Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jonas Leichsenring is active.

Publication


Featured researches published by Jonas Leichsenring.


Genes, Chromosomes and Cancer | 2016

Pan-cancer analysis of copy number changes in programmed death-ligand 1 (PD-L1, CD274) - associations with gene expression, mutational load, and survival.

Jan Budczies; Michael Bockmayr; Carsten Denkert; Frederick Klauschen; Stefan Gröschel; Silvia Darb-Esfahani; Nicole Pfarr; Jonas Leichsenring; Maristela L. Onozato; Jochen K. Lennerz; Manfred Dietel; Stefan Fröhling; Peter Schirmacher; A. John Iafrate; Wilko Weichert; Albrecht Stenzinger

Inhibition of the PD‐L1 (CD274) – PD‐1 axis has emerged as a powerful cancer therapy that prevents evasion of tumor cells from the immune system. While immunohistochemical detection of PD‐L1 was introduced as a predictive biomarker with variable power, much less is known about copy number alterations (CNA) affecting PD‐L1 and their associations with expression levels, mutational load, and survival. To gain insight, we employed The Cancer Genome Atlas (TCGA) datasets to comprehensively analyze 22 major cancer types for PD‐L1 CNAs. We observed a diverse landscape of PD‐L1 CNAs, which affected focal regions, chromosome 9p or the entire chromosome 9. Deletions of PD‐L1 were more frequent than gains (31% vs. 12%) with deletions being most prevalent in melanoma and non‐small cell lung cancer. Copy number gains most frequently occurred in ovarian cancer, head and neck cancer, bladder cancer, cervical and endocervical cancer, sarcomas, and colorectal cancers. Fine‐mapping of the genetic architecture revealed specific recurrently amplified and deleted core regions across cancers with putative biological and clinical consequences. PD‐L1 CNAs correlated significantly with PD‐L1 mRNA expression changes in many cancer types, and tumors with PD‐L1 gains harbored significantly higher mutational load compared to non‐amplified cases (median: 78 non‐synonymous mutations vs. 40, P = 7.1e‐69). Moreover, we observed that, in general, both PD‐L1 amplifications and deletions were associated with dismal prognosis. In conclusion, PD‐L1 CNAs, in particular PD‐L1 copy number gains, represent frequent genetic alterations across many cancers, which influence PD‐L1 expression levels, are associated with higher mutational loads, and may be exploitable as predictive biomarker for immunotherapy regimens.


Genes, Chromosomes and Cancer | 2017

Tubular, lactating, and ductal adenomas are devoid of MED12 Exon2 mutations, and ductal adenomas show recurrent mutations in GNAS and the PI3K–AKT pathway

Anna-Lena Volckmar; Jonas Leichsenring; Christa Flechtenmacher; Nicole Pfarr; Udo Siebolts; Martina Kirchner; Jan Budczies; Michael Bockmayr; Kathrin Ridinger; K Lorenz; Esther Herpel; Aurelia Noske; Wilko Weichert; Frederick Klauschen; Peter Schirmacher; Roland Penzel; Volker Endris; Albrecht Stenzinger

Adenomas of the breast are rare benign tumors although single cases with malignant behavior have been reported. However, the genetic basis of these tumors is unknown. Employing targeted next generation sequencing of 50 cancer‐related genes as well as Sanger sequencing, we profiled a cohort of 18 mammary adenomas comprising 9 ductal, 6 tubular, and 3 lactating adenoma. Missense mutations were detected in 8 of the 18 cases (44%). Specifically, five (56%) ductal adenomas and three (50%) tubular adenomas harbored mutated genes. No mutations were detected in lactating adenomas. Three of the nine ductal adenomas showed mutant AKT1 (p.E17K) with two of them harboring an additional GNAS mutation (p.R201C). One case had mutant PIK3CA (p.H1047R) and another case a mutation in GNAS (p.R201C). The three cases of mutated tubular adenomas showed mutations in either MET or FGFR3. Of note, we did not detect copy number changes and none of the cases including tubular adenomas had mutations in exon 2 of MED12. Our results suggest that ductal adenomas are related to papillomas of the breast and screening for mutations in exon 2 of MED12 might help to facilitate differential diagnosis between tubular adenoma and fibroadenoma in difficult cases. Lastly, our data exemplarily demonstrate that mutations in cancer‐related genes per se do not indicate malignancy but occur in benign tumors.


Genes, Chromosomes and Cancer | 2017

Targeted next-generation sequencing enables reliable detection of HER2 (ERBB2) status in breast cancer and provides ancillary information of clinical relevance.

Nicole Pfarr; Roland Penzel; Volker Endris; Clemens Lier; Christa Flechtenmacher; Anna-Lena Volckmar; Martina Kirchner; Jan Budczies; Jonas Leichsenring; Esther Herpel; Aurelia Noske; Wilko Weichert; Andreas Schneeweiss; Peter Schirmacher; Hans-Peter Sinn; Albrecht Stenzinger

HER2‐positive breast cancers are a heterogeneous group of tumors, which share amplification and overexpression of HER2. In routine diagnostics, the HER2 (ERBB2) status is currently assessed by immunohistochemistry (IHC) and in situ hybridization (ISH). Data on targeted next‐generation sequencing (NGS) approaches that could be used to determine the HER2 status are sparse. Employing two breast cancer‐related gene panels, we performed targeted NGS of 41 FFPE breast cancers for which full pathological work‐up including ISH and IHC results was available. Selected cases were analyzed by qPCR. Of the 41 cases, the HER2 status of the 4 HER2‐positive and 6 HER2‐negative tumors was independently detected by our NGS approach achieving a concordance rate of 100%. The remaining 31 cases were equivocal HER2 cases by IHC of which 5 showed amplification of HER2 by ISH. Our NGS approach classified all non‐amplified cases correctly as HER2 negative and corroborated all but one of the 5 cases with amplified HER2 as detected by ISH. For the overall cohort, concordance between the gold standard and NGS was 97.6% (sensitivity 88.9% and specificity 100%). Additionally, we observed mutations in PIK3CA (44%), HER2 (8%), and CDH1 (6%) among others. Amplifications were found in CCND1 (12%), followed by MYC (10%) and EGFR (2%) and deletions in CDKN2A (10%), MAP2K4 and PIK3R1 (2% each). We here show that targeted NGS data can be used to interrogate the HER2 status with high specificity and high concordance with gold standard methods. Moreover, this approach identifies additional genetic events that may be clinically exploitable.


Genes, Chromosomes and Cancer | 2017

Mutational Profiles of Brenner Tumors show Distinctive Features Uncoupling Urothelial Carcinomas and Ovarian Carcinoma with Transitional Cell Histology

Nicole Pfarr; Silvia Darb-Esfahani; Jonas Leichsenring; Eliane Tabea Taube; Melanie Boxberg; Ioana Braicu; Moritz Jesinghaus; Roland Penzel; Volker Endris; Aurelia Noske; Wilko Weichert; Peter Schirmacher; Carsten Denkert; Albrecht Stenzinger

Brenner tumors (BT) are rare ovarian tumors encompassing benign, borderline, and malignant variants. While the histopathology of BTs and their clinical course is well described, little is known about the underlying genetic defects. We employed targeted next generation sequencing to analyze the mutational landscape in a cohort of 23 BT cases (17 benign, 2 borderline, and 4 malignant) and 3 ovarian carcinomas with transitional cell histology (TCC). Copy number variations (CNV) were validated by fluorescence in‐situ hybridization (FISH) and quantitative PCR‐based copy number assays. Additionally, we analyzed the TERT promotor region by conventional Sanger sequencing. We identified 25 different point mutations in 23 of the analyzed genes in BTs and 10 mutations in 8 genes in TCCs. About 57% percent of mutations occurred in genes involved in cell cycle control, DNA repair, and epigenetic regulation processes. All TCC cases harbored TP53 mutations whereas all BTs were negative and none of the mutations observed in BTs were present in TCCs. CNV analysis revealed recurrent MDM2 amplifications in 3 out of 4 of the malignant BT cases with one case harboring a concomitant amplification of CCND1. No mutations were observed in the TERT promoter region in BTs and TCCs, which is mutated in about 50%‐75% of urothelial carcinoma and in 16% of ovarian clear‐cell carcinomas. In conclusion, our study highlights distinct genetic features of BTs, and detection of the triplet phenotype MDM2 amplification/TP53 wt/TERT wt may aid diagnosis of malignant BT in difficult cases. Moreover, selected genetic lesions may be clinically exploitable in a metastatic setting.


Genes, Chromosomes and Cancer | 2018

Targeted deep sequencing of effusion cytology samples is feasible, informs spatiotemporal tumor evolution, and has clinical and diagnostic utility

Jonas Leichsenring; Anna-Lena Volckmar; Martina Kirchner; Daniel Kazdal; Mark Kriegsmann; Fabian Stögbauer; Teresa Bockmayr; Frederick Klauschen; Felix Herth; Roland Penzel; Arne Warth; Peter Schirmacher; Volker Endris; Albrecht Stenzinger

During the course of disease, many cancer patients eventually present with metastatic disease including peritoneal or pleural spread. In this context, cytology specimens derived from ascites or pleural effusion may help to differentiate malignant from benign conditions and sometimes yield diagnosis of a malignancy. However, even when supported by immunohistochemistry, cytological interpretation can be challenging, especially if tumor cellularity is low. Here, we investigated whether targeted deep sequencing of formalin‐fixed and paraffin embedded (FFPE) cytology specimens of cancer patients is feasible, and has diagnostic and clinical impact. To this end, a cohort of 20 matched pairs was compiled, each comprising a cytology sample (FFPE cell block) and at least one biopsy/surgical resection specimen serving as benchmark. In addition, 5 non‐malignant effusions were sequenced serving as negative‐controls. All samples yielded sufficient libraries and were successfully subjected to targeted sequencing employing a semiconductor based next‐generation sequencing platform. Using gene panels of different size and composition, including the Oncomine Comprehensive Assay, for targeted sequencing, somatic mutations were detected in the tissue of all 20 cases. Of these, 15 (75%) harbored mutations that were also detected in the corresponding cytology samples. In four of these cases (20%), additional private mutations were detected in either cytology or tissue samples, reflecting spatiotemporal tumor evolution. Of the five remaining cases, three (15%) showed wild type alleles in cytology material whereas tumor tissue had mutations in interrogated genes. Two cases were discordant, showing different private mutations in the cytology and in the tissue sample, respectively. In summary, sequencing of cytology specimens (FFPE cell block) reflecting spatiotemporal tumor evolution is feasible and yields adjunct genetic information that may be exploitable for diagnostics and therapy.


Applied Immunohistochemistry & Molecular Morphology | 2017

Qualitative Comparison Between Carrier-based and Classical Tissue Microarrays

Katharina Lisenko; Jonas Leichsenring; Christiane Zgorzelski; Rémi Longuespée; Rita Casadonte; Alexander Harms; Daniel Kazdal; Albrecht Stenzinger; Arne Warth; Mark Kriegsmann

Tissue microarrays (TMAs) are commonly used in biomarker research. To enhance the efficacy of TMAs and to avoid floating or folding of tissue cores, various improvements such as the application of carriers and melting techniques have been proposed. Compared with classical TMAs (cTMAs), carrier-based TMAs (cbTMAs) have been shown to have several advantages including sample handling and sectioning. Up to now, little is known about the efficacy and quality of cbTMAs compared with cTMAs. Thus, we set out to compare both types systematically. We constructed 5 spleen-based TMAs and 5 cTMAs with 10×10 different tissue types each. The total number of available cores, the number of folded cores, and the total core area was measured and evaluated by digital pathology. About 2% of cores got lost due to floating in both, cbTMAs and cTMAs, respectively. The remaining cores showed significant differences with regard to core integrity as about 1% of cbTMA cores and 9% of cTMA cores were folded (P<0.01). Folding or rolling was associated with specific tissue types. The size of the cores was smaller and less variable in cbTMAs (0.86±0.06 mm2) compared with cTMAs (0.97±0.14 mm2). The application of cbTMAs is an easy, inexpensive, and effective way to improve TMA-based research.


Leukemia & Lymphoma | 2018

Oncogene-induced senescence: a potential breakpoint mechanism against malignant transformation in plasma cell disorders

Nicola Lehners; Elena Ellert; Jing Xu; Jens Hillengass; Jonas Leichsenring; Albrecht Stenzinger; Hartmut Goldschmidt; Mindaugas Andrulis; Marc S. Raab

Abstract Oncogene-induced senescence (OIS) is a cellular tumor-suppressive mechanism present in several premalignant conditions. Here, we analyze the possible impact of OIS on malignant transformation in plasma cell disorders. Tumor samples from 125 patients with different disease stages were analyzed immunohistochemically for expression of senescence markers. Protein expression of cyclin-dependent kinase inhibitor p21Cip1/Waf1 was significantly higher in smoldering multiple myeloma (SMM) compared to monoclonal gammopathy of undetermined significance (MGUS) (p = .02) or symptomatic multiple myeloma (MM) (p = .005). SMM plasma cells expressing p21Cip1/Waf1 were negative for Ki67, consistent with senescence. While p27Kip1 was highly expressed in healthy controls, MGUS and SMM, expression decreased significantly in MM (p = .02). SMM plasma cells displayed a mutually exclusive expression of p21Cip1/Waf1/p27Kip1 suggesting compensatory mechanisms of senescence. In conclusion, we found markers of cellular senescence differentially expressed in SMM compared to MGUS and MM supporting the hypothesis of OIS as a breakpoint mechanism against malignant transformation in plasma cell disorders.


International Journal of Cancer | 2018

Size Matters: Dissecting Key Parameters for Panel-Based Tumor Mutational Burden (TMB) Analysis: Panel-based TMB detection

Ivo Buchhalter; Eugen Rempel; Volker Endris; Michael Allgäuer; Olaf Neumann; Anna-Lena Volckmar; Martina Kirchner; Jonas Leichsenring; Amelie Lier; Moritz von Winterfeld; Roland Penzel; Petros Christopoulos; Michael Thomas; Stefan Fröhling; Peter Schirmacher; Jan Budczies; Albrecht Stenzinger

Tumor mutational burden (TMB) represents a new determinant of clinical benefit from immune checkpoint blockade that identifies responders independent of PD‐L1 expression levels and is currently being explored in clinical trials. Although TMB can be measured directly by comprehensive genomic approaches such as whole‐genome and exome sequencing, broad availability, short turnaround times, costs and amenability to formalin‐fixed and paraffin‐embedded tissue support the use of gene panel sequencing for approximating TMB in routine diagnostics. However, data on the parameters influencing panel‐based TMB estimation are limited. Here, we report an extensive in silico analysis of the TCGA data set that simulates various panel sizes and compositions. We demonstrate that panel size is a critical parameter that influences confidence intervals (CIs) and cutoff values as well as important test parameters including sensitivity, specificity, and positive predictive value. Moreover, we evaluate the Illumina TSO500 panel, which will be made available for TMB estimation, and propose dynamic, entity‐specific cutoff values based on current clinical trial data. Optimizing the cost–benefit ratio, our data suggest that panels between 1.5 and 3 Mbp are ideally suited to estimate TMB with small CIs, whereas smaller panels tend to deliver imprecise TMB estimates for low to moderate TMB (0–30 muts/Mbp), connected with insufficient separation of hypermutated tumors from non‐hypermutated tumors.


Genes, Chromosomes and Cancer | 2018

Next generation sequencing of the cellular and liquid fraction of pancreatic cyst fluid supports discrimination of IPMN from pseudocysts and reveals cases with multiple mutated driver clones: First findings from the prospective ZYSTEUS biomarker study

Anna-Lena Volckmar; Volker Endris; Matthias M. Gaida; Jonas Leichsenring; Fabian Stögbauer; Michael Allgäuer; Moritz von Winterfeld; Roland Penzel; Martina Kirchner; Regine Brandt; Olaf Neumann; Holger Sültmann; Peter Schirmacher; Jochen Rudi; Daniel Schmitz; Albrecht Stenzinger

Approximately half of all pancreatic cysts are neoplastic, mainly comprising intraductal papillary mucinous neoplasms (IPMN), which can progress to invasive carcinoma. Current Fukuoka guidelines have limited sensitivity and specificity in predicting progression of asymptomatic pancreatic cysts. We present first results of the prospective ZYSTEUS biomarker study investigating (i) whether detection of driver mutations in IPMN by liquid biopsy is technically feasible, (ii) which compartment of IPMN is most suitable for analysis, and (iii) implications for clinical diagnostics. Twenty‐two patients with clinical inclusion criteria were enrolled in ZYSTEUS. Fifteen cases underwent endoscopic ultrasound (EUS)‐guided fine‐needle aspiration and cytological diagnostics. Cellular and liquid fraction of the cysts of each case were separated and subjected to deep targeted next generation sequencing (NGS). Clinical parameters, imaging findings (EUS and MRI), and follow‐up data were collected continuously. All IPMN cases (n = 12) showed at least one mutation in either KRAS (n = 11) or GNAS (n = 4). Three cases showed both KRAS and GNAS mutations. Six cases harbored multiple KRAS/GNAS mutations. In the three cases with pseudocysts, no KRAS or GNAS mutations were detected. DNA yields were higher and showed higher mutation diversity in the cellular fraction. In conclusion, mutation detection in pancreatic cyst fluid is technically feasible with more robust results in the cellular than in the liquid fraction. Current results suggest that, together with imaging, targeted sequencing supports discrimination of IPMN from pseudocysts. The prospective design of ZYSTEUS will provide insight into diagnostic value of NGS in preoperative risk stratification. Our data provide evidence for an oligoclonal nature of IPMN.


Diagnostic Pathology | 2018

Expression of HMB45, MelanA and SOX10 is rare in non-small cell lung cancer

Mark Kriegsmann; Katharina Kriegsmann; Alexander Harms; Rémi Longuespée; Christiane Zgorzelski; Jonas Leichsenring; Thomas Muley; Hauke Winter; Daniel Kazdal; Benjamin Goeppert; Arne Warth

BackgroundNon-small cell lung cancer (NSCLC) and melanoma are frequent entities in routine diagnostics. Whereas the differential diagnosis is usually straight forward based on histomorphology, it can be challenging in poorly differentiated tumors as melanoma may mimic various histological patterns. Distinction of the two entities is of outmost importance as both are treated differently. HMB45 and MelanA are recommended immunohistological markers for melanoma in this scenario. SOX10 has been described as an additional marker for melanoma. However, comprehensive large-scale data about the expression of melanoma markers in NSCLC tumor tissue specimen are lacking so far.MethodsTherefore, we analyzed the expression of these markers in 1085 NSCLC tumor tissue samples. Tissue microarrays of NSCLC cases were immunohistochemically stained for HMB45, MelanA, and SOX10. Positivity of a marker was defined as ≥1% positive tumor cells.ResultsIn 1027 NSCLC tumor tissue samples all melanoma as well as conventional immunohistochemical markers for NSCLC could be evaluated. HMB45, MelanA, and SOX10 were positive in 1 (< 1%), 0 (0%) and 5 (< 1%) cases. The HMB45 positive case showed co-expression of SOX10 and was classified as large cell carcinoma. Three out of five SOX10 positive cases were SqCC and one case was an adenosquamous carcinoma.ConclusionsExpression of HMB45, MelanA and SOX10 is evident but exceedingly rare in NSCLC cases. Together with conventional immunomarkers a respective marker panel allows a clear-cut differential diagnosis even in poorly differentiated tumors.

Collaboration


Dive into the Jonas Leichsenring's collaboration.

Top Co-Authors

Avatar

Albrecht Stenzinger

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Peter Schirmacher

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Roland Penzel

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Volker Endris

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Anna-Lena Volckmar

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Martina Kirchner

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Arne Warth

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Jan Budczies

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Michael Allgäuer

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Olaf Neumann

University Hospital Heidelberg

View shared research outputs
Researchain Logo
Decentralizing Knowledge