humans ,Receptors, Somatostatin ,Octreotide ,Epigenesis, Genetic - Published
- 2022
7. Kidney absorbed radiation doses for [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T determined by 3D clinical dosimetry
- Author
-
Maike J.M. Uijen, Bastiaan Michael Privé, Carla M.L. Van Herpen, Harm Westdorp, Willemijn A. van Gemert, Maarten de Bakker, Martin Gotthardt, Mark Konijnenberg, Steffie M.B. Peters, and James Nagarajah
- Abstract
Purpose: For prostate-specific membrane antigen directed radioligand therapy (PSMA-RLT), [177Lu]Lu‑PSMA‑617 and [177Lu]Lu-PSMA-I&T are the currently preferred compounds. Recent preclinical studies suggested ~30x higher kidney absorbed dose for [177Lu]Lu-PSMA-I&T compared to [177Lu]Lu-PSMA-617, which may lead to an increased risk of kidney toxicity. We performed two single center, prospective dosimetry studies with either [177Lu]Lu-PSMA-617 or [177Lu]Lu-PSMA-I&T, using an identical dosimetry protocol. We evaluated the absorbed doses of both 177Lu labeled radioligands in human kidneys.Methods: 3D SPECT/CT imaging of the kidneys was performed after PSMA-RLT in cancer patients with PSMA-positive disease and an adequate glomerular filtration rate (GFR) (≥50 mL/min). Ten metastatic hormone-sensitive prostate cancer patients (mHSPC) were treated with [177Lu]Lu‑PSMA‑617 and ten advanced salivary gland cancer (SGC) patients were treated with [177Lu]Lu-PSMA-I&T. SPECT/CT imaging was performed at 5 time points (1h, 24h, 48h, 72h, and 168h post injection). In mHSPC patients SPECT/CT imaging was performed after cycles 1 and 2 (cumulative activity: 9 GBq) and in SGC patients only after cycle 1 (activity: 7.4 GBq). Kidney absorbed dose was calculated using organ-based dosimetry. Results: The median kidney absorbed dose was 0.49 Gy/GBq (range: 0.34-0.66) and 0.73 Gy/GBq (range: 0.42-1.31) for [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T, respectively (Wilcoxon-Mann-Whitney; p=0.002). Conclusion: This study shows that the kidney absorbed dose for [177Lu]Lu-PSMA-617 and [177Lu]Lu‑PSMA-I&T differs, with a ~1.5x higher median kidney absorbed dose for [177Lu]Lu-PSMA-I&T. This difference in the clinical setting is considerably smaller than observed in preclinical studies and may not hamper treatments with [177Lu]Lu-PSMA-I&T.
- Published
- 2022
- Full Text
- View/download PDF
8. Extensive preclinical evaluation of lutetium-177-labeled PSMA-specific tracers for prostate cancer radionuclide therapy
- Author
-
Simone U. Dalm, Debra Stuurman, Marion de Jong, Erik de Blois, Joost C. Haeck, Julie Nonnekens, Dik C. van Gent, Mark Konijnenberg, Corrina M.A. de Ridder, Nicole van Vliet, Wytske M. van Weerden, Eline A. M. Ruigrok, Radiology & Nuclear Medicine, Urology, and Molecular Genetics
- Subjects
Glutamate Carboxypeptidase II ,Male ,Biodistribution ,Lutetium ,urologic and male genital diseases ,Mice ,Prostate cancer ,SDG 3 - Good Health and Well-being ,Cell Line, Tumor ,PSMA ,medicine ,Animals ,Humans ,Tissue Distribution ,Radiology, Nuclear Medicine and imaging ,Binding site ,Radioisotopes ,Salivary gland ,Chemistry ,Prostatic Neoplasms ,General Medicine ,medicine.disease ,In vitro ,medicine.anatomical_structure ,Preclinical research ,Cell culture ,Antigens, Surface ,Radionuclide therapy ,Toxicity ,Cancer research ,Original Article ,Prostate cancer. Targeted radionuclide therapy - Abstract
Purpose Various radiolabeled prostate-specific membrane antigen (PSMA)–targeting tracers are clinically applied for prostate cancer (PCa) imaging and targeted radionuclide therapy. The PSMA binding affinities, biodistribution, and DNA-damaging capacities of these radiotracers have not yet been compared in detail. A major concern of PSMA-targeting radiotracers is the toxicity in other PSMA-expressing organs, such as the salivary glands, thus demanding careful evaluation of the most optimal and safest radiotracer. In this extensive preclinical study, we evaluated the clinically applied PSMA-targeting small molecule inhibitors DOTA-PSMA-617 (PSMA-617) and DOTAGA-PSMA-I&T (PSMA-I&T) and the PSMA nanobody DOTA-JVZ-007 (JVZ-007) using PSMA-expressing cell lines, a unique set of PCa patient-derived xenografts (PDX) and healthy human tissues. Methods and results In vitro displacement studies on PSMA-expressing cells and cryosections of a PSMA-positive PDX revealed high and specific binding affinity for all three tracers labeled with lutetium-177 with IC50 values in the nanomolar range. Interestingly, [177Lu]Lu-JVZ-007 could not be displaced by PSMA-617 or PSMA-I&T, suggesting that this tracer targets an alternative binding site. Autoradiography assays on cryosections of human salivary and renal tissues revealed [177Lu]Lu-PSMA-617 to have the lowest binding to these healthy organs compared with [177Lu]Lu-PSMA-I&T. In vivo biodistribution assays confirmed the in vitro results with comparable tumor uptake of [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T at all timepoints, resulting in induction of similar levels of DNA double-strand breaks in the tumors. However, [177Lu]Lu-PSMA-I&T demonstrated approximately 40× higher renal uptake at 4 and 8 h post injection resulting in an unfavorable tumor-to-kidney ratio. Conclusion [177Lu]Lu-PSMA-617 has the most favorable biodistribution in mice as well as more favorable binding characteristics in vitro in PSMA-positive cells and human kidney and salivary gland specimens compared with [177Lu]Lu-PSMA-I&T and [177Lu]Lu-JVZ-007. Based on our preclinical evaluation, [177Lu]Lu-PSMA-617 is the best performing tracer to be taken further into clinical evaluation for PSMA-targeted radiotherapeutic development although with careful evaluation of the tracer binding to PSMA-expressing organs.
- Published
- 2020
- Full Text
- View/download PDF
9. Intra-therapeutic dosimetry of [(177)Lu]Lu-PSMA-617 in low-volume hormone-sensitive metastatic prostate cancer patients and correlation with treatment outcome
- Author
-
Maarten de Bakker, Martin Gotthardt, Bastiaan M. Privé, Walter Jentzen, Mark Konijnenberg, Annemarie Eek, Steffie M. B. Peters, J. Alfred Witjes, Frank de Lange, James Nagarajah, Constantijn H.J. Muselaers, Niven Mehra, Radiotherapy, and Radiology & Nuclear Medicine
- Subjects
Male ,Organs at Risk ,medicine.medical_specialty ,Single Photon Emission Computed Tomography Computed Tomography ,Urology ,Lutetium ,Radionuclide therapy ,Radiation Dosage ,[177Lu]Lu-PSMA ,030218 nuclear medicine & medical imaging ,Lesion ,03 medical and health sciences ,Prostate cancer ,Tumours of the digestive tract Radboud Institute for Health Sciences [Radboudumc 14] ,0302 clinical medicine ,mHSPC ,SDG 3 - Good Health and Well-being ,Dosimetry ,Urological cancers Radboud Institute for Molecular Life Sciences [Radboudumc 15] ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Prospective Studies ,Prospective cohort study ,Index Lesion ,business.industry ,Other Research Radboud Institute for Health Sciences [Radboudumc 0] ,Prostatic Neoplasms ,General Medicine ,Prostate-Specific Antigen ,medicine.disease ,Hormones ,medicine.anatomical_structure ,Treatment Outcome ,030220 oncology & carcinogenesis ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,Toxicity ,Original Article ,Bone marrow ,medicine.symptom ,Radiopharmaceuticals ,business - Abstract
Introduction While [177Lu]Lu-PSMA radioligand therapy is currently only applied in end-stage metastatic castrate-resistant prostate cancer (mCRPC) patients, also low-volume hormone-sensitive metastatic prostate cancer (mHSPC) patients can benefit from it. However, there are toxicity concerns related to the sink effect in low-volume disease. This prospective study aims to determine the kinetics of [177Lu]Lu-PSMA in mHSPC patients, analyzing the doses to organs at risk (salivary glands, kidneys, liver, and bone marrow) and tumor lesions Methods Ten mHSPC patients underwent two cycles of [177Lu]Lu-PSMA therapy. Three-bed position SPECT/CT was performed at 5 time points after each therapy. Organ dosimetry and lesion dosimetry were performed using commercial software and a manual approach, respectively. Correlation between absorbed index lesion dose and treatment response (PSA drop of > 50% at the end of the study) was calculated and given as Spearman’s r and p-values. Results Kinetics of [177Lu]Lu-PSMA in mHSPC patients are comparable to those in mCRPC patients. Lesion absorbed dose was high (3.25 ± 3.19 Gy/GBq) compared to organ absorbed dose (salivary glands: 0.39 ± 0.17 Gy/GBq, kidneys: 0.49 ± 0.11 Gy/GBq, liver: 0.09 ± 0.01 Gy/GBq, bone marrow: 0.017 ± 0.008 Gy/GBq). A statistically significant correlation was found between treatment response and absorbed index lesion dose (p = 0.047). Conclusions We successfully performed small lesion dosimetry and showed that the tumor sink effect in mHSPC patients is of less concern than was expected. Tumor-to-organ ratio of absorbed dose was high and tumor uptake correlates with PSA response. Additional treatment cycles are legitimate in terms of organ toxicity and could lead to better tumor response.
- Published
- 2022
10. EURAMED ROCC-N-ROLL: DEVELOPING A EUROPEAN STRATEGIC RESEARCH AGENDA AND A CORRESPONDING ROADMAP FOR MEDICAL APPLICATIONS OF IONIZING RADIATION
- Author
-
Ms. Jing Ma, Guy Frija, Miss Monika Hierath, John Damilakis, Katrine Riklund, Mr. Alan Henry Tkaczyk, Mr. Mark Konijnenberg, Graciano Paulo, and Christoph Hoeschen
- Subjects
Biophysics ,General Physics and Astronomy ,Radiology, Nuclear Medicine and imaging ,General Medicine - Published
- 2022
- Full Text
- View/download PDF
11. Radionuclide therapy with the second-generation Affibody molecule [188Re]Re-Z41071 improves survival in mice bearing human HER2-expressing xenografts
- Author
-
Yongsheng Liu, Anzhelika Vorobyeva, Anna Orlova, Mark Konijnenberg, Tianqi Xu, Olga Bragina, Annika Loftenius, Erica Rosander, Fredrik Frejd, and Vladimir Tolmachev
- Subjects
Cancer Research ,Molecular Medicine ,Radiology, Nuclear Medicine and imaging - Published
- 2022
- Full Text
- View/download PDF
12. EANM procedure guidelines for radionuclide therapy with 177Lu-labelled PSMA-ligands (177Lu-PSMA-RLT)
- Author
-
Murat Bozkurt, Heiko Schöder, Samer Ezziddin, Hans-Jürgen Wester, Irene Virgolini, Clemens Kratochwil, Stefano Fanti, Felix M. Mottaghy, Flavio Forrer, Lisa Bodei, Matthias Eiber, Michael Lassmann, Mark Konijnenberg, Roberto C. Delgado Bolton, Klaus Kopka, Richard P. Baum, Wim J.G. Oyen, Kambiz Rahbar, Johannes Czernin, Wolfgang P. Fendler, Levant Kabasakal, Rodney J. Hicks, Uwe Haberkorn, Ken Herrmann, Thomas A. Hope, and Kratochwil C, Fendler WP, Eiber M, Baum R, Bozkurt MF, Czernin J, Delgado Bolton RC, Ezziddin S, Forrer F, Hicks RJ, Hope TA, Kabasakal L, Konijnenberg M, Kopka K, Lassmann M, Mottaghy FM, Oyen W, Rahbar K, Schöder H, Virgolini I, Wester HJ, Bodei L, Fanti S, Haberkorn U, Herrmann K.
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Lutetium ,Radionuclide therapy ,urologic and male genital diseases ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,Internal medicine ,PSMA ,medicine ,Radiology, Nuclear Medicine and imaging ,business.industry ,Cancer ,General Medicine ,Guideline ,Theranostics ,medicine.disease ,Radiation therapy ,Clinical trial ,Clinical research ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Nuclear medicine ,business - Abstract
Prostate-specific membrane antigen (PSMA) is expressed in most prostate cancers and can be identified by PSMA-ligand imaging, which has already become clinically accepted in several countries in- and outside Europe. PSMA-directed radioligand therapy (PSMA-RLT) with Lutetium-177 (177Lu-PSMA) is currently undergoing clinical validation. Retrospective observational data have documented favourable safety and striking clinical responses. Recent results from a prospective clinical trial (phase II) have been published confirming high response rates, low toxicity and reduction of pain in metastatic castration-resistant prostate cancer (mCRPC) patients who had progressed after conventional treatments. Such patients typically survive for periods less than 1.5 years. This has led some facilities to adopt compassionate or unproven use of this therapy, even in the absence of validation within a randomised-controlled trial. As a result, a consistent body of evidence exists to support efficacy and safety data of this treatment. The purpose of this guideline is to assist nuclear medicine specialists to deliver PSMA-RLT as an "unproven intervention in clinical practice", in accordance with the best currently available knowledge.
- Published
- 2019
- Full Text
- View/download PDF
13. Overcoming nephrotoxicity in peptide receptor radionuclide therapy using [
- Author
-
Lorain, Geenen, Julie, Nonnekens, Mark, Konijnenberg, Sarah, Baatout, Marion, De Jong, and An, Aerts
- Subjects
Heterocyclic Compounds, 1-Ring - Abstract
Peptide receptor radionuclide therapy (PRRT) is used for the treatment of patients with unresectable or metastasized somatostatin receptor type 2 (SSTR
- Published
- 2021
14. To 1000 Gy and back again: a systematic review on dose-response evaluation in selective internal radiation therapy for primary and secondary liver cancer
- Author
-
Lovisa E L Westlund Gotby, Mark Konijnenberg, Christiaan G. Overduin, Marcel Verheij, J. Frank W. Nijsen, Joey Roosen, and Nienke J. M. Klaassen
- Subjects
Oncology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Colorectal cancer ,medicine.medical_treatment ,Brachytherapy ,MEDLINE ,Review Article ,Cochrane Library ,TARE ,Tumours of the digestive tract Radboud Institute for Health Sciences [Radboudumc 14] ,Holmium ,SDG 3 - Good Health and Well-being ,Internal medicine ,Dosimetry ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Yttrium Radioisotopes ,SIRT ,Yttrium ,Prospective Studies ,Radioembolization ,Prospective cohort study ,Chemotherapy ,business.industry ,Selective internal radiation therapy ,Liver Neoplasms ,General Medicine ,medicine.disease ,Microspheres ,Hepatocellular carcinoma ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,business - Abstract
Purpose To systematically review all current evidence into the dose-response relation of yttrium-90 and holmium-166 selective internal radiation therapy (SIRT) in primary and secondary liver cancer. Methods A standardized search was performed in PubMed (MEDLINE), Embase, and the Cochrane Library in order to identify all published articles on dose-response evaluation in SIRT. In order to limit the results, all articles that investigated SIRT in combination with other therapy modalities (such as chemotherapy) were excluded. Results A total of 3038 records were identified of which 487 were screened based on the full text. Ultimately, 37 studies were included for narrative analysis. Meta-analysis could not be performed due to the large heterogeneity in study and reporting designs. Out of 37 studies, 30 reported a ‘mean dose threshold’ that needs to be achieved in order to expect a response. This threshold appears to be higher for hepatocellular carcinoma (HCC, 100–250 Gy) than for colorectal cancer metastases (CRC, 40–60 Gy). Reported thresholds tend to be lower for resin microspheres than when glass microspheres are used. Conclusion Although the existing evidence demonstrates a dose-response relationship in SIRT for both primary liver tumours and liver metastases, many pieces of the puzzle are still missing, hampering the definition of standardized dose thresholds. Nonetheless, most current evidence points towards a target mean dose of 100–250 Gy for HCC and 40–60 Gy for CRC. The field would greatly benefit from a reporting standard and prospective studies designed to elucidate the dose-response relation in different tumour types.
- Published
- 2020
15. GRPr Antagonist
- Author
-
Ingrid L, Bakker, Alida C, Fröberg, Martijn B, Busstra, J Fred, Verzijlbergen, Mark, Konijnenberg, Geert J L H, van Leenders, Ivo G, Schoots, Erik, de Blois, Wytske M, van Weerden, Simone U, Dalm, Theodosia, Maina, Berthold A, Nock, and Marion, de Jong
- Subjects
Male ,Receptors, Bombesin ,Positron Emission Tomography Computed Tomography ,Humans ,Prostatic Neoplasms ,Tissue Distribution ,Clinical Investigation ,Middle Aged - Abstract
The gastrin-releasing peptide receptor (GRPr) is overexpressed in prostate cancer (PCa) cells, making it an excellent tool for targeted imaging. The (68)Ga-labeled GRPr antagonist SB3 has shown excellent results in preclinical and clinical studies and was selected for further clinical investigation. The aims of this phase I study were to investigate (68)Ga-SB3 PET/CT imaging of primary PCa tumors and assess safety. More aims included an investigation of biodistribution and dosimetry and a comparison with pathology and GRPr expression. Methods: Ten therapy-naïve, biopsy-confirmed PCa patients planned for prostatectomy were included. A 3-h extensive PET/CT imaging protocol was performed within 2 wk before prostatectomy. Prostate tissue was evaluated for tumor localization and Gleason score, and in vitro autoradiography was performed to determine GRPr expression. Available MRI scans performed within 3 mo before the study were matched. For dosimetry, residence times were estimated and effective dose to the body as well as absorbed doses to organs were calculated using the IDAC dose model, version 2.1. Results: Administration of (68)Ga-SB3 (187.4 ± 40.0 MBq, 40 ± 5 μg) was well tolerated; no significant changes in vital signs or laboratory results were observed. (68)Ga-SB3 PET/CT showed lesions in 8 of 10 patients. Pathologic analysis revealed a total of 16 tumor lesions, of which PET/CT showed 14, resulting in a sensitivity of 88%. (68)Ga-SB3 PET/CT imaging showed uptake in 2 large prostatic intraepithelial neoplasia foci, considered a precursor to PCa, resulting in an 88% specificity. Autoradiography of tumor lesions revealed heterogeneous GRPr expression and was negative in 4 patients. Both PET/CT-negative patients had a GRPr-negative tumor. In autoradiography-positive tumors, the level of GRPr expression showed a significant correlation to tracer uptake on PET/CT. Dosimetry calculations estimated the effective dose to be 0.0144 mSv/MBq, similar to other (68)Ga-labeled radiopeptides. The highest absorbed dose was detected in the physiologic GRPr-expressing pancreas (0.198 mGy/MBq), followed by the bladder wall and kidneys. Conclusion: (68)Ga-SB3 PET/CT is a safe imaging method and a promising tool for early PCa imaging.
- Published
- 2020
16. EANM dosimetry committee series on standard operational procedures: a unified methodology for
- Author
-
Carlo, Chiesa, Katarina, Sjogreen-Gleisner, Stephan, Walrand, Lidia, Strigari, Glenn, Flux, Jonathan, Gear, Caroline, Stokke, Pablo Minguez, Gabina, Peter, Bernhardt, and Mark, Konijnenberg
- Subjects
90Y microspheres ,Lung shunt ,Prospective/retrospective dosimetry ,Radioembolization dosimetry ,99mTc-MAA ,Guideline ,90Y PET ,Liver dosimetry ,Tumour dosimetry - Abstract
The aim of this standard operational procedure is to standardize the methodology employed for the evaluation of pre- and post-treatment absorbed dose calculations in 90Y microsphere liver radioembolization. Basic assumptions include the permanent trapping of microspheres, the local energy deposition method for voxel dosimetry, and the patient–relative calibration method for activity quantification.The identity of 99mTc albumin macro-aggregates (MAA) and 90Y microsphere biodistribution is also assumed. The large observed discrepancies in some patients between 99mTc-MAA predictions and actual 90Y microsphere distributions for lesions is discussed. Absorbed dose predictions to whole non-tumoural liver are considered more reliable and the basic predictors of toxicity. Treatment planning based on mean absorbed dose delivered to the whole non-tumoural liver is advised, except in super-selective treatments. Given the potential mismatch between MAA simulation and actual therapy, absorbed doses should be calculated both pre- and post-therapy. Distinct evaluation between target tumours and non-tumoural tissue, including lungs in cases of lung shunt, are vital for proper optimization of therapy. Dosimetry should be performed first according to a mean absorbed dose approach, with an optional, but important, voxel level evaluation. Fully corrected 99mTc-MAA Single Photon Emission Computed Tomography (SPECT)/computed tomography (CT) and 90Y TOF PET/CT are regarded as optimal acquisition methodologies, but, for institutes where SPECT/CT is not available, non-attenuation corrected 99mTc-MAA SPECT may be used. This offers better planning quality than non dosimetric methods such as Body Surface Area (BSA) or mono-compartmental dosimetry. Quantitative 90Y bremsstrahlung SPECT can be used if dedicated correction methods are available. The proposed methodology is feasible with standard camera software and a spreadsheet. Available commercial or free software can help facilitate the process and improve calculation time. Supplementary Information The online version contains supplementary material available at 10.1186/s40658-021-00394-3.
- Published
- 2020
17. Radionuclide therapy using ABD-fused ADAPT scaffold protein: Proof of Principle
- Author
-
Anzhelika Vorobyeva, Mohamed Altai, Vladimir Tolmachev, Maryam Oroujeni, Mark Konijnenberg, Jesper Borin, Olga Vorontsova, Sophia Hober, Anna Orlova, Emma von Witting, Javad Garousi, and Radiology & Nuclear Medicine
- Subjects
Scaffold protein ,Scaffold ,Biodistribution ,Receptor, ErbB-2 ,Medical Biotechnology ,Biophysics ,Bioengineering ,02 engineering and technology ,Radionuclide therapy ,Biomaterials ,03 medical and health sciences ,Mice ,Molecular recognition ,HER2 ,Albumins ,Cell Line, Tumor ,Animals ,Tissue Distribution ,Medicinsk bioteknik ,030304 developmental biology ,Radioisotopes ,0303 health sciences ,biology ,Radiotherapy ,Chemistry ,Albumin ,Proteins ,177Lu ,021001 nanoscience & nanotechnology ,Cell biology ,Mechanics of Materials ,Ceramics and Composites ,biology.protein ,ABD (Albumin binding domain) ,Protein G ,Radiologi och bildbehandling ,Biodistribution modification ,0210 nano-technology ,ADAPT (Albumin-binding domain derived affinity ProTein) ,Binding domain ,Radiology, Nuclear Medicine and Medical Imaging - Abstract
Molecular recognition in targeted therapeutics is typically based on immunoglobulins. Development of engineered scaffold proteins (ESPs) has provided additional opportunities for the development of targeted therapies. ESPs offer inexpensive production in prokaryotic hosts, high stability and convenient approaches to modify their biodistribution. In this study, we demonstrated successful modification of the biodistribution of an ESP known as ADAPT (Albumin-binding domain Derived Affinity ProTein). ADAPTs are selected from a library based on the scaffold of ABD (Albumin Binding Domain) of protein G. A particular ADAPT, the ADAPT6, binds to human epidermal growth factor receptor type 2 (HER2) with high affinity. Preclinical and early clinical studies have demonstrated that radiolabeled ADAPT6 can image HER2-expression in tumors with high contrast. However, its rapid glomerular filtration and high renal reabsorption have prevented its use in radionuclide therapy. To modify the biodistribution, ADAPT6 was genetically fused to an ABD. The non-covalent binding to the host's albumin resulted in a 14-fold reduction of renal uptake and appreciable increase of tumor uptake for the best variant, 177Lu-DOTA-ADAPT6-ABD035. Experimental therapy in mice bearing HER2-expressing xenografts demonstrated more than two-fold increase of median survival even after a single injection of 18 MBq 177Lu-DOTA-ADAPT6-ABD035. Thus, a fusion with ABD and optimization of the molecular design provides ADAPT derivatives with attractive targeting properties for radionuclide therapy. De två första författarna delar förstaförfattarskapet
- Published
- 2020
18. Lutetium-177-PSMA-617 in low-volume hormone sensitive metastatic prostate cancer: a prospective study
- Author
-
D.M. Somford, I van Oort, Patrik Zamecnik, W R Gerritsen, Sandra Heskamp, Steffie M. B. Peters, J Verzijlbergen, Bastiaan M. Privé, Constantijn H.J. Muselaers, J.A. Witjes, Tom W. J. Scheenen, Martin Gotthardt, Marcel J.R. Janssen, James Nagarajah, Mark Konijnenberg, J.P.M. Sedelaar, J.O. Barentsz, and Niven Mehra
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,chemistry.chemical_element ,medicine.disease ,Lutetium ,Hormone-sensitive ,Low volume ,Prostate cancer ,chemistry ,Internal medicine ,medicine ,Prospective cohort study ,business - Published
- 2020
- Full Text
- View/download PDF
19. Radionuclide Therapy of HER2-Expressing Human Xenografts Using Affibody-Based Peptide Nucleic Acid–Mediated Pretargeting: In Vivo Proof of Principle
- Author
-
Christina Atterby, Anna Orlova, Maryam Oroujeni, Patrick Micke, Kristina Westerlund, Vladimir Tolmachev, Mohamed Altai, Johanna Sofia Margareta Mattsson, Marion de Jong, Amelie Eriksson Karlström, Mark Konijnenberg, Bogdan Mitran, and Radiology & Nuclear Medicine
- Subjects
Peptide Nucleic Acids ,0301 basic medicine ,Receptor, ErbB-2 ,Recombinant Fusion Proteins ,Pharmacology ,Kidney ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,DOTA ,Tissue Distribution ,Radiology, Nuclear Medicine and imaging ,Radiometry ,Pretargeting ,Ovarian Neoplasms ,Base Sequence ,Peptide nucleic acid ,Chemistry ,Proteins ,Survival Analysis ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Absorbed dose ,Radionuclide therapy ,Female ,Affibody molecule ,Bone marrow - Abstract
Affibody molecules are small proteins engineered using a nonantibody scaffold. Radiolabeled Affibody molecules are excellent imaging probes, but their application to radionuclide therapy has been prevented by high renal reabsorption. The aim of this study was to test the hypothesis that Affibody-based peptide nucleic acid (PNA)-mediated pretargeted therapy of human epidermal growth factor receptor 2 (HER2)-expressing cancer extends survival without accompanying renal toxicity. Methods: A HER2-targeting Affibody molecule ligated with an AGTCGTGATGTAGTC PNA hybridization probe (ZHER2:342-SR-HP1) was used as the primary pretargeting agent. A complementary AGTCGTGATGTAGTC PNA conjugated to the chelator DOTA and labeled with the radionuclide 177Lu (177Lu-HP2) was used as the secondary agent. The influence of different factors on pretargeting was investigated. Experimental radionuclide therapy in mice bearing SKOV-3 xenografts was performed in 6 cycles separated by 7 d. Results: Optimal tumor targeting was achieved when 16 MBq/3.5 μg (0.65 nmol) of 177Lu-HP2 was injected 16 h after injection of 100 μg (7.7 nmol) of ZHER2:342-SR-HP1. The calculated absorbed dose to tumors was 1,075 mGy/MBq, whereas the absorbed dose to kidneys was 206 mGy/MBq and the absorbed dose to blood (surrogate of bone marrow) was 4 mGy/MBq. Survival of mice was significantly longer (P < 0.05) in the treatment group (66 d) than in the control groups treated with the same amount of ZHER2:342-SR-HP1 only (37 d), the same amount and activity of 177Lu-HP2 only (32 d), or phosphate-buffered saline (37 d). Conclusion: The studied pretargeting system can deliver an absorbed dose to tumors appreciably exceeding absorbed doses to critical organs, making Affibody-based PNA-mediated pretargeted radionuclide therapy highly attractive.
- Published
- 2018
- Full Text
- View/download PDF
20. Modeling early radiation DNA damage occurring during [177Lu]Lu-DOTA-[Tyr3]octreotate radionuclide therapy
- Author
-
Marion de Jong, Giulia Tamborino, Mark Konijnenberg, Yann Perrot, Marijke De Saint-Hubert, Lara Struelens, Carmen Villagrasa, and Julie Nonnekens
- Subjects
Octreotate ,Chemistry ,DNA damage ,media_common.quotation_subject ,fungi ,Cell ,chemistry.chemical_compound ,medicine.anatomical_structure ,Cytoplasm ,Absorbed dose ,Radionuclide therapy ,medicine ,Biophysics ,Radiology, Nuclear Medicine and imaging ,Internalization ,Nucleus ,media_common - Abstract
Rationale: The aim of this study is to build a simulation framework to evaluate the number of DNA double strand breaks (DSBs) induced by in vitro targeted radionuclide therapy (TRT). This work represents the first step towards exploring underlying biological mechanisms and influence of physical/chemical parameters to enable a better response prediction in patients. We used this tool to characterize early DSB induction by [177Lu]Lu-DOTA-[Tyr3]octreotate (177Lu-DOTATATE), a commonly used TRT for neuroendocrine tumors. Methods: A multiscale approach is implemented to simulate the number of DSBs produced over 4 h by the cumulated decays of 177Lu distributed according the somatostatin receptor-binding. The approach involves 2 sequential simulations performed with Geant4/Geant4-DNA. The radioactive source is sampled according to uptake experiments on the distribution of activities within the medium and the planar cellular cluster, assuming instant and permanent internalization. A phase space (PHSP) is scored around the nucleus of the central cell. Then, the PHSP is used to generate particles entering the nucleus containing a multi-scale description of the DNA in order to score the number of DSBs per particle source. The final DSB computations are compared to experimental data, measured by immunofluorescent detection of 53BP1 foci. Results: The probability of electrons reaching the nucleus was significantly influenced by the shape of the cell compartment, causing large variance in the induction pattern of DSBs. A significant difference was found in the DSBs induced by activity distributions in cell and medium, which is explained by the specific energy (z) distributions. The average number of simulated DSBs is 14 DSBs/cell (range: 7-24 DSBs/cell) compared to 13 DSBs/cell (2-30) experimentally determined. We found a linear correlation between the mean absorbed dose to the nucleus and the number of DSBs/cell: 0.014 DSBs/cell mGy-1 for internalization in the Golgi apparatus and 0.017 DSBs/cell mGy-1 for internalization in the cytoplasm. Conclusion: This simulation tool can lead to more reliable absorbed dose to DNA correlation and help in prediction of biological response.
- Published
- 2021
- Full Text
- View/download PDF
21. Ga-68/Lu-177-NeoBOMB1, a Novel Radiolabeled GRPR Antagonist for Theranostic Use in Oncology
- Author
-
Erik de Blois, Donato Barbato, Mark Konijnenberg, Mattia Tedesco, Berthold A. Nock, Gabriela N. Doeswijk, Francesca Orlandi, Theodosia Maina, Marion de Jong, Simone U. Dalm, Ingrid L. Bakker, and Radiology & Nuclear Medicine
- Subjects
Male ,Biodistribution ,Mice, Nude ,Gallium Radioisotopes ,Pharmacology ,Theranostic Nanomedicine ,030218 nuclear medicine & medical imaging ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,In vivo ,Cell Line, Tumor ,Gastrin-releasing peptide receptor ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Molecular Targeted Therapy ,Receptor ,business.industry ,Antagonist ,Cancer ,Neoplasms, Experimental ,medicine.disease ,Receptors, Bombesin ,Treatment Outcome ,medicine.anatomical_structure ,Isotope Labeling ,030220 oncology & carcinogenesis ,Bombesin ,Radiopharmaceuticals ,Pancreas ,Nuclear medicine ,business - Abstract
Because overexpression of the gastrin-releasing peptide receptor (GRPR) has been reported on various cancer types, for example, prostate cancer and breast cancer, targeting this receptor with radioligands might have a significant impact on staging and treatment of GRPR-expressing tumors. NeoBOMB1 is a novel DOTAcoupled GRPR antagonist with high affinity for GRPR and excellent in vivo stability. The purpose of this preclinical study was to further explore the use of NeoBOMB1 for theranostic application by determining the biodistribution of 68Ga-NeoBOMB1 and 177Lu- NeoBOMB1. Methods: PC-3 tumor–xenografted BALB/c nu/nu mice were injected with either approximately 13 MBq/250 pmol 68Ga- NeoBOMB1 or a low (;1MBq/200pmol) versus high (;1MBq/10 pmol) peptide amount of 177Lu-NeoBOMB1, after which biodistribution and imaging studies were performed. At 6 time points (15, 30, 60, 120, 240, and 360 min for 68Ga-NeoBOMB1 and 1, 4, 24, 48, 96, and 168 h for 177Lu-NeoBOMB1) postinjection tumor and organ uptake was determined. To assess receptor specificity, additional groups of animals were coinjected with an excess of unlabeled NeoBOMB1. Results of the biodistribution studies were used to determine pharmacokinetics and dosimetry. Furthermore, PET/CT and SPECT/MRI were performed. Results: Injection of approximately 250 pmol 68Ga-NeoBOMB1 resulted in a tumor and pancreas uptake of 12.4 6 2.3 and 22.7 6 3.3 percentage injected dose per gram (%ID/g) of tissue, respectively, at 120 min after injection. 177Lu-NeoBOMB1 biodistribution studies revealed a higher tumor uptake (17.9 6 3.3 vs. 11.6 6 1.3 %ID/g of tissue at 240 min after injection) and a lower pancreatic uptake (19.8 6 6.9 vs. 105 6 13 %ID/g of tissue at 240 min after injection) with the higher peptide amount injected, leading to a significant increase in the absorbed dose to the tumor versus the pancreas (200 pmol, 570 vs. 265 mGy/ MBq; 10 pmol, 435 vs. 1393 mGy/MBq). Using these data to predict patient dosimetry, we found a kidney, pancreas, and liver exposure of 0.10, 0.65, and 0.06mGy/MBq, respectively. Imaging studies resulted in good visualization of the tumor with both 68Ga-NeoBOMB1 and 177Lu-NeoBOMB1. Conclusion: Our findings indicate that 68Ga- or 177Lu-labeled NeoBOMB1 is a promising radiotracer with excellent tumor uptake and favorable pharmacokinetics for imaging and therapy of GRPR-expressing tumors.
- Published
- 2017
22. EANM procedure guidelines for radionuclide therapy with
- Author
-
Clemens, Kratochwil, Wolfgang Peter, Fendler, Matthias, Eiber, Richard, Baum, Murat Fani, Bozkurt, Johannes, Czernin, Roberto C, Delgado Bolton, Samer, Ezziddin, Flavio, Forrer, Rodney J, Hicks, Thomas A, Hope, Levant, Kabasakal, Mark, Konijnenberg, Klaus, Kopka, Michael, Lassmann, Felix M, Mottaghy, Wim, Oyen, Kambiz, Rahbar, Heiko, Schöder, Irene, Virgolini, Hans-Jürgen, Wester, Lisa, Bodei, Stefano, Fanti, Uwe, Haberkorn, and Ken, Herrmann
- Subjects
Glutamate Carboxypeptidase II ,Male ,Radioisotopes ,Prostatic Neoplasms ,Documentation ,Lutetium ,Ligands ,Europe ,Editorial ,Antigens, Surface ,Practice Guidelines as Topic ,Humans ,Nuclear Medicine ,Safety ,Radiometry - Abstract
Prostate-specific membrane antigen (PSMA) is expressed in most prostate cancers and can be identified by PSMA-ligand imaging, which has already become clinically accepted in several countries in- and outside Europe. PSMA-directed radioligand therapy (PSMA-RLT) with Lutetium-177 (
- Published
- 2019
23. Radiochemical and analytical aspects of inter-institutional quality control measurements on radiopharmaceuticals
- Author
-
Mark Konijnenberg, Ho Sze Chan, Rory M. S. de Zanger, Erik de Blois, Wouter A.P. Breeman, Radiology & Nuclear Medicine, and Internal Medicine
- Subjects
lcsh:Medical physics. Medical radiology. Nuclear medicine ,Standardization ,Computer science ,lcsh:R895-920 ,Arbitrary unit ,030218 nuclear medicine & medical imaging ,Analytical Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Activity detection ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,Pharmacology ,lcsh:RM1-950 ,Methodology ,Reliability engineering ,RCP ,lcsh:Therapeutics. Pharmacology ,030220 oncology & carcinogenesis ,Multicenter trial ,Radiopharmaceuticals ,HPLC ,Validated conditions ,Smoothing ,Institutional quality - Abstract
Background Clinically applied radiopharmaceuticals have to meet quality release criteria like a high radiochemical yield and radiochemical purity. Many radiopharmaceuticals do not have marketing authorization and have no dedicated monograph within the European pharmacopeia, therefore general monographs on quality control have to be applied for clinical applications. These criteria require standardization and validation in labeling and preparation, including QC measurements according to well-defined standard operation procedures. QC measurements however, are often based on detection techniques specific for a certain LC-system. Multi-institutional research and development of new radiopharmaceuticals lead to an increase in multicenter trials. Although all institutes’ radiopharmacies are using the same standardized labeling and operation procedures, they often use different LC and radiodetection systems. Here we present a comparison of QC assessments for 3 radiopharmaceuticals with focus on the interpretation of chromatograms, data-output and potential differences in local practical performances of QC on (U)HPLC. Methods QC assessments for [111In]In-CCK, [68Ga]Ga-Bombesin and [177Lu]Lu-PSMA analogs were compared. Two of the radiopharmaceutical QC assessments were also applied in other institutes using their own HPLC-systems and concordant software. Data from the HPLC-injections and measurements is processed and summarized in chromatograms, based on a variety of smoothing algorithms for which different software programs are applied. Described radiopeptides were labeled and analyzed according their standardized labeling and operation procedures. Results Integration of main peaks on chromatograms resulted in a range of RCP, depending on the smoothing algorithm used. [111In]In-CCK(A), 68Ga-Bombesin(B) and [177Lu]Lu-PSMA(C) analogs had a RCP range of 88%–96%(A), 89–95%(B) and 92–99%(C) respectively. Important factors affecting final RCP value were site specific background radiation-levels, intrinsic system properties such as noise and sensitivity, personal interpretation e.g. peak-tailing and smoothing algorithms. Conclusion Measurement of RCP shows a strong method- and system-dependency, even when parameters are validated, standardized and SOP are followed. Release criteria are frequently based on RCP data from one central location. The lack of inter inter institutional validation and standardization in RCP determination makes the results therefore rather arbitrary. For multicenter trials, we recommend to compare locally determined RCP under validated and standardized conditions of in-line activity detection between institutes for each radiopharmaceutical.
- Published
- 2019
24. Nephrotoxicity after PRRT with 177Lu-DOTA-octreotate
- Author
-
Eric P. Krenning, Dik J. Kwekkeboom, Wouter A. van der Zwan, Hendrik Bergsma, P. P. M. Kooij, Boen L. R. Kam, Mark Konijnenberg, Jaap J.M. Teunissen, and Katya Mauff
- Subjects
Male ,medicine.medical_treatment ,177Lu-Octreotate ,Comorbidity ,Octreotide ,030218 nuclear medicine & medical imaging ,chemistry.chemical_compound ,0302 clinical medicine ,Coordination Complexes ,Risk Factors ,Neoplasms ,Molecular Targeted Therapy ,Nephrotoxicity ,Netherlands ,Aged, 80 and over ,Incidence ,Radiotherapy Dosage ,General Medicine ,Survival Rate ,Somatostatin ,Radiology Nuclear Medicine and imaging ,030220 oncology & carcinogenesis ,Toxicity ,Original Article ,Female ,Kidney Diseases ,PRRT ,medicine.medical_specialty ,Urology ,Renal function ,03 medical and health sciences ,Dosimetry ,Internal medicine ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,External beam radiotherapy ,Radiation Injuries ,Aged ,Creatinine ,Radiotherapy ,business.industry ,Kidneys ,Radiation therapy ,Endocrinology ,chemistry ,Radionuclide therapy ,Radiopharmaceuticals ,business - Abstract
Purpose After peptide receptor radionuclide therapy (PRRT), renal toxicity may occur, particular in PRRT with 90Y-labelled somatostatin analogues. Risk factors have been identified for increased probability of developing renal toxicity after PRRT, including hypertension, diabetes and age. We investigated the renal function over time, the incidence of nephrotoxicity and associated risk factors in patients treated with PRRT with [177Lu-DOTA0,Tyr3]-Octreotate (177Lu-Octreotate). Also, radiation dose to the kidneys was evaluated and compared with the accepted dose limits in external beam radiotherapy and PRRT with 90Y-radiolabelled somatostatin analogues. Methods The annual decrease in creatinine clearance (CLR) was determined in 209 Dutch patients and the incidence of grade 3 or 4 renal toxicity (according to CTCAE v4.03) was evaluated in 323 patients. Risk factors were analysed using a nonlinear mixed effects regression model. Also, radiation doses to the kidneys were calculated and their association with high annual decrease in renal function were analysed. Results Of the 323 patients, 3 (1 %) developed (subacute) renal toxicity grade 2 (increase in serum creatinine >1.5 – 3.0 times baseline or upper limit of normal). No subacute grade 3 or 4 nephrotoxicity was observed. The estimated average baseline CLR (± SD) was 108 ± 5 ml/min and the estimated average annual decrease in CLR (± SD) was 3.4 ± 0.4 %. None of the risk factors (hypertension, diabetes, high cumulative injected activity, radiation dose to the kidneys and CTCAE grade) at baseline had a significant effect on renal function over time. The mean absorbed kidney dose in 228 patients was 20.1 ± 4.9 Gy. Conclusion Nephrotoxicity in patients treated with 177Lu-octreotate was low. No (sub)acute grade 3 or 4 renal toxicity occurred and none of the patients had an annual decrease in renal function of >20 %. No risk factors for renal toxicity could be identified. Our data support the idea that the radiation dose threshold, adopted from external beam radiotherapy and PRRT with 90Y-labelled somatostatin analogues, does not seem valid for PRRT with 177Lu-octreotate. Electronic supplementary material The online version of this article (doi:10.1007/s00259-016-3382-9) contains supplementary material, which is available to authorized users.
- Published
- 2016
- Full Text
- View/download PDF
25. Towards Personalized Treatment of Prostate Cancer: PSMA I&T, a Promising Prostate-Specific Membrane Antigen-Targeted Theranostic Agent
- Author
-
Marian C. Clahsen-van Groningen, Hans-Jürgen Wester, Sandra Heskamp, Wytske M. van Weerden, Otto C. Boerman, Margret Schottelius, Marion de Jong, Kristell L.S. Chatalic, Mark Konijnenberg, Gerben M. Franssen, Janneke D.M. Molkenboer-Kuenen, Radiology & Nuclear Medicine, Urology, and Pathology
- Subjects
Glutamate Carboxypeptidase II ,Male ,Pathology ,medicine.medical_specialty ,Single Photon Emission Computed Tomography Computed Tomography ,Theranostic Nanomedicine ,Drug-Related Side Effects and Adverse Reactions ,medicine.medical_treatment ,Medicine (miscellaneous) ,Spleen ,urologic and male genital diseases ,030218 nuclear medicine & medical imaging ,Nephrotoxicity ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,medicine ,Glutamate carboxypeptidase II ,PSMA ,Animals ,CRPC ,Precision Medicine ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Radioisotopes ,2-PMPA ,Radiotherapy ,business.industry ,imaging ,Prostatic Neoplasms ,radionuclide therapy ,medicine.disease ,prostate cancer ,Radiation therapy ,Disease Models, Animal ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Absorbed dose ,SPECT ,Radionuclide therapy ,Antigens, Surface ,Cancer research ,business ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,Research Paper - Abstract
Contains fulltext : 167159.pdf (Publisher’s version ) (Open Access) Prostate-specific membrane antigen (PSMA) is a well-established target for nuclear imaging and therapy of prostate cancer (PCa). Radiolabeled small-molecule PSMA inhibitors are excellent candidates for PCa theranostics-they rapidly and efficiently localize in tumor lesions. However, high tracer uptake in kidneys and salivary glands are major concerns for therapeutic applications. Here, we present the preclinical application of PSMA I&T, a DOTAGA-chelated urea-based PSMA inhibitor, for SPECT/CT imaging and radionuclide therapy of PCa. (111)In-PSMA I&T showed dose-dependent uptake in PSMA-expressing tumors, kidneys, spleen, adrenals, lungs and salivary glands. Coadministration of 2-(phosphonomethyl)pentane-1,5-dioic acid (2-PMPA) efficiently reduced PSMA-mediated renal uptake of (111)In-PSMA I&T, with the highest tumor/kidney radioactivity ratios being obtained using a dose of 50 nmol 2-PMPA. SPECT/CT clearly visualized subcutaneous tumors and sub-millimeter intraperitoneal metastases; however, high renal and spleen uptake in control mice (no 2-PMPA) interfered with visualization of metastases in the vicinity of those organs. Coadministration of 2-PMPA increased the tumor-to-kidney absorbed dose ratio during (177)Lu-PSMA I&T radionuclide therapy. Hence, at equivalent absorbed dose to the tumor (36 Gy), coinjection of 2-PMPA decreased absorbed dose to the kidneys from 30 Gy to 12 Gy. Mice injected with (177)Lu-PSMA I&T only, showed signs of nephrotoxicity at 3 months after therapy, whereas mice injected with (177)Lu-PSMA I&T + 2-PMPA did not. These data indicate that PSMA I&T is a promising theranostic tool for PCa. PSMA-specific uptake in kidneys can be successfully tackled using blocking agents such as 2-PMPA.
- Published
- 2016
- Full Text
- View/download PDF
26. Localization of Radiolabeled Somatostatin Analogs in the Spleen
- Author
-
Mark Konijnenberg, Daniel Kaemmerer, Marleen Melis, Jan de Swart, Marion de Jong, Harald C. Groen, Harshad R. Kulkarni, Richard P. Baum, Jörg Sänger, Amelie Lupp, and Radiology & Nuclear Medicine
- Subjects
endocrine system ,Pathology ,medicine.medical_specialty ,Spleen ,Neuroendocrine tumors ,Octreotide ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Organometallic Compounds ,Neoplasm ,Humans ,Radiology, Nuclear Medicine and imaging ,Receptor ,Tomography, Emission-Computed, Single-Photon ,business.industry ,General Medicine ,medicine.disease ,medicine.anatomical_structure ,Somatostatin ,030220 oncology & carcinogenesis ,Red pulp ,Immunohistochemistry ,Radiopharmaceuticals ,business ,Ex vivo - Abstract
Radiolabeled somatostatin (SST) analogs, used to visualize and treat SST receptor (SSTR)-expressing neuroendocrine tumors, also accumulate in the spleen. There is a high interpatient variation and no significant radiation-induced splenic toxicity; however, an absorbed dose-related reduction in spleen size was detected. However, the exact localization of radioactivity and the role of SST receptors in splenic retention are unknown. Therefore, we performed ex vivo micro-SPECT of the isolated spleen from a patient with a pancreatic neoplasm after 1 GBq (27 mCi) Lu-DOTATATE administration, followed by autoradiography and immunohistochemistry. Ex vivo autoradiography demonstrated convincingly that most radioactivity accumulated in red pulp.
- Published
- 2016
27. Prostate cancer imaging and therapy
- Author
-
Lucia Zanoni, Cristina Nanni, Stefano Fanti, Martin Behe, Wolfgang Römer, Daniel Tempesta, David Gilmore, Giorgio Testanera, Giovanna Pepe, Yat Man Tsang, Michelle Leech, Sarah M. Schwarzenböck, Bernd Joachim Krause, Jens Kurth, Mark Konijnenberg, Domenico Albano, Rexhep Durmo, Laura Evangelista, and Federico Caobelli
- Published
- 2018
- Full Text
- View/download PDF
28. I-131 as adjuvant treatment for differentiated thyroid carcinoma may cause an increase in the incidence of secondary haematological malignancies: an 'inconvenient' truth?
- Author
-
Jasna Mihailovic, Luca Giovanella, Frederik A. Verburg, Ioannis Iakovou, Werner Langsteger, Markus Luster, Mark Konijnenberg, Michael Lassmann, and Radiology & Nuclear Medicine
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Incidence (epidemiology) ,medicine.medical_treatment ,Incidence ,General Medicine ,ORIGINAL REPORTS ,030218 nuclear medicine & medical imaging ,Thyroid carcinoma ,Iodine Radioisotopes ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,hemic and lymphatic diseases ,Hematologic Neoplasms ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Thyroid Neoplasms ,business ,Adjuvant - Abstract
PURPOSE: To investigate the risk and outcomes of second hematologic malignancies (SHMs) in a population-based cohort of patients with well-differentiated thyroid cancer (WDTC) treated or not with radioactive iodine (RAI). METHODS: Patients with WDTC were identified from SEER registries. Competing risk regression analysis was performed to calculate the risks of SHMs that occurred after WDTC treatment and outcomes after SHM development were assessed. RESULTS: Of 148,215 patients with WDTC, 53% received surgery alone and 47% received RAI. In total, 783 patients developed an SHM after a median interval of 6.5 years (interquartile range, 3.3 to 11.2 years) from WDTC diagnosis. In multivariable analysis, compared with those undergoing thyroidectomy alone, RAI treatment was associated with an increased early risk of developing acute myeloid leukemia (AML; hazard ratio, 1.79; 95% CI, 1.13 to 2.82; P = .01) and chronic myeloid leukemia (CML; hazard ratio, 3.44; 95% CI, 1.87 to 6.36; P < .001). This increased risk of AML and CML after RAI treatment was seen even in low-risk and intermediate-risk WDTC tumors. Occurrence of AML but not CML in patients with WDTC was associated with shorter median overall survival compared with matched controls (8.0 years v 31.0 years; P = .001). In addition, AML developing after RAI trended toward inferior survival compared with matched controls with de novo AML (median overall survival, 1.2 years v 2.9 years; P = .06). CONCLUSION: Patients with WDTC treated with RAI had an increased early risk of developing AML and CML but no other hematologic malignancies. AML that arises after RAI treatment has a poor prognosis. RAI use in patients with WDTC should be limited to patients with high-risk disease features, and patients with WDTC treated with adjuvant RAI should be monitored for myeloid malignancies as part of cancer surveillance.
- Published
- 2018
29. Persistent Hematologic Dysfunction after Peptide Receptor Radionuclide Therapy with Lu-177-DOTATATE: Incidence, Course, and Predicting Factors in Patients with Gastroenteropancreatic Neuroendocrine Tumors
- Author
-
B.L. Boen L.R. Kam, Dik J. Kwekkeboom, Eric P. Krenning, Wouter W. de Herder, Hendrik Bergsma, Kirsten van Lom, Marc H.G.P. Raaijmakers, Jaap J.M. Teunissen, Mark Konijnenberg, Radiology & Nuclear Medicine, Hematology, Internal Medicine, and Neurology
- Subjects
Oncology ,medicine.medical_specialty ,Pathology ,Cytopenia ,medicine.diagnostic_test ,business.industry ,Octreotide scan ,Bone metastasis ,Neuroendocrine tumors ,medicine.disease ,030218 nuclear medicine & medical imaging ,Cancer registry ,03 medical and health sciences ,Leukemia ,0302 clinical medicine ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,Radionuclide therapy ,medicine ,Radiology, Nuclear Medicine and imaging ,business ,Myeloproliferative neoplasm - Abstract
Peptide receptor radionuclide therapy (PRRT) may induce long-term toxicity to the bone marrow (BM). The aim of this study was to analyze persistent hematologic dysfunction (PHD) after PRRT with 177Lu-DOTATATE in patients with gastroenteropancreatic neuroendocrine tumors (GEP NETs). Methods: The incidence and course of PHD were analyzed in 274 GEP NET patients from a group of 367 patients with somatostatin receptor-positive tumors. PHD was defined as diagnosis of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML), myeloproliferative neoplasm (MPN), MDS/MPN, or otherwise unexplained cytopenia (for >6 mo). Using data from The Netherlands Cancer Registry, the expected number of hematopoietic neoplasms (MDS, AML, MPN, and MDS/MPN) was calculated and adjusted for sex, age, and follow-up period. The following risk factors were assessed: sex, age over 70 y, bone metastasis, prior chemotherapy, prior external-beam radiotherapy, uptake on the [111In-DTPA0]octreotide scan, tumor load, grade 3-4 hematologic toxicity during treatment, estimated absorbed BM dose, elevated plasma chromogranin A level, baseline blood counts, and renal function. Results: Eleven (4%) of the 274 patients had PHD after treatment with 177Lu-DOTATATE: 8 patients (2.9%) developed a hematopoietic neoplasm (4 MDS, 1 AML, 1 MPN, and 2 MDS/MPN) and 3 patients (1.1%) developed BM failure characterized by cytopenia and BM aplasia. The median latency period at diagnosis (or first suspicion of a PHD) was 41 mo (range, 15-84 mo). The expected number of hematopoietic neoplasms based on The Netherlands Cancer Registry data was 3.0, resulting in a relative risk of 2.7 (95% confidence interval, 0.7-10.0). No risk factors for PHD could be identified for the GEP NET patients, not even bone metastasis or estimated BM dose. Seven patients with PHD developed anemia in combination with a rise in mean corpuscular volume. Conclusion: The prevalence of PHD after PRRT with 177Lu-DOTATATE was 4% in our patient population. The median time at which PHD developed was 41 mo after the first PRRT cycle. The relative risk for developing a hematopoietic neoplasm was 2.7. No risk factors were found for the development of PHD in GEP NET patients.
- Published
- 2018
30. EANM practical guidance on uncertainty analysis for molecular radiotherapy absorbed dose calculations
- Author
-
Johan Gustafsson, Maurice G Cox, Jonathan Gear, Glenn D. Flux, Katarina Sjögreen Gleisner, Gerhard Glatting, Mark Konijnenberg, Iain Murray, and Radiology & Nuclear Medicine
- Subjects
Volume of interest ,medicine.medical_treatment ,Partial volume ,Guidelines ,030218 nuclear medicine & medical imaging ,Recovery coefficient ,03 medical and health sciences ,0302 clinical medicine ,Dosimetry ,Neoplasms ,Calibration ,medicine ,Humans ,Applied mathematics ,Yttrium Radioisotopes ,Radiology, Nuclear Medicine and imaging ,Uncertainty analysis ,Mathematics ,Radiotherapy Planning, Computer-Assisted ,Uncertainty ,Radiotherapy Dosage ,General Medicine ,Radiation therapy ,030220 oncology & carcinogenesis ,Absorbed dose ,Practice Guidelines as Topic ,Radiopharmaceuticals ,Algorithms - Abstract
A framework is proposed for modelling the uncertainty in the measurement processes constituting the dosimetry chain that are involved in internal absorbed dose calculations. The starting point is the basic model for absorbed dose in a site of interest as the product of the cumulated activity and a dose factor. In turn, the cumulated activity is given by the area under a time-activity curve derived from a time sequence of activity values. Each activity value is obtained in terms of a count rate, a calibration factor and a recovery coefficient (a correction for partial volume effects). The method to determine the recovery coefficient and the dose factor, both of which are dependent on the size of the volume of interest (VOI), are described. Consideration is given to propagating estimates of the quantities concerned and their associated uncertainties through the dosimetry chain to obtain an estimate of mean absorbed dose in the VOI and its associated uncertainty. This approach is demonstrated in a clinical example.
- Published
- 2018
31. Trastuzumab co-treatment improves survival of mice with PC-3 prostate cancer xenografts treated with [177Lu]Lu-DOTAGA-PEG2-RM26 GRPR antagonists
- Author
-
Theodosia Maina, Anzhelika Vorobyeva, Berthold A. Nock, Anna Orlova, Bogdan Mitran, Ulrika Rosenström, Mohamed Altai, Vladimir Tolmachev, Sara S. Rinne, and Mark Konijnenberg
- Subjects
Cancer Research ,Prostate cancer ,business.industry ,Trastuzumab ,Cancer research ,Molecular Medicine ,Medicine ,Radiology, Nuclear Medicine and imaging ,business ,medicine.disease ,medicine.drug - Published
- 2019
- Full Text
- View/download PDF
32. Subacute haematotoxicity after PRRT with 177Lu-DOTA-octreotate: prognostic factors, incidence and course
- Author
-
Dik J. Kwekkeboom, Wouter W. de Herder, Eric P. Krenning, Peter P. Kooij, Casper H.J. van Eijck, Hendrik Bergsma, Jaap J.M. Teunissen, Gaston J H Franssen, Mark Konijnenberg, Boen L. R. Kam, Radiology & Nuclear Medicine, Internal Medicine, and Surgery
- Subjects
Male ,medicine.medical_treatment ,Octreotide ,Lutetium ,Gastroenterology ,030218 nuclear medicine & medical imaging ,177Lu-DOTATATE ,0302 clinical medicine ,Bone Marrow ,Risk Factors ,Prospective Studies ,Prospective cohort study ,Netherlands ,Hematology ,General Medicine ,Middle Aged ,Prognosis ,Neuroendocrine Tumors ,Treatment Outcome ,medicine.anatomical_structure ,Radiology Nuclear Medicine and imaging ,030220 oncology & carcinogenesis ,Toxicity ,Original Article ,Female ,PRRT ,Iodine ,medicine.drug ,medicine.medical_specialty ,Receptors, Peptide ,Renal function ,03 medical and health sciences ,Dosimetry ,White blood cell ,Internal medicine ,Organometallic Compounds ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Radiation Injuries ,Radiometry ,Aged ,Radioisotopes ,Chemotherapy ,business.industry ,Radionuclide therapy ,Radiopharmaceuticals ,business ,Nuclear medicine - Abstract
Purpose In peptide receptor radionuclide therapy (PRRT), the bone marrow (BM) is one of the dose-limiting organs. The accepted dose limit for BM is 2 Gy, adopted from 131I treatment. We investigated the incidence and duration of haematological toxicity and its risk factors in patients treated with PRRT with 177Lu-DOTA0-Tyr3-octreotate (177Lu-DOTATATE). Also, absorbed BM dose estimates were evaluated and compared with the accepted 2 Gy dose limit. Methods The incidence and duration of grade 3 or 4 haematological toxicity (according to CTCAE v3.0) and risk factors were analysed. Mean BM dose per unit (gigabecquerels) of administered radioactivity was calculated and the correlations between doses to the BM and haematological risk factors were determined. Results Haematological toxicity (grade 3/4) occurred in 34 (11 %) of 320 patients. In 15 of the 34 patients, this lasted more than 6 months or blood transfusions were required. Risk factors significantly associated with haematological toxicity were: poor renal function, white blood cell (WBC) count
- Published
- 2015
- Full Text
- View/download PDF
33. Accurate assessment of whole-body retention for PRRT with 177Lu using paired measurements with external detectors
- Author
-
Hubert Th. Wolterbeek, Peter Bode, Mark Konijnenberg, Wouter A.P. Breeman, Boxue Liu, and Erik de Blois
- Subjects
Pharmacology ,Accuracy and precision ,Chromatography ,Chemistry ,Dose Calibrator ,business.industry ,Detector ,Urine ,Urine collection method ,Radiology, Nuclear Medicine and imaging ,Coverage factor ,Whole body ,Nuclear medicine ,business ,Urine collection - Abstract
The aim of this study was to assess the accuracy of the results of whole-body measurements by comparison with the urine collection method in the PRRT with 177 Lu and furthermore to develop a more accurate method of paired measurements. Excreted samples were collected at given intervals and activities were measured by a dose calibrator. Traditionally, whole-body activities during subsequent measurements are normalized individually to the administered activity. In order to correct for the effects of the activity in the bladder during the baseline measurement before the first voiding and activity redistributions in the patient body during subsequent measurements, a series of paired measurements before and after each voiding were carried out. Time-dependent detector responses at given times were derived and time-activity retentions were then determined. Compared to the results of the urine collection, whole-body activities by traditional whole-body measurements were overestimated by ca. 14% at 1 h after administration and randomly varied from -29% to 49% at 24 h. Measurement uncertainties of whole-body activities were from ±4% (the coverage factor k=2) at 1 h to >±20% at 24 h by the urine collection and ±7% by paired measurements, respectively. Whole-body activities at 1 h by paired measurements were validated using the results by measurements of the collected first urine. The new method of paired measurements has an equivalent measurement accuracy and even better during the later measurements with respect to the urine collection method and therefore can replace urine approach for assessing the time-activity remaining in the patient body.
- Published
- 2015
- Full Text
- View/download PDF
34. Consequences of meta-stable 177mLu admixture in 177Lu for patient dosimetry
- Author
-
Mark Konijnenberg
- Subjects
Pharmacology ,Physics ,business.industry ,Rare earth ,Radiation dose ,Radiochemistry ,Half-life ,Beta decay ,Excited state ,Patient dosimetry ,Absorbed dose ,Radiology, Nuclear Medicine and imaging ,Nuclear medicine ,business - Abstract
Lutetium-177 ((177)Lu) is a rare earth metal in the lanthanides series which decays by beta emission with a half life of 6.647 days to three excited states and the ground state of (177)Hf. When (177)Lu is produced by neutron capture in (176)Lu, inevitably an admixture is formed of the long-lived isomer (177)mLu. As its half-life of 160.4 days is so much longer than that of (177)Lu, concerns are raised on its possible enhancement in radiation dose to the patient treated with (177)Lu-DOTA-octreotate. This report evaluates this possible enhancement of the absorbed dose, based on the published pharmacokinetic profile of (177)Lu-DOTA-octreotate and assuming an admixture of 1 kBq (177)mLu /MBq (177)Lu (0.1%).
- Published
- 2015
- Full Text
- View/download PDF
35. Theranostic management of medullary thyroid cancer (MTC) with (111In/177Lu) CP04: how close are we to a clinical solution?
- Author
-
Clemens Decristoforo, Georg Goebel, Christine Rangger, Sebastijan Rep, Jong Marion de, Alide C Froberg, Mark Konijnenberg, Lorenza Scarpa, Katja Zaletel, Petra Kolenc-Peitl, Damijan Bergant, Helmut R. Maecke, Paola Anna Erba, Irene Virgolini, Renata Mikolajczak, Elwira Przybylik-Mazurek, Konrad Skorkiewicz, Luka Lezaic, Anna Sowa-Staszczak, Dariusz Pawlak, Bogusław Głowa, Alicja Hubalewska-Dydejczyk, Berthold A. Nock, Pirotr Garnuszek, Malgorzata Trofimiuk-Muldner, and Theodosia Maina-Nock
- Subjects
Pathology ,medicine.medical_specialty ,business.industry ,Medicine ,Medullary thyroid cancer ,Radiology ,business ,medicine.disease - Published
- 2017
- Full Text
- View/download PDF
36. In Vitro comparison of 213Bi- and 177Lu-radiation for peptide receptor radionuclide therapy
- Author
-
Ho Sze Chan, Marion de Jong, Wouter A.P. Breeman, Alfred Morgenstern, Mark Konijnenberg, Erik de Blois, and Frank Bruchertseifer
- Subjects
Cell Membranes ,lcsh:Medicine ,Lutetium ,Octreotide ,030218 nuclear medicine & medical imaging ,Binding Analysis ,0302 clinical medicine ,Mathematical and Statistical Techniques ,Linear Energy Transfer ,Receptors, Somatostatin ,lcsh:Science ,Incubation ,Liquid Chromatography ,Multidisciplinary ,Radiochemistry ,Radiation ,Chemistry ,Physics ,Chromatographic Techniques ,Absorption, Radiation ,Curve Fitting ,Dose–response relationship ,Radioactivity ,030220 oncology & carcinogenesis ,Absorbed dose ,Physical Sciences ,Cellular Structures and Organelles ,Monte Carlo Method ,Cell Binding Assay ,Research Article ,Cell Binding ,Cell Physiology ,Cell Survival ,Linear energy transfer ,Research and Analysis Methods ,Models, Biological ,Incubation period ,03 medical and health sciences ,Cell Line, Tumor ,Organometallic Compounds ,Dosimetry ,Humans ,Irradiation ,Radiometry ,Receptor Binding Assays ,Chemical Characterization ,Nuclear Physics ,Radioisotopes ,lcsh:R ,Biology and Life Sciences ,Dose-Response Relationship, Radiation ,Cell Biology ,High Performance Liquid Chromatography ,Kinetics ,Radionuclide therapy ,lcsh:Q ,Radiopharmaceuticals ,Bismuth ,Mathematical Functions - Abstract
Background Absorbed doses for α-emitters are different from those for β-emitters, as the high linear energy transfer (LET) nature of α-particles results in a very dense energy deposition over a relatively short path length near the point of emission. This highly localized and therefore high energy deposition can lead to enhanced cell-killing effects at absorbed doses that are non-lethal in low-LET type of exposure. Affinities of DOTA-DPhe1-Tyr3-octreotate (DOTATATE), 115In-DOTATATE, 175Lu-DOTATATE and 209Bi-DOTATATE were determined in the K562-SST2 cell line. Two other cell lines were used for radiation response assessment; BON and CA20948, with a low and high expression of somatostatin receptors, respectively. Cellular uptake kinetics of 111In-DOTATATE were determined in CA20948 cells. CA20948 and BON were irradiated with 137Cs, 177Lu-DTPA, 177Lu-DOTATATE, 213Bi-DTPA and 213Bi-DOTATATE. Absorbed doses were calculated using the MIRDcell dosimetry method for the specific binding and a Monte Carlo model of a cylindrical 6-well plate geometry for the exposure by the radioactive incubation medium. Absorbed doses were compared to conventional irradiation of cells with 137Cs and the relative biological effect (RBE) at 10% survival was calculated. Results IC50 of (labelled) DOTATATE was in the nM range. Absorbed doses up to 7 Gy were obtained by 5.2 MBq 213Bi-DOTATATE, in majority the dose was caused by α-particle radiation. Cellular internalization determined with 111In-DOTATATE showed a linear relation with incubation time. Cell survival after exposure of 213Bi-DTPA and 213Bi-DOTATATE to BON or CA20948 cells showed a linear-exponential relation with the absorbed dose, confirming the high LET character of 213Bi. The survival of CA20948 after exposure to 177Lu-DOTATATE and the reference 137Cs irradiation showed the typical curvature of the linear-quadratic model. 10% Cell survival of CA20948 was reached at 3 Gy with 213Bi-DOTATATE, a factor 6 lower than the 18 Gy found for 177Lu-DOTATATE and also below the 5 Gy after 137Cs external exposure. Conclusion 213Bi-DTPA and 213Bi-DOTATATE lead to a factor 6 advantage in cell killing compared to 177Lu-DOTATATE. The RBE at 10% survival by 213Bi-ligand compared to 137Cs was 2.0 whereas the RBE for 177Lu-DOTATATE was 0.3 in the CA20948 in vitro model.
- Published
- 2017
37. Variations in the practice of molecular radiotherapy and implementation of dosimetry: results from a European survey
- Author
-
Jill Tipping, Carlo Chiesa, Francesco Cicone, Glenn D. Flux, Caroline Stokke, Mark Konijnenberg, Boudewijn Brans, Maria Paphiti, Katarina Sjögreen Gleisner, Emiliano Spezi, Pablo Minguez Gabina, Pavel Solny, Mattias Sandström, and Radiology & Nuclear Medicine
- Subjects
medicine.medical_treatment ,MULTICENTER ,Radionuclide therapy ,Radiopharmaceutical therapy ,030218 nuclear medicine & medical imaging ,Dosimetry ,European survey ,Molecular radiotherapy ,Prostate cancer ,0302 clinical medicine ,INTERNAL DOSIMETRY ,Medicine and Health Sciences ,Instrumentation ,Original Research ,BONE METASTASES ,Radiation ,Thyroid ,3. Good health ,PROSTATE-CANCER ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Radiopharmaceutical ,Radiology, Nuclear Medicine and Medical Imaging ,lcsh:Medical physics. Medical radiology. Nuclear medicine ,medicine.medical_specialty ,lcsh:R895-920 ,Biomedical Engineering ,INDIVIDUALIZED DOSIMETRY ,Medical physicist ,03 medical and health sciences ,KIDNEY ,Internal medicine ,medicine ,RADIOIODINE TREATMENT ,Radiology, Nuclear Medicine and imaging ,Internal dosimetry ,Medical physics ,Medical prescription ,DIFFERENTIATED THYROID-CANCER ,therapy ,business.industry ,REFRACTORY ,medicine.disease ,Radiation therapy ,Radiologi och bildbehandling ,RECEPTOR RADIONUCLIDE THERAPY ,business ,DOSE-RESPONSE - Abstract
Background Currently, the implementation of dosimetry in molecular radiotherapy (MRT) is not well investigated, and in view of the Council Directive (2013/59/Euratom), there is a need to understand the current availability of dosimetry-based MRT in clinical practice and research studies. The aim of this study was to assess the current practice of MRT and dosimetry across European countries. Methods An electronic questionnaire was distributed to European countries. This addressed 18 explicitly considered therapies, and for each therapy, a similar set of questions were included. Questions covered the number of patients and treatments during 2015, involvement of medical specialties and medical physicists, implementation of absorbed dose planning, post-therapy imaging and dosimetry, and the basis of therapy prescription. Results Responses were obtained from 26 countries and 208 hospitals, administering in total 42,853 treatments. The most common therapies were 131I-NaI for benign thyroid diseases and thyroid ablation of adults. The involvement of a medical physicist (mean over all 18 therapies) was reported to be either minority or never by 32% of the responders. The percentage of responders that reported that dosimetry was included on an always/majority basis differed between the therapies and showed a median value of 36%. The highest percentages were obtained for 177Lu-PSMA therapy (100%), 90Y microspheres of glass (84%) and resin (82%), 131I-mIBG for neuroblastoma (59%), and 131I-NaI for benign thyroid diseases (54%). The majority of therapies were prescribed based on fixed-activity protocols. The highest number of absorbed-dose based prescriptions were reported for 90Y microsphere treatments in the liver (64% and 96% of responses for resin and glass, respectively), 131I-NaI treatment of benign thyroid diseases (38% of responses), and for 131I-mIBG treatment of neuroblastoma (18% of responses). Conclusions There is a wide variation in MRT practice across Europe and for different therapies, including the extent of medical-physicist involvement and the implementation of dosimetry-guided treatments. Electronic supplementary material The online version of this article (10.1186/s40658-017-0193-4) contains supplementary material, which is available to authorized users.
- Published
- 2017
38. The conflict between treatment optimization and registration of radiopharmaceuticals with fixed activity posology in oncological nuclear medicine therapy
- Author
-
Carlo Chiesa, Mark Konijnenberg, Eric P. Visser, Klaus Bacher, Lidia Strigari, Glenn D. Flux, Stephan Walrand, Michael Lassmann, K. Sjogreen Gleisner, Jonathan Gear, Michael Ljungberg, Nicolas Chouin, Uta Eberlein, Manuel Bardiès, Radiology & Nuclear Medicine, Nuclear Medicine [Milan, Italy], Foundation IRCCS Istituto Nazionale Tumori [Milan, Italy], Lund University [Lund], Molecular Carcinogenesis [Sutton], Institute of cancer research, Translational Cancer Genetics [Sutton], Université Catholique de Louvain (UCL), Department of Basic Medical Sciences [Ghent, Belgium], Division of Medical Physics [Ghent, Belgium], Ghent University [Belgium] (UGENT)-Ghent University [Belgium] (UGENT), Department of Radiology and Nuclear Medicine [Nijmegen], Radboud University Medical Center [Nijmegen], Centre de Recherche sur le Cancer Nantes-Angers (LUNAM), Université d'Angers (UA)-Université de Nantes (UN), Medical Radiation Physics - Department of Clinical Sciences [Lund, Sweden], Centre de Recherches en Cancérologie de Toulouse (CRCT), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Department of Nuclear Medicine [Würzburg, Germany], Julius-Maximilians-Universität Würzburg [Wurtzbourg, Allemagne] (JMU), Laboratory of Medical Physics and Expert Systems [Rome, Italy], National Cancer Institute Regina Elena [Rome, Italy], Department of Medical Oncology [Rotterdam], Erasmus University Medical Center [Rotterdam] (Erasmus MC), Université Catholique de Louvain = Catholic University of Louvain (UCL), Universiteit Gent = Ghent University [Belgium] (UGENT)-Universiteit Gent = Ghent University [Belgium] (UGENT), Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), Centre Hospitalier Universitaire d'Angers (CHU Angers), and PRES Université Nantes Angers Le Mans (UNAM)-PRES Université Nantes Angers Le Mans (UNAM)-Hôtel-Dieu de Nantes-Institut National de la Santé et de la Recherche Médicale (INSERM)-Hôpital Laennec-Centre National de la Recherche Scientifique (CNRS)-Faculté de Médecine d'Angers-Centre hospitalier universitaire de Nantes (CHU Nantes)
- Subjects
medicine.medical_specialty ,Radiobiology ,business.industry ,Other Research Radboud Institute for Health Sciences [Radboudumc 0] ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,General Medicine ,Neuroendocrine tumors ,medicine.disease ,030218 nuclear medicine & medical imaging ,3. Good health ,Microsphere ,03 medical and health sciences ,0302 clinical medicine ,Government regulation ,MESH: Neoplasms/radiotherapy ,Nuclear Medicine/legislation & jurisprudence ,Radiation Dosage ,Radiopharmaceuticals/therapeutic use ,030220 oncology & carcinogenesis ,medicine ,Radiology, Nuclear Medicine and imaging ,Medical physics ,Nuclear medicine ,business - Abstract
International audience; The “intended purpose” in all therapeutic exposures is treatment efficacy against malignant disease. The optimization principle (as low as reasonably achievable, ALARA) of article 56, when applied in a therapy situation, states that absorbed doses to nontarget tissues should be kept reasonably low, but not so low as to lose efficacy. We think that this applies above all to the fight against life-threatening cancer. As a consequence, we believe that to adhere to the optimization principle in oncological patients, nuclear medicine therapy should be based on individualized dosimetry.
- Published
- 2017
- Full Text
- View/download PDF
39. [In-111-DTPA]octreotide Tumor Uptake in GEPNET Liver Metastases After Intra-Arterial Administration: An Overview of Preclinical and Clinical Observations and Implications for Tumor Radiation Dose After Peptide Radionuclide Therapy
- Author
-
Woulter A P Breeman, Gerben A. Koning, Mark Konijnenberg, Eric P. Krenning, Marion de Jong, Timo L.M. ten Hagen, Casper H.J. van Eijck, Boen L. R. Kam, Stefan E. Pool, Radiology & Nuclear Medicine, and Surgery
- Subjects
Adult ,Male ,Cancer Research ,Biodistribution ,medicine.medical_specialty ,Urology ,Octreotide ,Peptide ,Liver Neoplasms, Experimental ,Stomach Neoplasms ,Intestinal Neoplasms ,medicine ,Animals ,Humans ,Infusions, Intra-Arterial ,Tissue Distribution ,Radiology, Nuclear Medicine and imaging ,Pharmacology ,chemistry.chemical_classification ,business.industry ,Somatostatin receptor ,Indium Radioisotopes ,General Medicine ,Middle Aged ,Pentetic Acid ,Rats ,Pancreatic Neoplasms ,Disease Models, Animal ,Neuroendocrine Tumors ,medicine.anatomical_structure ,Somatostatin ,Oncology ,chemistry ,Rats, Inbred Lew ,Radionuclide therapy ,Abdomen ,Female ,Radiopharmaceuticals ,business ,Nuclear medicine ,medicine.drug ,Artery - Abstract
Aims: With the aim to improve peptide receptor radionuclide therapy effects in patients with gastroenteropancreatic neuroendocrine tumor (GEPNET) liver metastases we explored the effect of intra-arterial (IA) administration of [In-111-DTPA]octreotide (In-111-DTPAOC) on tumor uptake in an animal model and in a patient study. Methods: Preclinical study: After administering In-111-DTPAOC intra-venously (IV) or IA, biodistribution studies were performed in rats with a hepatic somatostatin receptor subtype 2 (sst(2))-positive tumor. Clinical study: 3 patients with neuroendocrine liver metastases were injected twice with In-111-DTPAOC. The first injection was given IV, and 2 weeks later, the second was injected IA (hepatic artery). Planar images of the abdomen were made up to 72 hours after injection. Blood samples were taken and urine was collected. Pharmacokinetic modeling was performed on the IV and IA data of the same patient. Based on this model, additional Lu-177 dosimetry calculations for IV and IA administrations were performed. Results: The preclinical study showed a two-fold higher In-111-DTPAOC tumor uptake after IA administration than after IV injection. Patient data showed a large variability in radioactivity increment in liver metastases after IA administration compared with IV administration. Renal radioactivity was not significantly lower after IA administration; Lu-177 dosimetry simulations in 1 patient using a maximum kidney radiation dose of 23Gy showed IA administration resulted in a mean increase in tumor radiation dose of 2.9-fold. Conclusion: Preclinical and clinical data both indicate that IA administration of radiolabeled somatostatin analogs via the hepatic artery can significantly increase radionuclide uptake in GEPNET, sst(2)-positive, liver metastases up to 72 hours postinjection, although the effect of IA administration can differ between patients.
- Published
- 2014
- Full Text
- View/download PDF
40. Application of single-vial ready-for-use formulation of In-111- or Lu-177-labelled somatostatin analogs
- Author
-
Rory M. S. de Zanger, Erik de Blois, Wouter A.P. Breeman, Ho Sze Chan, Mark Konijnenberg, and Radiology & Nuclear Medicine
- Subjects
Radioisotopes ,Radiation ,Chromatography ,Ethanol ,Chemistry, Pharmaceutical ,Drug Storage ,Gentisates ,Indium Radioisotopes ,Ascorbic Acid ,Lutetium ,Ascorbic acid ,Vial ,digestive system ,Solutions ,chemistry.chemical_compound ,Somatostatin ,chemistry ,Drug Stability ,Organic chemistry ,Humans ,Gentisic acid ,Radiopharmaceuticals ,Chromatography, High Pressure Liquid - Abstract
For the sake of safety it would be desirable to store and transport the ready-for-use liquid formulation (diagnostics and therapeutics) of radiolabelled peptides. The use of ethanol, in combination with a mixture of gentisic- and ascorbic acid, has superior effects on stabilizing radiolabelled somatostatin analogs. As a consequence, In-111- and Lu-177-labelled somatostatin analogs can be stored and transported in a single-vial ready-for-use liquid formulation up to 7 days after radiolabelling. (C) 2013 Elsevier Ltd. All rights reserved.
- Published
- 2014
41. Effectiveness of Quenchers to Reduce Radiolysis of 111In- or 177Lu-Labelled Methionine-Containing Regulatory Peptides. Maintaining Radiochemical Purity as Measured by HPLC
- Author
-
Mark Konijnenberg, Wouter A.P. Breeman, Ho Sze Chan, Erik de Blois, Rory M. S. de Zanger, and Radiology & Nuclear Medicine
- Subjects
Lutetium ,Minigastrin ,digestive system ,High-performance liquid chromatography ,Excipients ,Heterocyclic Compounds, 1-Ring ,chemistry.chemical_compound ,Methionine ,Protein stability ,Drug Discovery ,Humans ,Organic chemistry ,Radiometry ,Chromatography, High Pressure Liquid ,Chelating Agents ,Chromatography ,Protein Stability ,Indium Radioisotopes ,Radiation dose ,General Medicine ,Pentetic Acid ,Radioactivity ,chemistry ,Isotope Labeling ,Radiolysis ,Radiopharmaceuticals ,Peptides - Abstract
An overview how to measure and to quantify radiolysis by the addition of quenchers and to maintain Radio-Chemical Purity (RCP) of vulnerable methionine-containing regulatory peptides is presented. High RCP was only achieved with a combination of quenchers. However, quantification of RCP is not standardized, and therefore comparison of radiolabelling and RCP of regulatory peptides between different HPLC-systems and between laboratories is cumbersome. Therefore we suggest a set of standardized requirements to quantify RCP by HPLC for radiolabelled DTPA- or DOTA-peptides. Moreover, a dosimetry model was developed to calculate the doses in the reaction vials during radiolabelling and storage of the radiopeptides, and to predict RCP in the presence and absence of quenchers. RCP was measured by HPLC, and a relation between radiation dose and radiolysis of RCP was established. The here described quenchers are tested individually as ƒ(concentration) to investigate efficacy to reduce radiolysis of radiolabelled methionine-containing regulatory peptides.
- Published
- 2013
- Full Text
- View/download PDF
42. Whole organ and islet of Langerhans dosimetry for calculation of absorbed doses resulting from imaging with radiolabeled exendin
- Author
-
Wietske Woliner-van der Weg, Lieke Joosten, Cathelijne Frielink, Maarten Brom, Martin Gotthardt, Inge van der Kroon, Mark Konijnenberg, Eric P. Visser, and Radiology & Nuclear Medicine
- Subjects
Male ,endocrine system diseases ,Cell Count ,Kidney ,030218 nuclear medicine & medical imaging ,Diabetes mellitus genetics ,0302 clinical medicine ,Insulin-Secreting Cells ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Medicine ,Multidisciplinary ,geography.geographical_feature_category ,Radiation ,digestive, oral, and skin physiology ,Indium Radioisotopes ,Middle Aged ,Islet ,3. Good health ,medicine.anatomical_structure ,Absorbed dose ,Intercellular Signaling Peptides and Proteins ,Female ,Beta cell ,Pancreas ,Adult ,medicine.medical_specialty ,endocrine system ,030209 endocrinology & metabolism ,Gallium Radioisotopes ,Radiation Dosage ,Rats, Mutant Strains ,Article ,03 medical and health sciences ,Islets of Langerhans ,Young Adult ,In vivo ,Internal medicine ,Diabetes Mellitus ,Dosimetry ,Animals ,Humans ,Radiation Injuries ,Radiometry ,geography ,business.industry ,Other Research Radboud Institute for Health Sciences [Radboudumc 0] ,Kidney metabolism ,Rats ,Disease Models, Animal ,Endocrinology ,business ,Peptides - Abstract
Radiolabeled exendin is used for non-invasive quantification of beta cells in the islets of Langerhans in vivo. High accumulation of radiolabeled exendin in the islets raised concerns about possible radiation-induced damage to these islets in man. In this work, islet absorbed doses resulting from exendin-imaging were calculated by combining whole organ dosimetry with small scale dosimetry for the islets. Our model contains the tissues with high accumulation of radiolabeled exendin: kidneys, pancreas and islets. As input for the model, data from a clinical study (radiolabeled exendin distribution in the human body) and from a preclinical study with Biobreeding Diabetes Prone (BBDP) rats (islet-to-exocrine uptake ratio, beta cell mass) were used. We simulated 111In-exendin and 68Ga-exendin absorbed doses in patients with differences in gender, islet size, beta cell mass and radiopharmaceutical uptake in the kidneys. In all simulated cases the islet absorbed dose was small, maximum 1.38 mGy for 68Ga and 66.0 mGy for 111In. The two sources mainly contributing to the islet absorbed dose are the kidneys (33–61%) and the islet self-dose (7.5–57%). In conclusion, all islet absorbed doses are low (
- Published
- 2016
43. Improved safety and efficacy of Bi-213-DOTATATE-targeted alpha therapy of somatostatin receptor-expressing neuroendocrine tumors in mice pre-treated with L-lysine
- Author
-
Robert W. Atcher, Mehran Makvandi, Erik de Blois, Tamara Daniels, Jeffrey P. Norenberg, Wouter A.P. Breeman, Monique Nysus, Mark Konijnenberg, Ho Sze Chan, Marion de Jong, and Radiology & Nuclear Medicine
- Subjects
medicine.medical_specialty ,business.industry ,Therapeutic effect ,Nephron ,Pharmacology ,Neuroendocrine tumors ,medicine.disease ,030218 nuclear medicine & medical imaging ,Nephrotoxicity ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,medicine.anatomical_structure ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Absorbed dose ,Internal medicine ,Radionuclide therapy ,Medicine ,Dosimetry ,Radiology, Nuclear Medicine and imaging ,business - Abstract
Targeted alpha therapy (TAT) offers advantages over current β-emitting conjugates for peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors. PRRT with 177Lu-DOTATATE or 90Y-DOTATOC has shown dose-limiting nephrotoxicity due to radiopeptide retention in the proximal tubules. Pharmacological protection can reduce renal uptake of radiopeptides, e.g., positively charged amino acids, to saturate in the proximal tubules, thereby enabling higher radioactivity to be safely administered. The aim of this preclinical study was to evaluate the therapeutic effect of 213Bi-DOTATATE with and without renal protection using L-lysine in mice. Tumor uptake and kinetics as a function of injected mass of peptide (range 0.03–3 nmol) were investigated using 111In-DOTATATE. These results allowed estimation of the mean radiation absorbed tumor dose for 213Bi-DOTATATE. Pharmacokinetics and dosimetry of 213Bi-DOTATATE was determined in mice, in combination with renal protection. A dose escalation study with 213Bi-DOTATATE was performed to determine the maximum tolerated dose (MTD) with and without pre-administration of l-lysine as for renal protection. Neutrophil gelatinase-associated lipocalin (NGAL) served as renal biomarker to determine kidney injury. The maximum mean radiation absorbed tumor dose occurred at 0.03 nmol and the minimum at 3 nmol. Similar mean radiation absorbed tumor doses were determined for 0.1 and 0.3 nmol with a mean radiation absorbed dose of approximately 0.5 Gy/MBq 213Bi-DOTATATE. The optimal mass of injected peptide was found to be 0.3 nmol. Tumor uptake was similar for 111In-DOTATATE and 213Bi-DOTATATE at 0.3 nmol peptide. Lysine reduced the renal uptake of 213Bi-DOTATATE by 50% with no effect on the tumor uptake. The MTD was
- Published
- 2016
- Full Text
- View/download PDF
44. In Vivo Stabilization of a Gastrin-Releasing Peptide Receptor Antagonist Enhances PET Imaging and Radionuclide Therapy of Prostate Cancer in Preclinical Studies
- Author
-
Jean Martinez, Wytske M. van Weerden, Jean-Alain Fehrentz, Sander Hoeben, Corrina M.A. de Ridder, Dik C. van Gent, Theodosia Maina, Erik de Blois, Luc Brunel, Berthold A. Nock, Marion de Jong, Julie Nonnekens, Kristell L.S. Chatalic, Mark Konijnenberg, Radiology & Nuclear Medicine, Urology, Molecular Genetics, Royal Marsden Hospital NHS Foundation TrustSarcoma/Melanoma UnitDept of Academic SurgeryFulham RoadLondon SW3 6JJUnited Kingdom, Institut des Biomolécules Max Mousseron [Pôle Chimie Balard] (IBMM), Centre National de la Recherche Scientifique (CNRS)-Institut de Chimie du CNRS (INC)-Université de Montpellier (UM)-Ecole Nationale Supérieure de Chimie de Montpellier (ENSCM), Institute of Physical Chemistry 'Demokritos' (IPC), National Center for Scientific Research 'Demokritos' (NCSR), Department of Radiology & Nuclear Medicine [Rotterdam, The Netherlands], and Erasmus University Medical Center [Rotterdam] (Erasmus MC)
- Subjects
phosphoramidon ,Male ,theranostics ,Pathology ,medicine.medical_specialty ,GRPR ,PET imaging ,Medicine (miscellaneous) ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Antineoplastic Agents ,030218 nuclear medicine & medical imaging ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,In vivo ,Gastrin-releasing peptide receptor ,Animals ,Medicine ,enzyme inhibition ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Neprilysin ,Radioisotopes ,Radiotherapy ,biology ,business.industry ,Phosphoramidon ,radionuclide therapy, prostate cancer ,Proteolytic enzymes ,Prostatic Neoplasms ,radionuclide therapy ,prostate cancer ,medicine.disease ,neutral endopeptidase ,3. Good health ,Receptors, Bombesin ,Disease Models, Animal ,chemistry ,Enzyme inhibitor ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Radionuclide therapy ,Cancer research ,biology.protein ,business ,Research Paper - Abstract
International audience; A single tool for early detection, accurate staging, and personalized treatment of prostate cancer (PCa) would be a major breakthrough in the field of PCa. Gastrin-releasing peptide receptor (GRPR) targeting peptides are promising probes for a theranostic approach for PCa overexpressing GRPR. However, the successful application of small peptides in a theranostic approach is often hampered by their fast in vivo degradation by proteolytic enzymes, such as neutral endopeptidase (NEP). Here we show for the first time that co-injection of a NEP inhibitor (phosphoramidon (PA)) can lead to an impressive enhancement of diagnostic sensitivity and therapeutic efficacy of the theranostic 68Ga-/177Lu-JMV4168 GRPR-antagonist. Co-injection of PA (300 µg) led to stabilization of 177Lu-JMV4168 in murine peripheral blood. In PC-3 tumor-bearing mice, PA co-injection led to a two-fold increase in tumor uptake of 68Ga-/177Lu-JMV4168, 1 h after injection. In positron emission tomography (PET) imaging with 68Ga-JMV4168, PA co-injection substantially enhanced PC-3 tumor signal intensity. Radionuclide therapy with 177Lu-JMV4168 resulted in significant regression of PC-3 tumor size. Radionuclide therapy efficacy was confirmed by production of DNA double strand breaks, decreased cell proliferation and increased apoptosis. Increased survival rates were observed in mice treated with 177Lu-JMV4168 plus PA as compared to those without PA. This data shows that co-injection of the enzyme inhibitor PA greatly enhances the theranostic potential of GRPR-radioantagonists for future application in PCa patients.
- Published
- 2016
- Full Text
- View/download PDF
45. Nephrotoxicity profiles and threshold dose values for [177Lu]-DOTATATE in nude mice
- Author
-
Johan Mölne, Johanna Svensson, Mark Konijnenberg, Eva Forssell-Aronsson, Peter Bernhardt, and Radiology & Nuclear Medicine
- Subjects
Cancer Research ,medicine.medical_specialty ,Kidney Cortex ,Renal cortex ,Mice, Nude ,Kidney ,Octreotide ,Radiation Dosage ,Injections ,Nephrotoxicity ,Kidney Tubules, Proximal ,Mice ,Bone Marrow ,Cortex (anatomy) ,Internal medicine ,Toxicity Tests ,Organometallic Compounds ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Radiometry ,business.industry ,Body Weight ,Kidney metabolism ,medicine.anatomical_structure ,Endocrinology ,Renal physiology ,Toxicity ,Radionuclide therapy ,Molecular Medicine ,Female ,business ,Biomarkers - Abstract
Introduction In peptide receptor radionuclide therapy for neuroendocrine tumors the main dose-limiting tissue is found in the kidneys because of tubular reabsorption and retention of radioactivity. The aim of this study was to quantify late effects in renal cortex of nude mice exposed to high amounts of [ 177 Lu]-DOTA-Tyr 3 -octreotate ([ 177 Lu]-DOTATATE), and to determine whether a threshold dose value exists for these findings. Methods Nude mice were exposed to 90, 120 or 150 MBq of [ 177 Lu]-DOTATATE. Renal toxicity was evaluated up to 6 months after injection. Blood samples were collected to examine renal functional markers, and after sacrifice at 6 months changes in renal morphology were explored. Tissue damage was estimated by quantifying the relative area of the different subunits in the renal cortex using point counting. Additional morphological signs of radiation damage were also noted. The absorbed doses to the kidneys were estimated by previously determined kidney pharmacokinetics and Monte Carlo simulations for different assumptions regarding the activity distribution. Results Increased serum creatinine and urea values indicated long-term renal toxicity. The tissue area occupied by proximal tubules decreased with increasing doses of [ 177 Lu]-DOTATATE, whereas the other subunits in cortex slightly increased. The mean absorbed dose in the renal cortex for [ 177 Lu]-DOTATATE was estimated to be 35–58 Gy for the different groups of animals. A dose–response relationship was observed for proximal tubular damage, and a threshold dose value of 24 Gy (BED 37 Gy) was determined. Conclusions Selective morphological changes in kidney cortex of nude mice were quantified and appeared in a dose dependent manner after injection of high amounts of [ 177 Lu]-DOTATATE.
- Published
- 2012
- Full Text
- View/download PDF
46. (68)Ga-labeled DOTA-Peptides and (68)Ga-labeled Radiopharmaceuticals for Positron Emission Tomography: Current Status of Research, Clinical Applications, and Future Perspectives
- Author
-
Mark Konijnenberg, Wouter A.P. Breeman, Dik J. Kwekkeboom, Eric P. Krenning, Ho Sze Chan, Erik de Blois, and Radiology & Nuclear Medicine
- Subjects
medicine.medical_specialty ,Gallium Radioisotopes ,Single-photon emission computed tomography ,Octreotide ,chemistry.chemical_compound ,medicine ,Organometallic Compounds ,DOTA ,Humans ,Radiology, Nuclear Medicine and imaging ,Medical physics ,In patient ,medicine.diagnostic_test ,Molecular Structure ,business.industry ,Radionuclide Generators ,United States ,Neuroendocrine Tumors ,Radioactivity ,chemistry ,Positron emission tomography ,Positron-Emission Tomography ,Radiopharmaceuticals ,business ,Peptides ,Forecasting - Abstract
In this review we give an overview of current knowledge of (68)Ga-labeled pharmaceuticals, with focus on imaging receptor-mediated processes. A major advantage of a (68)Ge/(68)Ga generator is its continuous source of (68)Ga, independently from an on-site cyclotron. The increase in knowledge of purification and concentration of the eluate and the complex ligand chemistry has led to (68)Ga-labeled pharmaceuticals with major clinical impact. (68)Ga-labeled pharmaceuticals have the potential to cover all today's clinical options with (99m)Tc, with the concordant higher resolution of positron emission tomography (PET) in comparison with single photon emission computed tomography. (68)Ga-labeled analogs of octreotide, such as DOTATOC, DOTANOC, and DOTA-TATE, are in clinical application in nuclear medicine, and these analogs are now the most frequently applied of all (68)Ga-labeled pharmaceuticals. All the above-mentioned items in favor of successful application of (68)Ga-labeled radiopharmaceuticals for imaging in patients are strong arguments for the development of a (68)Ge/(68)Ga generator with Marketing Authorization and thus to provide pharmaceutical grade eluate. Moreover, now not one United States Food and Drug Administration approved or European Medicines Agency approved (68)Ga-radiopharmaceutical is available. As soon as these are achieved, a whole new radiopharmacy providing PET radiopharmaceuticals might develop. Semin Nucl Med 41:314-321 (C) 2011 Elsevier Inc. All rights reserved.
- Published
- 2011
47. PO-132 Dissecting the radiobiology of targeted radionuclide therapy reveals an intra-tumoral heterogeneic response in a preclinical in vivo model
- Author
-
Gabriela N. Doeswijk, M. C. Clahsen-van Groningen, Joost C. Haeck, D C van Gent, Julie Nonnekens, Mark Konijnenberg, M. Hendriks-de Jong, and Danny Feijtel
- Subjects
Cancer Research ,Radiobiology ,DNA damage ,business.industry ,medicine.disease_cause ,medicine.anatomical_structure ,Somatostatin ,Oncology ,In vivo ,Radionuclide therapy ,medicine ,Cancer research ,Somatostatin receptor 2 ,Bone marrow ,Carcinogenesis ,business - Abstract
Introduction Neuroendocrine tumours (NETs) are a relatively rare, but deadly group of cancers. Since the cells of origin are neuroendocrine, diverse tumours can arise throughout the body. Often, patients are presented with metastases, making resection as a treatment strategy alone insufficient. In the clinic, localization of NETs has been assessed by targeting the somatostatin receptor 2 (SSTR2) which is overexpressed on tumour cells, with radiolabeled somatostatin analogues. Radiolabeling of these somatostatin analogues (e.g. DOTA-(Tyr3)-octreotate (DOTA-TATE)) with the DNA damaging nuclide (Lutetium-177) is currently utilised to treat the disease: peptide receptor radionuclide therapy (PRRT). PRRT with the use of 177Lu-DOTA-TATE proves effective in improving progression-free survival and life quality. However, mortality rates are still high. To increase the efficacy of PRRT we set out to dissect the cellular response of tumour cells to radionuclide therapy, the effect on dose-limiting organs (bone marrow and kidney) and mechanisms of therapy induced DNA damage in vitro and in vivo. Material and methods The SSTR2-expressing small cell lung cancer cell line NCI-H69 was treated with 177Lu-DOTA-TATE and fixed for assessment of DNA damage by means of 53 BP1 and γH2AX stainings on multiple time points. Also, BALB/c-nude mice were engrafted subcutaneously with NCI-H69 cells and were treated with 177Lu-DOTA-TATE after tumorigenesis. Mice were euthanized at different time points until 14 days post-treatment, and tumours, bone marrow and kidneys were excised for downstream analyses. Results and discussions After PRRT we observed a time-dependent intensity in DNA damage signalling and subsequent repair both in vitro and in vivo. We observed the peak of DNA double-strand break repair after two days. Although NCI-H69 tumours have a homogeneous nature, we observed intra-tumoral heterogeneity in cell death and activation of the DNA damage response. Furthermore, acute, but transient damage was observed in the kidneys and bone marrow. Conclusion With this investigation, we studied in detail the radiobiology of PRRT in a preclinical setting. Our results will contribute to a better understanding of PRRT effects. This might help to improve future patient treatment by finding the optimal therapeutic window for radiosensitization of the tumour without increase of side effects.
- Published
- 2018
- Full Text
- View/download PDF
48. Effect of time and exercise on the clearance rate of 201Tl in normal and ischemic myocardium
- Author
-
F.A. Verburg, Mark Konijnenberg, J.F. Verzijlbergen, Barbra E. Backus, Hezemans Re, and Keijsersa Rg
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Ischemic myocardium ,Washout ,chemistry.chemical_element ,General Medicine ,Single-photon emission computed tomography ,Myocardial perfusion imaging ,chemistry ,Internal medicine ,Cardiology ,Medicine ,Thallium ,Radiology, Nuclear Medicine and imaging ,business ,Perfusion ,Clearance rate ,Rest (music) - Abstract
Purpose Simultaneous dual isotope (SDI) acquisition of (201)Tl rest/(99m)Tc-sestamibi stress-myocardial perfusion single photon emission computed tomography is a desirable new procedure in nuclear cardiology. In this protocol (201)Tl is injected at rest but imaging is performed not earlier than after exercise. Therefore, one must be convinced that throughout exercise (201)Tl remains distributed in an identical pattern as at rest. Before SDI can be applied clinically, (201)Tl rest MPS before and after exercise test needs to be compared for equality. The aim of this study was to assess the effect of time and exercise on the clearance of preinjected (201)Tl in normal and ischemic myocardium. Methods In 122 patients rest (201)Tl and delayed (n =20) or poststress (n= 102) (201)Tl imaging was performed. Quantitative analysis of mean counts-per-pixel was performed for each segment in a 17-segment model. Differences between rest and delayed or poststress (201)Tl MPS were calculated. Patients with a poststress (201)Tl image were divided into normal (N= 66) and ischemic (N= 36) groups. Visual analysis was performed by two independent observers scoring the 17 segments on a scale of 0-4. Results The overall difference between rest (201)Tl and poststress (201)Tl MPS was - 15.4%. Normal and ischemic patients showed 16.2 and 14.0% (P =0.17) washout, respectively. Visual assessment by two independent observers revealed no regional differences between rest (201)Tl and delayed or poststress (201)Tl MPS. Conclusion (201)Tl poststress MPS shows significant washout of thallium. This washout is not segmental, but global over the myocardium. No significant differences are found between normal and ischemic myocardium. The poststress (201)Tl MPS is a reliable reflection of rest perfusion. SDI acquisition of (201)Tl rest/(99m)Tc-sestamibi stress-MPS is clinically applicable.
- Published
- 2010
- Full Text
- View/download PDF
49. Intra-patient reproducibility of myocardial SPECT imaging with 201Tl
- Author
-
R. Leo Romijn, Barbra E. Backus, J. Fred Verzijlbergen, Frederik A. Verburg, Mark Konijnenberg, and Freek J. Beekman
- Subjects
Male ,Thorax ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Heart Ventricles ,Sensitivity and Specificity ,Imaging phantom ,Spect imaging ,Image Interpretation, Computer-Assisted ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Thallium ,skin and connective tissue diseases ,Tomography, Emission-Computed, Single-Photon ,Reproducibility ,Phantoms, Imaging ,business.industry ,Reproducibility of Results ,nutritional and metabolic diseases ,Washout ,Middle Aged ,Image Enhancement ,Confidence interval ,Clinical diagnosis ,Female ,Radiology ,Radiopharmaceuticals ,Cardiology and Cardiovascular Medicine ,Nuclear medicine ,business ,Perfusion - Abstract
To define the physical and clinical reproducibility of (201)Tl myocardial perfusion SPECT (MPS), this study assesses the variation between two repeated rest (201)Tl MPS with repositioning only, with a two-hour time interval and with phantom measurements as a reference.Three anthropomorphic thorax phantoms were filled with (201)Tl. For each phantom five repeated (201)Tl MPS were obtained. In addition, in 20 patients repeated (201)Tl rest-MPS and in 26 patients early and delayed (201)Tl rest-MPS were performed. Quantitative analysis was done using MunichHeart. Statistical methods were used to calculate variability. Visual analysis was performed by 2 independent observers.The average variation between repeated phantom MPS was 0.5% (95% confidence interval (CI): -0.4% to 1.4%). For patient scans this was -5.0% (95% CI: -2.5% to -7.5%) and between early and delayed (201)Tl MPS -15.5% (95% CI: -11.7% to -19.3%). Visual assessment revealed no clinical significant differences between rest (201)Tl and repeated or delayed (201)Tl MPS.Repositioning in phantom (201)Tl MPS does not cause significant variation. Repeated (201)Tl MPS in patients shows 5.0% decrease of (201)Tl in 30 minutes, which increases to 15% during a two-hour time interval without quantitative or visual regional differences. This decrease indicates a time-related washout of (201)Tl, but does not change clinical diagnosis.
- Published
- 2009
- Full Text
- View/download PDF
50. Bone marrow dosimetry in peptide receptor radionuclide therapy with [Lu-177-DOTA(0),Tyr(3)]octreotate
- Author
-
P. P. M. Kooij, Eric P. Krenning, Bert F. Bernard, Willem H. Bakker, Mark Konijnenberg, Marion de Jong, Dik J. Kwekkeboom, Flavio Forrer, Jaap J.M. Teunissen, Wouter W. de Herder, Kirsten van Lom, Radiology & Nuclear Medicine, Hematology, and Internal Medicine
- Subjects
Male ,Receptors, Peptide ,medicine.medical_treatment ,Octreotide ,[177Lu-DOTA0,Tyr3]octreotate ,Somatostatin receptor ,chemistry.chemical_compound ,Bone Marrow ,Dosimetry ,Organometallic Compounds ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Receptors, Somatostatin ,Radiometry ,Tomography, Emission-Computed, Single-Photon ,Octreotate ,Platelet Count ,business.industry ,Radiotherapy Dosage ,General Medicine ,Radiation therapy ,Neuroendocrine Tumors ,Haematopoiesis ,Radioactivity ,medicine.anatomical_structure ,chemistry ,Radiology Nuclear Medicine and imaging ,Radionuclide therapy ,Female ,Original Article ,Therapy ,Bone marrow ,Stem cell ,Somatostatin ,Nuclear medicine ,business - Abstract
Purpose Adequate dosimetry is mandatory for effective and safe peptide receptor radionuclide therapy (PRRT). Besides the kidneys, the bone marrow is a potentially dose-limiting organ. The radiation dose to the bone marrow is usually calculated according to the MIRD scheme, where the accumulated activity in the bone marrow is calculated from the accumulated radioactivity of the radiopharmaceutical in the blood. This may underestimate the absorbed dose since stem cells express somatostatin receptors. We verified the blood-based method by comparing the activity in the blood with the radioactivity in bone marrow aspirates. Also, we evaluated the absorbed cross-dose from the source organs (liver, spleen, kidneys and blood), tumours and the so-called “remainder of the body” to the bone marrow. Methods Bone marrow aspirates were drawn in 15 patients after treatment with [177Lu-DOTA0,Tyr3]octreotate. Radioactivity in the bone marrow was compared with radioactivity in the blood drawn simultaneously. The nucleated cell fraction was isolated from the bone marrow aspirate and radioactivity was measured. The absorbed dose to the bone marrow was calculated. The results were correlated to the change in platelet counts 6 weeks after treatment. Results A strong linear correlation and high agreement between the measured radioactivities in the bone marrow aspirates and in the blood was found (r=0.914, p
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.