Christoph Heining
German Cancer Research Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Christoph Heining.
Cancer Discovery | 2013
Mindaugas Andrulis; Nicola Lehners; David Capper; Roland Penzel; Christoph Heining; Jennifer Huellein; Thorsten Zenz; Andreas von Deimling; Peter Schirmacher; Anthony D. Ho; Hartmut Goldschmidt; Kai Neben; Marc S. Raab
In multiple myeloma, there has been little progress in the specific therapeutic targeting of oncogenic mutations. Whole-genome sequencing data have recently revealed that a subset of patients carry an activating mutation (V600E) in the BRAF kinase. To uncover the clinical relevance of this mutation in multiple myeloma, we correlated the mutation status in primary tumor samples from 379 patients with myeloma with disease outcome. We found a significantly higher incidence of extramedullary disease and a shorter overall survival in mutation carriers when compared with controls. Most importantly, we report on a patient with confirmed BRAF V600E mutation and relapsed myeloma with extensive extramedullary disease, refractory to all approved therapeutic options, who has rapidly and durably responded to low doses of the mutation-specific BRAF inhibitor vermurafenib. Collectively, we provide evidence for the development of the BRAF V600E mutation in the context of clonal evolution and describe the prognostic and therapeutic relevance of this targetable mutation.
Leukemia | 2016
M Kordes; Michael Röring; Christoph Heining; Sandra Braun; Barbara Hutter; Daniela Richter; Christina Geörg; Claudia Scholl; Stefan Gröschel; W Roth; Andreas Rosenwald; E Geissinger; C. Von Kalle; Dirk Jäger; Benedikt Brors; Wilko Weichert; C. Grüllich; Hanno Glimm; Tilman Brummer; Stefan Fröhling
Activating BRAF mutations, in particular V600E/K, drive many cancers and are considered mutually exclusive with mutant RAS, whereas inactivating BRAF mutations in the D594F595G596 motif cooperate with RAS via paradoxical MEK/ERK activation. Due to the increasing use of comprehensive tumor genomic profiling, many non-V600 BRAF mutations are being detected whose functional consequences and therapeutic actionability are often unknown. We investigated an atypical BRAF mutation, F595L, which was identified along with mutant HRAS in histiocytic sarcoma and also occurs in epithelial cancers, melanoma and neuroblastoma, and determined its interaction with mutant RAS. Unlike other DFG motif mutants, BRAFF595L is a gain-of-function variant with intermediate activity that does not act paradoxically, but nevertheless cooperates with mutant RAS to promote oncogenic signaling, which is efficiently blocked by pan-RAF and MEK inhibitors. Mutation data from patients and cell lines show that BRAFF595L, as well as other intermediate-activity BRAF mutations, frequently coincide with mutant RAS in various cancers. These data define a distinct class of activating BRAF mutations, extend the spectrum of patients with systemic histiocytoses and other malignancies who are candidates for therapeutic blockade of the RAF-MEK-ERK pathway and underscore the value of comprehensive genomic testing for uncovering the vulnerabilities of individual tumors.
Cold Spring Harb Mol Case Stud | 2016
Stefan Gröschel; Martin Bommer; Barbara Hutter; Jan Budczies; David Bonekamp; Christoph Heining; Peter Horak; Martina Fröhlich; Sebastian Uhrig; Daniel Hübschmann; Christina Geörg; Daniela Richter; Nicole Pfarr; Katrin Pfütze; Stephan Wolf; Peter Schirmacher; Dirk Jäger; Christof von Kalle; Benedikt Brors; Hanno Glimm; Wilko Weichert; Albrecht Stenzinger; Stefan Fröhling
Identification of the tissue of origin in cancer of unknown primary (CUP) poses a diagnostic challenge and is critical for directing site-specific therapy. Currently, clinical decision-making in patients with CUP primarily relies on histopathology and clinical features. Comprehensive molecular profiling has the potential to contribute to diagnostic categorization and, most importantly, guide CUP therapy through identification of actionable lesions. We here report the case of an advanced-stage malignancy initially mimicking poorly differentiated soft-tissue sarcoma that did not respond to multiagent chemotherapy. Molecular profiling within a clinical whole-exome and transcriptome sequencing program revealed a heterozygous, highly amplified KRAS G12S mutation, compound-heterozygous TP53 mutation/deletion, high mutational load, and focal high-level amplification of Chromosomes 9p (including PDL1 [CD274] and JAK2) and 10p (including GATA3). Integrated analysis of molecular data and histopathology provided a rationale for immune checkpoint inhibitor (ICI) therapy with pembrolizumab, which resulted in rapid clinical improvement and a lasting partial remission. Histopathological analyses ruled out sarcoma and established the diagnosis of a poorly differentiated adenocarcinoma. Although neither histopathology nor molecular data were able to pinpoint the tissue of origin, our analyses established several differential diagnoses including triple-negative breast cancer (TNBC). We analyzed 157 TNBC samples from The Cancer Genome Atlas, revealing PDL1 copy number gains coinciding with excessive PDL1 mRNA expression in 24% of cases. Collectively, these results illustrate the impact of multidimensional tumor profiling in cases with nondescript histology and immunophenotype, show the predictive potential of PDL1 amplification for immune checkpoint inhibitors (ICIs), and suggest a targeted therapeutic strategy in Chromosome 9p24.1/PDL1-amplified cancers.
International Journal of Cancer | 2017
Peter Horak; Barbara Klink; Christoph Heining; Stefan Gröschel; Barbara Hutter; Martina Fröhlich; Sebastian Uhrig; Daniel Hübschmann; Matthias Schlesner; Roland Eils; Daniela Richter; Katrin Pfütze; Christina Geörg; Bettina Meißburger; Stephan Wolf; Angela Schulz; Roland Penzel; Esther Herpel; Martina Kirchner; Amelie Lier; Volker Endris; Stephan Singer; Peter Schirmacher; Wilko Weichert; Albrecht Stenzinger; Richard F. Schlenk; Evelin Schröck; Benedikt Brors; Christof von Kalle; Hanno Glimm
Precision oncology implies the ability to predict which patients will likely respond to specific cancer therapies based on increasingly accurate, high‐resolution molecular diagnostics as well as the functional and mechanistic understanding of individual tumors. While molecular stratification of patients can be achieved through different means, a promising approach is next‐generation sequencing of tumor DNA and RNA, which can reveal genomic alterations that have immediate clinical implications. Furthermore, certain genetic alterations are shared across multiple histologic entities, raising the fundamental question of whether tumors should be treated by molecular profile and not tissue of origin. We here describe MASTER (Molecularly Aided Stratification for Tumor Eradication Research), a clinically applicable platform for prospective, biology‐driven stratification of younger adults with advanced‐stage cancer across all histologies and patients with rare tumors. We illustrate how a standardized workflow for selection and consenting of patients, sample processing, whole‐exome/genome and RNA sequencing, bioinformatic analysis, rigorous validation of potentially actionable findings, and data evaluation by a dedicated molecular tumor board enables categorization of patients into different intervention baskets and formulation of evidence‐based recommendations for clinical management. Critical next steps will be to increase the number of patients that can be offered comprehensive molecular analysis through collaborations and partnering, to explore ways in which additional technologies can aid in patient stratification and individualization of treatment, to stimulate clinically guided exploratory research projects, and to gradually move away from assessing the therapeutic activity of targeted interventions on a case‐by‐case basis toward controlled clinical trials of genomics‐guided treatments.
Annals of Oncology | 2016
Sebastian M. Dieter; Christoph Heining; Abbas Agaimy; Daniel Huebschmann; David Bonekamp; Barbara Hutter; K. R. Ehrenberg; Martina Fröhlich; Matthias Schlesner; Claudia Scholl; H. P. Schlemmer; Stephan Wolf; A. Mavratzas; C. S. Jung; Stefan Gröschel; C. Von Kalle; Roland Eils; Benedikt Brors; Roland Penzel; Mark Kriegsmann; D. E. Reuss; Peter Schirmacher; Albrecht Stenzinger; P. A. Federspil; Wilko Weichert; Hanno Glimm; Stefan Fröhling
Background Sinonasal carcinomas (SNCs) comprise various rare tumor types that are characterized by marked histologic diversity and largely unknown molecular profiles, yet share an overall poor prognosis owing to an aggressive clinical course and frequent late-stage diagnosis. The lack of effective systemic therapies for locally advanced or metastatic SNC poses a major challenge to therapeutic decision making for individual patients. We here aimed to identify actionable genetic alterations in a patient with metastatic SNC whose tumor, despite all diagnostic efforts, could not be assigned to any known SNC category and was refractory to multimodal therapy. Patients and methods We used whole-exome and transcriptome sequencing to identify a KIT exon 11 mutation (c.1733_1735del, p.D579del) as potentially druggable target in this patient and carried out cancer hotspot panel sequencing to detect secondary resistance-conferring mutations in KIT. Furthermore, as a step towards clinical exploitation of the recently described signatures of mutational processes in cancer genomes, we established and applied a novel bioinformatics algorithm that enables supervised analysis of the mutational catalogs of individual tumors. Results Molecularly guided treatment with imatinib in analogy to the management of gastrointestinal stromal tumor (GIST) resulted in a dramatic and durable response with remission of nearly all tumor manifestations, indicating a dominant driver function of mutant KIT in this tumor. KIT dependency was further validated by a secondary KIT exon 17 mutation (c.2459_2462delATTCinsG, p.D820_S821delinsG) that was detected upon tumor progression after 10 months of imatinib treatment and provided a rationale for salvage therapy with regorafenib, which has activity against KIT exon 11/17 mutant GIST. Conclusions These observations highlight the potential of unbiased genomic profiling for uncovering the vulnerabilities of individual malignancies, particularly in rare and unclassifiable tumors, and underscore that KIT exon 11 mutations represent tractable therapeutic targets across different histologies.BACKGROUND Sinonasal carcinomas (SNCs) comprise various rare tumor types that are characterized by marked histologic diversity and largely unknown molecular profiles, yet share an overall poor prognosis owing to an aggressive clinical course and frequent late-stage diagnosis. The lack of effective systemic therapies for locally advanced or metastatic SNC poses a major challenge to therapeutic decision making for individual patients. We here aimed to identify actionable genetic alterations in a patient with metastatic SNC whose tumor, despite all diagnostic efforts, could not be assigned to any known SNC category and was refractory to multimodal therapy. PATIENTS AND METHODS We used whole-exome and transcriptome sequencing to identify a KIT exon 11 mutation (c.1733_1735del, p.D579del) as potentially druggable target in this patient and carried out cancer hotspot panel sequencing to detect secondary resistance-conferring mutations in KIT. Furthermore, as a step towards clinical exploitation of the recently described signatures of mutational processes in cancer genomes, we established and applied a novel bioinformatics algorithm that enables supervised analysis of the mutational catalogs of individual tumors. RESULTS Molecularly guided treatment with imatinib in analogy to the management of gastrointestinal stromal tumor (GIST) resulted in a dramatic and durable response with remission of nearly all tumor manifestations, indicating a dominant driver function of mutant KIT in this tumor. KIT dependency was further validated by a secondary KIT exon 17 mutation (c.2459_2462delATTCinsG, p.D820_S821delinsG) that was detected upon tumor progression after 10 months of imatinib treatment and provided a rationale for salvage therapy with regorafenib, which has activity against KIT exon 11/17 mutant GIST. CONCLUSIONS These observations highlight the potential of unbiased genomic profiling for uncovering the vulnerabilities of individual malignancies, particularly in rare and unclassifiable tumors, and underscore that KIT exon 11 mutations represent tractable therapeutic targets across different histologies.
Clinical Cancer Research | 2017
Priya Chudasama; Marcus Renner; Melanie Straub; Sadaf S. Mughal; Barbara Hutter; Zeynep Kosaloglu; Ron Schweßinger; Matthias Scheffler; Ingo Alldinger; Simon Schimmack; Thorsten Persigehl; Carsten Kobe; Dirk Jäger; Christof von Kalle; Peter Schirmacher; Marie Beckhaus; Stephan Wolf; Christoph Heining; Stefan Gröschel; Jürgen Wolf; Benedikt Brors; Wilko Weichert; Hanno Glimm; Claudia Scholl; Gunhild Mechtersheimer; Katja Specht; Stefan Fröhling
Purpose: Altered FGFR1 signaling has emerged as a therapeutic target in epithelial malignancies. In contrast, the role of FGFR1 in soft-tissue sarcoma (STS) has not been established. Prompted by the detection and subsequent therapeutic inhibition of amplified FGFR1 in a patient with metastatic leiomyosarcoma, we investigated the oncogenic properties of FGFR1 and its potential as a drug target in patients with STS. Experimental Design: The frequency of FGFR1 amplification and overexpression, as assessed by FISH, microarray-based comparative genomic hybridization and mRNA expression profiling, SNP array profiling, and RNA sequencing, was determined in three patient cohorts. The sensitivity of STS cell lines with or without FGFR1 alterations to genetic and pharmacologic FGFR1 inhibition and the signaling pathways engaged by FGFR1 were investigated using viability assays, colony formation assays, and biochemical analysis. Results: Increased FGFR1 copy number was detected in 74 of 190 (38.9%; cohort 1), 13 of 79 (16.5%; cohort 2), and 80 of 254 (31.5%; cohort 3) patients. FGFR1 overexpression occurred in 16 of 79 (20.2%, cohort 2) and 39 of 254 (15.4%; cohort 3) patients. Targeting of FGFR1 by RNA interference and small-molecule inhibitors (PD173074, AZD4547, BGJ398) revealed that the requirement for FGFR1 signaling in STS cells is dictated by FGFR1 expression levels, and identified the MAPK–ERK1/2 axis as critical FGFR1 effector pathway. Conclusions: These data identify FGFR1 as a driver gene in multiple STS subtypes and support FGFR1 inhibition, guided by patient selection according to the FGFR1 expression and monitoring of MAPK–ERK1/2 signaling, as a therapeutic option in this challenging group of diseases. Clin Cancer Res; 23(4); 962–73. ©2016 AACR.
Genome Medicine | 2018
Júlia Perera-Bel; Barbara Hutter; Christoph Heining; Annalen Bleckmann; Martina Fröhlich; Stefan Fröhling; Hanno Glimm; Benedikt Brors; Tim Beißbarth
BackgroundA comprehensive understanding of cancer has been furthered with technological improvements and decreasing costs of next-generation sequencing (NGS). However, the complexity of interpreting genomic data is hindering the implementation of high-throughput technologies in the clinical context: increasing evidence on gene–drug interactions complicates the task of assigning clinical significance to genomic variants.MethodsHere we present a method that automatically matches patient-specific genomic alterations to treatment options. The method relies entirely on public knowledge of somatic variants with predictive evidence on drug response. The output report is aimed at supporting clinicians in the task of finding the clinical meaning of genomic variants. We applied the method to 1) The Cancer Genome Atlas (TCGA) and Genomics Evidence Neoplasia Information Exchange (GENIE) cohorts and 2) 11 patients from the NCT MASTER trial whose treatment discussions included information on their genomic profiles.ResultsOur reporting strategy showed a substantial number of patients with actionable variants in the analyses of TCGA and GENIE samples. Notably, it was able to reproduce experts’ treatment suggestions in a retrospective study of 11 patients from the NCT MASTER trial. Our results establish a proof of concept for comprehensive, evidence-based reports as a supporting tool for discussing treatment options in tumor boards.ConclusionsWe believe that a standardized method to report actionable somatic variants will smooth the incorporation of NGS in the clinical context. We anticipate that tools like the one we present here will become essential in summarizing for clinicians the growing evidence in the field of precision medicine. The R code of the presented method is provided in Additional file 6 and available at https://github.com/jperera-bel/MTB-Report.
Hematological Oncology | 2016
Iris Breitkreutz; Natalia Becker; Axel Benner; Florentina Kosely; Christoph Heining; Jens Hillengass; Gerlinde Egerer; Anthony D. Ho; Hartmut Goldschmidt; Marc S. Raab
Therapeutic options in heavily pretreated relapsed/refractory multiple myeloma patients are often very limited because of impaired bone marrow function. Bendamustine is effective in multiple myeloma and has a favourable toxicity profile. We hypothesized that dose‐intensified bendamustine (180 mg/m2, day 1 and 2) followed by autologous blood stem cell support (ASCS) would improve bone marrow function with low post‐transplant toxicity in patients with severely impaired haematopoiesis. We analyzed 28 consecutive myeloma patients, with a median of three prior lines of therapy (range 2–7), who had relapsed from the last treatment with very limited bone marrow function and were therefore ineligible for conventional chemotherapy, novel agents or trial enrolment. Dose‐intensified bendamustine with ASCS improved haematopoiesis as reflected by increased platelet counts (median 40/nl vs 94/nl, p = 0.0004) and white blood cell counts (3.0/nl vs 4.8/nl, p = 0.02) at day +100. The median time until engraftment of platelets (>50/nl) was 11 days (0–24 days) and of white cell counts (>1.0/nl) 0 days (0–24 days). At least, a minimal response was achieved in 36% of patients. The disease stabilization rate was 50% while the median progression‐free survival rate was limited to 2.14 months. Most importantly, patients were once again eligible for alternative treatments including enrolment into clinical trials. We conclude that dose‐intensified bendamustine followed by ASCS is safe and feasible for multiple myeloma patients with very limited bone marrow reserve. Copyright
Cancer Discovery | 2018
Christoph Heining; Peter Horak; Sebastian Uhrig; Paula L. Codo; Barbara Klink; Barbara Hutter; Martina Fröhlich; David Bonekamp; Daniela Richter; Katja Steiger; Roland Penzel; Volker Endris; Karl Roland Ehrenberg; Stephanie Frank; Kortine Kleinheinz; Umut H Toprak; Matthias Schlesner; Ranadip Mandal; Lothar Schulz; Helmut Lambertz; Sebastian Fetscher; Michael Bitzer; Nisar P. Malek; Marius Horger; Nathalia A. Giese; Oliver Strobel; Thilo Hackert; Christoph Springfeld; Lars Feuerbach; Frank Bergmann
We used whole-genome and transcriptome sequencing to identify clinically actionable genomic alterations in young adults with pancreatic ductal adenocarcinoma (PDAC). Molecular characterization of 17 patients with PDAC enrolled in a precision oncology program revealed gene fusions amenable to pharmacologic inhibition by small-molecule tyrosine kinase inhibitors in all patients with KRAS wild-type (KRASWT) tumors (4 of 17). These alterations included recurrent NRG1 rearrangements predicted to drive PDAC development through aberrant ERBB receptor-mediated signaling, and pharmacologic ERBB inhibition resulted in clinical improvement and remission of liver metastases in 2 patients with NRG1-rearranged tumors that had proved resistant to standard treatment. Our findings demonstrate that systematic screening of KRASWT tumors for oncogenic fusion genes will substantially improve the therapeutic prospects for a sizeable fraction of patients with PDAC.Significance: Advanced PDAC is a malignancy with few treatment options that lacks molecular mechanism-based therapies. Our study uncovers recurrent gene rearrangements such as NRG1 fusions as disease-driving events in KRASwt tumors, thereby providing novel insights into oncogenic signaling and new therapeutic options in this entity. Cancer Discov; 8(9); 1087-95. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 1047.
Onkologe | 2016
Christoph Heining; C. von Kalle
ZusammenfassungNeue wissenschaftliche Erkenntnisse im Hinblick auf bislang unbekannte genomische Alterationen maligner Zellen und zunehmend detailliertere Einblicke in therapeutisch adressierbare immunregulatorische Vorgänge bei Krebserkrankungen haben die diagnostischen und therapeutischen Strategien ebenso wie auch die Präventionsmöglichkeiten in den letzten Jahren erheblich bereichert. Um einen schnellstmöglichen Transfer dieser grundlagenwissenschaftlichen Ergebnisse in präventiv oder klinisch wirksame Anwendungen zu gewährleisten, bedarf es effizienter translationaler Programme. Eine Infrastruktur, die eine erfolgreiche Umsetzung derartiger Projekte an der Schnittstelle von Wissenschaft und Klinik erlaubt, erfordert zum einen den engen Austausch zwischen Grundlagenwissenschaftlern, translationalen Onkologen und klinisch spezialisierten Ärzten, zum anderen die notwendigen technischen und personellen Voraussetzungen für sowohl eine international kompetitive Forschung als auch eine hochklassige klinische Versorgung. Um diesen stetig wachsenden Anforderungen gerecht werden zu können, initiierte die Deutsche Krebshilfe (DKH) ein Programm zur Förderung onkologischer Spitzenzentren, das derzeit 13 Spitzenzentren, die definierte Voraussetzungen erfüllen müssen und regelmäßig durch internationale Expertengremien evaluiert werden, unterstützt. Gerade mit Blick auf die ermutigenden und rasanten Fortschritte der genomischen und immunologischen Grundlagenforschung ist es zwingend notwendig, den Prozess von der grundlagenwissenschaftlichen Entdeckung hin zur praktischen Anwendung gezielt zu fördern und langfristige Kooperationen translationaler Forschungseinrichtungen zu schaffen. Vor diesem Hintergrund wurde das Deutsche Konsortium für Translationale Onkologie (DKTK) gegründet. Gemeinsame Kommunikations- und Technikplattformen ermöglichen hier eine Bündelung von Ressourcen und komplementären Expertisen, zudem fördert das Programm die Ausbildung junger translationaler Onkologen und Wissenschaftler. Nur durch eine kontinuierliche Weiterentwicklung dieser lokalen Infrastrukturen und nationaler wie auch internationaler Kooperationen können Bedingungen geschaffen werden, die dauerhaft innovative translationale Forschungsprogramme sowie erfolgreiche Präventions- und Behandlungsstrategien ermöglichen.AbstractNew scientific knowledge concerning the detection of formerly unknown genomic alterations of malignant cells as well as an increasingly more detailed characterization of therapeutically relevant immunoregulatory mechanisms in cancer have substantially influenced the preventive, diagnostic and therapeutic strategies in cancer treatment. To accelerate the transfer of basic scientific data into preventive algorithms and clinical applications, the implementation of competitive and efficient translational oncology programs is critical. Nevertheless, setting up an infrastructure for successful programs at the interface between science and clinical applications requires a close collaboration of scientists, translational oncologists and clinical physicians as well as adequate technical and personnel resources that enable successful basic research to interact with outstanding clinical care. To meet these requirements the German Cancer Aid launched an oncological centers of excellence program and currently provides funding for 13 centers of excellence that have to fulfill defined criteria and undergo periodic reevaluation and certification by international experts. With respect to the encouraging and rapid progress in molecular sciences it is necessary to accelerate the process of transferring research discoveries to clinical implementation and to encourage long-term collaboration between translational research programs; therefore, the German Consortium for Translational Cancer Research, a nationwide network of cancer centers focusing on translational research was launched. Communication and technical platforms provide the possibility to bundle resources and to bring together complementary expertise. Furthermore, the program provides training of young translational oncologists and scientists. Only a continuous improvement of local infrastructures and nationwide as well as international collaborative interactions will provide the conditions to generate successful cancer care strategies and innovative translational research programs.