287 results on '"Bahce, Idris"'
Search Results
102. Additional file 1: of Effects of erlotinib therapy on [11C]erlotinib uptake in EGFR mutated, advanced NSCLC
- Author
-
Bahce, Idris, Yaqub, Maqsood, Errami, Hanane, Schuit, Robert, Schober, Patrick, Thunnissen, Erik, Windhorst, Albert, Lammertsma, Adriaan, Smit, Egbert, and N. Hendrikse
- Abstract
Supplementary data. Table S1. Parent fractions (%). Table S2. Whole blood SUV. Table S3. Tumor [11C]erlotinib V T and K1 values. Table S4. Tumor [15O]H2O flow values. Table S5. SUV and TBR values (unitless). (DOC 170 kb)
- Published
- 2016
- Full Text
- View/download PDF
103. A multi species evaluation of the radiation dosimetry of [ 11 C]erlotinib, the radiolabeled analog of a clinically utilized tyrosine kinase inhibitor
- Author
-
Petrulli, J. Ryan, primary, Hansen, Søren B., additional, Abourbeh, Galith, additional, Yaqub, Maqsood, additional, Bahce, Idris, additional, Holden, Daniel, additional, Huang, Yiyun, additional, Nabulsi, Nabeel B., additional, Contessa, Joseph N., additional, Mishani, Eyal, additional, Lammertsma, Adriaan A., additional, and Morris, Evan D., additional
- Published
- 2017
- Full Text
- View/download PDF
104. Quantitative and Simplified Analysis of 11C-Erlotinib Studies
- Author
-
Yaqub, Maqsood, primary, Bahce, Idris, additional, Voorhoeve, Charlotte, additional, Schuit, Robert C., additional, Windhorst, Albert D., additional, Hoekstra, Otto S., additional, Boellaard, Ronald, additional, Hendrikse, N. Harry, additional, Smit, Egbert F., additional, and Lammertsma, Adriaan A., additional
- Published
- 2016
- Full Text
- View/download PDF
105. Treatment and survival of patients with EGFR-mutated non-small cell lung cancer and leptomeningeal metastasis: A retrospective cohort analysis
- Author
-
Kuiper, Justine L., Kuiper, Justine L., Hendriks, Lizza E., van der Wekken, Anthonie J., de Langen, Adrianus J., Bahce, Idris, Thunnissen, Erik, Heideman, Danielle A. M., Berk, Yvonne, Buijs, Ed J. M., Speel, Ernst-Jan M., Krouwels, Frans H., Smit, Hans J. M., Groen, Harry J. M., Dingemans, Anne-Marie C., Smit, Egbert F., Kuiper, Justine L., Kuiper, Justine L., Hendriks, Lizza E., van der Wekken, Anthonie J., de Langen, Adrianus J., Bahce, Idris, Thunnissen, Erik, Heideman, Danielle A. M., Berk, Yvonne, Buijs, Ed J. M., Speel, Ernst-Jan M., Krouwels, Frans H., Smit, Hans J. M., Groen, Harry J. M., Dingemans, Anne-Marie C., and Smit, Egbert F.
- Abstract
Objectives: Development of leptomeningeal metastasis (LM) in non-small cell lung cancer (NSCLC)patients is associated with a poor prognosis. It has been suggested that LM-patients with epidermal growth factor receptor mutated (EGER+) NSCLC have a superior prognosis compared to EGFR-wild type NSCLC. Studies in EGFR+ NSCLC-patients with LM are scarce. We retrospectively evaluated a multiinstitutional cohort of EGER+ NSCLC-patients for LM to assess clinical outcome in relation to patient characteristics and treatment modalities. Material and methods: Medical records of advanced-stage EGFR+ NSCLC-patients (diagnosed between August 2000 and June 2014) from 11 Dutch hospitals were evaluated for LM as diagnosed by MRI and/or cytopathological liquor analysis. Data on patient characteristics, treatment and outcome were collected. Results: Thirty-two of 356 (9.0%) advanced-stage EGFR+ NSCLC-patients (median follow-up 21.0 months), were diagnosed with LM between 2006 and 2014. LM was diagnosed by MM (59.4%), liquor analysis (9.4%) or by both MRI and liquor analysis (31.3%). Median survival after LM-diagnosis was 3.1 months (95% Cl: 0.0-7.3). Six- and 12-month survival rates were 43.8% and 18.8%, respectively. Patients with performance status (PS) 0-1 at time of diagnosis of LM had a significantly higher chance to be alive after 6 months and had a significantly longer survival after diagnosis of LM compared to patients with PS >= 2. Age, treatment with high-dose EGFR-TKI, radiotherapy and whether LM was the only site of progressive disease did not influence survival after LM-diagnosis. Conclusion: Although median survival after LM-diagnosis in EGFR-mutated NSCLC-patients was poor, a substantial part of the patients had a prolonged survival of more than 6 months. PS of 0-1 at time of diagnosis of LM was associated with prolonged survival. No other patient- or treatment-related characteristics were identified. Further research is warranted to identify treatment strategies that
- Published
- 2015
106. Multiparametric Analysis of the Relationship Between Tumor Hypoxia and Perfusion with 18F-Fluoroazomycin Arabinoside and 15O-H2O PET
- Author
-
Iqbal, Ramsha, primary, Kramer, Gem M., additional, Verwer, Eline E., additional, Huisman, Marc C., additional, de Langen, Adrianus J., additional, Bahce, Idris, additional, van Velden, Floris H.P., additional, Windhorst, Albert D., additional, Lammertsma, Adriaan A., additional, Hoekstra, Otto S., additional, and Boellaard, Ronald, additional
- Published
- 2015
- Full Text
- View/download PDF
107. Bringing third and second harmonic generation microscopy into the clinic for the assessment of fresh lung tissue.
- Author
-
van Huizen, Laura M. G., Seinstra, Daniëlle, Dickhoff, Chris, Radonic, Teodora, Bahce, Idris, van Mourik, Frank, Annema, Jouke T., Daniels, Johannes M. A., van Boven, Wim-Jan P., and Groot, Marie Louise
- Published
- 2019
- Full Text
- View/download PDF
108. Bringing third and second harmonic generation microscopy into the clinic for the assessment of fresh lung tissue
- Author
-
Lilge, Lothar D., Philipp, Carsten M., van Huizen, Laura M. G., Seinstra, Daniëlle, Dickhoff, Chris, Radonic, Teodora, Bahce, Idris, van Mourik, Frank, Annema, Jouke T., Daniels, Johannes M. A., van Boven, Wim-Jan P., and Groot, Marie Louise
- Published
- 2019
- Full Text
- View/download PDF
109. Artificial intelligence algorithm developed to predict immune checkpoint inhibitors efficacy in non–small-cell lung cancer.
- Author
-
Rakaee, Mehrdad, Tafavvoghi, Masoud, Adib, Elio, Ricciuti, Biagio, Alessi, Joao Victor Machado, Cortellini, Alessio, Fulgenzi, Claudia A.M., Møllersen, Kajsa, Bongo, Lars Ailo, Hashemi, Sayed MS, Houda, Ilias, Busund, Lill-Tove Rasmussen, Donnem, Tom, Bahce, Idris, Pinato, David J. James, Sholl, Lynette M., Awad, Mark M., and Kwiatkowski, David J.
- Published
- 2023
- Full Text
- View/download PDF
110. Treatment and survival of patients with EGFR -mutated non-small cell lung cancer and leptomeningeal metastasis: A retrospective cohort analysis
- Author
-
Kuiper, Justine L., primary, Hendriks, Lizza E., additional, van der Wekken, Anthonie J., additional, de Langen, Adrianus J., additional, Bahce, Idris, additional, Thunnissen, Erik, additional, Heideman, Daniëlle A.M., additional, Berk, Yvonne, additional, Buijs, Ed J.M., additional, Speel, Ernst-Jan M., additional, Krouwels, Frans H., additional, Smit, Hans J.M., additional, Groen, Harry J.M., additional, Dingemans, Anne-Marie C., additional, and Smit, Egbert F., additional
- Published
- 2015
- Full Text
- View/download PDF
111. Detecting resistance in EGFR-mutated non-small-cell lung cancer after clonal selection through targeted therapy
- Author
-
Kuiper, Justine L, primary, Bahce, Idris, additional, Voorhoeve, Charlotte, additional, Yaqub, Maqsood, additional, Heideman, Daniëlle AM, additional, Thunnissen, Erik, additional, Paul, Marinus A, additional, Postmus, Pieter E, additional, Hendrikse, N Harry, additional, and Smit, Egbert F, additional
- Published
- 2015
- Full Text
- View/download PDF
112. Parametric Methods for Quantification of 18F-FAZA Kinetics in Non–Small Cell Lung Cancer Patients
- Author
-
Verwer, Eline E., primary, Bahce, Idris, additional, van Velden, Floris H.P., additional, Yaqub, Maqsood, additional, Schuit, Robert C., additional, Windhorst, Albert D., additional, Raijmakers, Pieter, additional, Hoekstra, Otto S., additional, Lammertsma, Adriaan A., additional, Smit, Egbert F., additional, and Boellaard, Ronald, additional
- Published
- 2014
- Full Text
- View/download PDF
113. A randomized phase II study of paclitaxel-carboplatin-bevacizumab (PCB) with or without nitroglycerin patches (NTG) in patients (pts) with stage IV nonsquamous non-small cell lung cancer (NSCLC): Nvalt 12 (NCT01171170).
- Author
-
Dingemans, Anne-Marie C., primary, Groen, Harry J.M., additional, Herder, Judith, additional, Stigt, Jos, additional, Smit, Egbert F., additional, Bahce, Idris, additional, Dalesio, Otilia, additional, Burgers, Sjaak A., additional, Codrington, Henk, additional, VAN Den Borne, BEN, additional, Biesma, Bonne, additional, Vincent, Andrew D., additional, van de Noort, Vincent, additional, and Aerts, Joachim, additional
- Published
- 2014
- Full Text
- View/download PDF
114. Development of [11C]erlotinib Positron Emission Tomography for In Vivo Evaluation of EGF Receptor Mutational Status
- Author
-
Bahce, Idris, Smit, Egbert F, Lubberink, Mark, van der Veldt, Astrid A M, Yaqub, Maqsood, Windhorst, Albert D, Schuit, Robert C, Thunnissen, Erik, Heideman, Daniëlle A M, Postmus, Pieter E, Lammertsma, Adriaan A, Hendrikse, N Harry, Bahce, Idris, Smit, Egbert F, Lubberink, Mark, van der Veldt, Astrid A M, Yaqub, Maqsood, Windhorst, Albert D, Schuit, Robert C, Thunnissen, Erik, Heideman, Daniëlle A M, Postmus, Pieter E, Lammertsma, Adriaan A, and Hendrikse, N Harry
- Abstract
PURPOSE: To evaluate whether, in patients with non-small cell lung carcinoma (NSCLC), tumor uptake of [(11)C]erlotinib can be quantified and imaged using positron emission tomography and to assess whether the level of tracer uptake corresponds with the presence of activating tumor EGF receptor (EGFR) mutations.EXPERIMENTAL DESIGN: Ten patients with NSCLCs, five with an EGFR exon 19 deletion, and five without were scanned twice (test retest) on the same day with an interval of at least 4 hours. Each scanning procedure included a low-dose computed tomographic scan, a 10-minute dynamic [(15)O]H(2)O scan, and a 1-hour dynamic [(11)C]erlotinib scan. Data were analyzed using full tracer kinetic modeling. EGFR expression was evaluated using immunohistochemistry.RESULTS: The quantitative measure of [(11)C]erlotinib uptake, that is, volume of distribution (V(T)), was significantly higher in tumors with activating mutations, that is, all with exon 19 deletions (median V(T), 1.76; range, 1.25-2.93), than in those without activating mutations (median V(T), 1.06; range, 0.67-1.22) for both test and retest data (P = 0.014 and P = 0.009, respectively). Good reproducibility of [(11)C]erlotinib V(T) was seen (intraclass correlation coefficient = 0.88). Intergroup differences in [(11)C]erlotinib uptake were not correlated with EGFR expression levels, nor tumor blood flow.CONCLUSION: [(11)C]erlotinib V(T) was significantly higher in NSCLCs tumors with EGFR exon 19 deletions.
- Published
- 2013
- Full Text
- View/download PDF
115. Rapid Decrease in Delivery of Chemotherapy to Tumors after Anti-VEGF Therapy : Implications for Scheduling of Anti-Angiogenic Drugs
- Author
-
van der Veldt, Astrid A. M., Lubberink, Mark, Bahce, Idris, Walraven, Maudy, de Boer, Michiel P., Greuter, Henri N. J. M., Hendrikse, N. Harry, Eriksson, Jonas, Windhorst, Albert D., Postmus, Pieter E., Verheul, Henk M., Serne, Erik H., Lammertsma, Adriaan A., Smit, Egbert F., van der Veldt, Astrid A. M., Lubberink, Mark, Bahce, Idris, Walraven, Maudy, de Boer, Michiel P., Greuter, Henri N. J. M., Hendrikse, N. Harry, Eriksson, Jonas, Windhorst, Albert D., Postmus, Pieter E., Verheul, Henk M., Serne, Erik H., Lammertsma, Adriaan A., and Smit, Egbert F.
- Abstract
Current strategies combining anti-angiogenic drugs with chemotherapy provide clinical benefit in cancer patients. It is assumed that anti-angiogenic drugs, such as bevacizumab, transiently normalize abnormal tumor vasculature and contribute to improved delivery of subsequent chemotherapy. To investigate this concept, a study was performed in non-small cell lung cancer (NSCLC) patients using positron emission tomography (PET) and radiolabeled docetaxel ([11C]docetaxel). In NSCLC, bevacizumab reduced both perfusion and net influx rate of [11C]docetaxel within 5 hr. These effects persisted after 4 days. The clinical relevance of these findings is notable, as there was no evidence for a substantial improvement in drug delivery to tumors. These findings highlight the importance of drug scheduling and advocate further studies to optimize scheduling of anti-angiogenic drugs.
- Published
- 2012
- Full Text
- View/download PDF
116. Development of [11C]erlotinib Positron Emission Tomography for In Vivo Evaluation of EGF Receptor Mutational Status
- Author
-
Bahce, Idris, primary, Smit, Egbert F., additional, Lubberink, Mark, additional, van der Veldt, Astrid A. M., additional, Yaqub, Maqsood, additional, Windhorst, Albert D., additional, Schuit, Robert C., additional, Thunnissen, Erik, additional, Heideman, Daniëlle A. M., additional, Postmus, Pieter E., additional, Lammertsma, Adriaan A., additional, and Hendrikse, N. Harry, additional
- Published
- 2013
- Full Text
- View/download PDF
117. Quantitative and Simplified Analysis of 11C-Erlotinib Studies.
- Author
-
Yaqub, Maqsood, Bahce, Idris, Voorhoeve, Charlotte, Schuit, Robert C., Windhorst, Albert D., Hoekstra, Otto S., Boellaard, Ronald, Hendrikse, N. Harry, Smit, Egbert F., and Lammertsma, Adriaan A.
- Published
- 2016
- Full Text
- View/download PDF
118. Multiparametric Analysis of the Relationship Between Tumor Hypoxia and Perfusion with 18F-Fluoroazomycin Arabinoside and 15O-H2O PET.
- Author
-
Iqbal, Ramsha, Kramer, Gem M., Verwer, Eline E., Huisman, Marc C., de Langen, Adrianus J., Bahce, Idris, van Velden, Floris H. P., Windhorst, Albert D., Lammertsma, Adriaan A., Hoekstra, Otto S., and Boellaard, Ronald
- Published
- 2016
- Full Text
- View/download PDF
119. Effects of erlotinib therapy on [C]erlotinib uptake in EGFR mutated, advanced NSCLC.
- Author
-
Bahce, Idris, Yaqub, Maqsood, Errami, Hanane, Schuit, Robert, Schober, Patrick, Thunnissen, Erik, Windhorst, Albert, Lammertsma, Adriaan, Smit, Egbert, and Hendrikse, N.
- Abstract
Background: In non-small cell lung cancer (NSCLC) patients off erlotinib therapy, positron emission tomography (PET) using [C]erlotinib distinguished epidermal growth factor receptor (EGFR) mutations from wild-type EGFR. However, tumor uptake of [C]erlotinib during erlotinib therapy is unknown. Therefore, the aims of this study were to evaluate tumor [C]erlotinib uptake in NSCLC patients both on and off erlotinib therapy, to evaluate the effect of erlotinib therapy on tumor perfusion and its correlation to tumor [C]erlotinib uptake, and also, to investigate simplified uptake parameters using arterial and venous blood samples. Methods: Ten patients were to be scanned twice with a 1-2-week interval, i.e., on (E+) and off (E−) erlotinib therapy. Each procedure consisted of a low-dose CT scan, a 10-min dynamic [O]HO PET scan, and a 60-min dynamic [C]erlotinib PET scan with arterial and venous sampling at six time points. In patients(E+), the optimal compartment model was analyzed using Akaike information criterion. In patients(E−), the uptake parameter was the volume of distribution ( V), estimated by using metabolite-corrected plasma input curves based on image-derived input functions and discrete arterial and venous blood samples. Tumor blood flow (TBF) was determined by rate constant of influx (K1) of [O]HO using the 1T2k model and correlated with V and K1 values of [C]erlotinib. The investigated simplified parameters were standardized uptake value (SUV) and tumor-to-blood ratio (TBR) at 40-60 min pi interval. Results: Of the 13 patients included, ten were scanned twice. In patients(E+), [C]erlotinib best fitted the 2T4k model with V. In all patients, tumor V(E+) was lower than V(E−) (median V(E−) = 1.61, range 0.77-3.01; median V(E+) = 1.17, range 0.53-1.74; P = 0.004). Using [O]HO, five patients were scanned twice. TBF did not change with erlotinib therapy, TBF showed a positive trend towards correlation with [C]erlotinib K1, but not with V. TBR and TBR, using both arterial and venous sampling, correlated with V(E−) (all r >0.9, P < 0.001), while SUV did not. In patients off and on therapy, venous TBR underestimated arterial TBR by 26 ± 12 and 9 ± 9 %, respectively. Conclusions: In patients on erlotinib in therapeutic dose, tumor V decreases with high variability, independent of tumor perfusion. For simplification of [C]erlotinib PET scanning protocols, both arterial and venous TBR 40-60 min post injection can be used; however, arterial and venous TBR values should not be interchanged as venous values underestimate arterial values. Trial registration: Registered at the Netherlands Trial Registry: . [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
120. Superior sulcus tumors invading the spine: multi-modal treatment outcomes from the pre-immunotherapy era
- Author
-
Ünal, Semih, Feller, Ricardo, Stadhouder, Agnita, Heineman, David.J., Jiya, Timothy U., van Dorp, Martijn, Bahce, Idris, Braun, Jerry A., Senan, Suresh, Dahele, Max, and Dickhoff, Chris
- Abstract
Curative-intent treatment of superior sulcus tumors of the lung invading the spine presents significant challenges. We retrospectively studied outcomes in a single center, uniformly staged patient cohort treated with induction concurrent chemoradiotherapy (CRT) followed by surgical resection (trimodality therapy).
- Published
- 2023
- Full Text
- View/download PDF
121. Parametric Methods for Quantification of 18F-FAZA Kinetics in Non-Small Cell Lung Cancer Patients.
- Author
-
Verwer, Eline E., Bahce, Idris, van Velden, Floris H. P., Yaqub, Maqsood, Schuit, Robert C., Windhorst, Albert D., Raijmakers, Pieter, Hoekstra, Otto S., Lammertsma, Adriaan A., Smit, Egbert F., and Boellaard, Ronald
- Published
- 2014
- Full Text
- View/download PDF
122. Translation of third and second harmonic generation microscopy into the clinic for the assessment of fresh lung tumor tissue.
- Author
-
van Huizen, Laura M. G., Daniels, Johannes M. A., Radonic, Teodora, van Mourik, Frank, Dickhoff, Chris, Bahce, Idris, Annema, Jouke T., and Groot, Marie Louise
- Published
- 2021
- Full Text
- View/download PDF
123. Pharmacokinetic analysis of [F]FAZA in non-small cell lung cancer patients.
- Author
-
Verwer, Eline, Velden, Floris, Bahce, Idris, Yaqub, Maqsood, Schuit, Robert, Windhorst, Albert, Raijmakers, Pieter, Lammertsma, Adriaan, Smit, Egbert, and Boellaard, Ronald
- Subjects
PHARMACOKINETICS ,POSITRON emission tomography ,LUNG cancer patients ,ADIPOSE tissues ,BLOOD volume - Abstract
Purpose: [F]Fluoroazomycin arabinoside (FAZA) is a positron emission tomography (PET) tracer developed to enable identification of hypoxic regions within a tumour. The aims of this study were to determine the optimal kinetic model along with validation of using alternatives to arterial blood sampling for analysing [F]FAZA studies and to assess the validity of simplified analytical methods. Methods: Dynamic 70-min [F]FAZA PET/CT scans were obtained from nine non-small cell lung cancer patients. Continuous arterial blood sampling, together with manual arterial and venous sampling, was performed to derive metabolite-corrected plasma input functions. Volumes of interest (VOIs) were defined for tumour, healthy lung muscle and adipose tissue generating [F]FAZA time-activity curves (TACs). TACs were analysed using one- and two-tissue compartment models using both metabolite-corrected blood sampler plasma input functions (BSIF) and image-derived plasma input functions (IDIF). Results: The reversible two-tissue compartment model with blood volume parameter (2T4k+V) best described kinetics of [F]FAZA in tumours. Volumes of distribution (V) obtained using IDIF correlated well with those derived using BSIF ( R = 0.82). Venous samples yielded the same radioactivity concentrations as arterial samples for times >50 min post-injection (p.i.). In addition, both plasma to whole blood ratios and parent fractions were essentially the same for venous and arterial samples. Both standardised uptake value (SUV), normalised to lean body mass, and tumour to blood ratio correlated well with V ( R = 0.77 and R = 0.87, respectively, at 50-60 min p.i.), although a bias was observed at low V. Conclusion: The 2T4k+V model provided the best fit to the dynamic [F]FAZA data. IDIF with venous blood samples can be used as input function. Further data are needed to validate the use of simplified methods. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
124. The Analysis of Platelet-Derived circRNA Repertoire as Potential Diagnostic Biomarker for Non-Small Cell Lung Cancer.
- Author
-
D'Ambrosi, Silvia, Visser, Allerdien, Antunes-Ferreira, Mafalda, Poutsma, Ankie, Giannoukakos, Stavros, Sol, Nik, Sabrkhany, Siamack, Bahce, Idris, Kuijpers, Marijke J. E., Oude Egbrink, Mirjam G. A., Griffioen, Arjan W., Best, Myron G., Koppers-Lalic, Danijela, Oudejans, Cees, and Würdinger, Thomas
- Subjects
RNA analysis ,LUNG cancer diagnosis ,CIRCULAR RNA ,PILOT projects ,COMPUTER software ,LUNG cancer ,SEQUENCE analysis ,BLOOD platelets ,GENE expression profiling ,DESCRIPTIVE statistics ,TUMOR markers ,BODY fluid examination ,POLYMERASE chain reaction ,CARRIER proteins ,LONGITUDINAL method - Abstract
Simple Summary: Interaction between blood platelets and cancer cells play an important role in various steps of cancer development and progression. These interactions lead to changes in the platelets' RNA content, resulting in tumor-mediated "education" of platelets. Tumor-educated platelets (TEPs) can be used as a non-invasive biomarker source for cancer detection and progression monitoring. Our lab has previously identified that spliced mRNA TEP signatures provide specific information on the presence, location, and molecular features of cancers. Next to mRNA, other RNA types are present in platelets, and their repertoire can potentially be subjected to cancer-mediated alterations. Despite the evidence that circRNA could be a promising cancer biomarker, they have not yet been analyzed in blood platelets of cancer patients. In this proof-of-concept study, we aim to evaluate whether platelets' circRNA signature could be used as a biomarker for cancer detection and progression. Tumor-educated Platelets (TEPs) have emerged as rich biosources of cancer-related RNA profiles in liquid biopsies applicable for cancer detection. Although human blood platelets have been found to be enriched in circular RNA (circRNA), no studies have investigated the potential of circRNA as platelet-derived biomarkers for cancer. In this proof-of-concept study, we examine whether the circRNA signature of blood platelets can be used as a liquid biopsy biomarker for the detection of non-small cell lung cancer (NSCLC). We analyzed the total RNA, extracted from the platelet samples collected from NSCLC patients and asymptomatic individuals, using RNA sequencing (RNA-Seq). Identification and quantification of known and novel circRNAs were performed using the accurate CircRNA finder suite (ACFS), followed by the differential transcript expression analysis using a modified version of our thromboSeq software. Out of 4732 detected circRNAs, we identified 411 circRNAs that are significantly (p-value < 0.05) differentially expressed between asymptomatic individuals and NSCLC patients. Using the false discovery rate (FDR) of 0.05 as cutoff, we selected the nuclear receptor-interacting protein 1 (NRIP1) circRNA (circNRIP1) as a potential biomarker candidate for further validation by reverse transcription–quantitative PCR (RT-qPCR). This analysis was performed on an independent cohort of platelet samples. The RT-qPCR results confirmed the RNA-Seq data analysis, with significant downregulation of circNRIP1 in platelets derived from NSCLC patients. Our findings suggest that circRNAs found in blood platelets may hold diagnostic biomarkers potential for the detection of NSCLC using liquid biopsies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
125. Translation of third and second harmonic generation microscopy into the clinic for the assessment of fresh lung tumor tissue
- Author
-
Huang, Zhiwei, Lilge, Lothar D., van Huizen, Laura M. G., Daniels, Johannes M. A., Radonic, Teodora, van Mourik, Frank, Dickhoff, Chris, Bahce, Idris, Annema, Jouke T., and Groot, Marie Louise
- Published
- 2021
- Full Text
- View/download PDF
126. A Clinical Imaging Study of the Changes in [18F]F-AraG Uptake Following Anti-PD-1 Therapy in Non-small Cell Lung Cancer (SHARP)
- Author
-
Boehringer Ingelheim and Idris Bahce, Idris Bahce, MD, PhD, Principal Investigator
- Published
- 2023
127. Pharmacokinetic analysis and simplified uptake measures for tumour lesion [18F]F-AraG PET imaging in patients with non-small cell lung cancer.
- Author
-
Wijngaarden, Jessica E., Slebe, Maarten, Pouw, Johanna E. E., Oprea-Lager, Daniela E., Schuit, Robert C., Dickhoff, Chris, Levi, Jelena, Windhorst, Albert D., Oordt, C. Willemien Menke-van der Houven van, Thiele, Andrea, Bahce, Idris, Boellaard, Ronald, and Yaqub, Maqsood
- Subjects
- *
LEAN body mass , *NON-small-cell lung carcinoma , *POSITRON emission tomography , *BODY surface area , *BODY weight - Abstract
Introduction: The novel positron emission tomography (PET) imaging tracer, [18F]F-AraG, targets activated T-cells, offering a potential means to improve our understanding of immune-oncological processes. The aim of this study was to determine the optimal pharmacokinetic model to quantify tumour lesion [18F]F-AraG uptake in patients with non-small cell lung cancer (NSCLC), and to validate simplified measures at different time intervals against the pharmacokinetic uptake parameter.Ten patients with early-stage NSCLC and three patients with advanced NSCLC underwent a dynamic PET scan of minimal 60 min. Venous and/or arterial blood sampling was obtained at maximum seven time points. Tumour lesion time activity curves and metabolite-corrected input functions were analysed using single-tissue reversible (1T2k), two-tissue irreversible (2T3k) and two-tissue reversible (2T4k) plasma input models. Simplified uptake measures, such as standardised uptake value (SUV) and tumour-to-blood (TBR) or tumour-to-plasma ratio (TPR), were evaluated for different time intervals.Whole-blood and plasma radioactivity concentrations showed rapid clearance of [18F]F-AraG. Metabolite analysis revealed a low rate of metabolism, at 70 min p.i., on average, 79% (SD = 9.8%) of the total radioactivity found in blood corresponded to intact [18F]F-AraG. The time activity curves were best fitted by the 2T3k model. Strong positive correlations were found for SUV (body weight (BW), lean body mass (LBM) or body surface area (BSA) corrected), TBR and TPR for any time interval between 20 and 70 min p.i. against the 2T3k-derived
K i . The correlation of TBR at 60–70 min p.i. with 2T3K-derivedK i (r (df = 20) = 0.87,p < 0.01), was stronger than for SUVBW (r (df = 20) = 0.80,p < 0.01).Tumour lesion [18F]F-AraG uptake in patients with NSCLC is characterised by a 2T3k model. TBR and TPR show most potential for simplified quantification of tumour lesion [18F]F-AraG uptake in patients with NSCLC.Methods: The novel positron emission tomography (PET) imaging tracer, [18F]F-AraG, targets activated T-cells, offering a potential means to improve our understanding of immune-oncological processes. The aim of this study was to determine the optimal pharmacokinetic model to quantify tumour lesion [18F]F-AraG uptake in patients with non-small cell lung cancer (NSCLC), and to validate simplified measures at different time intervals against the pharmacokinetic uptake parameter.Ten patients with early-stage NSCLC and three patients with advanced NSCLC underwent a dynamic PET scan of minimal 60 min. Venous and/or arterial blood sampling was obtained at maximum seven time points. Tumour lesion time activity curves and metabolite-corrected input functions were analysed using single-tissue reversible (1T2k), two-tissue irreversible (2T3k) and two-tissue reversible (2T4k) plasma input models. Simplified uptake measures, such as standardised uptake value (SUV) and tumour-to-blood (TBR) or tumour-to-plasma ratio (TPR), were evaluated for different time intervals.Whole-blood and plasma radioactivity concentrations showed rapid clearance of [18F]F-AraG. Metabolite analysis revealed a low rate of metabolism, at 70 min p.i., on average, 79% (SD = 9.8%) of the total radioactivity found in blood corresponded to intact [18F]F-AraG. The time activity curves were best fitted by the 2T3k model. Strong positive correlations were found for SUV (body weight (BW), lean body mass (LBM) or body surface area (BSA) corrected), TBR and TPR for any time interval between 20 and 70 min p.i. against the 2T3k-derivedK i . The correlation of TBR at 60–70 min p.i. with 2T3K-derivedK i (r (df = 20) = 0.87,p < 0.01), was stronger than for SUVBW (r (df = 20) = 0.80,p < 0.01).Tumour lesion [18F]F-AraG uptake in patients with NSCLC is characterised by a 2T3k model. TBR and TPR show most potential for simplified quantification of tumour lesion [18F]F-AraG uptake in patients with NSCLC.Results: The novel positron emission tomography (PET) imaging tracer, [18F]F-AraG, targets activated T-cells, offering a potential means to improve our understanding of immune-oncological processes. The aim of this study was to determine the optimal pharmacokinetic model to quantify tumour lesion [18F]F-AraG uptake in patients with non-small cell lung cancer (NSCLC), and to validate simplified measures at different time intervals against the pharmacokinetic uptake parameter.Ten patients with early-stage NSCLC and three patients with advanced NSCLC underwent a dynamic PET scan of minimal 60 min. Venous and/or arterial blood sampling was obtained at maximum seven time points. Tumour lesion time activity curves and metabolite-corrected input functions were analysed using single-tissue reversible (1T2k), two-tissue irreversible (2T3k) and two-tissue reversible (2T4k) plasma input models. Simplified uptake measures, such as standardised uptake value (SUV) and tumour-to-blood (TBR) or tumour-to-plasma ratio (TPR), were evaluated for different time intervals.Whole-blood and plasma radioactivity concentrations showed rapid clearance of [18F]F-AraG. Metabolite analysis revealed a low rate of metabolism, at 70 min p.i., on average, 79% (SD = 9.8%) of the total radioactivity found in blood corresponded to intact [18F]F-AraG. The time activity curves were best fitted by the 2T3k model. Strong positive correlations were found for SUV (body weight (BW), lean body mass (LBM) or body surface area (BSA) corrected), TBR and TPR for any time interval between 20 and 70 min p.i. against the 2T3k-derivedK i . The correlation of TBR at 60–70 min p.i. with 2T3K-derivedK i (r (df = 20) = 0.87,p < 0.01), was stronger than for SUVBW (r (df = 20) = 0.80,p < 0.01).Tumour lesion [18F]F-AraG uptake in patients with NSCLC is characterised by a 2T3k model. TBR and TPR show most potential for simplified quantification of tumour lesion [18F]F-AraG uptake in patients with NSCLC.Conclusion: The novel positron emission tomography (PET) imaging tracer, [18F]F-AraG, targets activated T-cells, offering a potential means to improve our understanding of immune-oncological processes. The aim of this study was to determine the optimal pharmacokinetic model to quantify tumour lesion [18F]F-AraG uptake in patients with non-small cell lung cancer (NSCLC), and to validate simplified measures at different time intervals against the pharmacokinetic uptake parameter.Ten patients with early-stage NSCLC and three patients with advanced NSCLC underwent a dynamic PET scan of minimal 60 min. Venous and/or arterial blood sampling was obtained at maximum seven time points. Tumour lesion time activity curves and metabolite-corrected input functions were analysed using single-tissue reversible (1T2k), two-tissue irreversible (2T3k) and two-tissue reversible (2T4k) plasma input models. Simplified uptake measures, such as standardised uptake value (SUV) and tumour-to-blood (TBR) or tumour-to-plasma ratio (TPR), were evaluated for different time intervals.Whole-blood and plasma radioactivity concentrations showed rapid clearance of [18F]F-AraG. Metabolite analysis revealed a low rate of metabolism, at 70 min p.i., on average, 79% (SD = 9.8%) of the total radioactivity found in blood corresponded to intact [18F]F-AraG. The time activity curves were best fitted by the 2T3k model. Strong positive correlations were found for SUV (body weight (BW), lean body mass (LBM) or body surface area (BSA) corrected), TBR and TPR for any time interval between 20 and 70 min p.i. against the 2T3k-derivedK i . The correlation of TBR at 60–70 min p.i. with 2T3K-derivedK i (r (df = 20) = 0.87,p < 0.01), was stronger than for SUVBW (r (df = 20) = 0.80,p < 0.01).Tumour lesion [18F]F-AraG uptake in patients with NSCLC is characterised by a 2T3k model. TBR and TPR show most potential for simplified quantification of tumour lesion [18F]F-AraG uptake in patients with NSCLC. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
128. EGFR TKI PET/CT in advanced stage non-small cell lung cancer patients
- Author
-
van de Stadt, Eveline Annette, Hendrikse, N.H., Bahce, Idris, Yaqub, Mohammed Maqsood, Bartelink, I.H., and VUmc - School of Medical Sciences
- Subjects
PET/CT ,EGFR TKI ,NSCLC ,EGFR TKI, PET/CT, NSCLC - Abstract
An overview of biomarker development is provided in chapter 2.PET tracer-based biomarkers can be used to monitor different biological or clinical metrics. A clinically important biomarker, especially in lung cancer, is the epidermal growth factor receptor (EGFR) abundance. In this chapter we give an overview of current EGFR-directed PET tracers to visualize the tumors’ EGFR binding capacity, and discuss the challenges and opportunities regarding their clinical application. One of the major challenges is the necessity of highly complex and invasive acquisition protocols for quantitative assessment of tracer uptake. Another important challenge for the current tracer developmental process is the rapidly changing treatment landscape. Development of a TKI-based PET tracer can be relatively long and with the current rate of change in treatment options, developed tracers should be brought quickly into clinical usage before they lose their clinical relevance. This chapter also highlights some opportunities. The total body PET scanner could greatly decrease the level of complexity of protocols. This would decrease the time needed for a tracer to be ready for clinical use, which in turn could improve the clinical relevance of EGFR-directed tracers. In chapter 3 we describe the process of quantification of 18F-afatinib. Ten NSCLC patients underwent dynamic PET scanning using 18F-afatinib. Three pharmacokinetic compartment models were assessed using both plasma-derived input functions and image-derived input functions: a single-tissue model (1T2K), a two-tissue reversible model (2T4K) and a two-tissue irreversible model (2T3K). The preferred model was the two-tissue irreversible model. This is consistent with in vitro data showing irreversible binding of afatinib to the EGF receptor. The relationship between 18F-afatinib tumor uptake, EGFR mutational status and response to treatment using afatinib was investigated in chapter 4. In this chapter we compared tracer uptake of EGFR wild type tumors with both EGFR common and uncommon tumors, hypothesizing that uncommon EGFR mutations behave similarly to common mutations. A significant difference was observed between tracer uptake of wild type tumors versus both common and uncommon mutations. Furthermore, a TBR60-90 value of >6 was shown to be predictive of response to treatment using afatinib. Chapter 5 focuses on the biodistribution and image quality of three generations of EGFR TKI PET tracers: 11C-erlotinib, 18F-afatinib and 11C-osimertinib based. The image quality derived from patients in a recently started clinical trial of the 11C-osimertinib derived tracer was remarkably different from the first two generation of TKI tracers. The tumor tissue showed a low tracer uptake compared to the surrounding lung tissue. To investigate this phenomenon, we (re-)analyzed data from three previously published prospective studies and one ongoing clinical trial. Image quality was also quantified for each tracer by calculating the tumor-to-lung contrast and background noise for each tracer. 11C-erlotinib and 18F-afatinib showed the best image quality based on both tumor-to-lung contrast and noise, whereas 11C-osimertinib showed an inverse tumor-to-lung contrast: lung tissue showed a higher level of tracer uptake when compared to tumor tissue. Differences in physicochemical, pharmacological and pharmacokinetic parameters of the three generation of TKI’s may explain the observed the differences in tumor-to-lung contrast. The results of the comparison that we conducted in chapter 5 lead to chapter 6. In this chapter we developed a physiologically-based pharmacokinetic (PBPK) model that accurately predicted tissue uptake for the three EGFR TKIs (erlotinib, afatinib and osimertinib) using physicochemical and drug specific properties. This model was validated using PET data as obtained and described in the previous chapter. Our model yielded an adequate prediction of tumor-to-lung contrast and whole-body distribution of the three EGFR TKIs.
- Published
- 2023
129. EGFR TKI PET/CT in advanced stage non-small cell lung cancer patients
- Author
-
van de Stadt, E.A., Hendrikse, Harry, Bahce, Idris, Yaqub, Mohammed, Bartelink, Imke, and Pulmonary medicine
- Published
- 2023
130. Multidisciplinary decision-making in the care of patients with lung cancer
- Author
-
Ronden, Merle Ilona, Senan, Suresh, Bahce, Idris, Pulmonary medicine, APH - Quality of Care, APH - Personalized Medicine, Radiation Oncology, and Senan, S.
- Subjects
interstitial lung disease ,lung cancer ,SDG 16 - Peace ,high-risk radiological features ,locally advanced ,SDG 16 - Peace, Justice and Strong Institutions ,multidisciplinary decision making ,patterns of care ,early stage ,Justice and Strong Institutions ,SABR - Abstract
The work performed in this thesis addresses some of the challenges facing multidisciplinary tumor boards (MDT) when assessing patients who present with a non-metastatic non-small cell lung cancer (NSCLC). Challenges in patients with early-stage NSCLC treated with stereotactic ablative radiotherapy Acute and late fibrotic changes following stereotactic ablative radiotherapy (SABR) are commonly observed, and mass-like fibrosis is sometimes difficult to distinguish from tumor recurrence. An overview of the patterns of radiological changes seen after SABR were highlighted, and high-risk radiological features (HRF’s) associated with a higher risk of tumor recurrence were presented. In addition, the incidence and patterns of change of HRF’s were studied on 747 follow-up CT scans of 88 patients who were known to have no local recurrence. More than half of these patients developed HRF’s, with (sequential) enlarging opacities being observed in most of these cases. In nearly 25% of these patients 3 or more HRF’s were present. Considerable inter-observer variability in scoring these features was observed, and multidisciplinary assessment of patients with HRF’s is therefore recommended. Another challenge is the treatment of patients with lung cancer and co-existing interstitial lung disease (ILD) as they have an increased risk on toxicity following cancer treatments. We presented our institutional experience with SABR in 19 patients with early stage NSCLC and co-existing ILD, with 42% having idiopathic pulmonary fibrosis. Grade ≥2 lung toxicity was observed in 32% of patients, with 21% of cases classified as a possible, probable or definite grade 5 radiation pneumonitis. Median overall survival was only 17 months, a finding which was predicted by a high ILD-GAP score of ≥4 in 63% of patients. Factors influencing multidisciplinary decision-making in patients with locally advanced NSCLC Treatment decision-making was studied in 197 patients presenting with locally advanced NSCLC between 2015-2017 at our regional network comprising 5 hospitals, using data from the Netherlands Cancer Registry, hospital records and weekly MDT reports. MDT assessment was performed in 95% of all patients, and radical intent treatments (RIT) including surgery or concurrent chemoradiotherapy (CCRT) were recommended in 61%. Finally, 48% actually received a RIT, and the most important factors associated with a failure to do so were an age of ≥70 years and a WHO-performance score of ≥2. The commonest cause of death for all patients was progressive lung cancer, and more comorbidity related deaths were observed in the patients who did not undergo a RIT. In a subsequent study, we extended this analysis of treatment decis
- Published
- 2021
131. Pharmacokinetic analysis and simplified uptake measures for tumour lesion [ 18 F]F-AraG PET imaging in patients with non-small cell lung cancer.
- Author
-
Wijngaarden JE, Slebe M, Pouw JEE, Oprea-Lager DE, Schuit RC, Dickhoff C, Levi J, Windhorst AD, Oordt CWMHV, Thiele A, Bahce I, Boellaard R, and Yaqub M
- Abstract
Introduction: The novel positron emission tomography (PET) imaging tracer, [
18 F]F-AraG, targets activated T-cells, offering a potential means to improve our understanding of immune-oncological processes. The aim of this study was to determine the optimal pharmacokinetic model to quantify tumour lesion [18 F]F-AraG uptake in patients with non-small cell lung cancer (NSCLC), and to validate simplified measures at different time intervals against the pharmacokinetic uptake parameter., Methods: Ten patients with early-stage NSCLC and three patients with advanced NSCLC underwent a dynamic PET scan of minimal 60 min. Venous and/or arterial blood sampling was obtained at maximum seven time points. Tumour lesion time activity curves and metabolite-corrected input functions were analysed using single-tissue reversible (1T2k), two-tissue irreversible (2T3k) and two-tissue reversible (2T4k) plasma input models. Simplified uptake measures, such as standardised uptake value (SUV) and tumour-to-blood (TBR) or tumour-to-plasma ratio (TPR), were evaluated for different time intervals., Results: Whole-blood and plasma radioactivity concentrations showed rapid clearance of [18 F]F-AraG. Metabolite analysis revealed a low rate of metabolism, at 70 min p.i., on average, 79% (SD = 9.8%) of the total radioactivity found in blood corresponded to intact [18 F]F-AraG. The time activity curves were best fitted by the 2T3k model. Strong positive correlations were found for SUV (body weight (BW), lean body mass (LBM) or body surface area (BSA) corrected), TBR and TPR for any time interval between 20 and 70 min p.i. against the 2T3k-derived Ki . The correlation of TBR at 60-70 min p.i. with 2T3K-derived Ki (r (df = 20) = 0.87, p < 0.01), was stronger than for SUVBW (r (df = 20) = 0.80, p < 0.01)., Conclusion: Tumour lesion [18 F]F-AraG uptake in patients with NSCLC is characterised by a 2T3k model. TBR and TPR show most potential for simplified quantification of tumour lesion [18 F]F-AraG uptake in patients with NSCLC., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
132. Stereologic consequences of iatrogenic collapse: The morphology of adenocarcinoma in situ overlaps with invasive patterns. Proposal for a necessary modified classification of pulmonary adenocarcinomas.
- Author
-
Filipello F, Blaauwgeers H, Lissenberg-Witte B, Schonau A, Doglioni C, Arrigoni G, Radonic T, Bahce I, Smit A, Dickhoff C, Nuccio A, Bulotta A, Minami Y, Noguchi M, Ambrosi F, and Thunnissen E
- Abstract
Recognizing non-invasive growth patterns is necessary for correct diagnosis, invasive size determination and pT-stage in resected non-small cell lung carcinoma. Due to iatrogenic collapse after resection, the distinction between adenocarcinoma in-situ (AIS) and invasive adenocarcinoma may be difficult. The aim of this study is to investigate the complex morphology of non-mucinous non-invasive patterns of AIS in resection specimen with iatrogenic collapse, and to relate this to follow-up. The effects of iatrogenic collapse on the morphology of collapsed AIS were simulated in a mathematical model. Three dimensional related criteria applied in a modified classification, using also cytokeratin 7 and elastin as additional stains, in two independent retrospective cohorts of primary pulmonary adenocarcinomas ≤3 cm resection specimen with available follow-up information. The model demonstrated that infolding of alveolar walls occurs during iatrogenic collapse and lead to a significant increase in tumor cell heights in maximal collapse areas, compared to less collapsed areas. The morphology of infolded AIS overlaps with patterns described as papillary and acinar adenocarcinoma according to the WHO classification, necessitating an adaptation. The modified classification incorporates recognition of iatrogenic and biologic collapse, tangential cutting effect true invasion and surrogate markers of invasion i.e. grey zone, covering a multilayering falling short of micropapillary, cribriform and solid alveolar filling growth. The use of elastin and CK7 staining aids in the morphologic recognition of iatrogenic collapsed AIS and the distinction from invasive adenocarcinoma. Out of a total of 70 resection specimens 1 case was originally classified as AIS and 9 were reclassified as iatrogenic collapsed AIS. Patients with collapsed AIS showed a 100 % recurrence-free survival after a mean follow-up time of 69.5 months. With the current WHO classification, AIS is overdiagnosed as invasive adenocarcinoma due to infolding. The modified classification facilitates the diagnosis of AIS., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Andreas schonau is developer of the pathogate website. None of the other authors declares a conflict of interest., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
133. Single-arm trial of neoadjuvant ipilimumab plus nivolumab with chemoradiotherapy in patients with resectable and borderline resectable lung cancer: the INCREASE study.
- Author
-
Bahce I, Dickhoff C, Schneiders FL, Veltman J, Heineman DJ, Hashemi SMS, Vrijmoet A, Houda I, Ulas EB, Bakker J, van de Ven P, Bouwhuis N, Meijboom LJ, Oprea-Lager DE, van Maldegem F, Fransen MF, de Gruijl TD, Radonic T, and Senan S
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Chemoradiotherapy methods, Chemoradiotherapy adverse effects, Adult, Carcinoma, Non-Small-Cell Lung therapy, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung drug therapy, Ipilimumab therapeutic use, Ipilimumab administration & dosage, Lung Neoplasms therapy, Lung Neoplasms pathology, Lung Neoplasms drug therapy, Nivolumab therapeutic use, Nivolumab administration & dosage, Nivolumab adverse effects, Neoadjuvant Therapy methods
- Abstract
Background: In non-small cell lung cancer (NSCLC), chemoradiotherapy (CRT) yields pathological complete response (pCR) rates of approximately 30%. We investigated using ipilimumab plus nivolumab (IPI-NIVO) with neoadjuvant CRT in resectable, and borderline resectable NSCLC., Methods: This single-arm, phase-II trial enrolled operable T3-4N0-2 patients with NSCLC without oncogenic drivers. Primary study endpoints were safety, major pathological response (MPR) and pCR. Treatment encompassed platinum-doublet concurrent CRT, IPI 1 mg/kg intravenous and NIVO 360 mg intravenous on day-1, followed by chemotherapy plus NIVO 360 mg 3 weeks later. Thoracic radiotherapy was 50 or 60 Gy, in once-daily doses of 2 Gy. Resections were 6 weeks post-radiotherapy., Results: In a total of 30 patients in the intention-to-treat (ITT) population, grades 3-4 treatment-related adverse events (TRAEs) occurred in 70%, one TRAE grade 5 late-onset pneumonitis on day 96 post-surgery (1/30, 3.3%) occurred, and one non-TRAE COVID-19 death (1/30, 3.3%). pCR and MPR were achieved in 50% (15/30) and 63% (19/30) of the ITT; and in 58% (15/26) and 73% (19/26) of the 26 patients who underwent surgery, respectively. Postoperative melanoma was seen in one non-pCR patient. The R0 rate was 100% (26/26), and no patient failed surgery due to TRAEs. In peripheral blood, proliferative CD8
+ T cells were increased, while proliferative regulatory T cells (Tregs) were not. On-treatment, pCR-positives had higher CD8+ CD39+ T cells and lower HLA-DR+ Tregs., Conclusions: Neoadjuvant IPI-NIVO-CRT in T3-4N0-2 NSCLC showed acceptable safety with pCR and MPR in 58% and 73% of operated patients, respectively. No patient failed surgery due to TRAEs., Trial Registration Number: NCT04245514., Competing Interests: Competing interests: The authors have disclosed the following conflicts of interest: DEO-L received grants from Janssen and Curium, honoraria from EANM, EAU, ESMO, and Curium, travel support from EANM, ESMO, EAU, and Bayer, and serves on a Bayer advisory board. CD received institutional fees for advisory roles with BMS, AstraZeneca, and MSD. SMSH has research contracts with several pharmaceutical companies and received honoraria from Janssen. IB’s institution received support from BMS, AstraZeneca, and Boehringer Ingelheim, and he received honoraria from AstraZeneca, MSD, and BMS. FLS received a research grant from ViewRay and honoraria from AstraZeneca. SS’s institution received grants from Varian, ViewRay, and AstraZeneca; he received consulting fees and honoraria from AstraZeneca and MSD, and serves on advisory boards for AstraZeneca and MSD. TDdG’s institution received grants from Idera Pharmaceuticals, consulting fees from LAVA Therapeutics and Mendus, and she holds stock in LAVA Therapeutics. All other authors have declared no conflicts of interest., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
134. Comparison of cell-free and small extracellular-vesicle-associated DNA by sequencing plasma of lung cancer patients.
- Author
-
Moldovan N, Verkuijlen S, van der Pol Y, Bosch L, van Weering JRT, Bahce I, Pegtel DM, and Mouliere F
- Abstract
Blood contains multiple analytes that can be used as liquid biopsy to analyze cancer. Mutations have been detected in DNA associated with small extracellular vesicles (sEVs). The genome-wide composition and structure of sEV DNA remains poorly characterized, and whether sEVs are enriched in tumor signal compared to cell-free DNA (cfDNA) is unclear. Here, using whole-genome sequencing from lung cancer patients we determined that the tumor fraction and heterogeneity are comparable between DNA associated with sEV (<200 nm) and matched plasma cfDNA. sEV DNA, obtained with size-exclusion chromatography, is composed of short ∼150-180 bp fragments and long >1000 bp fragments poor in tumor signal. The structural patterns of sEV DNA are related to plasma cfDNA. Mitochondrial DNA is relatively enriched in the sEV fractions. Our results suggest that DNA associated to sEV (including exosomes) is not preferentially enriched in tumor signal and is less abundant than cfDNA., Competing Interests: F.M. is coinventor on patents related to cfDNA fragmentation analysis. F.M. has consulted for Roche Dx. D.M.P. is co-founder and CSO of ExBiome BV. Other coauthors have no relevant conflict of interests., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
135. Association Between PD-L1 Score and the Outcomes of Consolidation Durvalumab in a Large Nationwide Series of Patients With Stage III NSCLC Treated With Chemoradiotherapy.
- Author
-
Damhuis R, Bahce I, and Senan S
- Abstract
Background: The Pacific trial reported improved outcomes when durvalumab was administered following concurrent chemoradiotherapy (CRT) for stage III NSCLC. Post-hoc subgroup analysis did not show favorable results for PD-L1 negative cases. We compared nationwide survival data with the trial outcomes, and evaluated the influence of PD-L1., Patients and Methods: Data from the Netherlands Cancer Registry were queried regarding patients with clinical stage III who underwent CRT, either by concurrent or sequential administration. Predictors for the use of consolidation treatment with durvalumab were evaluated by tabulations and logistic regression analysis. Overall survival (OS) was calculated from start of radiation or start of durvalumab and was stratified by PD-L1 score., Results: Between 2017 and 2021, application of consolidation durvalumab increased from 2% to 21%, 40%, 57%, 62%, respectively. In the period 2020-2021, durvalumab use was more frequent among patients with younger age, concurrent CRT, better performance score and proton radiation, but was irrespective of PD-L1 score. For patients receiving durvalumab (n = 1639), the 4-year OS was 53% overall (95%CI 50-57), and it was 56% (95%CI 52-60) after concurrent CRT. Four-year OS was considerably better for the PD-L1 subgroup ≧50% at 67% (95%CI 59-73), and it was similar for PD-L1 subgroups 0 and 1-49, at 51% (95%CI 42-58) and 46% (95%CI 39-54), respectively., Conclusion: In real-world clinical practice, survival outcomes were equivalent to results from trial series. Overall survival in patients with negative PD-L1 was similar to the survival in patients with PD-L1 1-49, questioning the restrictions imposed by the European Medicines Agency., Competing Interests: Disclosure IB reports research grants from AstraZeneca, Bristol-Myers Squibb and Boehringer Ingelheim, consulting fees from AstraZeneca, Bristol-Myers Squibb and Boehringer Ingelheim. SS reports participating on advisory boards for AstraZeneca, Merck Sharp & Dohme, and Varian Medical Systems; and institutional research grants from AstraZeneca, Varian Medical Systems, and ViewRay. RD declares no conflicts of interest., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
136. The Surgical Resection Difficulty From Neoadjuvant Chemoimmunotherapy Is Minimal and Neoadjuvant Therapy Should Be the Standard.
- Author
-
Dickhoff C, Heineman DJ, van Dorp M, Senan S, and Bahce I
- Subjects
- Humans, Lung Neoplasms pathology, Lung Neoplasms drug therapy, Lung Neoplasms surgery, Lung Neoplasms therapy, Neoadjuvant Therapy methods, Immunotherapy methods
- Abstract
Competing Interests: Disclosure Dr. Dickhoff reports receiving research grants and consulting fees from AstraZeneca and Bristol Myers Squibb. Dr. Bahce reports receiving research grants and consulting fees from AstraZeneca, Bristol Myers Squibb, and Boehringer Ingelheim. Dr. Senan reports receiving research grants from AstraZeneca, ViewRay Inc., and Varian Medical Systems; and consulting fees from ViewRay Inc., AstraZeneca, and Merck Sharp & Dohme. The remaining authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
137. Identification of protein biomarkers for prediction of response to platinum-based treatment regimens in patients with non-small cell lung cancer.
- Author
-
Böttger F, Radonic T, Bahce I, Monkhorst K, Piersma SR, Pham TV, Dingemans AC, Hillen LM, Santarpia M, Giovannetti E, Smit EF, Burgers SA, and Jimenez CR
- Subjects
- Humans, Female, Male, Aged, Middle Aged, Proteomics methods, HMGB1 Protein metabolism, Platinum therapeutic use, Cohort Studies, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Lung Neoplasms pathology, Biomarkers, Tumor metabolism
- Abstract
The majority of patients with resected stage II-IIIA non-small cell lung cancer (NSCLC) are treated with platinum-based adjuvant chemotherapy (ACT) in a one-size-fits-all approach. However, a significant number of patients do not derive clinical benefit, and no predictive patient selection biomarker is currently available. Using mass spectrometry-based proteomics, we have profiled tumour resection material of 2 independent, multi-centre cohorts of in total 67 patients with NSCLC who underwent ACT. Unsupervised cluster analysis of both cohorts revealed a poor response/survival sub-cluster composed of ~ 25% of the patients, that displayed a strong epithelial-mesenchymal transition signature and stromal phenotype. Beyond this stromal sub-population, we identified and validated platinum response prediction biomarker candidates involved in pathways relevant to the mechanism of action of platinum drugs, such as DNA damage repair, as well as less anticipated processes such as those related to the regulation of actin cytoskeleton. Integration with pre-clinical proteomics data supported a role for several of these candidate proteins in platinum response prediction. Validation of one of the candidates (HMGB1) in a third independent patient cohort using immunohistochemistry highlights the potential of translating these proteomics results to clinical practice., (© 2023 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
138. The development process of 'fit-for-purpose' imaging biomarkers to characterize the tumor microenvironment.
- Author
-
Eertink JJ, Bahce I, Waterton JC, Huisman MC, Boellaard R, Wunder A, Thiele A, and Menke-van der Houven van Oordt CW
- Abstract
Immune-based treatment approaches are successfully used for the treatment of patients with cancer. While such therapies can be highly effective, many patients fail to benefit. To provide optimal therapy choices and to predict treatment responses, reliable biomarkers for the assessment of immune features in patients with cancer are of significant importance. Biomarkers (BM) that enable a comprehensive and repeatable assessment of the tumor microenvironment (TME), the lymphoid system, and the dynamics induced by drug treatment can fill this gap. Medical imaging, notably positron emission tomography (PET) and magnetic resonance imaging (MRI), providing whole-body imaging BMs, might deliver such BMs. However, those imaging BMs must be well characterized as being 'fit for purpose' for the intended use. This review provides an overview of the key steps involved in the development of 'fit-for-purpose' imaging BMs applicable in drug development, with a specific focus on pharmacodynamic biomarkers for assessing the TME and its modulation by immunotherapy. The importance of the qualification of imaging BMs according to their context of use (COU) as defined by the Food and Drug Administration ( FDA ) and National Institutes of Health Biomarkers, EndpointS, and other Tools ( BEST ) glossary is highlighted. We elaborate on how an imaging BM qualification for a specific COU can be achieved., Competing Interests: AT and AW were employees of Boehringer Ingelheim. JW holds stock in Quantitative Imaging Ltd. and is a Director of, and has received compensation from, Bioxydyn Ltd., a for-profit company engaged in the discovery and development of MR biomarkers and the provision of imaging biomarker services. CM reports research funding from Bristol-Myers Squibb, Boehringer Ingelheim, GSK, Pfizer, and AstraZeneca. Consultancy: GE Health Care, Novartis, Eli Lilly. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Eertink, Bahce, Waterton, Huisman, Boellaard, Wunder, Thiele and Menke-van der Houven van Oordt.)
- Published
- 2024
- Full Text
- View/download PDF
139. Loose Tumor Cells in Pulmonary Arteries of Lung Adenocarcinoma Resection Specimens: No Correlation With Survival, Despite High Prevalence.
- Author
-
Blaauwgeers H, Filipello F, Lissenberg-Witte B, Doglioni C, Radonic T, Bahce I, Minami Y, Schonau A, Vincenten JPL, Smit AAJ, Dickhoff C, and Thunnissen E
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Retrospective Studies, Pilot Projects, Prognosis, Prevalence, Neoplastic Cells, Circulating pathology, Adenocarcinoma pathology, Adenocarcinoma surgery, Adenocarcinoma mortality, Aged, 80 and over, Keratin-7 metabolism, Keratin-7 analysis, Lung Neoplasms surgery, Lung Neoplasms pathology, Lung Neoplasms mortality, Pulmonary Artery pathology, Pulmonary Artery surgery, Adenocarcinoma of Lung surgery, Adenocarcinoma of Lung pathology, Adenocarcinoma of Lung mortality
- Abstract
Context: Loose tumor cells and tumor cell clusters can be recognized in the lumen of intratumoral pulmonary arteries of resected non-small cell lung cancer specimens. It is unclear whether these should be considered tumor-emboli, and as such could predict a worsened prognosis., Objective: To investigate the nature and prognostic impact of pulmonary artery intraluminal tumor cells., Design: This multicenter study involved an exploratory pilot study and a validation study from 3 institutions. For the exploratory pilot study, a retrospective pulmonary resection cohort of primary adenocarcinomas, diagnosed between November 2007 and November 2010, were scored for the presence of tumor cells, as well as potentially other cells in the intravascular spaces, using hematoxylin-eosin and cytokeratin 7 (CK7) stains. In the validation part, 2 retrospective cohorts of resected pulmonary adenocarcinomas, between January 2011 and December 2016, were included. Recurrence-free survival (RFS) and overall survival (OS) data were collected., Results: In the pilot study, CK7+ intravascular cells, mainly tumor cells, were present in 23 of 33 patients (69.7%). The 5-year OS for patients with intravascular tumor cells was 61%, compared with 40% for patients without intravascular tumor cells (P = .19). In the validation study, CK7+ intravascular tumor cells were present in 41 of 70 patients (58.6%). The 5-year RFS for patients with intravascular tumor cells was 80.0%, compared with 80.6% in patients without intravascular tumor cells (P = .52). The 5-year OS rates were, respectively, 82.8% and 71.6% (P = .16)., Conclusions: Loose tumor cells in pulmonary arterial lumina were found in most non-small cell lung cancer resection specimens and were not associated with a worse RFS or OS. Therefore, most probably they represent an artifact., Competing Interests: The authors have no relevant financial interest in the products or companies described in this article., (© 2024 College of American Pathologists.)
- Published
- 2024
- Full Text
- View/download PDF
140. New systemic treatment paradigms in resectable non-small cell lung cancer and variations in patient access across Europe.
- Author
-
Houda I, Dickhoff C, Uyl-de Groot CA, Reguart N, Provencio M, Levy A, Dziadziuszko R, Pompili C, Di Maio M, Thomas M, Brunelli A, Popat S, Senan S, and Bahce I
- Abstract
The treatment landscape of resectable early-stage non-small cell lung cancer (NSCLC) is set to change significantly due to encouraging results from randomized trials evaluating neoadjuvant and adjuvant immunotherapy, as well as adjuvant targeted therapy. As of January 2024, marketing authorization has been granted for four new indications in Europe, and regulatory approvals for other study regimens are expected. Because cost-effectiveness and reimbursement criteria for novel treatments often differ between European countries, access to emerging developments may lead to inequalities due to variations in recommended and available lung cancer care throughout Europe. This Series paper (i) highlights the clinical studies reshaping the treatment landscape in resectable early-stage NSCLC, (ii) compares and contrasts approaches taken by the European Medicines Agency (EMA) for drug approval to that taken by the United States Food and Drug Administration (FDA), and (iii) evaluates the differences in access to emerging treatments from an availability perspective across European countries., Competing Interests: I.H., C.D., N.R., and I.B. declare no competing interests. C.A.G. has received grants or contracts from Boehringer Ingelheim, Astellas, Celgene, Sanofi, Janssen-Cilag, Bayer, Amgen, Genzyme, Merck, Gilead, Novartis, AstraZeneca, Roche, NIH, and ASCERTAIN, all payments were made to the institute, outside the submitted work. M.P. has received research funding from MSD, AstraZeneca, Roche, Boehringer Ingelheim, and Takeda, outside the submitted work; consulting fees from Bristol-Meyers, Roche, MSD, AstraZeneca, Takeda, Eli Lilly, F Hoffman-La Roche, Janssen, Pfizer, and Takeda, outside the submitted work; honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Bristol-Meyers, Roche, MSD, AstraZeneca, Takeda, Eli Lilly, F Hoffman-La Roche, Janssen, and Pfizer, outside the submitted work; and support for attending meetings and/or travel from AstraZeneca, Boehringer Ingelheim, Bristol-Meyers Eli Lilly, F Hoffman-La Roche, Phierre Fabre Pharmaceuticals, and Takeda, outside the submitted work. A.L. has received grants for academic research from PharMamar, Beigene, Roche, AstraZeneca, and Amgen, outside the submitted work. R.D. has received payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Roche, AstraZeneca, Takeda, Novartis, BMS, MSD, Pfizer, and Amgen, outside the submitted work; support for attending meetings and/or travel from Pfizer, outside the submitted work; drug samples from Novartis, outside the submitted work; and participated on a Data Safety Monitoring Board or Advisory Board of GlaxoSmithKline, outside the submitted work. C.P. has received payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from AstraZeneca, outside the submitted work. M.D.M. has received institutional research funding from Tesaro/GlaxoSmithKline and institutional funding for work in clinical trials/contracted research from Beigene, Exelixis, MSD, Pfizer and Roche, outside the submitted work; and personal fees for consultancy or participation to advisory boards from AstraZeneca, Boehringer Ingelheim, Janssen, Merck Sharp & Dohme (MSD), Novartis, Pfizer, Roche, GlaxoSmithKline, Amgen, Merck, and Takeda, outside the submitted work. M.T. has received institutional research funding from AstraZeneca, BMS, MSD, Roche, and Takeda, outside the submitted work; payment or honoraria (personal) for speakers bureaus from Amgen, AstraZeneca, Beigene, BMS, Boehringer Ingelheim, Celgene, Chugai, Daiichi Sankyo, GlaxoSmithKline, Janssen Oncology, Lilly, MSD, Novartis, Pfizer, Roche, Sanofi, and Takeda, outside the submitted work; and support for attending meetings and/or travel from AstraZeneca, BMS, Boehringer Ingelheim, Daiichi Sankyo, Janssen Oncology, Lilly, Merck, MSD, Novartis, Pfizer, Roche, Sanofi, and Takeda, outside the submitted work. A.B. has received consulting fees (personal) from AstraZeneca, BMS, MSD, and Roche, outside the submitted work; and payment or honoraria (personal) for lectures, presentations, speakers bureaus, manuscript writing or educational events from AstraZeneca, BMS, MSD, and Roche, outside the submitted work. S.P. has received consulting fees (personal) from Amgen, AstraZeneca, Bayer, Blueprint, BMS, Boehringer Ingelheim, Daiichi Sankyo, GSK, Guardant Health, Incyte, Janssen, Lilly, Merck Serono, MSD, Novartis, Roche, Takeda, Pfizer, Seattle Genetics, Turning Point Therapeutics, and EQRx, outside the submitted work; payment or honoraria (personal) for lectures, presentations, speakers bureaus, manuscript writing or educational events from AstraZeneca, Bayer, Guardant Health, Janssen, Merck Serono, Roche and Takeda, outside the submitted work; payment for expert testimony from Roche and Merck Serono, outside the submitted work; support for travel from Janssen and Roche, outside the submitted work; consulting fees for participation on an Advisory Board, outside the submitted work; unpaid leadership role in the British Thoracic Oncology Group, ALK Positive UK, Lung Cancer Europe, Ruth Strauss Foundation, Mesothelioma Applied Research Foundation, and ETOP-IBCSG Partners Foundation Board, outside the submitted work. S.S. has received research grants (institution) from AstraZeneca and personal honoraria for participating in the trial steering committee for immunotherapy in small cell lung cancer from AstraZeneca, outside the submitted work; consulting fees from AstraZeneca and BMS, outside the submitted work; speaker honoraria (self) from AstraZeneca, outside the submitted work; participates on a Data Safety Monitoring Board as a review panel member for lung toxicity adjudication with immunotherapy (MSD), outside the submitted work; and has a leadership role as an ETOP member of the scientific committee for lung cancer, outside the submitted work., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
141. Challenges and controversies in resectable non-small cell lung cancer: a clinician's perspective.
- Author
-
Houda I, Dickhoff C, Uyl-de Groot CA, Damhuis RAM, Reguart N, Provencio M, Levy A, Dziadziuszko R, Pompili C, Di Maio M, Thomas M, Brunelli A, Popat S, Senan S, and Bahce I
- Abstract
The treatment landscape of resectable early-stage non-small cell lung cancer (NSCLC) is transforming due to the approval of novel adjuvant and neoadjuvant systemic treatments. The European Medicines Agency (EMA) recently approved adjuvant osimertinib, adjuvant atezolizumab, adjuvant pembrolizumab, and neoadjuvant nivolumab combined with chemotherapy, and the approval of other agents or new indications may follow soon. Despite encouraging results, many unaddressed questions remain. Moreover, the transformed treatment paradigm in resectable NSCLC can pose major challenges to healthcare systems and magnify existing disparities in care as differences in reimbursement may vary across different European countries. This Viewpoint discusses the challenges and controversies in resectable early-stage NSCLC and how existing inequalities in access to these treatments could be addressed., Competing Interests: I.H., C.D., R.A.M.D., N.R., and I.B. declare no competing interests. C.A.G. has received grants or contracts from Boehringer Ingelheim, Astellas, Celgene, Sanofi, Janssen-Cilag, Bayer, Amgen, Genzyme, Merck, Gilead, Novartis, AstraZeneca, Roche, NIH, and ASCERTAIN, all payments were made to the institute, outside the submitted work. M.P. has received research funding from MSD, AstraZeneca, Roche, Boehringer Ingelheim, and Takeda, outside the submitted work; consulting fees from Bristol-Meyers, Roche, MSD, AstraZeneca, Takeda, Eli Lilly, F Hoffman-La Roche, Janssen, Pfizer, and Takeda, outside the submitted work; honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Bristol-Meyers, Roche, MSD, AstraZeneca, Takeda, Eli Lilly, F Hoffman-La Roche, Janssen, and Pfizer, outside the submitted work; and support for attending meetings and/or travel from AstraZeneca, Boehringer Ingelheim, Bristol-Meyers Eli Lilly, F Hoffman-La Roche, Phierre Fabre Pharmaceuticals, and Takeda, outside the submitted work. A.L. has received grants for academic research from PharMamar, Beigene, Roche, AstraZeneca, and Amgen, outside the submitted work. R.D. has received payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Roche, AstraZeneca, Takeda, Novartis, BMS, MSD, Pfizer, and Amgen, outside the submitted work; support for attending meetings and/or travel from Pfizer, outside the submitted work; drug samples from Novartis, outside the submitted work; and participated on a Data Safety Monitoring Board or Advisory Board of GlaxoSmithKline, outside the submitted work. C.P. has received payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from AstraZeneca, outside the submitted work. M.D.M. has received institutional research funding from Tesaro/GlaxoSmithKline and institutional funding for work in clinical trials/contracted research from Beigene, Exelixis, MSD, Pfizer and Roche, outside the submitted work; and personal fees for consultancy or participation to advisory boards from AstraZeneca, Boehringer Ingelheim, Janssen, Merck Sharp & Dohme (MSD), Novartis, Pfizer, Roche, GlaxoSmithKline, Amgen, Merck, and Takeda, outside the submitted work. M.T. has received institutional research funding from AstraZeneca, BMS, MSD, Roche, and Takeda, outside the submitted work; payment or honoraria (personal) for speakers bureaus from Amgen, AstraZeneca, Beigene, BMS, Boehringer Ingelheim, Celgene, Chugai, Daiichi Sankyo, GlaxoSmithKline, Janssen Oncology, Lilly, MSD, Novartis, Pfizer, Roche, Sanofi, and Takeda, outside the submitted work; and support for attending meetings and/or travel from AstraZeneca, BMS, Boehringer Ingelheim, Daiichi Sankyo, Janssen Oncology, Lilly, Merck, MSD, Novartis, Pfizer, Roche, Sanofi, and Takeda, outside the submitted work. A.B. has received consulting fees (personal) from AstraZeneca, BMS, MSD, and Roche, outside the submitted work; and payment or honoraria (personal) for lectures, presentations, speakers bureaus, manuscript writing or educational events from AstraZeneca, BMS, MSD, and Roche, outside the submitted work. S.P. has received consulting fees (personal) from Amgen, AstraZeneca, Bayer, Blueprint, BMS, Boehringer Ingelheim, Daiichi Sankyo, GSK, Guardant Health, Incyte, Janssen, Lilly, Merck Serono, MSD, Novartis, Roche, Takeda, Pfizer, Seattle Genetics, Turning Point Therapeutics, and EQRx, outside the submitted work; payment or honoraria (personal) for lectures, presentations, speakers bureaus, manuscript writing or educational events from AstraZeneca, Bayer, Guardant Health, Janssen, Merck Serono, Roche and Takeda, outside the submitted work; payment for expert testimony from Roche and Merck Serono, outside the submitted work; support for travel from Janssen and Roche, outside the submitted work; consulting fees for participation on an Advisory Board, outside the submitted work; unpaid leadership role in the British Thoracic Oncology Group, ALK Positive UK, Lung Cancer Europe, Ruth Strauss Foundation, Mesothelioma Applied Research Foundation, and ETOP-IBCSG Partners Foundation Board, outside the submitted work. S.S. has received research grants (institution) from AstraZeneca and personal honoraria for participating in the trial steering committee for immunotherapy in small cell lung cancer from AstraZeneca, outside the submitted work; consulting fees from AstraZeneca and BMS, outside the submitted work; speaker honoraria (self) from AstraZeneca, outside the submitted work; participates on a Data Safety Monitoring Board as a review panel member for lung toxicity adjudication with immunotherapy (MSD), outside the submitted work; and has a leadership role as an ETOP member of the scientific committee for lung cancer, outside the submitted work., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
142. How to obtain the image-derived blood concentration from 89 Zr-immuno-PET scans.
- Author
-
Wijngaarden JE, Ahbari A, Pouw JEE, Greuter HNJM, Bahce I, Zwezerijnen GJC, Vugts DJ, van Dongen GAMS, Boellaard R, Menke-van der Houven van Oordt CW, and Huisman MC
- Abstract
Background: PET scans using zirconium-89 labelled monoclonal antibodies (
89 Zr-mAbs), known as89 Zr-immuno-PET, are made to measure uptake in tumour and organ tissue. Uptake is related to the supply of89 Zr-mAbs in the blood. Measuring activity concentrations in blood, however, requires invasive blood sampling. This study aims to identify the best delineation strategy to obtain the image-derived blood concentration (IDBC) from89 Zr-immuno-PET scans., Methods: PET imaging and blood sampling of two89 Zr-mAbs were included,89 Zr-cetuximab and89 Zr-durvalumab. For seven patients receiving89 Zr-cetuximab, PET scans on 1-2 h, 2 and 6 days post-injection (p.i.) were analysed. Five patients received three injections of89 Zr-durvalumab. The scanning protocol for the first two injections consisted of PET scanning on 2, 5 and 7 days p.i. and for the third injection only on 7 days p.i. Blood samples were drawn with every PET scan and the sample-derived blood concentration (SDBC) was used as gold standard for the IDBC. According to an in-house developed standard operating procedure, the aortic arch, ascending aorta, descending aorta and left ventricle were delineated. Bland-Altman analyses were performed to assess the bias (mean difference) and variability (1.96 times the standard deviation of the differences) between IDBC and SDBC., Results: Overall, the activity concentration obtained from the IDBC was lower than from the SDBC. When comparing IDBC with SDBC, variability was smallest for the ascending aorta (20.3% and 17.0% for89 Zr-cetuximab and89 Zr-durvalumab, respectively). Variability for the other regions ranged between 17.9 and 30.8%. Bias for the ascending aorta was - 10.9% and - 11.4% for89 Zr-cetuximab and89 Zr-durvalumab, respectively., Conclusions: Image-derived blood concentrations should be obtained from delineating the ascending aorta in89 Zr-immuno-PET scans, as this results in the lowest variability with respect to sample-derived blood concentrations., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
143. First exploration of the on-treatment changes in tumor and organ uptake of a radiolabeled anti PD-L1 antibody during chemoradiotherapy in patients with non-small cell lung cancer using whole body PET.
- Author
-
Pouw JEE, Hashemi SMS, Huisman MC, Wijngaarden JE, Slebe M, Oprea-Lager DE, Zwezerijnen GJC, Vugts D, Ulas EB, de Gruijl TD, Radonic T, Senan S, Menke-van der Houven van Oordt CW, and Bahce I
- Subjects
- Humans, B7-H1 Antigen metabolism, Positron-Emission Tomography methods, Chemoradiotherapy, Carcinoma, Non-Small-Cell Lung diagnostic imaging, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy
- Abstract
Background: In patients with locally advanced unresectable non-small cell lung cancer (NSCLC), durvalumab, an anti-programmed cell death ligand-1 (PD-L1) antibody, has shown improved overall survival when used as consolidation therapy following concurrent chemoradiotherapy (CRT). However, it is unclear whether CRT itself upregulates PD-L1 expression. Therefore, this study aimed to explore the changes in the uptake of the anti PD-L1 antibody [
89 Zr]Zr-durvalumab in tumors and healthy organs during CRT in patients with NSCLC., Methods: Patients with NSCLC scheduled to undergo CRT were scanned 7±1 days after administration of 37±1 MBq [89 Zr]Zr-durvalumab at baseline, 1-week on-treatment and 1 week after finishing 6 weeks of CRT. First, [89 Zr]Zr-durvalumab uptake was visually assessed in a low dose cohort with a mass dose of 2 mg durvalumab (0.13% of therapeutic dose) and subsequently, quantification was done in a high dose cohort with a mass dose of 22.5 mg durvalumab (1.5% of therapeutic dose). Tracer pharmacokinetics between injections were compared using venous blood samples drawn in the 22.5 mg cohort. Visual assessment included suspected lesion detectability. Positron emission tomography (PET) uptake in tumoral and healthy tissues was quantified using tumor to plasma ratio (TPR) and organ to plasma ratio, respectively., Results: In the 2 mg dose cohort, 88% of the 17 identified tumor lesions were positive at baseline, compared with 69% (9/13) for the 22.5 mg cohort. Although the absolute plasma concentrations between patients varied, the intrapatient variability was low. The ten quantitatively assessed lesions in the 22.5 mg cohort had a median TPR at baseline of 1.3 (IQR 0.7-1.5), on-treatment of 1.0 (IQR 0.7-1.4) and at the end of treatment of 0.7 (IQR 0.6-0.7). On-treatment, an increased uptake in bone marrow was seen in three out of five patients together with a decreased uptake in the spleen in four out of five patients., Conclusions: This study successfully imaged patients with NSCLC with [89 Zr]Zr-durvalumab PET before and during CRT. Our data did not show any increase in [89 Zr]Zr-durvalumab uptake in the tumor 1-week on-treatment and at the end of treatment. The changes observed in bone marrow and spleen may be due to an CRT-induced effect on immune cells., Trial Registration Number: EudraCT number: 2019-004284-51., Competing Interests: Competing interests: SMSH is on the advisory board and/or received institutional research support from AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Janssen, GlaxoSmithKline, Merck Sharp & Dohme, Novartis, Roche and Takeda. DV received institutional research support from Bristol-Myers Squibb, Daiichi Sankyo, GlaxoSmithKline, Boehringer Ingelheim, Roche and Aplagon. SS is on the advisory board of AstraZeneca, receives institutional research support and a speakers fee from AstraZeneca. SS is on the advisory board of Bristol-Myers Squibb and receives institutional research support from Bristol-Myers Squibb. CWM-vdHvO is advisor for GEHealthcare and Eli Lilly and received institutional research support from GlaxoSmithKline, Boehringer Ingelheim, AstraZeneca, G1 Therapeutics and Bristol-Myers Squibb., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
144. Multi-modal cell-free DNA genomic and fragmentomic patterns enhance cancer survival and recurrence analysis.
- Author
-
Moldovan N, van der Pol Y, van den Ende T, Boers D, Verkuijlen S, Creemers A, Ramaker J, Vu T, Bootsma S, Lenos KJ, Vermeulen L, Fransen MF, Pegtel M, Bahce I, van Laarhoven H, and Mouliere F
- Subjects
- Humans, Biomarkers, Tumor genetics, Genomics, Liquid Biopsy, ROC Curve, Cell-Free Nucleic Acids genetics, Neoplasms
- Abstract
The structure of cell-free DNA (cfDNA) is altered in the blood of patients with cancer. From whole-genome sequencing, we retrieve the cfDNA fragment-end composition using a new software (FrEIA [fragment end integrated analysis]), as well as the cfDNA size and tumor fraction in three independent cohorts (n = 925 cancer from >10 types and 321 control samples). At 95% specificity, we detect 72% cancer samples using at least one cfDNA measure, including 64% early-stage cancer (n = 220). cfDNA detection correlates with a shorter overall (p = 0.0086) and recurrence-free (p = 0.017) survival in patients with resectable esophageal adenocarcinoma. Integrating cfDNA measures with machine learning in an independent test set (n = 396 cancer, 90 controls) achieve a detection accuracy of 82% and area under the receiver operating characteristic curve of 0.96. In conclusion, harnessing the biological features of cfDNA can improve, at no extra cost, the diagnostic performance of liquid biopsies., Competing Interests: Declaration of interests F.M. is co-inventor on multiple patents related to cfDNA analysis. Other co-authors have no relevant conflict of interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
145. Real-time analysis of the cancer genome and fragmentome from plasma and urine cell-free DNA using nanopore sequencing.
- Author
-
van der Pol Y, Tantyo NA, Evander N, Hentschel AE, Wever BM, Ramaker J, Bootsma S, Fransen MF, Lenos KJ, Vermeulen L, Schneiders FL, Bahce I, Nieuwenhuijzen JA, Steenbergen RD, Pegtel DM, Moldovan N, and Mouliere F
- Subjects
- Humans, Genomics methods, Sequence Analysis, DNA, DNA genetics, Biomarkers, Tumor genetics, Cell-Free Nucleic Acids genetics, Nanopore Sequencing, Lung Neoplasms diagnosis, Lung Neoplasms genetics
- Abstract
Cell-free DNA (cfDNA) can be isolated and sequenced from blood and/or urine of cancer patients. Conventional short-read sequencing lacks deployability and speed and can be biased for short cfDNA fragments. Here, we demonstrate that with Oxford Nanopore Technologies (ONT) sequencing we can achieve delivery of genomic and fragmentomic data from liquid biopsies. Copy number aberrations and cfDNA fragmentation patterns can be determined in less than 24 h from sample collection. The tumor-derived cfDNA fraction calculated from plasma of lung cancer patients and urine of bladder cancer patients was highly correlated (R = 0.98) with the tumor fraction calculated from short-read sequencing of the same samples. cfDNA size profile, fragmentation patterns, fragment-end composition, and nucleosome profiling near transcription start sites in plasma and urine exhibited the typical cfDNA features. Additionally, a high proportion of long tumor-derived cfDNA fragments (> 300 bp) are recovered in plasma and urine using ONT sequencing. ONT sequencing is a cost-effective, fast, and deployable approach for obtaining genomic and fragmentomic results from liquid biopsies, allowing the analysis of previously understudied cfDNA populations., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
146. The landscape of cell-free mitochondrial DNA in liquid biopsy for cancer detection.
- Author
-
van der Pol Y, Moldovan N, Ramaker J, Bootsma S, Lenos KJ, Vermeulen L, Sandhu S, Bahce I, Pegtel DM, Wong SQ, Dawson SJ, Chandrananda D, and Mouliere F
- Subjects
- Male, Humans, Liquid Biopsy, Mitochondria genetics, DNA, Mitochondrial genetics, Biomarkers, Tumor genetics, Cell-Free Nucleic Acids, Prostatic Neoplasms genetics
- Abstract
Background: Existing methods to detect tumor signal in liquid biopsy have focused on the analysis of nuclear cell-free DNA (cfDNA). However, non-nuclear cfDNA and in particular mitochondrial DNA (mtDNA) has been understudied. We hypothesize that an increase in mtDNA in plasma could reflect the presence of cancer, and that leveraging cell-free mtDNA could enhance cancer detection., Results: We survey 203 healthy and 664 cancer plasma samples from three collection centers covering 12 cancer types with whole genome sequencing to catalogue the plasma mtDNA fraction. The mtDNA fraction is increased in individuals with cholangiocarcinoma, colorectal, liver, pancreatic, or prostate cancer, in comparison to that in healthy individuals. We detect almost no increase of mtDNA fraction in individuals with other cancer types. The mtDNA fraction in plasma correlates with the cfDNA tumor fraction as determined by somatic mutations and/or copy number aberrations. However, the mtDNA fraction is also elevated in a fraction of patients without an apparent increase in tumor-derived cfDNA. A predictive model integrating mtDNA and copy number analysis increases the area under the curve (AUC) from 0.73 when using copy number alterations alone to an AUC of 0.81., Conclusions: The mtDNA signal retrieved by whole genome sequencing has the potential to boost the detection of cancer when combined with other tumor-derived signals in liquid biopsies., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
147. Superior Sulcus Tumors Invading the Spine: Multimodal Treatment Outcomes From the Preimmunotherapy Era.
- Author
-
Unal S, Feller R, Stadhouder A, Heineman DJ, Jiya TU, van Dorp M, Bahce I, Braun J, Senan S, Dahele M, and Dickhoff C
- Abstract
Introduction: Curative-intent treatment of superior sulcus tumors (SSTs) of the lung invading the spine presents considerable challenges. We retrospectively studied outcomes in a single center, uniformly staged patient cohort treated with induction concurrent chemoradiotherapy followed by surgical resection (trimodality therapy)., Methods: An institutional surgical database from the period between 2002 and 2021 was accessed to identify SSTs in which the resection included removal of at least part of the vertebral body. All patients were staged using fluorodeoxyglucose positron emission tomography (/computed tomography), computed tomography scan of the chest/upper abdomen, and brain imaging. Surgical morbidity was assessed using the Clavien-Dindo classification. Overall and disease-free survival were calculated using the Kaplan-Meier method., Results: A total of 18 patients were included: 8 complete and 10 partial vertebrectomies were performed, with six of the eight complete vertebrectomies involving two vertebral levels, resulting in Complete surgical resection (R0) in 94%. Nine patients had a 1-day procedure, and nine were staged over 2 days. The median follow-up was 30 months (interquartile range 11-57). The 90-day postoperative morbidity was 44% (grade III/IV), with no 90-day surgery-related mortality. There were 83% who had a major pathologic response, associated with improved survival ( p = 0.044). The 5-year overall and disease-free survival were 55% and 40%, respectively. Disease progression occurred in 10 patients, comprising locoregional recurrences in two and distant metastases in eight patients., Conclusions: Multimodality treatment in selected patients with a superior sulcus tumor invading the spine is safe and results in good survival. Such patients should be referred to expert centers. Future research should focus on improving distant control (e.g. [neo]adjuvant immunotherapy)., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
148. Unresectable Stage III NSCLC Can Be Reevaluated for Resectability After Initial Treatment.
- Author
-
Dickhoff C, Heineman DJ, Bahce I, and Senan S
- Subjects
- Humans, Lung Neoplasms surgery, Carcinoma, Non-Small-Cell Lung surgery
- Published
- 2023
- Full Text
- View/download PDF
149. Neoadjuvant immunochemotherapy in resectable non-small cell lung cancer: the more cycles, the better?
- Author
-
Ulas EB and Bahce I
- Abstract
Competing Interests: Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-329/coif). IB reports that he participated in an advisory board of MSD. EBU has no conflicts of interest to declare.
- Published
- 2023
- Full Text
- View/download PDF
150. Tumor-educated platelet blood tests for Non-Small Cell Lung Cancer detection and management.
- Author
-
Antunes-Ferreira M, D'Ambrosi S, Arkani M, Post E, In 't Veld SGJG, Ramaker J, Zwaan K, Kucukguzel ED, Wedekind LE, Griffioen AW, Oude Egbrink M, Kuijpers MJE, van den Broek D, Noske DP, Hartemink KJ, Sabrkhany S, Bahce I, Sol N, Bogaard HJ, Koppers-Lalic D, Best MG, and Wurdinger T
- Subjects
- Humans, Biomarkers, Tumor genetics, Algorithms, RNA metabolism, Blood Platelets metabolism, Hematologic Tests, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms genetics
- Abstract
Liquid biopsy approaches offer a promising technology for early and minimally invasive cancer detection. Tumor-educated platelets (TEPs) have emerged as a promising liquid biopsy biosource for the detection of various cancer types. In this study, we processed and analyzed the TEPs collected from 466 Non-small Cell Lung Carcinoma (NSCLC) patients and 410 asymptomatic individuals (controls) using the previously established thromboSeq protocol. We developed a novel particle-swarm optimization machine learning algorithm which enabled the selection of an 881 RNA biomarker panel (AUC 0.88). Herein we propose and validate in an independent cohort of samples (n = 558) two approaches for blood samples testing: one with high sensitivity (95% NSCLC detected) and another with high specificity (94% controls detected). Our data explain how TEP-derived spliced RNAs may serve as a biomarker for minimally-invasive clinical blood tests, complement existing imaging tests, and assist the detection and management of lung cancer patients., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.