62 research outputs found

    On-site Cytology for Development of Patient-Derived Three-dimensional Organoid Cultures - A Pilot Study

    Full text link
    BACKGROUND/AIM Development of patient-derived three-dimensional (3D) organoid cultures is an emerging technique in the field of precision oncology. We aimed to integrate on-site adequacy evaluation using cytology into the tumor organoid development workflow to ensure precise characterization and growth of these cultures. PATIENTS AND METHODS Cancer patients were consented to a Precision Medicine trial. Fresh tissue was procured for genomic analyses as well as organoid development. Fresh tissue destined for organoid development was evaluated by preparing on-site cytology smears to ensure that only lesional tissue would be submitted for further cell culture work. RESULTS Cytology preparations were made from 64 different tumor samples and evaluated prior to tissue submission for organoid development. In 53 (82.2%) of those tumor samples, the cytology preparation was diagnostic, thus providing adequate material for organoid development. CONCLUSION Characterizing the tissue prior to submission for organoid development ensures submission of lesional tissue only. Furthermore, it is a cost-effective method that can help document patient diagnosis. This can be of importance in biopsies, since the tissue submitted for organoid development cannot be retrieved for clinical diagnosis afterwards. Our findings in this pilot study led to the implementation of on-site cytological evaluation in the tumor organoid development workflow at the Englander Institute for Precision Medicine, NY, USA

    Expression of Prostate-Specific Membrane Antigen (PSMA) on Biopsies Is an Independent Risk Stratifier of Prostate Cancer Patients at Time of Initial Diagnosis

    Get PDF
    Background: Stratifying prostate cancer (PCa) patients into risk groups at time of initial diagnosis enabling a risk-adapted disease management is still a major clinical challenge. Existing studies evaluating the prognostic potential of PSMA (prostate-specific membrane antigen) for PCa were performed on radical prostatectomy specimens (RPE), i.e., decision making for disease management was already completed at time of sample analysis. Aim of our study was to assess the prognostic value of PSMA expression for PCa patients on biopsies at time of initial diagnosis.Methods: PSMA expression was assessed by immunohistochemistry on 294 prostate biopsies with corresponding RPE, 621 primary tumor foci from 242 RPE, 43 locally advanced or recurrent tumors, 34 lymph node metastases, 78 distant metastases and 52 benign prostatic samples. PSMA expression was correlated with clinico-pathologic features. Primary endpoint was recurrence free survival. Other clinicopathologic features included WHO/ISUP grade groups, PSA serum level, TNM-stage, and R-status. Chi-square test, ANOVA-analyses, Cox-regression, and log-rank tests were performed for statistical analyses.Results: High PSMA expression on both biopsy and RPE significantly associates with a higher risk of disease recurrence following curative surgery. The 5-year-recurrence free survival rates were 88.2, 74.2, 67.7 and 26.8% for patients exhibiting no, low, medium, or high PSMA expression on biopsy, respectively. High PSMA expression on biopsy was significant in multivariate analysis predicting a 4-fold increased risk of disease recurrence independently from established prognostic markers. PSMA significantly increases during PCa progression.Conclusion: PSMA is an independent prognostic marker on biopsies at time of initial diagnosis and can predict disease recurrence following curative therapy for PCa. Our study proposes the application of the routinely used IHC marker PSMA for outcome prediction and decision making in risk-adapted PCa management on biopsies at time of initial diagnosis

    Delta-like protein 3 expression in paired chemonaive and chemorelapsed small cell lung cancer samples

    Get PDF
    Rovalpituzumab tesirine (Rova-T), an antibody-drug conjugate directed against Delta-like protein 3 (DLL3), is under development for patients with small cell lung cancer (SCLC). DLL3 is expressed on the majority of SCLC samples. Because SCLC is rarely biopsied in the course of disease, data regarding DLL3 expression in relapses is not available. The aim of this study was to investigate the expression of DLL3 in chemorelapsed (but untreated with Rova-T) SCLC samples and compare the results with chemonaive counterparts. Two evaluation methods to assess DLL3 expression were explored. Additionally, we assessed if DLL3 expression of chemorelapsed and/or chemonaive samples has prognostic impact and if it correlates with other clinicopathological data. The study included 30 paired SCLC samples, which were stained with an anti DLL3 antibody. DLL3 expression was assessed using tumor proportion score (TPS) and H-score and was categorized as DLL3 low (TPS < 50%, H-score ≀ 150) and DLL3 high (TPS ≄ 50%, H-score > 150). Expression data were correlated with clinicopathological characteristics. Kaplan-Meier curves were used to illustrate overall survival (OS) depending on DLL3 expression in chemonaive and chemorelapsed samples, respectively, and depending on dynamics of expression during course of therapy. DLL3 was expressed in 86.6% chemonaive and 80% chemorelapsed SCLC samples without significant differences between the two groups. However, the extent of expression varied in a substantial proportion of pairs (36.6% with TPS, 43.3% with H-score), defined as a shift from low to high or high to low expression. TPS and H-score provided comparable results. There were no profound correlations with clinicopathological data. Survival analysis revealed a trend toward a more favorable OS in DLL low-expressing chemonaive SCLC (p = 0.57) and, in turn, in DLL3 high-expressing chemorelapsed SCLC (p = 0.42) as well as in SCLC demonstrating a shift from low to high expression (p = 0.56) without being statistically significant. This is the first study to investigate DLL3 expression in a large cohort of rare paired chemonaive-chemorelapsed SCLC specimens. Comparative analysis revealed that DLL3 expression was not stable during the course of therapy, suggesting therapy-based alterations. Unlike in chemonaive samples, a high DLL3 expression in chemorelapsed samples indicated a trend for a more favorable prognosis. Our results highlight the importance to investigate DLL3 in latest chemorelapsed SCLC tumor tissue

    Cell-free DNA profiling of metastatic prostate cancer reveals microsatellite instability, structural rearrangements and clonal hematopoiesis.

    Get PDF
    This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.BACKGROUND: There are multiple existing and emerging therapeutic avenues for metastatic prostate cancer, with a common denominator, which is the need for predictive biomarkers. Circulating tumor DNA (ctDNA) has the potential to cost-efficiently accelerate precision medicine trials to improve clinical efficacy and diminish costs and toxicity. However, comprehensive ctDNA profiling in metastatic prostate cancer to date has been limited. METHODS: A combination of targeted and low-pass whole genome sequencing was performed on plasma cell-free DNA and matched white blood cell germline DNA in 364 blood samples from 217 metastatic prostate cancer patients. RESULTS: ctDNA was detected in 85.9% of baseline samples, correlated to line of therapy and was mirrored by circulating tumor cell enumeration of synchronous blood samples. Comprehensive profiling of the androgen receptor (AR) revealed a continuous increase in the fraction of patients with intra-AR structural variation, from 15.4% during first-line metastatic castration-resistant prostate cancer therapy to 45.2% in fourth line, indicating a continuous evolution of AR during the course of the disease. Patients displayed frequent alterations in DNA repair deficiency genes (18.0%). Additionally, the microsatellite instability phenotype was identified in 3.81% of eligible samples (≄ 0.1 ctDNA fraction). Sequencing of non-repetitive intronic and exonic regions of PTEN, RB1, and TP53 detected biallelic inactivation in 47.5%, 20.3%, and 44.1% of samples with ≄ 0.2 ctDNA fraction, respectively. Only one patient carried a clonal high-impact variant without a detectable second hit. Intronic high-impact structural variation was twice as common as exonic mutations in PTEN and RB1. Finally, 14.6% of patients presented false positive variants due to clonal hematopoiesis, commonly ignored in commercially available assays. CONCLUSIONS: ctDNA profiles appear to mirror the genomic landscape of metastatic prostate cancer tissue and may cost-efficiently provide somatic information in clinical trials designed to identify predictive biomarkers. However, intronic sequencing of the interrogated tumor suppressors challenges the ubiquitous focus on coding regions and is vital, together with profiling of synchronous white blood cells, to minimize erroneous assignments which in turn may confound results and impede true associations in clinical trials.The Belgian Foundation Against Cancer (grant number C/2014/227); Kom op tegen Kanker (Stand up to Cancer), the Flemish Cancer Society (grant number 00000000116000000206); Royal College of Surgeons/Cancer Research UK (C19198/A1533); The Cancer Research Funds of Radiumhemmet, through the PCM program at KI (grant number 163012); The Erling-Persson family foundation (grant number 4-2689-2016); the Swedish Research Council (grant number K2010-70X-20430-04-3), and the Swedish Cancer Foundation (grant number 09-0677)

    Inhibition des Epidermal Growth Factor Rezeptors in chemisch induzierten prÀneoplastischen Leberherden und hepatozellulÀren Karzinomen der Ratte

    No full text
    Das hepatozellulĂ€re Karzinom stellt weltweit einen der am hĂ€ufigsten auftretenden malignen Tumoren mit limitierten therapeutischen Optionen dar. Ein besseres VerstĂ€ndnis der Tumorigenese durch tierexperimentell gewonnene Erkenntnisse kann zur Entwicklung zielgerichteter Medikamente beitragen. Ein langjĂ€hrig etabliertes Modell zur Erzeugung hepatozellulĂ€rer Karzinome im Tierexperiment ist die tĂ€gliche orale Gabe von N-Nitrosomorpholin (NNM). Die chemische Hepatokarzinogenese durch NNM fĂŒhrt abhĂ€ngig von Dosis und Dauer in der Rattenleber zur Entstehung von PrĂ€neoplasien, hepatozellulĂ€ren Adenomen und Karzinomen. Zu diesem Prozess trĂ€gt der Transforming Growth Factor alpha (TGF-alpha) ĂŒber die Aktivierung des transmembranĂ€ren Epidermal Growth Factor Rezeptors (EGFR) im Sinne einer Proliferationsaktivierung der Hepatozyten bei. In der vorliegenden Arbeit wurde die Methode der chemischen Hepatokarzinogenese in Kombination mit der oralen Gabe des Tyrosinkinaseinhibitors Gefitinib, der selektiv die intrazellulĂ€re Tyrosinkinase des EGFR blockiert und somit zu einer Unterbrechung der Signalkaskade in der Zelle fĂŒhrt, angewandt. Die Tiere erhielten NNM ĂŒber einen Zeitraum von entweder drei oder sechs Monaten. Es sollte ĂŒberprĂŒft werden, ob sich durch die parallele Applikation von Gefitinib ĂŒber zwei Wochen oder drei Monate die Entwicklung von PrĂ€neoplasien, Adenomen und Karzinomen beeinflussen lĂ€sst. Es entwickelten sich in allen ĂŒber drei Monate mit NNM behandelten Tieren klarzellige und auch gemischtzellige prĂ€neoplastische Leberherde. Die Gruppe, die gleichzeitig ĂŒber drei Monate Gefitinib erhielt, entwickelte statistisch signifikant weniger prĂ€neoplastische Leberherde. Die Tiere der ĂŒber sechs Monate mit NNM behandelten Gruppen wiesen neben klarzelligen und gemischtzelligen Herden auch basophile PrĂ€neoplasien sowie hepatozellulĂ€re Adenome und Karzinome auf. Die Anzahl der hepatozellulĂ€ren Adenome und Karzinome war in den Gruppen, die neben NNM ĂŒber sechs Monate auch ĂŒber drei Monate Gefitinib erhielten, statistisch signifikant geringer. Die ebenfalls statistisch signifikante geringere Proliferation in dieser Gruppe zeigt, dass sich die Progression einmal initiierter Herde verlangsamen lĂ€sst. Mit diesen Ergebnissen korrelierte bei den Gruppen beider ZeitrĂ€ume die Verminderung der immunhistochemischen Expression von EGFR und TGF-alpha der PrĂ€neoplasien. Gleichzeitig beleuchtet diese Arbeit, dass TGF-alpha und EGFR in der Entstehung frĂŒher prĂ€neoplastischer Herde sowie spĂ€terer Adenome und Karzinome eine wichtige Rolle spielen.The hepatocellular carcinoma is worldwide one of the most common malignant tumors with limited therapeutic options. Improved knowledge of hepatocarcinogenesis as gained in animal experiments can help to develop new targeted drugs. N-Nitrosomorpholine (NNM) administered daily orally leads to a well characterized sequence of preneoplastic lesions, hepatocellular adenoma and carcinoma in the rat liver. Among others, the Epidermal Growth Factor Receptor (EGFR) and its ligand Transforming Growth Factor alpha (TGF-alpha) is implicated in this process. Here we combined a method of chemical hepatocarcinogenesis in the rat with an inhibition of the intracellular tyrosinkinase of the EGFR by the drug Gefitinib. Rats were given NNM either for a period of three or six months. Gefitinib was concurrently administered either for two weeks or three months before sacrificing. We saw mostly clear cell preneoplastic lesion in the three months groups and in addition to that hepatocellular adenomas and carcinomas in the six months groups. The animals which received NNM and Gefinib for three months developed a lower portion of preneoplastic hepatic lesions as compared to the lesions that developed in the other two groups, which received NNM for three months. The animals, which received NNM for six months and concurrently three months gefitinib showed a lower portion of hepatocellular adenoma and fewer hepatocellular carcinoma. Those differences were statistically significant. Immunohistochemical staining revealed lower EGFR and TGF-&alpha expression in the preneoplastic hepatic lesions of the Gefitinib-treated animals. No differences were observed in the EGFR- and TGF-alpha-Expression of hepatocellular adenomas and carcinomas. These results show the efficacy of EGFR-Inhibition in preventing and delaying the development of preneoplastic lesions, hepatocellular adenoma and carcinoma in a model of rodent chemical hepatocarcinogenesis

    On-site Cytology for Development of Patient-Derived Three-dimensional Organoid Cultures - A Pilot Study

    Full text link
    BACKGROUND/AIM Development of patient-derived three-dimensional (3D) organoid cultures is an emerging technique in the field of precision oncology. We aimed to integrate on-site adequacy evaluation using cytology into the tumor organoid development workflow to ensure precise characterization and growth of these cultures. PATIENTS AND METHODS Cancer patients were consented to a Precision Medicine trial. Fresh tissue was procured for genomic analyses as well as organoid development. Fresh tissue destined for organoid development was evaluated by preparing on-site cytology smears to ensure that only lesional tissue would be submitted for further cell culture work. RESULTS Cytology preparations were made from 64 different tumor samples and evaluated prior to tissue submission for organoid development. In 53 (82.2%) of those tumor samples, the cytology preparation was diagnostic, thus providing adequate material for organoid development. CONCLUSION Characterizing the tissue prior to submission for organoid development ensures submission of lesional tissue only. Furthermore, it is a cost-effective method that can help document patient diagnosis. This can be of importance in biopsies, since the tissue submitted for organoid development cannot be retrieved for clinical diagnosis afterwards. Our findings in this pilot study led to the implementation of on-site cytological evaluation in the tumor organoid development workflow at the Englander Institute for Precision Medicine, NY, USA

    Clinical performance validation of PITX2 DNA methylation as prognostic biomarker in patients with head and neck squamous cell carcinoma.

    No full text
    Despite advances in combined modality therapy, outcomes in head and neck squamous cell cancer (HNSCC) remain dismal with five-year overall survival rates of less than 50%. Prognostic biomarkers are urgently needed to identify patients with a high risk of death after initial curative treatment. Methylation status of the paired-like homeodomain transcription factor 2 (PITX2) has recently emerged as a powerful prognostic biomarker in various cancers. In the present study, the clinical performance of PITX2 methylation was validated in a HNSCC cohort by means of an independent analytical platform (Infinium HumanMethylation450 BeadChip, Illumina, Inc.).A total of 528 HNSCC patients from The Cancer Genome Atlas (TCGA) were included in the study. Death was defined as primary endpoint. PITX2 methylation was correlated with overall survival and clinicopathological parameters.PITX2 methylation was significantly associated with sex, tumor site, p16 status, and grade. In univariate Cox proportional hazards analysis, PITX2 hypermethylation analyzed as continuous and dichotomized variable was significantly associated with prolonged overall survival of HNSCC patients (continuous: hazard ratio (HR) = 0.19 [95%CI: 0.04-0.88], p = 0.034; dichotomized: HR = 0.52 [95%CI: 0.33-0.84], p = 0.007). In multivariate Cox analysis including established clinicopathological parameters, PITX2 promoter methylation was confirmed as prognostic factor (HR = 0.28 [95%CI: 0.09-0.84], p = 0.023).Using an independent analytical platform, PITX2 methylation was validated as a prognostic biomarker in HNSCC patients, identifying patients that potentially benefit from intensified surveillance and/or administration of adjuvant/neodjuvant treatment, i.e. immunotherapy

    Improved PCR performance using template DNA from formalin-fixed and paraffin-embedded tissues by overcoming PCR inhibition.

    Get PDF
    Formalin-fixed and paraffin-embedded (FFPE) tissues represent a valuable source for biomarker studies and clinical routine diagnostics. However, they suffer from degradation of nucleic acids due to the fixation process. Since genetic and epigenetic studies usually require PCR amplification, this degradation hampers its use significantly, impairing PCR robustness or necessitating short amplicons. In routine laboratory medicine a highly robust PCR performance is mandatory for the clinical utility of genetic and epigenetic biomarkers. Therefore, methods to improve PCR performance using DNA from FFPE tissue are highly desired and of wider interest. The effect of template DNA derived from FFPE tissues on PCR performance was investigated by means of qPCR and conventional PCR using PCR fragments of different sizes. DNA fragmentation was analyzed via agarose gel electrophoresis. This study showed that poor PCR amplification was partly caused by inhibition of the DNA polymerase by fragmented DNA from FFPE tissue and not only due to the absence of intact template molecules of sufficient integrity. This PCR inhibition was successfully minimized by increasing the polymerase concentration, dNTP concentration and PCR elongation time thereby allowing for the robust amplification of larger amplicons. This was shown for genomic template DNA as well as for bisulfite-converted template DNA required for DNA methylation analyses. In conclusion, PCR using DNA from FFPE tissue suffers from inhibition which can be alleviated by adaptation of the PCR conditions, therefore allowing for a significant improvement of PCR performance with regard to variability and the generation of larger amplicons. The presented solutions to overcome this PCR inhibition are of tremendous value for clinical chemistry and laboratory medicine
    • 

    corecore