Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Urol Oncol ; 41(4): 205.e17-205.e24, 2023 04.
Article in English | MEDLINE | ID: mdl-36588019

ABSTRACT

PURPOSE: Prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) is an emerging staging tool for patients with primary high-risk prostate cancer (PCa). Patients with primary metastatic disease are staged using PSMA-PET/CT imaging, while previously published randomized clinical trials relied on conventional imaging (i.e., bone scintigraphy (BS) results. The aim of this study was to compare the ability of bone metastatic lesion detection and changes in staging for 18F-PSMA-PET/CT versus BS in high-risk PCa patients. METHODS: 79 patients with high-risk PCa were prospectively staged using BS and subsequent 18F-PSMA-PET/CT before initial therapy. Patients who presented with a BS showing no metastases represented Group 1, and patients with a BS showing low-volume disease according to the CHAARTED criteria (<4 bone metastases, no metastases outside vertebral column or pelvis and no visceral metastases) represented Group 2. Metastatic risk group according to CHAARTED and treatment strategies based on both imaging modalities were assessed. RESULTS: A change of CHAARTED risk group was observed in 9/70 (12.8%) of patients in Group 1. In Group 2, a change of risk group was found in 66.7% of patients, due to either upstaging (4/9 patients (44.4%)) and downstaging (2/9 patients (22.2%)). Treatment changes due to use of a different imaging modality occurred in almost 20% of patients. CONCLUSION: In patients with negative for cancer results on BS, upstaging on 18F-PSMA-PET/CT occurred only infrequently. Moreover, 18F-PSMA-PET/CT resulted in both upstaging and downstaging in a substantial subset of patients with low-volume metastatic disease on BS. Treatment changes occurred in almost 20% of cases depending on imaging results.


Subject(s)
Bone Neoplasms , Prostatic Neoplasms , Male , Humans , Positron Emission Tomography Computed Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Prospective Studies , Gallium Radioisotopes , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/secondary
2.
Pharmaceuticals (Basel) ; 15(7)2022 Jun 27.
Article in English | MEDLINE | ID: mdl-35890095

ABSTRACT

INTRODUCTION: Epidermal growth factor receptor (EGFR) mutated NSCLC is best treated using an EGFR tyrosine kinase inhibitor (TKI). The presence and accessibility of EGFR overexpression and mutation in NSCLC can be determined using radiolabeled EGFR TKI PET/CT. However, recent research has shown a significant difference between image qualities (i.e., tumor-to-lung contrast) in three generation EGFR TKIs: 11C-erlotinib, 18F-afatinib and 11C-osimertinib. In this research we aim to develop a physiological pharmacokinetic (PBPK)-model to predict tumor-to-lung contrast and as a secondary outcome the uptake of healthy tissue of the three tracers. METHODS: Relevant physicochemical and drug specific properties (e.g., pKa, lipophilicity, target binding) for each TKI were collected and applied in established base PBPK models. Key hallmarks of NSCLC include: immune tumor deprivation, unaltered tumor perfusion and an acidic tumor environment. Model accuracy was demonstrated by calculating the prediction error (PE) between predicted tissue-to-blood ratios (TBR) and measured PET-image-derived TBR. Sensitivity analysis was performed by excluding each key component and comparing the PE with the final mechanistical PBPK model predictions. RESULTS: The developed PBPK models were able to predict tumor-to-lung contrast for all EGFR-TKIs within threefold of observed PET image ratios (PE tumor-to-lung ratio of -90%, +44% and -6.3% for erlotinib, afatinib and osimertinib, respectively). Furthermore, the models depicted agreeable whole-body distribution, showing high tissue distribution for osimertinib and afatinib and low tissue distribution at high blood concentrations for erlotinib (mean PE, of -10.5%, range -158%-+190%, for all tissues). CONCLUSION: The developed PBPK models adequately predicted the image quality of afatinib and osimertinib and erlotinib. Some deviations in predicted whole-body TBR lead to new hypotheses, such as increased affinity for mutated EGFR and active influx transport (erlotinib into excreting tissues) or active efflux (afatinib from brain), which is currently unaccounted for. In the future, PBPK models may be used to predict the image quality of new tracers.

3.
Urol Oncol ; 40(2): 58.e1-58.e7, 2022 02.
Article in English | MEDLINE | ID: mdl-34404590

ABSTRACT

PURPOSE: To assess the diagnostic performance of prostate specific membranous antigen (PSMA) positron emission tomography/computed tomography (PET/CT) imaging to localize primary prostate cancer (PCa) in men with persistent elevated prostate-specific antigen (PSA) levels and previous prostate biopsies that were negative for PCa. METHODS: In this study, 34 men with persistently elevated PSA-levels, previous negative for PCa biopsies and who subsequently underwent diagnostic PSMA-PET/CT imaging were retrospectively evaluated. Men were divided into 3 groups: 1. 12 men with a previous negative mpMRI scan (PI-RADS 1-2) 2. 17 men with a positive mpMRI scan (PI-RADS 3-5), but negative MRI-targeted biopsies and 3. Four men in whom mpMRI was contraindicated. If PSMA-avid lesions were seen, patients underwent 2-4 cognitive targeted biopsies in combination with systematic biopsies. The detection rate of PSMA-PET/CT for PCa, and the accuracy of (possible) targeted biopsies were calculated. RESULTS: Included men had a median PSA-level of 22.8 ng/mL (Interquartile Range 15.6-30.0) at the time of PSMA-PET/CT. Elevated PSMA-ligand uptake in the prostate suspicious for PCa was observed in 22/34 patients (64.7%). In 18/22 patients (54.5%), PSMA-targeted prostate biopsies were performed. In 3/18 patients (16.6%), the targeted biopsies showed International Society of Urological Pathology (ISUP) score 1-2 PCa. The other men had inflammation or benign findings after histopathological examination of the biopsy cores. CONCLUSION: In this study, the clinical value of PSMA-PET/CT for patients with an elevated PSA-level, and negative for PCa biopsies was low. Only very few men were diagnosed with PCa, and no clinically significant PCa was found.


Subject(s)
Biopsy/methods , Positron Emission Tomography Computed Tomography/methods , Prostate-Specific Antigen/analysis , Prostate/pathology , Humans , Male , Surveys and Questionnaires
4.
World J Urol ; 39(7): 2439-2446, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33079250

ABSTRACT

PURPOSE: In primary prostate cancer (PCa) patients, accurate staging and histologic grading are crucial to guide treatment decisions. 18F-DCFPyL (PSMA)-PET/CT has been successfully introduced for (re)staging PCa, showing high accuracy to localise PCa in lymph nodes and/or osseous structures. The diagnostic performance of 18F-DCFPyL-PET/CT in localizing primary PCa within the prostate gland was assessed, allowing for PSMA-guided targeted-prostate biopsy. METHODS: Thirty patients with intermediate-/high-risk primary PCa were prospectively enrolled between May 2018 and May 2019 and underwent 18F-DCFPyL-PET/CT prior to robot-assisted radical prostatectomy (RARP). Two experienced and blinded nuclear medicine physicians assessed tumour localisation within the prostate gland on PET/CT, using a 12-segment mapping model of the prostate. The same model was used by a uro-pathologist for the RARP specimens. Based on PET/CT imaging, a potential biopsy recommendation was given per patient, based on the size and PET-intensity of the suspected PCa localisations. The biopsy recommendation was correlated to final histopathology in the RARP specimen. Sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV) for clinically significant PCa (csPCa, Gleason score ≥ 3 + 4 = 7) were assessed. RESULTS: The segments recommended for potential targeted biopsy harboured csPCA in 28/30 patients (93%), and covered the highest Gleason score PCa segment in 26/30 patient (87%). Overall, 122 of 420 segments (29.0%) contained csPCa at final histopathological examination. Sensitivity, specificity, PPV and NPV for csPCa per segment using 18F-DCFPyL-PET/CT were 61.4%, 88.3%, 68.1% and 84.8%, respectively. CONCLUSIONS: When comparing the PCa-localisation on 18F-DCFPyL-PET/CT with the RARP specimens, an accurate per-patient detection (93%) and localisation of csPCa was found. Thus, 18F-DCFPyL-PET/CT potentially allows for accurate PSMA-targeted biopsy.


Subject(s)
Antigens, Surface , Glutamate Carboxypeptidase II , Lysine/analogs & derivatives , Positron Emission Tomography Computed Tomography , Prostatectomy , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Urea/analogs & derivatives , Aged , Biopsy , Feasibility Studies , Humans , Male , Middle Aged , Positron Emission Tomography Computed Tomography/methods , Prospective Studies , Prostatic Neoplasms/surgery
5.
EJNMMI Res ; 10(1): 97, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32804306

ABSTRACT

INTRODUCTION: Only a subgroup of non-small cell lung cancer (NSCLC) patients benefit from treatment using epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) such as afatinib. Tumour uptake of [18F]afatinib using positron emission tomography (PET) may identify those patients that respond to afatinib therapy. Therefore, the aim of this study was to find the optimal tracer kinetic model for quantification of [18F]afatinib uptake in NSCLC tumours. METHODS: [18F]Afatinib PET scans were performed in 10 NSCLC patients. The first patient was scanned for the purpose of dosimetry. Subsequent patients underwent a 20-min dynamic [15O]H2O PET scan (370 MBq) followed by a dynamic [18F]afatinib PET scan (342 ± 24 MBq) of 60 or 90 min. Using the Akaike information criterion (AIC), three pharmacokinetic plasma input models were evaluated with both metabolite-corrected sampler-based input and image-derived (IDIF) input functions in combination with discrete blood samples. Correlation analysis of arterial on-line sampling versus IDIF was performed. In addition, perfusion dependency and simplified measures were assessed. RESULTS: Ten patients were included. The injected activity of [18F]afatinib was 341 ± 37 MBq. Fifteen tumours could be identified in the field of view of the scanner. Based on AIC, tumour kinetics were best described using an irreversible two-tissue compartment model and a metabolite-corrected sampler-based input function (Akaike 50%). Correlation of plasma-based input functions with metabolite-corrected IDIF was very strong (r2 = 0.93). The preferred simplified uptake parameter was the tumour-to-blood ratio over the 60- to 90-min time interval (TBR60-90). Tumour uptake of [18F]afatinib was independent of perfusion. CONCLUSION: The preferred pharmacokinetic model for quantifying [18F]afatinib uptake in NSCLC tumours was the 2T3K_vb model. TBR60-90 showed excellent correlation with this model and is the best candidate simplified method. TRIAL REGISTRATION: https://eudract.ema.europa.eu/ nr 2012-002849-38.

7.
Nat Commun ; 9(1): 4664, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30405135

ABSTRACT

PD-L1 immunohistochemistry correlates only moderately with patient survival and response to PD-(L)1 treatment. Heterogeneity of tumor PD-L1 expression might limit the predictive value of small biopsies. Here we show that tumor PD-L1 and PD-1 expression can be quantified non-invasively using PET-CT in patients with non-small-cell lung cancer. Whole body PD-(L)1 PET-CT reveals significant tumor tracer uptake heterogeneity both between patients, as well as within patients between different tumor lesions.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Positron-Emission Tomography , Programmed Cell Death 1 Receptor/metabolism , Whole Body Imaging , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Lung Neoplasms/drug therapy , Nivolumab/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Treatment Outcome
8.
Lung Cancer ; 107: 1-13, 2017 05.
Article in English | MEDLINE | ID: mdl-27319335

ABSTRACT

Non-small cell lung cancer (NSCLC) therapy has entered a rapidly advancing era of precision medicine with an ever increasing number of drugs directed against a variety of specific tumor targets. Amongst these new agents, tyrosine kinase inhibitors (TKIs) and monoclonal antibodies (mAbs) are most frequently used. However, as only a sensitive subgroup of patients benefits from targeting drugs, predictive biomarkers are needed. Positron emission tomography (PET) may offer such a biomarker for predicting therapy efficacy. Some of the TKIs and mAbs that are in clinical use can be radioactively labeled and used as tracers. PET can visualize and quantify tumor specific uptake of radiolabeled targeting drugs, allowing for characterization of their pharmacokinetic behavior. In this review, the clinical potential of PET using radiolabeled TKIs (TKI-PET) and mAbs (immuno-PET) in NSCLC is discussed, and an overview is provided of the most relevant preclinical and clinical studies.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/pharmacokinetics , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Drug Evaluation, Preclinical , ErbB Receptors/metabolism , Heterografts , Humans , Immunoconjugates/therapeutic use , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Mice , Molecular Targeted Therapy/methods , Precision Medicine , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases
9.
Clin Pharmacol Ther ; 98(3): 234-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26095095

ABSTRACT

Important information gaps remain on the efficacy and safety of drugs in children. Pediatric drug development encounters several ethical, practical, and scientific challenges. One barrier to the evaluation of medicines for children is a lack of innovative methodologies that have been adapted to the needs of children. This article presents our successful experience of pediatric microdose and microtracer studies using (14) C-labeled probes in Europe to illustrate the strengths and limitations of these approaches.


Subject(s)
Carbon Radioisotopes/administration & dosage , Clinical Trials, Phase I as Topic , Drug Approval , Pharmaceutical Preparations/administration & dosage , Age Factors , Carbon Radioisotopes/adverse effects , Carbon Radioisotopes/economics , Carbon Radioisotopes/pharmacokinetics , Child , Child, Preschool , Clinical Trials, Phase I as Topic/economics , Clinical Trials, Phase I as Topic/ethics , Clinical Trials, Phase I as Topic/legislation & jurisprudence , Dose-Response Relationship, Drug , Drug Approval/economics , Drug Approval/legislation & jurisprudence , Drug Costs , Drug Dosage Calculations , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/prevention & control , Europe , Government Regulation , Humans , Infant , Infant, Newborn , Patient Safety , Pharmaceutical Preparations/economics , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Risk Assessment , Risk Factors
10.
Clin Pharmacol Ther ; 98(2): 196-204, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25869840

ABSTRACT

Preclinical development of new biological entities (NBEs), such as human protein therapeutics, requires considerable expenditure of time and costs. Poor prediction of pharmacokinetics in humans further reduces net efficiency. In this study, we show for the first time that pharmacokinetic data of NBEs in humans can be successfully obtained early in the drug development process by the use of microdosing in a small group of healthy subjects combined with ultrasensitive accelerator mass spectrometry (AMS). After only minimal preclinical testing, we performed a first-in-human phase 0/phase 1 trial with a human recombinant therapeutic protein (RESCuing Alkaline Phosphatase, human recombinant placental alkaline phosphatase [hRESCAP]) to assess its safety and kinetics. Pharmacokinetic analysis showed dose linearity from microdose (53 µg) [(14) C]-hRESCAP to therapeutic doses (up to 5.3 mg) of the protein in healthy volunteers. This study demonstrates the value of a microdosing approach in a very small cohort for accelerating the clinical development of NBEs.


Subject(s)
Alkaline Phosphatase/administration & dosage , Alkaline Phosphatase/pharmacokinetics , Carbon Radioisotopes , Isoenzymes/administration & dosage , Isoenzymes/pharmacokinetics , Administration, Intravenous , Adolescent , Adult , Alkaline Phosphatase/adverse effects , Area Under Curve , Double-Blind Method , Drug Dosage Calculations , GPI-Linked Proteins/administration & dosage , GPI-Linked Proteins/adverse effects , GPI-Linked Proteins/pharmacokinetics , Half-Life , Healthy Volunteers , Humans , Isoenzymes/adverse effects , Linear Models , Male , Mass Spectrometry/methods , Metabolic Clearance Rate , Models, Biological , Netherlands , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Young Adult
11.
Nucl Med Biol ; 42(5): 465-469, 2015 May.
Article in English | MEDLINE | ID: mdl-25662844

ABSTRACT

INTRODUCTION: (188)Rhenium-HEDP is an effective bone-targeting therapeutic radiopharmaceutical, for treatment of osteoblastic bone metastases. It is known that the presence of carrier (non-radioactive rhenium as ammonium perrhenate) in the reaction mixture during labeling is a prerequisite for adequate bone affinity, but little is known about the optimal carrier concentration. METHODS: We investigated the influence of carrier concentration in the formulation on the radiochemical purity, in-vitro hydroxyapatite affinity and the in-vivo bone accumulation of (188)Rhenium-HEDP in mice. RESULTS: The carrier concentration influenced hydroxyapatite binding in-vitro as well as bone accumulation in-vivo. Variation in hydroxyapatite binding with various carrier concentrations seemed to be mainly driven by variation in radiochemical purity. The in-vivo bone accumulation appeared to be more complex: satisfactory radiochemical purity and hydroxyapatite affinity did not necessarily predict acceptable bio-distribution of (188)Rhenium-HEDP. CONCLUSIONS: For development of new bisphosphonate-based radiopharmaceuticals for clinical use, human administration should not be performed without previous animal bio-distribution experiments. Furthermore, our clinical formulation of (188)Rhenium-HEDP, containing 10 µmol carrier, showed excellent bone accumulation that was comparable to other bisphosphonate-based radiopharmaceuticals, with no apparent uptake in other organs. ADVANCES IN KNOWLEDGE: Radiochemical purity and in-vitro hydroxyapatite binding are not necessarily predictive of bone accumulation of (188)Rhenium-HEDP in-vivo. IMPLICATIONS FOR PATIENT CARE: The formulation for (188)Rhenium-HEDP as developed by us for clinical use exhibits excellent bone uptake and variation in carrier concentration during preparation of this radiopharmaceutical should be avoided.


Subject(s)
Durapatite/chemistry , Etidronic Acid/chemistry , Radiochemistry/methods , Radioisotopes/chemistry , Radiopharmaceuticals/chemistry , Rhenium/chemistry , Animals , Bone and Bones/metabolism , Durapatite/pharmacokinetics , Durapatite/therapeutic use , Etidronic Acid/pharmacokinetics , Etidronic Acid/therapeutic use , Male , Mice , Mice, Inbred C57BL , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Tissue Distribution
14.
Br J Cancer ; 97(3): 322-6, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17609666

ABSTRACT

The blood-brain barrier (BBB) hampers delivery of several drugs including chemotherapeutics to the brain. The drug efflux pump P-glycoprotein (P-gp), expressed on brain capillary endothelial cells, is part of the BBB. P-gp expression on capillary endothelium decreases 5 days after brain irradiation, which may reduce P-gp function and increase brain levels of P-gp substrates. To elucidate whether radiation therapy reduces P-gp expression and function in the brain, right hemispheres of rats were irradiated with single doses of 2-25 Gy followed by 10 mg kg(-1) of the P-gp substrate cyclosporine A (CsA) intravenously (i.v.), with once 15 Gy followed by CsA (10, 15 or 20 mg kg(-1)), or with fractionated irradiation (4 x 5 Gy) followed by CsA (10 mg kg(-1)) 5 days later. Additionally, four groups of three rats received 25 Gy once and were killed 10, 15, 20 or 25 days later. The brains were removed and P-gp detected immunohistochemically. P-gp function was assessed by [(11)C]carvedilol uptake using quantitative autoradiography. Irradiation increased [(11)C]carvedilol uptake dose-dependently, to a maximum of 20% above non irradiated hemisphere. CsA increased [(11)C]carvedilol uptake dose-dependently in both hemispheres, but more (P<0.001) in the irradiated hemisphere. Fractionated irradiation resulted in a lost P-gp expression 10 days after start irradiation, which coincided with increased [(11)C]carvedilol uptake. P-gp expression decreased between day 15 and 20 after single dose irradiation, and increased again thereafter. Rat brain irradiation results in a temporary decreased P-gp function.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/radiation effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Animals , Autoradiography , Brain/metabolism , Carbazoles/metabolism , Carvedilol , Immunohistochemistry , Male , Propanolamines/metabolism , Radioligand Assay , Rats , Rats, Wistar
15.
Toxicol Appl Pharmacol ; 207(2 Suppl): 257-60, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16043202

ABSTRACT

P-glycoprotein (P-gp) is a transmembrane drug efflux pump encoded by the MDR-1 gene in humans. Most likely P-gp protects organs against endogenous and exogenous toxins by extruding toxic compounds such as chemotherapeutics and other drugs. Many drugs are substrates for P-gp. Since P-gp is also expressed in the blood-brain barrier, P-gp substrates reach lower concentrations in the brain than in P-gp-negative tissues. Failure of response to chemotherapy of malignancies can be due to intrinsic or acquired drug resistance. Many tumors are multidrug resistant (MDR); resistant to several structurally unrelated chemotherapeutic agents. Several mechanisms are involved in MDR of which P-gp is studied most extensively. P-gp extrudes drugs out of tumor cells resulting in decreased intracellular drug concentrations, leading to the MDR phenotype. Furthermore, the MDR-1 gene exhibits several single nucleotide polymorphisms, some of which result in different transport capabilities. P-gp functionality and the effect of P-gp modulation on the pharmacokinetics of novel and established drugs can be studied in vivo by positron emission tomography (PET) using carbon-11 and fluorine-18-labeled P-gp substrates and modulators. PET may demonstrate the consequences of genetic differences on tissue pharmacokinetics. Inhibitors such as calcium-channel blockers (verapamil), cyclosporin A, ONT-093, and XR9576 can modulate the P-gp functionality. With PET the effect of P-gp modulation on the bioavailability of drugs can be investigated in humans in vivo. PET also allows the measurement of the efficacy of newly developed P-gp modulators.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Polymorphism, Genetic , Positron-Emission Tomography , Radioligand Assay
16.
Eur J Cancer ; 40(14): 2064-70, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15341980

ABSTRACT

The drug-efflux pumps P-glycoprotein (P-gp) and multidrug resistance-associated protein 1 (MRP1) are present in the blood-testis barrier (BTB) and may hamper the delivery of cytotoxic drugs to the testis. The precise localisation of P-gp and MRP1 in testicular tissue and the presence of the efflux pumps MRP2 and breast cancer resistance protein (BCRP) in the BTB are unknown. We therefore studied the localisation of these pumps in the BTB in normal testis (n = 12), in non-seminoma (n = 10) seminoma (n = 10), and testicular lymphoma (n = 9). Slides were scored semi-quantitatively for P-gp, MRP1, MRP2 and BCRP and blood vessels with factor VIII antibody. In normal testis, P-gp and BCRP were strongly expressed by myoid cells and luminal capillary endothelial wall and P-gp also by Leydig cells. MRP1 was observed at the basal side of Sertoli cells and on Leydig cells. MRP2 was only weakly expressed by myoid cells. Seminomas and non-seminomas expressed P-gp and/or BCRP and/or MRP1, lymphomas strongly expressed P-gp, weakly expressed BCRP and did not or showed weak expression of MRP1. There was very little staining for MRP2 in the tumours. Newly formed vessels in all tumours only expressed P-gp and BCRP. P-gp, BCRP and MRP1 are present in different cell layers of the normal testis, suggesting the optimal protection of spermatogenesis. In germ cell tumours, this expression pattern may explain the chemoresistance observed to P-gp, BCRP and MRP1 substrates. In germ cell tumours and testicular lymphomas, P-gp and BCRP expression by tumour cells and by newly formed vessels may also contribute to chemoresistance. These findings underscore the importance of removing the affected testis in cases of primary germ cell tumours and testicular lymphomas, irrespective of whether the patient has already undergone chemotherapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , ATP-Binding Cassette Transporters/analysis , Blood-Testis Barrier/chemistry , Multidrug Resistance-Associated Proteins/analysis , Seminoma/chemistry , Testicular Neoplasms/chemistry , Humans , Immunohistochemistry , Male
17.
Methods ; 27(3): 228-33, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12183111

ABSTRACT

Multidrug resistance (MDR) is characterized by the occurrence of cross-resistance to a broad range of structurally and functionally unrelated drugs. Several mechanisms are involved in MDR. One of the most well-known mechanisms is the overexpression of P-glycoprotein (P-gp), encoded by the MDR1 gene in humans and by the mdr1a and mdr1b genes in rodents. P-gp is extensively expressed in the human body, e.g., in the blood-brain barrier and also in solid tumor tissue. Overexpression of P-gp on tumor membranes might result in MDR of human tumors. To circumvent this resistant phenotype, several P-gp modulators such as cyclosporin A (CsA) are available. Competition between P-gp drugs and modulators results in decreased transport of the drug out of tumor tissue and an increased cellular level of these drugs. For effective clinical treatment it is important to have knowledge about P-gp functionality in tumors. Therefore, we have developed a method to measure the P-gp functionality in vivo with PET and [(11)C]verapamil as a positron-emitting P-gp substrate. The results obtained in rodents and in cancer patients are described in this article.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Drug Resistance, Multiple/physiology , Tomography, Emission-Computed/methods , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Animals , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Rats , Rats, Nude , Tissue Distribution , Tumor Cells, Cultured , Verapamil/pharmacokinetics
18.
Novartis Found Symp ; 243: 137-45; discussion 145-8, 180-5, 2002.
Article in English | MEDLINE | ID: mdl-11990773

ABSTRACT

P glycoprotein (Pgp) is expressed on cell membranes of various organs in the body, such as the capillary endothelial cells of the brain. Furthermore, Pgp can also be expressed on the cell membrane of tumour cells. Because of Pgp-mediated efflux, tissue levels of several Pgp substrates are lower than in Pgp-negative tissues. Drug levels in Pgp-expressing organs may be increased by modulation of this Pgp-facilitated transport with several compounds, such as cyclosporin A. Up to now, the presence of drug efflux pumps in tissues could only be examined at the mRNA and protein level. However, this gives no insight into the important question of the functionality of these drug efflux pumps. Information about the transport function of Pgp and the effect of modulating this function may improve the therapeutic treatment of these patients. Positron emission tomography (PET) gives us a unique opportunity to study non-invasively (patho)physiological dynamic processes in vivo. We have therefore developed and validated a method for studying Pgp-mediated transport and its modulation in vivo with PET.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Tomography, Emission-Computed , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Animals , Biological Transport, Active , Blood-Brain Barrier , Colchicine/pharmacokinetics , Daunorubicin/pharmacokinetics , Drug Resistance, Multiple , Electrons , Humans , Mice , Neoplasm Proteins/analysis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Verapamil/pharmacology
20.
Eur J Clin Pharmacol ; 56(11): 827-9, 2001.
Article in English | MEDLINE | ID: mdl-11294373

ABSTRACT

OBJECTIVE: To evaluate [11C]verapamil kinetics in humans. METHODS: After intravenous injection of [11C]verapamil (370 MBq, specific activity >1,600 GBq/mmol), kinetics were evaluated in five cancer patients using positron emission tomography (PET). RESULTS: One hour after injection, accumulation of [11C] in lungs, heart and tumour was 43.0%, 1.3% and 0.9% of the injected verapamil dose, respectively. Half-lives of [11C]verapamil in these tissues were 46.2 min, 73.8 min and 23.7 min, respectively. CONCLUSIONS: Intravenously administered [11C] was mainly extracted by the lungs. Transport of a [11C]verapamil bolus out of solid tumour tissue is relatively fast.


Subject(s)
Calcium Channel Blockers/pharmacokinetics , Neoplasms/metabolism , Verapamil/pharmacokinetics , Adult , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/blood , Female , Half-Life , Humans , Injections, Intravenous , Male , Middle Aged , Tissue Distribution , Tomography, Emission-Computed , Verapamil/administration & dosage , Verapamil/blood
SELECTION OF CITATIONS
SEARCH DETAIL