88 results on '"Griewank KG"'
Search Results
2. GNA11 (guanine nucleotide binding protein (G protein), alpha 11 (Gq class))
- Author
-
Griewank, KG, primary, Vemula, S, additional, and Bastian, BC, additional
- Published
- 2012
- Full Text
- View/download PDF
3. Shortened progression free and overall survival to immune-checkpoint inhibitors in BRAF-, RAS- and NF1- ("Triple") wild type melanomas.
- Author
-
Jansen P, Galetzka W, Lodde GC, Standl F, Zaremba A, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Mohr P, Gutzmer R, Meier F, Dippel E, Weichenthal M, Placke JM, Landsberg J, Möller I, Sucker A, Paschen A, Hadaschik E, Zimmer L, Livingstone E, Schadendorf D, Ugurel S, Stang A, and Griewank KG
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Skin Neoplasms genetics, Skin Neoplasms drug therapy, Skin Neoplasms mortality, Skin Neoplasms pathology, Skin Neoplasms immunology, Neurofibromin 1 genetics, Prospective Studies, Progression-Free Survival, Aged, 80 and over, Programmed Cell Death 1 Receptor antagonists & inhibitors, Telomerase genetics, GTP Phosphohydrolases genetics, Promoter Regions, Genetic, Membrane Proteins, Melanoma drug therapy, Melanoma genetics, Melanoma mortality, Melanoma pathology, Melanoma immunology, Immune Checkpoint Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf genetics, Mutation
- Abstract
Background: Melanomas lacking mutations in BRAF, NRAS and NF1 are frequently referred to as "triple wild-type" (tWT) melanomas. They constitute 5-10 % of all melanomas and remain poorly characterized regarding clinical characteristics and response to therapy. This study investigates the largest multicenter collection of tWT-melanomas to date., Methods: Targeted next-generation sequencing of the TERT promoter and 29 melanoma-associated genes were performed on 3109 melanoma tissue samples of the prospective multicenter study ADOREG/TRIM of the DeCOG revealing 292 patients suffering from tWT-melanomas. Clinical characteristics and mutational patterns were analyzed. As subgroup analysis, we analyzed 141 tWT-melanoma patients receiving either anti-CTLA4 plus anti-PD1 or anti PD1 monotherapy as first line therapy in AJCC stage IV., Results: 184 patients with cutaneous melanomas, 56 patients with mucosal melanomas, 34 patients with acral melanomas and 18 patients with melanomas of unknown origin (MUP) were included. A TERT promoter mutation could be identified in 33.2 % of all melanomas and 70.5 % of all tWT-melanomas harbored less than three mutations per sample. For the 141 patients with stage IV disease, mPFS independent of melanoma type was 6.2 months (95 % CI: 4-9) and mOS was 24.8 months (95 % CI: 14.2-53.4) after first line anti-CTLA4 plus anti-PD1 therapy. After first-line anti-PD1 monotherapy, mPFS was 4 months (95 %CI: 2.9-8.5) and mOS was 29.18 months (95 % CI: 17.5-46.2)., Conclusions: While known prognostic factors such as TERT promoter mutations and TMB were equally distributed among patients who received either anti-CTLA4 plus anti-PD1 combination therapy or anti-PD1 monotherapy as first line therapy, we did not find a prolonged mPFS or mOS in either of those. For both therapy concepts, mPFS and mOS were considerably shorter than reported for melanomas with known oncogene mutations., Competing Interests: Declaration of Competing Interest All other authors declare no conflicts of interest for the submitted work., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Clinical and genetic characteristics of BAP1 -mutated non-uveal and uveal melanoma.
- Author
-
Matull J, Placke JM, Lodde G, Zaremba A, Utikal J, Terheyden P, Pföhler C, Herbst R, Kreuter A, Welzel J, Kretz J, Möller I, Sucker A, Paschen A, Livingstone E, Zimmer L, Hadaschik E, Ugurel S, Schadendorf D, Thielmann CM, and Griewank KG
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Retrospective Studies, Adult, Aged, 80 and over, Prognosis, Ubiquitin Thiolesterase genetics, Melanoma genetics, Melanoma mortality, Melanoma therapy, Uveal Neoplasms genetics, Uveal Neoplasms mortality, Uveal Neoplasms therapy, Tumor Suppressor Proteins genetics, Mutation
- Abstract
Background: Screening for gene mutations has become routine clinical practice across numerous tumor entities, including melanoma. BAP1 gene mutations have been identified in various tumor types and acknowledged as a critical event in metastatic uveal melanoma, but their role in non-uveal melanoma remains inadequately characterized., Methods: A retrospective analysis of all melanomas sequenced in our department from 2014-2022 (n=2650) was conducted to identify BAP1 mutated samples. Assessment of clinical and genetic characteristics was performed as well as correlations with treatment outcome., Results: BAP1 mutations were identified in 129 cases and distributed across the entire gene without any apparent hot spots. Inactivating BAP1 mutations were more prevalent in uveal (55%) compared to non-uveal (17%) melanomas. Non-uveal BAP1 mutated melanomas frequently exhibited UV-signature mutations and had a significantly higher mutation load than uveal melanomas. GNAQ and GNA11 mutations were common in uveal melanomas, while MAP-Kinase mutations were frequent in non-uveal melanomas with NF1 , BRAF V600 and NRAS Q61 mutations occurring in decreasing frequency, consistent with a strong UV association. Survival outcomes did not differ among non-uveal melanoma patients based on whether they received targeted or immune checkpoint therapy, or if their tumors harbored inactivating BAP1 mutations., Conclusion: In contrast to uveal melanomas, where BAP1 mutations serve as a significant prognostic indicator of an unfavorable outcome, BAP1 mutations in non-uveal melanomas are primarily considered passenger mutations and do not appear to be relevant from a prognostic or therapeutic perspective., Competing Interests: JM: Declares travel support from Bristol Myers Squibb, Novartis and Sun Pharmaceutical Industries, outside the submitted work. J-MP: served as consultant and/or has received honoraria from Bristol-Myers Squibb, Novartis, Sanofi and received travel support from Bristol-Myers Squibb, Novartis, Pierre Fabre and Therakos, outside the submitted work. GL: Declares travel support from Sun Pharma, outside the submitted work. AZ: Declares travel support from Novartis, Sanofi Grenzyme, and Bristol-Myers Squibb, outside the submitted work. JU: Is on the advisory board or has received honoraria and travel support from Amgen, Bristol Myers Squibb, GSK, Immunocore, LeoPharma, Merck Sharp and Dohme, Novartis, Pierre Fabre, Roche, Sanofi outside the submitted work. PT: served as consultant and/or received honoraria form Almirall, Bristol Myers Squibb, Biofrontera, Curevac, Kyowa Kirin, Merck, Merck Sharp & Dohme, Novartis, Pierre-Fabre, Roche, Sanofi, 4SC, and travel support from Bristol Myers Squibb outside the submitted work. CP: Received honoraria speaker honoraria and advisory-board honoraria and travel support from BMS, MSD, Novartis, Merck Serono, Pierre Fabre, Sunpharma, AbbVie, LEO, and Kyona Kirin, outside the submitted work. RH: Is an employee of Helios Kliniken Erfurt GmbH. JW: Received honoraria and travel support from Almirall, Bristol Myers Squibb, Novartis, Pierre Fabre and Merck Sharp & Dohme, outside the submitted work. EL: Served as consultant and/or has received honoraria from Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Pierre-Fabre, Sanofi, Sunpharma, Takeda and travel support from Bristol-Myers Squibb, Pierre Fabre, Sunpharma and Novartis, outside the submitted work. LZ: Served as consultant and/or has received honoraria from Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Pierre-Fabre, Sunpharma and Sanofi; Research funding to institution: Novartis; travel support from Merck Sharp & Dohme, Bristol- Myers Squibb, Amgen, Pierre-Fabre, Sunpharma and Novartis, outside the submitted work. SU: Research support from Bristol Myers Squibb and Merck Serono; speakers and advisory board honoraria from Bristol Myers Squibb, Merck Sharp & Dohme, Merck Serono, and Novartis; meeting and travel support from Almirall, Bristol-Myers Squibb, IGEA Clinical Biophysics, Merck Sharp & Dohme, Novartis, Pierre Fabre, and Sun Pharma, outside the submitted work. DS: Reports personal fees and non-financial support from Roche/Genentech, grants, personal fees, non-financial support and other from BMS, personal fees from Merck Sharp & Dohme, personal fees and non-financial support from Merck Serono, grant, personal fees and non-financial support from Amgen, personal fees from Immunocore, personal fees from Incyte, personal fees from 4SC, personal fees from Pierre Fabre, personal fees and non-financial support from Sanofi/Regeneron, personal fees from Array BioPharma, personal fees from Pfizer, personal fees from Philogen, personal fees from Regeneron, personal fees from Nektar, personal fees from Sandoz, grants, personal fees and non-financial support from Novartis, personal fees and non-financial support from SunPharma, Replimune, Helsinn, OncoSec and InFlaRx outside the submitted work. 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., (Copyright © 2024 Matull, Placke, Lodde, Zaremba, Utikal, Terheyden, Pföhler, Herbst, Kreuter, Welzel, Kretz, Möller, Sucker, Paschen, Livingstone, Zimmer, Hadaschik, Ugurel, Schadendorf, Thielmann and Griewank.)
- Published
- 2024
- Full Text
- View/download PDF
5. Deep learning based histological classification of adnex tumors.
- Author
-
Jansen P, Arrastia JL, Baguer DO, Schmidt M, Landsberg J, Wenzel J, Emberger M, Schadendorf D, Hadaschik E, Maass P, and Griewank KG
- Subjects
- Humans, Artificial Intelligence, Algorithms, Neural Networks, Computer, Deep Learning, Skin Neoplasms pathology
- Abstract
Background: Cutaneous adnexal tumors are a diverse group of tumors arising from structures of the hair appendages. Although often benign, malignant entities occur which can metastasize and lead to patients´ death. Correct diagnosis is critical to ensure optimal treatment and best possible patient outcome. Artificial intelligence (AI) in the form of deep neural networks has recently shown enormous potential in the field of medicine including pathology, where we and others have found common cutaneous tumors can be detected with high sensitivity and specificity. To become a widely applied tool, AI approaches will also need to reliably detect and distinguish less common tumor entities including the diverse group of cutaneous adnexal tumors., Methods: To assess the potential of AI to recognize cutaneous adnexal tumors, we selected a diverse set of these entities from five German centers. The algorithm was trained with samples from four centers and then tested on slides from the fifth center., Results: The neural network was able to differentiate 14 different cutaneous adnexal tumors and distinguish them from more common cutaneous tumors (i.e. basal cell carcinoma and seborrheic keratosis). The total accuracy on the test set for classifying 248 samples into these 16 diagnoses was 89.92 %. Our findings support AI can distinguish rare tumors, for morphologically distinct entities even with very limited case numbers (< 50) for training., Conclusion: This study further underlines the enormous potential of AI in pathology which could become a standard tool to aid pathologists in routine diagnostics in the foreseeable future. The final diagnostic responsibility will remain with the pathologist., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Applying an artificial intelligence deep learning approach to routine dermatopathological diagnosis of basal cell carcinoma.
- Author
-
Duschner N, Baguer DO, Schmidt M, Griewank KG, Hadaschik E, Hetzer S, Wiepjes B, Le'Clerc Arrastia J, Jansen P, Maass P, and Schaller J
- Subjects
- Humans, Artificial Intelligence, Sensitivity and Specificity, Skin Neoplasms pathology, Deep Learning, Carcinoma, Squamous Cell pathology, Carcinoma, Basal Cell pathology
- Abstract
Background: Institutes of dermatopathology are faced with considerable challenges including a continuously rising numbers of submitted specimens and a shortage of specialized health care practitioners. Basal cell carcinoma (BCC) is one of the most common tumors in the fair-skinned western population and represents a major part of samples submitted for histological evaluation. Digitalizing glass slides has enabled the application of artificial intelligence (AI)-based procedures. To date, these methods have found only limited application in routine diagnostics. The aim of this study was to establish an AI-based model for automated BCC detection., Patients and Methods: In three dermatopathological centers, daily routine practice BCC cases were digitalized. The diagnosis was made both conventionally by analog microscope and digitally through an AI-supported algorithm based on a U-Net architecture neural network., Results: In routine practice, the model achieved a sensitivity of 98.23% (center 1) and a specificity of 98.51%. The model generalized successfully without additional training to samples from the other centers, achieving similarly high accuracies in BCC detection (sensitivities of 97.67% and 98.57% and specificities of 96.77% and 98.73% in centers 2 and 3, respectively). In addition, automated AI-based basal cell carcinoma subtyping and tumor thickness measurement were established., Conclusions: AI-based methods can detect BCC with high accuracy in a routine clinical setting and significantly support dermatopathological work., (© 2023 Deutsche Dermatologische Gesellschaft (DDG).)
- Published
- 2023
- Full Text
- View/download PDF
7. Einsatz künstlicher Intelligenz mittels Deep Learning in der dermatopathologischen Routinediagnostik des Basalzellkarzinoms: Applying an artificial intelligence deep learning approach to routine dermatopathological diagnosis of basal cell carcinoma.
- Author
-
Duschner N, Baguer DO, Schmidt M, Griewank KG, Hadaschik E, Hetzer S, Wiepjes B, Le'Clerc Arrastia J, Jansen P, Maass P, and Schaller J
- Published
- 2023
- Full Text
- View/download PDF
8. Deep learning detection of melanoma metastases in lymph nodes.
- Author
-
Jansen P, Baguer DO, Duschner N, Arrastia JL, Schmidt M, Landsberg J, Wenzel J, Schadendorf D, Hadaschik E, Maass P, Schaller J, and Griewank KG
- Subjects
- Humans, Sentinel Lymph Node Biopsy methods, Artificial Intelligence, Lymph Nodes pathology, Lymphatic Metastasis pathology, Lymph Node Excision, Deep Learning, Melanoma pathology, Lymphadenopathy, Skin Neoplasms pathology
- Abstract
Background: In melanoma patients, surgical excision of the first draining lymph node, the sentinel lymph node (SLN), is a routine procedure to evaluate lymphogenic metastases. Metastasis detection by histopathological analysis assesses multiple tissue levels with hematoxylin and eosin and immunohistochemically stained glass slides. Considering the amount of tissue to analyze, the detection of metastasis can be highly time-consuming for pathologists. The application of artificial intelligence in the clinical routine has constantly increased over the past few years., Methods: In this multi-center study, a deep learning method was established on histological tissue sections of sentinel lymph nodes collected from the clinical routine. The algorithm was trained to highlight potential melanoma metastases for further review by pathologists, without relying on supplementary immunohistochemical stainings (e.g. anti-S100, anti-MelanA)., Results: The established method was able to detect the existence of metastasis on individual tissue cuts with an area under the curve of 0.9630 and 0.9856 respectively on two test cohorts from different laboratories. The method was able to accurately identify tumour deposits>0.1 mm and, by automatic tumour diameter measurement, classify these into 0.1 mm to -1.0 mm and>1.0 mm groups, thus identifying and classifying metastasis currently relevant for assessing prognosis and stratifying treatment., Conclusions: Our results demonstrate that AI-based SLN melanoma metastasis detection has great potential and could become a routinely applied aid for pathologists. Our current study focused on assessing established parameters; however, larger future AI-based studies could identify novel biomarkers potentially further improving SLN-based prognostic and therapeutic predictions for affected patients., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
9. Human Papillomavirus 42 Drives Digital Papillary Adenocarcinoma and Elicits a Germ Cell-like Program Conserved in HPV-Positive Cancers.
- Author
-
Leiendecker L, Neumann T, Jung PS, Cronin SM, Steinacker TL, Schleiffer A, Schutzbier M, Mechtler K, Kervarrec T, Laurent E, Bachiri K, Coyaud E, Murali R, Busam KJ, Itzinger-Monshi B, Kirnbauer R, Cerroni L, Calonje E, Rütten A, Stubenrauch F, Griewank KG, Wiesner T, and Obenauf AC
- Subjects
- Female, Humans, Human Papillomavirus Viruses, Papillomaviridae genetics, Germ Cells pathology, Papillomavirus Infections complications, Uterine Cervical Neoplasms, Skin Neoplasms, Bone Neoplasms, Breast Neoplasms, Adenocarcinoma, Clear Cell, Adenocarcinoma, Papillary
- Abstract
The skin is exposed to viral pathogens, but whether they contribute to the oncogenesis of skin cancers has not been systematically explored. Here we investigated 19 skin tumor types by analyzing off-target reads from commonly available next-generation sequencing data for viral pathogens. We identified human papillomavirus 42 (HPV42) in 96% (n = 45/47) of digital papillary adenocarcinoma (DPA), an aggressive cancer occurring on the fingers and toes. We show that HPV42, so far considered a nononcogenic, "low-risk" HPV, recapitulates the molecular hallmarks of oncogenic, "high-risk" HPVs. Using machine learning, we find that HPV-driven transformation elicits a germ cell-like transcriptional program conserved throughout all HPV-driven cancers (DPA, cervical carcinoma, and head and neck cancer). We further show that this germ cell-like transcriptional program, even when reduced to the top two genes (CDKN2A and SYCP2), serves as a fingerprint of oncogenic HPVs with implications for early detection, diagnosis, and therapy of all HPV-driven cancers., Significance: We identify HPV42 as a uniform driver of DPA and add a new member to the short list of tumorigenic viruses in humans. We discover that all oncogenic HPVs evoke a germ cell-like transcriptional program with important implications for detecting, diagnosing, and treating all HPV-driven cancers. See related commentary by Starrett et al., p. 17. This article is highlighted in the In This Issue feature, p. 1., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
10. Deep Learning Assisted Diagnosis of Onychomycosis on Whole-Slide Images.
- Author
-
Jansen P, Creosteanu A, Matyas V, Dilling A, Pina A, Saggini A, Schimming T, Landsberg J, Burgdorf B, Giaquinta S, Müller H, Emberger M, Rose C, Schmitz L, Geraud C, Schadendorf D, Schaller J, Alber M, Klauschen F, and Griewank KG
- Abstract
Background: Onychomycosis numbers among the most common fungal infections in humans affecting finger- or toenails. Histology remains a frequently applied screening technique to diagnose onychomycosis. Screening slides for fungal elements can be time-consuming for pathologists, and sensitivity in cases with low amounts of fungi remains a concern. Convolutional neural networks (CNNs) have revolutionized image classification in recent years. The goal of our project was to evaluate if a U-NET-based segmentation approach as a subcategory of CNNs can be applied to detect fungal elements on digitized histologic sections of human nail specimens and to compare it with the performance of 11 board-certified dermatopathologists., Methods: In total, 664 corresponding H&E- and PAS-stained histologic whole-slide images (WSIs) of human nail plates from four different laboratories were digitized. Histologic structures were manually annotated. A U-NET image segmentation model was trained for binary segmentation on the dataset generated by annotated slides., Results: The U-NET algorithm detected 90.5% of WSIs with fungi, demonstrating a comparable sensitivity with that of the 11 board-certified dermatopathologists (sensitivity of 89.2%)., Conclusions: Our results demonstrate that machine-learning-based algorithms applied to real-world clinical cases can produce comparable sensitivities to human pathologists. Our established U-NET may be used as a supportive diagnostic tool to preselect possible slides with fungal elements. Slides where fungal elements are indicated by our U-NET should be reevaluated by the pathologist to confirm or refute the diagnosis of onychomycosis.
- Published
- 2022
- Full Text
- View/download PDF
11. Evaluation of a Deep Learning Approach to Differentiate Bowen's Disease and Seborrheic Keratosis.
- Author
-
Jansen P, Baguer DO, Duschner N, Le'Clerc Arrastia J, Schmidt M, Wiepjes B, Schadendorf D, Hadaschik E, Maass P, Schaller J, and Griewank KG
- Abstract
Background: Some of the most common cutaneous neoplasms are Bowen’s disease and seborrheic keratosis, a malignant and a benign proliferation, respectively. These entities represent a significant fraction of a dermatopathologists’ workload, and in some cases, histological differentiation may be challenging. The potential of deep learning networks to distinguish these diseases is assessed. Methods: In total, 1935 whole-slide images from three institutions were scanned on two different slide scanners. A U-Net-based segmentation deep learning algorithm was trained on data from one of the centers to differentiate Bowen’s disease, seborrheic keratosis, and normal tissue, learning from annotations performed by dermatopathologists. Optimal thresholds for the class distinction of diagnoses were extracted and assessed on a test set with data from all three institutions. Results: We aimed to diagnose Bowen’s diseases with the highest sensitivity. A good performance was observed across all three centers, underlining the model’s robustness. In one of the centers, the distinction between Bowen’s disease and all other diagnoses was achieved with an AUC of 0.9858 and a sensitivity of 0.9511. Seborrheic keratosis was detected with an AUC of 0.9764 and a sensitivity of 0.9394. Nevertheless, distinguishing irritated seborrheic keratosis from Bowen’s disease remained challenging. Conclusions: Bowen’s disease and seborrheic keratosis could be correctly identified by the evaluated deep learning model on test sets from three different centers, two of which were not involved in training, and AUC scores > 0.97 were obtained. The method proved robust to changes in the staining solution and scanner model. We believe this demonstrates that deep learning algorithms can aid in clinical routine; however, the results should be confirmed by qualified histopathologists.
- Published
- 2022
- Full Text
- View/download PDF
12. Genetic characterization of advanced conjunctival melanoma and response to systemic treatment.
- Author
-
Lodde GC, Jansen P, Möller I, Sucker A, Hassel JC, Forschner A, Eckardt J, Meier F, Reinhardt L, Kähler KC, Ziemer M, Schlaak M, Rahimi F, Schatton K, Meiss F, Gutzmer R, Pföhler C, Terheyden P, Schilling B, Sachse M, Heppt MV, Sindrilaru A, Leiter U, Zaremba A, Thielmann CM, Ugurel S, Zimmer L, Hadaschik E, Bechrakis NE, Schadendorf D, Westekemper H, Livingstone E, and Griewank KG
- Subjects
- Conjunctiva pathology, DNA Copy Number Variations, Humans, Immune Checkpoint Inhibitors, Mitogen-Activated Protein Kinase Kinases, Mutation, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf genetics, Retrospective Studies, Melanoma, Cutaneous Malignant, Eye Neoplasms drug therapy, Eye Neoplasms genetics, Melanoma drug therapy, Melanoma genetics, Skin Neoplasms pathology
- Abstract
Background: Conjunctival melanoma is a rare type of ocular melanoma, which is prone to local recurrence and metastasis and can lead to patient death. Novel therapeutic strategies have revolutionized cutaneous melanoma management. The efficacy of these therapies in conjunctival melanoma, however, has not been evaluated in larger patient cohorts., Methods: In this multi-center retrospective cohort study with additional screening of the ADOREG database, data were collected from 34 patients with metastatic conjunctival melanoma who received targeted therapy (TT) (BRAF ± MEK inhibitors) or immune checkpoint inhibitors (ICI) (anti-PD-1 ± anti-CTLA4). In 15 cases, tissue was available for targeted next-generation-sequencing (611 genes) and RNA sequencing. Driver mutations, tumor mutational burden, copy number variations and inflammatory/IFNγ gene expression signatures were determined., Results: Genetic characterization identified frequent BRAF (46.7%, 7/15), NRAS (26.7%, 4/15), NF1 (20%, 3/15), and TERT promoter (46.7%, 7/15) mutations. UV associated C>T and CC>TT mutations were common. Median follow-up time after start of first TT or ICI therapy was 13.2 months. In 26 patients receiving first-line ICI, estimated one-year progression-free survival (PFS) rate was 42.0%, PFS and overall survival (OS) 6.2 and 18.0 months, respectively. First-line TT was given to 8 patients, estimated one-year PFS rate was 54.7%, median PFS and OS 12.6 and 29.1 months, respectively., Conclusions: Our findings support the role of UV irradiation in conjunctival melanoma and the genetic similarity with cutaneous melanoma. Conjunctival melanoma patients with advanced disease benefit from both targeted therapies (BRAF ± MEK inhibitors) and immune checkpoint inhibitors., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: GL has received travel support for congress participation: Sun Pharma. PJ declares no conflicts of interest. IM declares no conflicts of interest. AS declares no conflicts of interest. JCH receives a scientific grant from BMS, payments ad consultant from Pierre Fabre, MSD, Sunpharma and speaker honoraria from BMS, MSD, Roche, Novartis, Pfizer, Sanofi, GSK, Amgen, Almirall. AF Advisory board: Pierre-Fabre, Roche, Novartis, BMS, MSD, travel support for congress participation: Pierre-Fabre, Roche, Novartis, BMS, honoraria for talks: Roche, Novartis, BMS, MSD, CeGaT, research funding: Stiftung Immunonkologie BMS. All outside the submitted work. JE declars no conflicts of interest. FM has received travel support or/and speaker's fees or/and advisor's honoraria by Novartis, Roche, BMS, MSD and Pierre Fabre and research funding from Novartis and Roche. LR declares no conflicts of interest. KCK has served as consultant or/and has received honoraria from Amgen, Roche, Bristol Myers Squibb, Merck Sharp and Dohme, Pierre Fabre, and Novartis, and received travel support from Amgen, Merck Sharp and Dohme, Bristol Myers Squibb, Amgen, Pierre Fabre, Medac and Novartis. MZ has served as consultant or/and has received honoraria from Bristol Myers Squibb, Merck Sharp and Dohme, Pierre Fabre, Novartis, Sanofi Genzyme and SUN Pharma. MS Consultancy, speaker fees or travel grants: BMS, MSD, Roche, Kyowa Kirin, Novartis, Sanofi Genzyme, Pierre Fabre, Sun Pharma, Immunocore. FR declares no conflicts of interest. AS declares no conflicts of interest. KS declares no conflicts of interest. FM served as consultant and/or has received honoraria from Novartis, Roche, Bristol-Myers Squibb, Merck Sharp & Dohme, Pierre Fabre, Sanofi Genzyme and travel support from Novartis, Sunpharma and Bristol-Myers Squibb, outside the submitted work. RG Honoraria for lectures (personally): Roche, BMS, MSD, Novartis, Amgen, Merck Serono, Almirall Hermal, SUN, Sanofi, Pierre-Fabre. Honoraria for advice (personally): BMS, Roche, Novartis, Almirall Hermal, MSD, Amgen, SUN, Sanofi, Pierre-Fabre, 4SC, Bayer, MerckSerono, Pfizer, Immunocore. Research grants (to institution): Novartis, Pfizer, Johnson & Johnson, Amgen, Merck-Serono, SUN Pharma, Sanofi, Almirall Hermal. Support for meeting participation: Roche, BMS, SUN, Merck-Serono, Pierre-Fabre. CP received honoraria (speaker honoraria or honoraria as a consultant) and travel support from: Novartis, BMS, Roche, Merck Serono, MSD, Celgene, AbbVie, SUNPHARMA, UCB, Allergy Therapeutics, Pierre Fabre, Kyowa Kirin and LEO. PT declares honoraria from Bristol-Myers Squibb, Novartis, Merck Sharp & Dohme, Pierre-Fabre, CureVac, Merck Serono, Sanofi, Roche, Kyowa Kirin, Biofrontera, Almirall, and 4SC; travel support from Bristol-Myers Squibb and Pierre-Fabre, outside the submitted work.BS is on the advisory board or has received honoraria from Almirall, Pfizer, Sanofi, Incyte, Novartis, Roche, Bristol-Myers Squibb (BMS) and MSD Sharp & Dohme, research funding from Novartis, Pierre Fabre Pharmaceuticals, Bristol-Myers Squibb and MSD Sharp & Dohme and travel support from Novartis, Roche, Bristol-Myers Squibb, Pierre Fabre Pharmaceuticals, MSD Sharp & Dohme and Amgen, outside the submitted work. MS declares no conflict of interest. MH has been a member of advisory boards of Almirall Hermal, Sanofi-Aventis, MSD, 622 BMS, Novartis, and received speaker's honoraria from Galderma and Biofrontera. UL speaker fees: Roche, Novartis, Sanofi, MSD; consultancy: Roche, Sanofi, MSD, Novartis. AZ received travel support from Novartis, Sanofi Grenzyme, Sun Pharma, and Bristol-Myers Squibb, outside the submitted work. CM declares no conflicts of interest. SU declares research support from Bristol Myers Squibb and Merck Serono; speakers and advisory board honoraria from Bristol Myers Squibb, Merck Sharp & Dohme, Merck Serono, Novartis and Roche, and travel support from Bristol Myers Squibb, Merck Sharp & Dohme, and Pierre Fabre. LZ served as consultant and/or has received honoraria from Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Pierre-Fabre, Sunpharma and Sanofi; Research funding to institution: Novartis; travel support from Merck Sharp & Dohme, Bristol-Myers Squibb, Amgen, Pierre-Fabre, Sanofi, Sunpharma and Novartis, outside the submitted work. EH declares no conflicts of interest. NB declares no conflicts of interest. DS D.S. Served as consultant, advisory board and/or speaker and has received honoraria from Amgen, Array, Bristol-Myers Squibb, Helsinn, Immunocore, InflarX, Merck Sharp & Dohme, Merck-Serono, Nektar, Novartis, OncoSec, Pfizer, Philogen, Regeneron, Replimune, Sandoz, Sanofi, Sunpharma, 4SC and travel support from Bristol-Myers Squibb, Merck Sharp & Dohme, Merck-Serono, Nektar, Novartis, Pierre Fabre, Sandoz, Sanofi, and 4SC, outside the submitted work. HW declares no conflicts of interest. EL served as consultant and/or has received honoraria from Amgen, Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Medac, Sanofi, Sunpharma and travel support from Medac, Merck Sharp & Dohme, Bristol-Myers Squibb, Pierre Fabre, Sunpharma and Novartis, outside the submitted work. KG declares no conflicts of interest., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
13. Genetic and Clinical Characteristics of ARID1A Mutated Melanoma Reveal High Tumor Mutational Load without Implications on Patient Survival.
- Author
-
Thielmann CM, Matull J, Roth S, Placke JM, Chorti E, Zaremba A, Lodde G, Jansen P, Krefting F, Kretz J, Möller I, Sucker A, Paschen A, Livingstone E, Zimmer L, Ugurel S, Schadendorf D, Hadaschik E, and Griewank KG
- Abstract
(1) Background: Melanoma has the highest mortality of all cutaneous tumors, despite recent treatment advances. Many relevant genetic events have been identified in the last decade, including recurrent ARID1A mutations, which in various tumors have been associated with improved outcomes to immunotherapy. (2) Methods: Retrospective analysis of 116 melanoma samples harboring ARID1A mutations. Assessment of clinical and genetic characteristics was performed as well as correlations with treatment outcome applying Kaplan-Meier (log-rank test), Fisher's exact and Chi-squared tests. (3) Results: The majority of ARID1A mutations were in cutaneous and occult melanoma. ARID1A mutated samples had a higher number of mutations than ARID1A wild-type samples and harbored UV-mutations. A male predominance was observed. Many samples also harbored NF1 mutations. No apparent differences were noted between samples harboring genetically inactivating (frame-shift or nonsense) mutations and samples with other mutations. No differences in survival or response to immunotherapy of patients with ARID1A mutant melanoma were observed. (4) Conclusions: ARID1A mutations primarily occur in cutaneous melanomas with a higher mutation burden. In contrast to findings in other tumors, our data does not support ARID1A mutations being a biomarker of favorable response to immunotherapies in melanoma. Larger prospective studies would still be warranted.
- Published
- 2022
- Full Text
- View/download PDF
14. The Prognostic Relevance of PMCA4 Expression in Melanoma: Gender Specificity and Implications for Immune Checkpoint Inhibition.
- Author
-
Hegedüs L, Livingstone E, Bánkfalvi Á, Viehof J, Enyedi Á, Bilecz Á, Győrffy B, Baranyi M, Tőkés AM, Gil J, Marko-Varga G, Griewank KG, Zimmer L, Váraljai R, Sucker A, Zaremba A, Schadendorf D, Aigner C, and Hegedüs B
- Subjects
- Animals, Female, Humans, Immune Checkpoint Inhibitors, Mammals metabolism, Plasma Membrane Calcium-Transporting ATPases metabolism, Prognosis, Programmed Cell Death 1 Receptor metabolism, RNA, Messenger, Melanoma, Cutaneous Malignant, Melanoma genetics, Nevus, Skin Neoplasms genetics
- Abstract
PMCA4 is a critical regulator of Ca
2+ homeostasis in mammalian cells. While its biological and prognostic relevance in several cancer types has already been demonstrated, only preclinical investigations suggested a metastasis suppressor function in melanoma. Therefore, we studied the expression pattern of PMCA4 in human skin, nevus, as well as in primary and metastatic melanoma using immunohistochemistry. Furthermore, we analyzed the prognostic power of PMCA4 mRNA levels in cutaneous melanoma both at the non-metastatic stage as well as after PD-1 blockade in advanced disease. PMCA4 localizes to the plasma membrane in a differentiation dependent manner in human skin and mucosa, while nevus cells showed no plasma membrane staining. In contrast, primary cutaneous, choroidal and conjunctival melanoma cells showed specific plasma membrane localization of PMCA4 with a wide range of intensities. Analyzing the TCGA cohort, PMCA4 mRNA levels showed a gender specific prognostic impact in stage I-III melanoma. Female patients with high transcript levels had a significantly longer progression-free survival. Melanoma cell specific PMCA4 protein expression is associated with anaplasticity in melanoma lung metastasis but had no impact on survival after lung metastasectomy. Importantly, high PMCA4 transcript levels derived from RNA-seq of cutaneous melanoma are associated with significantly longer overall survival after PD-1 blockade. In summary, we demonstrated that human melanoma cells express PMCA4 and PMCA4 transcript levels carry prognostic information in a gender specific manner.- Published
- 2022
- Full Text
- View/download PDF
15. TERT promoter mutations are associated with longer progression-free and overall survival in patients with BRAF-mutant melanoma receiving BRAF and MEK inhibitor therapy.
- Author
-
Thielmann CM, Matull J, Zaremba A, Murali R, Chorti E, Lodde G, Jansen P, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Mohr P, Gutzmer R, Meier F, Dippel E, Weichenthal M, Kretz J, Möller I, Sucker A, Paschen A, Livingstone E, Zimmer L, Hadaschik E, Ugurel S, Schadendorf D, and Griewank KG
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Biomarkers, Tumor genetics, Female, Humans, Male, Middle Aged, Progression-Free Survival, Retrospective Studies, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Melanoma drug therapy, Melanoma genetics, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mutation genetics, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Telomerase genetics
- Abstract
Background: Around 50% of cutaneous melanomas harbour therapeutically targetable BRAF V600 mutations. Reliable clinical biomarkers predicting duration of response to BRAF-targeted therapies are still lacking. Recent in vitro studies demonstrated that BRAF-MEK inhibitor therapy response is associated with tumour TERT promoter mutation status. We assessed this potential association in a clinical setting., Methods: The study cohort comprised 232 patients with metastatic or unresectable BRAF V600-mutated melanoma receiving combined BRAF/MEK inhibitor treatment, including a single-centre retrospective discovery cohort (N = 120) and a prospectively collected multicenter validation cohort (N = 112). Patients were excluded if they received BRAF or MEK inhibitors in an adjuvant setting, as monotherapy, or in combination with immunotherapy. Kaplan-Meier and univariate/multivariate Cox regression analyses were performed as appropriate., Results: median age at first diagnosis was 54 years (range 16-84 years). The majority of patients were men 147/232 (63.4%). Most tumours harboured TERT promoter mutations (72%, N = 167). A survival advantage was observed in both progression-free survival (PFS) and overall survival (OS) for patients with TERT promoter-mutant versus wild-type tumours in both the discovery cohort (mPFS of 9.6 months [N = 87] vs 5.0 months [N = 33]; hazard ratio [HR] = 0.56 [95% confidence interval {CI} 0.33-0.96] and mOS of 33.6 months vs 15.0 months; HR = 0.47 [95%CI 0.32-0.70]) as well as the validation cohort (mPFS of 7.3 months [N = 80] vs 5.8 months [N = 32]; HR = 0.67 [95%CI 0.41-1.10] and mOS of 51.1 months vs 15.0 months; HR = 0.33 [95%CI 0.18-0.63]). In the pooled cohort of TERT promoter-mutant (N = 167) versus wild-type (N = 65) tumours, respectively, PFS was 8.9 versus 5.5 months, (HR = 0.62; 95%CI 0.45-0.87; P = 0.004), and OS was 33.6 versus 17.0 months, (HR = 0.51; 95%CI 0.35-0.75, P = 0.0001)., Conclusions: In patients with melanoma receiving BRAF/MEK-targeted therapies, TERT promoter mutations are associated with longer survival. If validated in larger studies, TERT promoter mutation status should be included as a predictive biomarker in treatment algorithms for advanced melanoma., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: C.M.T. reported no relevant conflicts of interest. J.M. declares travel support from Bristol Myers Squibb, Novartis and Sun Pharmaceutical Industries, outside the submitted work. A.Z. declares travel support from Novartis, Sanofi Genzyme and Bristol-Myers Squibb, outside the submitted work. R.M. reported no relevant conflicts of interest. E.C. reported no relevant conflicts of interest. G.L. declares travel support from Sun Pharma, outside the submitted work. P.J. reported no relevant conflicts of interest. R.H. declares speakers and advisory board honoraria from BMS, MSD, Novartis, Pierre Fabre, Roche and SUN-Pharma, outside the submitted work. P.T. declares honoraria from Bristol-Myers Squibb, Novartis, Merck Sharp & Dohme, Pierre-Fabre, CureVac, Merck Serono, Sanofi, Roche, Kyowa Kirin and Biofrontera; travel support from Bristol-Myers Squibb and Pierre-Fabre, outside the submitted work. J.U. declares advisory board or has received honoraria and travel support from Amgen, Bristol Myers Squibb, GSK, LeoPharma, Merck Sharp and Dohme, Novartis, Pierre Fabre, Roche and Sanofi, outside the submitted work. C.P. served as consultant and/or has received honoraria from Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Sanofi, Sun Pharma, Pierre Fabre, AbbVie, Kyona Kirin and Amgen and received travel support from Amgen, Merck Sharp & Dohme, Bristol-Myers Squibb, Pierre Fabre, Sun Pharma and Novartis, outside the submitted work. J.Ul declares travel support: Medac, Sun Pharma; consulting: Bristol-Myers Squibb, Sun Pharma; lectures: Bristol-Myers Squibb, MSD, Merck, Novartis, Roche, Sanofi, Sun Pharma; grants: Novartis, outside the submitted work. A.K. reported no relevant conflicts of interest. P.M. declares research support (to institution): Bristol-Myers Squibb, Novartis, MSD. Honoraria for lectures (personally): Roche Pharma, Bristol-Myers Squibb, Novartis, MSD, Almirall-Hermal, Amgen, Merck-Serono, Bayer, Pierre-Fabre, Sanofi. Honoraria for advisory boards: Bayersdorf, Roche Pharma, Bristol-Myers Squibb, Novartis, MSD, Almirall-Hermal, Amgen, Pierre-Fabre, Merck-Serono, SUN, Merck-Serono, Sanofi, outside the submitted work. R.G. invited speaker: Roche, BMS, MSD, Novartis, Amgen, Merck Serono, Almirall Hermal, SUN, Sanofi, Pierre-Fabre. Advisory board: BMS, Roche, Novartis, Almirall Hermal, MSD, Amgen, SUN, Sanofi, Pierre-Fabre, 4SC, Bayer, Merck Serono, Pfizer, Immunocore. Research grants: Novartis, Pfizer, Johnson & Johnson, Amgen, Merck-Serono, SUN Pharma, Sanofi. Travel/meeting support: Roche, BMS, SUN, Merck-Serono, Pierre-Fabre, outside the submitted work. F.M. declares travel support or/and speaker's fees or/and advisor's honoraria by Novartis, Roche, BMS, MSD and Pierre Fabre and research funding from Novartis and Roche, outside the submitted work. E.D. reported no relevant conflicts of interest. M.W. reported no relevant conflicts of interest. J.K. reported no relevant conflicts of interest. I.M. reported no relevant conflicts of interest. A.S. reported no relevant conflicts of interest. A.P. reported no relevant conflicts of interest. E.L. served as a consultant and/or has received honoraria from Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Medac, Sanofi, Sun Pharma and travel support from Amgen, Merck Sharp & Dohme, Bristol-Myers Squibb, Pierre Fabre, Sun Pharma and Novartis, outside the submitted work. L.Z. served as consultant and/or has received honoraria from Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Pierre-Fabre, Sun Pharma and Sanofi; Research funding to institution: Novartis; travel support from Merck Sharp & Dohme, Bristol-Myers Squibb, Amgen, Pierre-Fabre, Sanofi, Sun Pharma and Novartis, outside the submitted work.E.H. reported no relevant conflicts of interest. S.U. declares research support from Bristol Myers Squibb and Merck Serono; speakers and advisory board honoraria from Bristol Myers Squibb, Merck Sharp & Dohme, Merck Serono, Novartis and Roche, and travel support from Bristol Myers Squibb, and Merck Sharp & Dohme, outside the submitted work. D.S. reports personal fees and non-financial support from Roche/Genentech, grants, personal fees, non-financial support and other from BMS, personal fees from Merck Sharp & Dohme, personal fees and non-financial support from Merck Serono, grant, personal fees and non-financial support from Amgen, personal fees from Immunocore, personal fees from Incyte, personal fees from 4SC, personal fees from Pierre Fabre, personal fees and non-financial support from Sanofi/Regeneron, personal fees from Array BioPharma, personal fees from Pfizer, personal fees from Philogen, personal fees from Regeneron, personal fees from Nektar, personal fees from Sandoz, grants, personal fees and non-financial support from Novartis, personal fees and non-financial support from Sun Pharma, Replimune, Helsinn, OncoSec and InFlaRx outside the submitted work. K.G.: No relevant conflicts of interest., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. Management of partial and non-responding cutaneous squamous cell carcinoma.
- Author
-
Jansen P, Lodde GC, Griewank KG, Hadaschik E, Roesch A, Ugurel S, Zimmer L, Livingstone E, and Schadendorf D
- Subjects
- Humans, Carcinoma, Basal Cell, Carcinoma, Squamous Cell drug therapy, Skin Neoplasms drug therapy
- Abstract
Cutaneous squamous cell carcinoma (cSCC) and basal cell carcinoma are the most common types of skin cancer. For patients with locally advanced and metastatic cSCC, the programmed cell death 1 (PD-1) inhibitor cemiplimab is approved for systemic treatment. Despite this revolutionary immunomodulatory therapeutic approach, tumours may fail to respond either completely or partially. In addition to the previously established local treatment with radiotherapy or systemic treatment with chemotherapy and epidermal growth factor receptor inhibitors, ongoing trials are currently focussed on re-stimulating the antitumour immune response in patients with advanced cSCC refractory to PD-1 inhibitors. In this review, ongoing and recently finished trials with different therapeutic approaches will be discussed., (© 2021 European Academy of Dermatology and Venereology.)
- Published
- 2022
- Full Text
- View/download PDF
17. Checkpoint immunotherapy of cutaneous squamous cell carcinoma in patients suffering from chronic lymphocytic leukaemia: divergent outcomes in two men treated with PD-1 inhibitors.
- Author
-
Jansen P, Lodde GC, Wetter A, Welt A, Stuschke M, Dührsen U, Stoffels I, Klode J, Livingstone E, Zimmer L, Roesch A, Hadaschik E, Griewank KG, Schadendorf D, and Ugurel S
- Subjects
- Humans, Immune Checkpoint Inhibitors, Immunotherapy, Male, United States, Carcinoma, Squamous Cell drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Skin Neoplasms drug therapy
- Abstract
Cutaneous squamous cell carcinoma (cSCC) numbers among the most common types of skin cancer and is known as one of the cancer entities with the highest mutational burden among all solid tumours. Due to the positive correlation between mutational burden and response rate to inhibitors of the programmed cell death 1 (PD-1), those inhibitors are considered promising candidates for the systemic therapy of cSCC. Recently, the PD-1 inhibitors pembrolizumab, nivolumab and cemiplimab demonstrated efficacy in the systemic treatment of locally advanced or metastatic cSCC leading to the approval of cemiplimab by the FDA (U.S. Food and Drug Administration) in 2018 and the EMA (European Medicines Agency) in 2019. Patients with haematological malignancies tend to develop skin cancers of high aggressiveness, enhanced cumulative recurrence rate and higher rates of metastases with subsequent death. Chronic lymphocytic leukaemia (CLL) is the most frequent type of leukaemia in the United States and Europe with the majority of patients older than 50 years of age. This neoplasm predominantly originates from B -cells leading to an impaired immune system of the patient. Although CLL is a B-cell malignancy, studies have also described the involvement of T cells in the pathogenesis and progression of the disease with contradictory findings on the effects of PD-1 inhibitors in CLL. Due to their underlying hematologic malignancy, these patients have commonly no access to PD-1 inhibitor trials for treatment of advanced cSCC. We report on two patients with locally advanced or metastatic cSCC. Both patients had been suffering from a CLL for many years without indication for treatment. Despite a potential immunosuppressive state of the patients due to their CLL, both were treated with the PD-1 inhibitor pembrolizumab resulting in different therapy outcomes., (© 2021 European Academy of Dermatology and Venereology.)
- Published
- 2022
- Full Text
- View/download PDF
18. NF1-mutated melanomas reveal distinct clinical characteristics depending on tumour origin and respond favourably to immune checkpoint inhibitors.
- Author
-
Thielmann CM, Chorti E, Matull J, Murali R, Zaremba A, Lodde G, Jansen P, Richter L, Kretz J, Möller I, Sucker A, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Mohr P, Gutzmer R, Meier F, Dippel E, Weichenthal M, Paschen A, Livingstone E, Zimmer L, Schadendorf D, Hadaschik E, Ugurel S, and Griewank KG
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Mutation, Young Adult, Melanoma, Cutaneous Malignant, Immune Checkpoint Inhibitors therapeutic use, Melanoma drug therapy, Melanoma genetics, Melanoma pathology, Neurofibromin 1 genetics, Skin Neoplasms drug therapy, Skin Neoplasms genetics, Skin Neoplasms pathology
- Abstract
Background: NF1-mutated tumours represent a small subset (10-15%) of melanomas, not sufficiently analysed in large clinical cohorts. This study investigated the largest multicentre collection of NF1-mutated melanomas to date., Methods: This study analysed a multicentre tumour tissue sample cohort from 266 patients with NF1-mutated melanoma. Targeted next-generation sequencing of the TERT promoter and 29 relevant melanoma genes was performed. Survival was compared with NF1 wild-type cohorts from the Tissue Registry in Melanoma project (n = 432)., Results: Most NF1-mutated melanoma arose in the head-and-neck region of patients >60 years. NF1 alterations were frequently inactivating, primarily non-sense, less frequently truncating mutations. Non-inactivating NF1 mutations more frequently co-occurred with activating BRAF and RAS mutations. NF1-mutated tumours had higher numbers of gene mutations and UV signature C>T and CC>TT transitions than BRAF, RAS and triple wild-type melanomas. NF1-mutated acral and mucosal melanomas harboured a different mutation signature and were frequent in women (69% and 83%, respectively), differing from non-acral cutaneous NF1-mutated melanomas (men 73%, women 27%). Overall survival in stage IV disease was comparable for patients with NF1-mutated or wild-type melanoma. However, in patients receiving first-line immune checkpoint inhibitor treatment, better median overall survival (mOS) was observed for NF1-mutated than wild-type tumours (mOS = not reached vs mOS = 25.82, p = 0.0154, n = 80 and 432, respectively)., Conclusions: Cutaneous, acral and mucosal NF1-mutated melanomas vary in clinical and genetic characteristics and demonstrate a favourable outcome on immune checkpoint inhibition therapy., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
19. GNA14, GNA11, and GNAQ Mutations Are Frequent in Benign but Not Malignant Cutaneous Vascular Tumors.
- Author
-
Jansen P, Müller H, Lodde GC, Zaremba A, Möller I, Sucker A, Paschen A, Esser S, Schaller J, Gunzer M, Standl F, Bauer S, Schadendorf D, Mentzel T, Hadaschik E, and Griewank KG
- Abstract
Cutaneous vascular tumors consist of a heterogeneous group of benign proliferations, including a range of hemangiomas and vascular malformations, as well as heterogeneous groups of both borderline and malignant neoplasms such as Kaposi's sarcoma and angiosarcomas. The genetics of these tumors have been assessed independently in smaller individual cohorts making comparisons difficult. In our study, we analyzed a representative cohort of benign vascular proliferations observed in a clinical routine setting as well as a selection of malignant vascular proliferations. Our cohort of 104 vascular proliferations including hemangiomas, malformations, angiosarcomas and Kaposi's sarcoma were screened by targeted next-generation sequencing for activating genetic mutations known or assumed to be potentially relevant in vascular proliferations. An association analysis was performed for mutation status and clinico-pathological parameters. Frequent activating hotspot mutations in GNA genes, including GNA14 Q205, GNA11 and GNAQ Q209 were identified in 16 of 64 benign vascular tumors (25%). GNA gene mutations were particularly frequent (52%) in cherry (senile) hemangiomas (13 of 25). In angiosarcomas, activating RAS mutations ( HRAS and NRAS ) were identified in three samples (16%). No activating GNA or RAS gene mutations were identified in Kaposi's sarcomas. Our study identifies GNA14 Q205, GNA11 and GNAQ Q209 mutations as being the most common and mutually exclusive mutations in benign hemangiomas. These mutations were not identified in malignant vascular tumors, which could be of potential diagnostic value in distinguishing these entities., Competing Interests: The 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 reviewer BS declared a past co-authorship with the authors DS, PJ, AP, AS, KG, EH, and AZ to the handling editor., (Copyright © 2021 Jansen, Müller, Lodde, Zaremba, Möller, Sucker, Paschen, Esser, Schaller, Gunzer, Standl, Bauer, Schadendorf, Mentzel, Hadaschik and Griewank.)
- Published
- 2021
- Full Text
- View/download PDF
20. GNAQ and GNA11 mutant nonuveal melanoma: a subtype distinct from both cutaneous and uveal melanoma.
- Author
-
Livingstone E, Zaremba A, Horn S, Ugurel S, Casalini B, Schlaak M, Hassel JC, Herbst R, Utikal JS, Weide B, Gutzmer R, Meier F, Koelsche C, Hadaschik E, Sucker A, Reis H, Merkelbach-Bruse S, Siewert M, Sahm F, von Deimling A, Cosgarea I, Zimmer L, Schadendorf D, Schilling B, and Griewank KG
- Subjects
- DNA Mutational Analysis, GTP-Binding Protein alpha Subunits genetics, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Humans, Mutation genetics, Prospective Studies, Retrospective Studies, Tumor Suppressor Proteins, Ubiquitin Thiolesterase, Melanoma genetics, Skin Neoplasms genetics, Uveal Neoplasms genetics, Uveal Neoplasms therapy
- Abstract
Background: GNAQ and GNA11 mutant nonuveal melanoma represent a poorly characterized rare subgroup of melanoma with a gene mutation profile similar to uveal melanoma., Objectives: To characterize these tumours in terms of clinical behaviour and genetic characteristics., Methods: Patients with nonuveal GNAQ/11 mutated melanoma were identified from the prospective multicentre tumour tissue registry ADOREG, Tissue Registry in Melanoma (TRIM) and additional cooperating skin cancer centres. Extensive data on patient, tumour and treatment characteristics were collected retrospectively. Targeted sequencing was used to determine tumour mutational burden. Immunohistochemistry staining was performed for programmed death-ligand 1 and BRCA1-associated protein (BAP)1. Existing whole-exome cutaneous and uveal melanoma data were analysed for mutation type and burden., Results: We identified 18 patients with metastatic GNAQ/11 mutant nonuveal melanoma. Tumours had a lower tumour mutational burden and fewer ultraviolet signature mutations than cutaneous melanomas. In addition to GNAQ and GNA11 mutations (nine each), six splicing factor 3b subunit 1 (SF3B1), three eukaryotic translation initiation factor 1A X-linked (EIF1AX) and four BAP1 mutations were detected. In contrast to uveal melanoma, GNAQ/11 mutant nonuveal melanomas frequently metastasized lymphatically and concurrent EIF1AX, SF3B1 and BAP1 mutations showed no apparent association with patient prognosis. Objective response to immunotherapy was poor with only one partial response observed in 10 treated patients (10%)., Conclusions: Our findings suggest that GNAQ/11 mutant nonuveal melanomas are a subtype of melanoma that is both clinically and genetically distinct from cutaneous and uveal melanoma. As they respond poorly to available treatment regimens, novel effective therapeutic approaches for affected patients are urgently needed. What is already known about this topic? The rare occurrence of GNAQ/11 mutations in nonuveal melanoma has been documented. GNAQ/11 mutant nonuveal melanomas also harbour genetic alterations in EIF1AX, SF3B1 and BAP1 that are of prognostic relevance in uveal melanoma. What does this study add? GNAQ/11 mutant nonuveal melanomas show metastatic spread reminiscent of cutaneous melanoma, but not uveal melanoma. GNAQ/11 mutant nonuveal melanomas have a low tumour mutational burden that is higher than uveal melanoma, but lower than cutaneous melanoma. What is the translational message? Primary GNAQ/11 mutant nonuveal melanomas are a subtype of melanoma that is clinically and genetically distinct from both cutaneous and uveal melanoma. As metastatic GNAQ/11 mutant nonuveal melanomas respond poorly to available systemic therapies, including immune checkpoint inhibition, novel therapeutic approaches for these tumours are urgently needed. Linked Comment: Rafei-Shamsabadi. Br J Dermatol 2020; 183:806-807., (© 2020 The Authors. British Journal of Dermatology published by John Wiley & Sons Ltd on behalf of British Association of Dermatologists.)
- Published
- 2020
- Full Text
- View/download PDF
21. GNA11 Q209L Mouse Model Reveals RasGRP3 as an Essential Signaling Node in Uveal Melanoma.
- Author
-
Moore AR, Ran L, Guan Y, Sher JJ, Hitchman TD, Zhang JQ, Hwang C, Walczak EG, Shoushtari AN, Monette S, Murali R, Wiesner T, Griewank KG, Chi P, and Chen Y
- Published
- 2020
- Full Text
- View/download PDF
22. [Immunotherapy for malignant melanoma].
- Author
-
Zaremba A, Zimmer L, Griewank KG, Ugurel S, Roesch A, Schadendorf D, and Livingstone E
- Subjects
- Combined Modality Therapy, Humans, Melanoma pathology, Skin Neoplasms pathology, Immunotherapy, Melanoma therapy, Molecular Targeted Therapy methods, Skin Neoplasms therapy
- Abstract
Although cutaneous melanoma accounts for only about 4% of all skin cancers (including nonmelanocytic skin cancer), it is responsible for 80% of all deaths caused by skin cancer. The introduction of immune checkpoint inhibitors led to a significant improvement in long-term survival of patients in an advanced stage regardless of BRAF mutation status. In addition to targeted therapy for patients with BRAF-mutated melanoma, immunotherapies are the therapies of choice in advanced stages and, since 2018, also in the adjuvant setting. The effectiveness of combination therapies and sequences of targeted and immunotherapies are currently being tested.
- Published
- 2020
- Full Text
- View/download PDF
23. Deep Penetrating Nevus and Borderline-Deep Penetrating Nevus: A Literature Review.
- Author
-
Cosgarea I, Griewank KG, Ungureanu L, Tamayo A, and Siepmann T
- Abstract
Deep penetrating nevi (DPN) are rare melanocytic nevi, which can exhibit atypical histological features hampering the differentiation from malignant melanoma. DPN are considered benign melanocytic lesions, but rare spread to lymph nodes and unfavorable clinical outcomes associated with borderline/atypical DPN (B-DPN) has been reported. Since no guidelines are available for DPN and B-DPN, we aimed to review the literature on DPN and B-DPN to assess the management and prognosis. We screened 3,513 references from EMBASE, Scopus and Medline databases, and included 15 studies with a total of 355 DPN patients and 48 B-DPN patients. Therapeutic interventions ranged from simple excision to wide excisions and sentinel lymph node biopsy (SLNB), with block lymph node dissection in some positive SLNB cases. Follow-up periods ranged from 3 months to 23 years during which a total of five recurrences, two in DPN and three in B-DPN group, and three metastases, in B-DPN group, were reported. While some of the included studies comprised clinical and histopathological correlation, few included genetic assessment. The present review highlights the need for prospective cohort studies applying composite measures to identify effective regimens of diagnostic workup and treatment in DPN and B-DPN., (Copyright © 2020 Cosgarea, Griewank, Ungureanu, Tamayo and Siepmann.)
- Published
- 2020
- Full Text
- View/download PDF
24. Re: Deep learning outperformed 11 pathologists in the classification of histopathological melanoma images.
- Author
-
Géraud C and Griewank KG
- Subjects
- Humans, Pathologists, Deep Learning, Melanoma, Skin Neoplasms
- Published
- 2020
- Full Text
- View/download PDF
25. An Animal Model of Cutaneous Cyst Development Enables the Identification of Three Quantitative Trait Loci, Including the Homologue of a Human Locus (TRICY1).
- Author
-
Cosgarea I, Koelsch B, Fischer C, Griewank KG, van den Berg L, Kutritz A, Schadendorf D, and Kindler-Röhrborn A
- Subjects
- Animals, Biopsy, Needle, Disease Models, Animal, Epidermal Cyst pathology, Female, Humans, Immunohistochemistry, Locus Control Region genetics, Male, Random Allocation, Rats, Rats, Inbred BN, Sensitivity and Specificity, Epidermal Cyst genetics, Gene Expression Regulation, Quantitative Trait Loci genetics
- Published
- 2019
- Full Text
- View/download PDF
26. Clinical and genetic analysis of melanomas arising in acral sites.
- Author
-
Zaremba A, Murali R, Jansen P, Möller I, Sucker A, Paschen A, Zimmer L, Livingstone E, Brinker TJ, Hadaschik E, Franklin C, Roesch A, Ugurel S, Schadendorf D, Griewank KG, and Cosgarea I
- Subjects
- Adult, Aged, Aged, 80 and over, Cohort Studies, Female, High-Throughput Nucleotide Sequencing methods, Humans, Male, Melanoma diagnosis, Middle Aged, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins B-raf genetics, Skin Neoplasms diagnosis, Telomerase genetics, Young Adult, Extremities, Genetic Testing methods, Melanoma genetics, Mutation, Skin Neoplasms genetics
- Abstract
Study Aim: Melanomas arising in acral sites are associated with a poorer prognosis than other melanoma subtypes. The aim of this study was to evaluate clinical-pathological and genetic characteristics as well as therapeutic responses of a larger cohort of patients with melanomas arising in acral sites., Methods: Clinical data of 134 patients with melanomas arising in acral sites from the Dept. of Dermatology Essen were collected and analysed with regard to clinicopathological characteristics and treatment responses. Genetic analysis with targeted next-generation sequencing was done on 50 samples., Results: In our cohort, BRAF (30%), NRAS (28%), TERT promoter (26%), NF1 (14%) and KIT (6%) were frequently identified mutations. Comparing tumours situated on palms and soles with melanomas arising on dorsal acral sites, a higher frequency of NRAS (39.1% versus 25%) and NF1 (17.3% versus 0%) and lower frequencies of BRAF (21.7% versus 75%) and TERT promoter (8.6% versus 50%) mutations were observed. MAPK activating mutations were identified in 64% of tumours. Overall survival was longer in patients treated with immune checkpoint inhibitors as first-line treatment than in patients receiving other systemic therapies (i.e. BRAF/MEK inhibitors and chemotherapy)., Conclusion: Our data suggest that the genetics of melanomas arising in acral sites varies by tumour location and may influence biological behaviour., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
27. Assessment of Nonradioactive Multispectral Optoacoustic Tomographic Imaging With Conventional Lymphoscintigraphic Imaging for Sentinel Lymph Node Biopsy in Melanoma.
- Author
-
Stoffels I, Jansen P, Petri M, Goerdt L, Brinker TJ, Griewank KG, Poeppel TD, Schadendorf D, and Klode J
- Subjects
- Adult, Aged, Coloring Agents administration & dosage, Cross-Sectional Studies, Female, Humans, Indocyanine Green administration & dosage, Male, Middle Aged, Radiopharmaceuticals administration & dosage, Technetium Tc 99m Aggregated Albumin administration & dosage, Tomography, X-Ray Computed, Lymphoscintigraphy methods, Melanoma pathology, Photoacoustic Techniques methods, Sentinel Lymph Node Biopsy methods
- Abstract
Importance: The metastatic status of sentinel lymph nodes (SLNs) is the most relevant prognostic factor in breast cancer, melanoma, and other tumors. The conventional standard to label SLNs is lymphoscintigraphy with technetium Tc 99m. A worldwide shortage and known disadvantages of Tc 99m have intensified efforts to establish alternative, nonradioactive imaging techniques., Objective: To assess a new nonradioactive method using multispectral optoacoustic tomographic (MSOT) imaging in comparison with conventional lymphoscintigraphic imaging for SLN biopsy (SLNB) in melanoma., Design, Setting, and Participants: Analysis of a cross-sectional study was conducted at the University Hospital-Essen, Skin Cancer Center, Essen, Germany. Between June 2, 2014, and February 22, 2019, 83 patients underwent SLNB with an additional preoperative indocyanine green (ICG) application. Sentinel lymph node basins were preoperatively identified by MSOT imaging, and ICG-labeled SLNs were intraoperatively detected using a near-infrared camera. The surgeons were blinded to the lymphoscintigraphic imaging results in the beginning of the SLNB. Use of a γ probe was restricted until the SLNB procedure was attempted by the nonradioactive method., Main Outcomes and Measures: Concordance of SLN basins and SLNs identified by MSOT imaging plus near-infrared camera vs lymphoscintigraphic imaging plus single-photon emission computed tomographic or computed tomographic imaging was assessed., Results: Of the 83 patients (mean [SD] age, 54.61 [17.53] years), 47 (56.6%) were men. In 83 surgical procedures, 165 SLNs were excised. The concordance rate of ICG-labeled and Tc 99m-marked detected SLN basins was 94.6% (n = 106 of 112). Intraoperatively, 159 SLNs were detected using a near-infrared camera and 165 were detected by a γ probe, resulting in a concordance rate of 96.4%. Multispectral optoacoustic tomographic imaging visualized SLNs in all anatomic regions with high penetration depth (5 cm)., Conclusions and Relevance: The findings of this study suggest that nonradioactive SLN detection via MSOT imaging allows identification of SLNs at a frequency equivalent to that of the current radiotracer conventional standard. Multispectral optoacoustic tomographic imaging appears to be a viable nonradioactive alternative to detect SLNs in malignant tumors.
- Published
- 2019
- Full Text
- View/download PDF
28. Proton radiotherapy in advanced malignant melanoma of the conjunctiva.
- Author
-
Scholz SL, Hérault J, Stang A, Griewank KG, Meller D, Thariat J, Steuhl KP, Westekemper H, and Sauerwein W
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Conjunctival Neoplasms pathology, Female, Follow-Up Studies, Humans, Male, Melanoma pathology, Middle Aged, Neoplasm Staging, Retrospective Studies, Time Factors, Treatment Outcome, Young Adult, Conjunctiva pathology, Conjunctival Neoplasms radiotherapy, Melanoma radiotherapy, Proton Therapy methods
- Abstract
Background: The management of conjunctival melanoma is challenging and frequently ends in exenteration. The aim of this retrospective study was to evaluate the long-term results of proton beam radiation with regard to various clinical parameters., Methods: Eighty-nine patients with extended conjunctival melanoma (≥T2) and multifocal bulbar located tumors (T1c/d) were treated consecutively with proton radiotherapy (dose 45 Gy). The following parameters were assessed: TNM stage, tumor origin, local recurrence, performance of exenteration, occurrence of metastases, overall survival, and potential complications. A time-to-event analysis was preformed to the primary endpoints: relapse, metastasis, exenteration, and death by use of Kaplan-Meier cumulative survival estimates and Cox proportional hazards regression that provides hazard ratios and 95% confidence intervals., Results: The median follow-up time was 4.2 years (max. 21.7 years). Local recurrence and metastatic disease occurred in 33% and 16% of patients, respectively. Exenteration-free survival and overall survival tended to be worse in T3 melanoma. No association between tumor origin and local recurrence, metastatic disease, or overall survival was observed. Main complications after proton radiotherapy were sicca-syndrome (30%), secondary glaucoma (11%), and limbal stem cell deficiency (8%)., Conclusions: In summary, proton radiotherapy in conjunctival melanoma is an effective alternative to exenteration, with a 5-year cumulative probability of eye preservation of 69%.
- Published
- 2019
- Full Text
- View/download PDF
29. Frequent Occurrence of NRAS and BRAF Mutations in Human Acral Naevi.
- Author
-
Jansen P, Cosgarea I, Murali R, Möller I, Sucker A, Franklin C, Paschen A, Zaremba A, Brinker TJ, Stoffels I, Schadendorf D, Klode J, Hadaschik E, and Griewank KG
- Abstract
Acral naevi are benign melanocytic tumors occurring at acral sites. Occasionally they can progress to become malignant tumors (melanomas). The genetics of acral naevi have not been assessed in larger studies. In our study, a large cohort of 130 acral naevi was screened for gene mutations known to be important in other naevi and melanoma subtypes by targeted next-generation sequencing. Mutation status was correlated with clinicopathological parameters. Frequent mutations in genes activating the MAP kinase pathway were identified, including n = 87 (67%) BRAF , n = 24 (18%) NRAS , and one (1%) MAP2K1 mutations. BRAF mutations were almost exclusively V600E ( n = 86, 99%) and primarily found in junctional and compound naevi. NRAS mutations were either Q61K or Q61R and frequently identified in dermal naevi. Recurrent non-V600E BRAF , KIT , NF1 , and TERT promoter mutations, present in acral melanoma, were not identified. Our study identifies BRAF and NRAS mutations as the primary pathogenic event in acral naevi, however, distributed differently to those in non-acral naevi. The mutational profile of acral naevi is distinct from acral melanoma, which may be of diagnostic value in distinguishing these entities.
- Published
- 2019
- Full Text
- View/download PDF
30. [Digital papillary adenocarcinoma : Four case reports with brief literature review].
- Author
-
Held L, Mentzel T, Paredes BE, Griewank KG, Itzlinger-Monshi B, and Rütten A
- Subjects
- Adenocarcinoma, Papillary surgery, Adult, Female, Fingers pathology, Humans, Male, Middle Aged, Neoplasm Recurrence, Local, Sweat Gland Neoplasms surgery, Treatment Outcome, Adenocarcinoma, Papillary pathology, Sweat Gland Neoplasms pathology, Sweat Glands pathology
- Abstract
Digital papillary adenocarcinoma is a rare but well characterized carcinoma of the sweat glands, which apart from very few exceptions is localized in acral skin. This type of sweat gland carcinoma tends to recur locally and may cause delayed metastases in a few cases. We describe the clinical findings and the broad histopathologic spectrum of four cases of this rare adnexal carcinoma and give a short summary of the literature.
- Published
- 2019
- Full Text
- View/download PDF
31. Genome-wide methylation profiling and copy number analysis in atypical fibroxanthomas and pleomorphic dermal sarcomas indicate a similar molecular phenotype.
- Author
-
Koelsche C, Stichel D, Griewank KG, Schrimpf D, Reuss DE, Bewerunge-Hudler M, Vokuhl C, Dinjens WNM, Petersen I, Mittelbronn M, Cuevas-Bourdier A, Buslei R, Pfister SM, Flucke U, Mechtersheimer G, Mentzel T, and von Deimling A
- Abstract
Background: Atypical fibroxanthomas (AFX) and pleomorphic dermal sarcomas (PDS) are lesions of the skin with overlapping histologic features and unspecific molecular traits. PDS behaves aggressive compared to AFX. Thus, a precise delineation, although challenging in some instances, is relevant., Methods: We examined the value of DNA-methylation profiling and copy number analysis for separating these tumors. DNA-methylation data were generated from 17 AFX and 15 PDS using the Illumina EPIC array. These were compared with DNA-methylation data generated from 196 tumors encompassing potential histologic mimics like cutaneous squamous carcinomas (cSCC; n = 19), basal cell carcinomas (n = 10), melanoma metastases originating from the skin (n = 11), leiomyosarcomas (n = 11), angiosarcomas of the skin and soft tissue (n = 11), malignant peripheral nerve sheath tumors (n = 19), dermatofibrosarcomas protuberans (n = 13), extraskeletal myxoid chondrosarcomas (n = 9), myxoid liposarcomas (n = 14), schwannomas (n = 10), neurofibromas (n = 21), alveolar (n = 19) and embryonal (n = 17) rhabdomyosarcomas as well as undifferentiated pleomorphic sarcomas (n = 12)., Results: DNA-methylation profiling did not separate AFX from PDS. The DNA-methylation profiles of the other cases, however, were distinct from AFX/PDS. They reliably assigned to subtype-specific DNA-methylation clusters, although overlap occurred between some AFX/PDS and cSCC. Copy number profiling revealed alterations in a similar frequency and distribution between AFX and PDS. They involved losses of 9p (22/32) and 13q (25/32). Gains frequently involved 8q (8/32). Notably, a homozygous deletion of CDKN2A was more frequent in PDS (6/15) than in AFX (2/17), whereas amplifications were non-recurrent and overall rare (5/32)., Conclusions: Our findings support the concept that AFX and PDS belong to a common tumor spectrum. We could demonstrate the diagnostic value of DNA-methylation profiling to delineating AFX/PDS from potential mimics. However, the assessment of certain histologic features remains crucial for separating PDS from AFX.
- Published
- 2019
- Full Text
- View/download PDF
32. Re: van Poppelen et al.: Genetic background of iris melanomas and iris melanocytic tumors of uncertain malignant potential (Ophthalmology. 2018;125:904-912).
- Author
-
Scholz SL, Murali R, Westekemper H, Schadendorf D, and Griewank KG
- Subjects
- Genetic Background, Humans, Iris, Iris Neoplasms, Melanoma, Uveal Neoplasms
- Published
- 2018
- Full Text
- View/download PDF
33. Melanoma.
- Author
-
Schadendorf D, van Akkooi ACJ, Berking C, Griewank KG, Gutzmer R, Hauschild A, Stang A, Roesch A, and Ugurel S
- Subjects
- Age Distribution, Antineoplastic Agents therapeutic use, Humans, Incidence, Mass Screening, Neoplasm Staging, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Mas, Randomized Controlled Trials as Topic, Risk Factors, Sunlight adverse effects, Ultraviolet Rays adverse effects, Melanoma, Cutaneous Malignant, Melanoma epidemiology, Melanoma etiology, Melanoma mortality, Melanoma therapy, Neoplasm Metastasis therapy, Skin Neoplasms epidemiology, Skin Neoplasms etiology, Skin Neoplasms mortality, Skin Neoplasms therapy
- Abstract
Cutaneous melanoma causes 55 500 deaths annually. The incidence and mortality rates of the disease differ widely across the globe depending on access to early detection and primary care. Once melanoma has spread, this type of cancer rapidly becomes life-threatening. For more than 40 years, few treatment options were available, and clinical trials during that time were all unsuccessful. Over the past 10 years, increased biological understanding and access to innovative therapeutic substances have transformed advanced melanoma into a new oncological model for treating solid cancers. Treatments that target B-Raf proto-oncogene serine/threonine-kinase (BRAF)
V600 (Val600) mutations using selected BRAF inhibitors combined with mitogen-activated protein kinase inhibitors have significantly improved response and overall survival. Furthermore, advanced cutaneous melanoma has developed into a prototype for testing checkpoint-modulating agents, which has increased hope for long-term tumour containment and a potential cure. These expectations have been sustained by clinical success with targeted agents and antibodies that block programmed cell-death protein 1 in locoregional disease, which induces prolongation of relapse-free, distant-metastasis-free, and overall survival times., (Copyright © 2018 Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
34. Integrated Genomic Classification of Melanocytic Tumors of the Central Nervous System Using Mutation Analysis, Copy Number Alterations, and DNA Methylation Profiling.
- Author
-
Griewank KG, Koelsche C, van de Nes JAP, Schrimpf D, Gessi M, Möller I, Sucker A, Scolyer RA, Buckland ME, Murali R, Pietsch T, von Deimling A, and Schadendorf D
- Subjects
- Adult, Aged, Aged, 80 and over, Central Nervous System Neoplasms classification, Central Nervous System Neoplasms pathology, Chromosomes, Human, Pair 3 genetics, DNA Copy Number Variations genetics, DNA Methylation genetics, DNA Mutational Analysis, Female, Genomics, Humans, Male, Melanoma classification, Melanoma pathology, Middle Aged, Mutation genetics, Neoplasm Metastasis, Nevus, Blue classification, Nevus, Blue genetics, Nevus, Blue pathology, Sequence Analysis, DNA, Skin Neoplasms classification, Skin Neoplasms pathology, Tumor Suppressor Proteins genetics, Ubiquitin Thiolesterase genetics, Uveal Neoplasms classification, Uveal Neoplasms pathology, Melanoma, Cutaneous Malignant, Central Nervous System Neoplasms genetics, Melanoma genetics, Skin Neoplasms genetics, Uveal Neoplasms genetics
- Abstract
Purpose: In the central nervous system, distinguishing primary leptomeningeal melanocytic tumors from melanoma metastases and predicting their biological behavior solely using histopathologic criteria may be challenging. We aimed to assess the diagnostic and prognostic value of integrated molecular analysis. Experimental Design: Targeted next-generation sequencing, array-based genome-wide methylation analysis, and BAP1 IHC were performed on the largest cohort of central nervous system melanocytic tumors analyzed to date, including 47 primary tumors of the central nervous system, 16 uveal melanomas, 13 cutaneous melanoma metastases, and 2 blue nevus-like melanomas. Gene mutation, DNA-methylation, and copy-number profiles were correlated with clinicopathologic features. Results: Combining mutation, copy-number, and DNA-methylation profiles clearly distinguished cutaneous melanoma metastases from other melanocytic tumors. Primary leptomeningeal melanocytic tumors, uveal melanomas, and blue nevus-like melanoma showed common DNA-methylation, copy-number alteration, and gene mutation signatures. Notably, tumors demonstrating chromosome 3 monosomy and BAP1 alterations formed a homogeneous subset within this group. Conclusions: Integrated molecular profiling aids in distinguishing primary from metastatic melanocytic tumors of the central nervous system. Primary leptomeningeal melanocytic tumors, uveal melanoma, and blue nevus-like melanoma share molecular similarity with chromosome 3 and BAP1 alterations, markers of poor prognosis. Clin Cancer Res; 24(18); 4494-504. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
35. Intraventricular melanocytoma diagnosis confirmed by gene mutation profile.
- Author
-
Knappe UJ, Tischoff I, Tannapfel A, Reinbold WD, Möller I, Sucker A, Schadendorf D, Griewank KG, and van de Nes JAP
- Subjects
- Aged, 80 and over, Brain pathology, Cerebral Ventricle Neoplasms complications, Eukaryotic Initiation Factor-1 genetics, Humans, Male, Meningeal Neoplasms complications, Mutation, Receptors, Leukotriene genetics, Cerebral Ventricle Neoplasms diagnosis, Cerebral Ventricle Neoplasms genetics, Melanocytes pathology, Meningeal Neoplasms diagnosis, Meningeal Neoplasms genetics
- Abstract
Primary leptomeningeal melanocytic tumors (PLMTs) are rare. They usually arise along the spinal cord and at the skull base. Here we report on a patient with a very rare intraventricular melanocytoma. Histologically, a melanocytic tumor was clearly diagnosed. However, to make the uncommon diagnosis of an intraventricular melanocytoma, metastatic melanoma needed to be excluded. Next generation sequencing covering gene mutations that may occur in PLMTs and cutaneous melanoma was performed. The unique gene mutation profile detected, consisting of an activating CYSLTR2 L129Q mutation and EIF1AX G9R mutation and a lack of mutations in genes known to occur in metastatic melanoma (i.e. BRAF or NRAS) confirmed the diagnosis of an intraventricular melanocytoma. This case report is the second intraventricular melanocytoma published to date and demonstrates the value of applying novel genetic assays to make this diagnosis., (© 2017 Japanese Society of Neuropathology.)
- Published
- 2018
- Full Text
- View/download PDF
36. Tumor CDKN2A-Associated JAK2 Loss and Susceptibility to Immunotherapy Resistance.
- Author
-
Horn S, Leonardelli S, Sucker A, Schadendorf D, Griewank KG, and Paschen A
- Subjects
- Cell Line, Tumor, Gene Deletion, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Genetic Predisposition to Disease, High-Throughput Nucleotide Sequencing, Humans, Melanoma genetics, Melanoma pathology, Melanoma therapy, Mutation, Neoplasms genetics, Neoplasms pathology, Polymorphism, Single Nucleotide, Skin Neoplasms genetics, Skin Neoplasms pathology, Skin Neoplasms therapy, Treatment Failure, Cyclin-Dependent Kinase Inhibitor p16 genetics, Drug Resistance, Neoplasm genetics, Immunologic Factors therapeutic use, Immunotherapy, Janus Kinase 2 genetics, Neoplasms therapy
- Abstract
Poor clinical responses to checkpoint blockade with anti-CTLA-4 and anti-PD-1 antibodies in melanoma have recently been associated with acquired IFNγ resistance that protects tumor cells from the antiproliferative and pro-apoptotic cytokine activity. IFNγ-resistant melanoma cells very often lack functional expression of the IFNγ signaling pathway gene JAK2 due to gene deletions or inactivating gene mutations. Analyzing melanoma cell lines (n = 46, applying next-generation targeted sequencing and single nucleotide polymorphism arrays) as well as available genomic data sets from The Cancer Genome Atlas (TCGA) tumor tissue samples (cutaneous melanoma n = 367, lung squamous cell carcinoma n = 501, bladder urothelial carcinoma n = 408, breast invasive carcinoma n = 768, colorectal adenocarcinoma n = 257), we demonstrate that the frequent chromosomal losses of the tumor suppressor CDKN2A in melanoma and other tumor entities enhance the susceptibility to IFNγ resistance by concomitant deletion of the JAK2 gene (odds ratio = 223.17, 95% confidence interval = 66.91 to 1487.38, two-sided P = 7.6×10-46). Tumors with JAK2 mutations or homozygous JAK2 deletions demonstrate allelic losses covering both CDKN2A and JAK2. This suggests that patients with tumor chromosomal CDKN2A losses are susceptible to developing immunotherapy resistance and should be screened for JAK2 deficiency prior to and under immune checkpoint blocking therapy.
- Published
- 2018
- Full Text
- View/download PDF
37. NF1 mutations in conjunctival melanoma.
- Author
-
Scholz SL, Cosgarea I, Süßkind D, Murali R, Möller I, Reis H, Leonardelli S, Schilling B, Schimming T, Hadaschik E, Franklin C, Paschen A, Sucker A, Steuhl KP, Schadendorf D, Westekemper H, and Griewank KG
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor genetics, Cohort Studies, Conjunctival Neoplasms pathology, DNA Mutational Analysis methods, Female, High-Throughput Nucleotide Sequencing, Humans, Male, Melanoma pathology, Middle Aged, Proto-Oncogene Proteins B-raf genetics, ras Proteins genetics, Conjunctival Neoplasms genetics, Melanoma genetics, Mutation, Neurofibromin 1 genetics
- Abstract
Background: Conjunctival melanoma is a potentially deadly eye tumour. Despite effective local therapies, tumour recurrence and metastasis remain frequent. The genetics of conjunctival melanomas remain incompletely understood., Methods: A large cohort of 63 conjunctival melanomas was screened for gene mutations known to be important in other melanoma subtypes by targeted next-generation sequencing. Mutation status was correlated with patient prognosis., Results: Frequent mutations in genes activating the MAP kinase pathway were identified. NF1 mutations were most frequent (n = 21, 33%). Recurrent activating mutations were also identified in BRAF (n = 16, 25%) and RAS genes (n = 12, 19%; 11 NRAS and 1 KRAS)., Conclusions: Similar to cutaneous melanomas, conjunctival melanomas can be grouped genetically into four groups: BRAF-mutated, RAS-mutated, NF1-mutated and triple wild-type melanomas. This genetic classification may be useful for assessment of therapeutic options for patients with metastatic conjunctival melanoma.
- Published
- 2018
- Full Text
- View/download PDF
38. The genetic basis for most patients with pustular skin disease remains elusive.
- Author
-
Mössner R, Wilsmann-Theis D, Oji V, Gkogkolou P, Löhr S, Schulz P, Körber A, Prinz JC, Renner R, Schäkel K, Vogelsang L, Peters KP, Philipp S, Reich K, Ständer H, Jacobi A, Weyergraf A, Kingo K, Kõks S, Gerdes S, Steinz K, Schill T, Griewank KG, Müller M, Frey S, Ebertsch L, Uebe S, Sticherling M, Sticht H, and Hüffmeier U
- Subjects
- Adult, CARD Signaling Adaptor Proteins genetics, Female, Genetic Predisposition to Disease genetics, Genetic Testing, Guanylate Cyclase genetics, Heterozygote, Humans, Male, Membrane Proteins genetics, Middle Aged, Vesicular Transport Proteins genetics, Interleukins genetics, Mutation genetics, Psoriasis genetics
- Abstract
Background: Rare variants in the genes IL36RN, CARD14 and AP1S3 have been identified to cause or contribute to pustular skin diseases, primarily generalized pustular psoriasis (GPP)., Objectives: To better understand the disease relevance of these genes, we screened our cohorts of patients with pustular skin diseases [primarily GPP and palmoplantar pustular psoriasis (PPP)] for coding changes in these three genes. Carriers of single heterozygous IL36RN mutations were screened for a second mutation in IL36RN., Methods: Coding exons of IL36RN, CARD14 and AP1S3 were sequenced in 67 patients - 61 with GPP, two with acute generalized exanthematous pustulosis and four with acrodermatitis continua of Hallopeau. We screened IL36RN and AP1S3 for intragenic copy-number variants and 258 patients with PPP for coding changes in AP1S3. Eleven heterozygous IL36RN mutations carriers were analysed for a second noncoding IL36RN mutation. Genotype-phenotype correlations in carriers/noncarriers of IL36RN mutations were assessed within the GPP cohort., Results: The majority of patients (GPP, 64%) did not carry rare variants in any of the three genes. Biallelic and monoallelic IL36RN mutations were identified in 15 and five patients with GPP, respectively. Noncoding rare IL36RN variants were not identified in heterozygous carriers. The only significant genotype-phenotype correlation observed for IL36RN mutation carriers was early age at disease onset. Additional rare CARD14 or AP1S3 variants were identified in 15% of IL36RN mutation carriers., Conclusions: The identification of IL36RN mutation carriers harbouring additional rare variants in CARD14 or AP1S3 indicates a more complex mode of inheritance of pustular psoriasis. Our results suggest that, in heterozygous IL36RN mutation carriers, there are additional disease-causing genetic factors outside IL36RN., (© 2017 The Authors. British Journal of Dermatology published by John Wiley & Sons Ltd on behalf of British Association of Dermatologists.)
- Published
- 2018
- Full Text
- View/download PDF
39. Atypical fibroxanthoma and pleomorphic dermal sarcoma harbor frequent NOTCH1/2 and FAT1 mutations and similar DNA copy number alteration profiles.
- Author
-
Griewank KG, Wiesner T, Murali R, Pischler C, Müller H, Koelsche C, Möller I, Franklin C, Cosgarea I, Sucker A, Schadendorf D, Schaller J, Horn S, Brenn T, and Mentzel T
- Subjects
- Aged, Aged, 80 and over, Comparative Genomic Hybridization, Cyclin-Dependent Kinase Inhibitor p16 genetics, DNA Copy Number Variations, Female, Humans, Male, Middle Aged, Mutation, Promoter Regions, Genetic, Telomerase genetics, Tumor Suppressor Protein p53 genetics, Cadherins genetics, Receptor, Notch1 genetics, Receptor, Notch2 genetics, Sarcoma genetics, Skin Neoplasms genetics
- Abstract
Atypical fibroxanthomas and pleomorphic dermal sarcomas are tumors arising in sun-damaged skin of elderly patients. They have differing prognoses and are currently distinguished using histological criteria, such as invasion of deeper tissue structures, necrosis and lymphovascular or perineural invasion. To investigate the as-yet poorly understood genetics of these tumors, 41 atypical fibroxanthomas and 40 pleomorphic dermal sarcomas were subjected to targeted next-generation sequencing approaches as well as DNA copy number analysis by comparative genomic hybridization. In an analysis of the entire coding region of 341 oncogenes and tumor suppressor genes in 13 atypical fibroxanthomas using an established hybridization-based next-generation sequencing approach, we found that these tumors harbor a large number of mutations. Gene alterations were identified in more than half of the analyzed samples in FAT1, NOTCH1/2, CDKN2A, TP53, and the TERT promoter. The presence of these alterations was verified in 26 atypical fibroxanthoma and 35 pleomorphic dermal sarcoma samples by targeted amplicon-based next-generation sequencing. Similar mutation profiles in FAT1, NOTCH1/2, CDKN2A, TP53, and the TERT promoter were identified in both atypical fibroxanthoma and pleomorphic dermal sarcoma. Activating RAS mutations (G12 and G13) identified in 3 pleomorphic dermal sarcoma were not found in atypical fibroxanthoma. Comprehensive DNA copy number analysis demonstrated a wide array of different copy number gains and losses, with similar profiles in atypical fibroxanthoma and pleomorphic dermal sarcoma. In summary, atypical fibroxanthoma and pleomorphic dermal sarcoma are highly mutated tumors with recurrent mutations in FAT1, NOTCH1/2, CDKN2A, TP53, and the TERT promoter, and a range of DNA copy number alterations. These findings suggest that atypical fibroxanthomas and pleomorphic dermal sarcomas are genetically related, potentially representing two ends of a common tumor spectrum and distinguishing these entities is at present still best performed using histological criteria.
- Published
- 2018
- Full Text
- View/download PDF
40. GNA11 Q209L Mouse Model Reveals RasGRP3 as an Essential Signaling Node in Uveal Melanoma.
- Author
-
Moore AR, Ran L, Guan Y, Sher JJ, Hitchman TD, Zhang JQ, Hwang C, Walzak EG, Shoushtari AN, Monette S, Murali R, Wiesner T, Griewank KG, Chi P, and Chen Y
- Subjects
- Animals, Cell Line, Tumor, Cell Lineage drug effects, Cell Proliferation drug effects, Central Nervous System Neoplasms pathology, Disease Models, Animal, Female, Humans, Male, Melanocytes drug effects, Melanocytes pathology, Mitogen-Activated Protein Kinase Kinases metabolism, Mitogen-Activated Protein Kinases metabolism, Neoplasm Invasiveness, Protein Kinase Inhibitors pharmacology, Skin Neoplasms pathology, Tumor Suppressor Proteins metabolism, Ubiquitin Thiolesterase metabolism, GTP-Binding Protein alpha Subunits metabolism, Melanocytes metabolism, Melanoma metabolism, Melanoma pathology, Signal Transduction drug effects, Uveal Neoplasms metabolism, Uveal Neoplasms pathology, ras Guanine Nucleotide Exchange Factors metabolism
- Abstract
Uveal melanoma (UM) is characterized by mutually exclusive activating mutations in GNAQ, GNA11, CYSLTR2, and PLCB4, four genes in a linear pathway to activation of PLCβ in almost all tumors and loss of BAP1 in the aggressive subset. We generated mice with melanocyte-specific expression of GNA11
Q209L with and without homozygous Bap1 loss. The GNA11Q209L mice recapitulated human Gq-associated melanomas, and they developed pigmented neoplastic lesions from melanocytes of the skin and non-cutaneous organs, including the eye and leptomeninges, as well as at atypical sites, including the lymph nodes and lungs. The addition of Bap1 loss increased tumor proliferation and cutaneous melanoma size. Integrative transcriptome analysis of human and murine melanomas identified RasGRP3 to be specifically expressed in GNAQ/GNA11-driven melanomas. In human UM cell lines and murine models, RasGRP3 is specifically required for GNAQ/GNA11-driven Ras activation and tumorigenesis. This implicates RasGRP3 as a critical node and a potential target in UM., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
41. Integrative Analysis Identifies Four Molecular and Clinical Subsets in Uveal Melanoma.
- Author
-
Robertson AG, Shih J, Yau C, Gibb EA, Oba J, Mungall KL, Hess JM, Uzunangelov V, Walter V, Danilova L, Lichtenberg TM, Kucherlapati M, Kimes PK, Tang M, Penson A, Babur O, Akbani R, Bristow CA, Hoadley KA, Iype L, Chang MT, Cherniack AD, Benz C, Mills GB, Verhaak RGW, Griewank KG, Felau I, Zenklusen JC, Gershenwald JE, Schoenfield L, Lazar AJ, Abdel-Rahman MH, Roman-Roman S, Stern MH, Cebulla CM, Williams MD, Jager MJ, Coupland SE, Esmaeli B, Kandoth C, and Woodman SE
- Published
- 2018
- Full Text
- View/download PDF
42. Trametinib-Induced Remission of an MEK1 -Mutated Langerhans Cell Histiocytosis.
- Author
-
Papapanagiotou M, Griewank KG, Hillen U, Schimming TT, Moeller LC, Führer D, Zimmer L, Roesch A, Sucker A, Schadendorf D, Livingstone E, and Schilling B
- Published
- 2017
- Full Text
- View/download PDF
43. Activating CYSLTR2 and PLCB4 Mutations in Primary Leptomeningeal Melanocytic Tumors.
- Author
-
van de Nes JAP, Koelsche C, Gessi M, Möller I, Sucker A, Scolyer RA, Buckland ME, Pietsch T, Murali R, Schadendorf D, and Griewank KG
- Subjects
- Adult, Aged, DNA Mutational Analysis, Female, Genetic Predisposition to Disease, Humans, Male, Melanoma metabolism, Meningeal Neoplasms metabolism, Middle Aged, Phospholipase C beta metabolism, Receptors, Leukotriene metabolism, Young Adult, DNA, Neoplasm genetics, Melanoma genetics, Meningeal Neoplasms genetics, Mutation, Phospholipase C beta genetics, Receptors, Leukotriene genetics
- Published
- 2017
- Full Text
- View/download PDF
44. Integrative Analysis Identifies Four Molecular and Clinical Subsets in Uveal Melanoma.
- Author
-
Robertson AG, Shih J, Yau C, Gibb EA, Oba J, Mungall KL, Hess JM, Uzunangelov V, Walter V, Danilova L, Lichtenberg TM, Kucherlapati M, Kimes PK, Tang M, Penson A, Babur O, Akbani R, Bristow CA, Hoadley KA, Iype L, Chang MT, Cherniack AD, Benz C, Mills GB, Verhaak RGW, Griewank KG, Felau I, Zenklusen JC, Gershenwald JE, Schoenfield L, Lazar AJ, Abdel-Rahman MH, Roman-Roman S, Stern MH, Cebulla CM, Williams MD, Jager MJ, Coupland SE, Esmaeli B, Kandoth C, and Woodman SE
- Subjects
- DNA Copy Number Variations, Eukaryotic Initiation Factor-1 genetics, Humans, Melanoma classification, Monosomy, Phosphoproteins genetics, Prognosis, RNA Splicing Factors genetics, Serine-Arginine Splicing Factors genetics, Tumor Suppressor Proteins genetics, Ubiquitin Thiolesterase genetics, Uveal Neoplasms classification, Biomarkers, Tumor genetics, DNA Methylation, Gene Expression Regulation, Neoplastic, Melanoma genetics, Mutation, Uveal Neoplasms genetics
- Abstract
Comprehensive multiplatform analysis of 80 uveal melanomas (UM) identifies four molecularly distinct, clinically relevant subtypes: two associated with poor-prognosis monosomy 3 (M3) and two with better-prognosis disomy 3 (D3). We show that BAP1 loss follows M3 occurrence and correlates with a global DNA methylation state that is distinct from D3-UM. Poor-prognosis M3-UM divide into subsets with divergent genomic aberrations, transcriptional features, and clinical outcomes. We report change-of-function SRSF2 mutations. Within D3-UM, EIF1AX- and SRSF2/SF3B1-mutant tumors have distinct somatic copy number alterations and DNA methylation profiles, providing insight into the biology of these low- versus intermediate-risk clinical mutation subtypes., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
45. SF3B1 and BAP1 mutations in blue nevus-like melanoma.
- Author
-
Griewank KG, Müller H, Jackett LA, Emberger M, Möller I, van de Nes JA, Zimmer L, Livingstone E, Wiesner T, Scholz SL, Cosgarea I, Sucker A, Schimming T, Hillen U, Schilling B, Paschen A, Reis H, Mentzel T, Kutzner H, Rütten A, Murali R, Scolyer RA, and Schadendorf D
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Diagnosis, Differential, GTP-Binding Protein alpha Subunits genetics, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, Humans, Melanoma genetics, Melanoma pathology, Middle Aged, Mutation, Nevus, Blue genetics, Nevus, Blue pathology, Skin Neoplasms genetics, Skin Neoplasms pathology, Young Adult, Melanoma diagnosis, Nevus, Blue diagnosis, Phosphoproteins genetics, RNA Splicing Factors genetics, Skin Neoplasms diagnosis, Tumor Suppressor Proteins genetics, Ubiquitin Thiolesterase genetics
- Abstract
Blue nevi are melanocytic tumors originating in the cutaneous dermis. Malignant tumors may arise in association with or resembling blue nevi, so called 'blue nevus-like melanoma', which can metastasize and result in patient death. Identifying which tumors will behave in a clinically aggressive manner can be challenging. Identifying genetic alterations in such tumors may assist in their diagnosis and prognostication. Blue nevi are known to be genetically related to uveal melanomas (eg, both harboring GNAQ and GNA11 mutations). In this study, we analyzed a large cohort (n=301) of various morphologic variants of blue nevi and related tumors including tumors diagnosed as atypical blue nevi (n=21), and blue nevus-like melanoma (n=12), screening for all gene mutations known to occur in uveal melanoma. Similar to published reports, we found the majority of blue nevi harbored activating mutations in GNAQ (53%) or GNA11 (15%). In addition, rare CYSLTR2 (1%) and PLCB4 (1%) mutations were identified. EIF1AX, SF3B1, and BAP1 mutations were also detected, with BAP1 and SF3B1 R625 mutations being present only in clearly malignant tumors (17% (n=2) and 25% (n=3) of blue nevus-like melanoma, respectively). In sequencing data from a larger cohort of cutaneous melanomas, this genetic profile was also identified in tumors not originally diagnosed as blue nevus-like melanoma. Our findings suggest that the genetic profile of coexistent GNAQ or GNA11 mutations with BAP1 or SF3B1 mutations can aid the histopathological diagnosis of blue nevus-like melanoma and distinguish blue nevus-like melanoma from conventional epidermal-derived melanomas. Future studies will need to further elucidate the prognostic implications and appropriate clinical management for patients with tumors harboring these mutation profiles.
- Published
- 2017
- Full Text
- View/download PDF
46. Frequent GNAQ, GNA11, and EIF1AX Mutations in Iris Melanoma.
- Author
-
Scholz SL, Möller I, Reis H, Süßkind D, van de Nes JAP, Leonardelli S, Schilling B, Livingstone E, Schimming T, Paschen A, Sucker A, Murali R, Steuhl KP, Schadendorf D, Westekemper H, and Griewank KG
- Subjects
- Aged, DNA Mutational Analysis, Eukaryotic Initiation Factor-1 metabolism, Female, GTP-Binding Protein alpha Subunits metabolism, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Humans, Iris Neoplasms metabolism, Iris Neoplasms pathology, Male, Melanoma metabolism, Melanoma pathology, Middle Aged, DNA, Neoplasm genetics, Eukaryotic Initiation Factor-1 genetics, GTP-Binding Protein alpha Subunits genetics, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, Iris Neoplasms genetics, Melanoma genetics, Mutation
- Abstract
Purpose: The most common malignant intraocular tumors with a high mortality in adults are uveal melanomas. Uveal melanomas arise most frequently in the choroid or ciliary body (97%) and rarely in the iris (3%). Whereas conjunctival and posterior uveal (ciliary body and choroidal) melanomas have been studied in more detail genetically, little data exist regarding iris melanomas., Methods: In our study, we genetically analyzed 19 iris melanomas, 8 ciliary body melanomas, 3 ring melanomas, and 4 iris nevi. A targeted next-generation sequencing approach was applied, covering the mutational hotspot regions of nine genes known to be mutated in conjunctival and uveal melanoma (BRAF, NRAS, KIT, GNAQ, GNA11, CYSLTR2, SF3B1, EIF1AX, and BAP1)., Results: Activating GNAQ or GNA11 hotspot mutations were detected in a mutually exclusive fashion in 84% (16/19) of iris melanomas. EIF1AX gene mutations also were frequent, detected in 42% (8/19) of iris melanomas. In 4 iris nevi, one GNAQ mutation was identified. GNAQ, GNA11, EIF1AX, and BAP1 mutations were identified at varying frequencies in ciliary body and ring melanomas., Conclusions: In this most comprehensive genetic analysis of iris melanomas published to date, we find iris melanomas to be related genetically to choroidal and ciliary body melanomas, frequently harboring GNAQ, GNA11, and EIF1AX mutations. Future studies will need to assess if screening mutation profiles in iris melanomas may be of diagnostic or prognostic value.
- Published
- 2017
- Full Text
- View/download PDF
47. Targeted next generation sequencing of mucosal melanomas identifies frequent NF1 and RAS mutations.
- Author
-
Cosgarea I, Ugurel S, Sucker A, Livingstone E, Zimmer L, Ziemer M, Utikal J, Mohr P, Pfeiffer C, Pföhler C, Hillen U, Horn S, Schadendorf D, Griewank KG, and Roesch A
- Subjects
- Adult, Aged, Aged, 80 and over, Base Sequence, Female, Gene Frequency, Genetic Predisposition to Disease genetics, High-Throughput Nucleotide Sequencing methods, Humans, Male, Middle Aged, Mucous Membrane pathology, GTP Phosphohydrolases genetics, Melanoma genetics, Membrane Proteins genetics, Mucous Membrane metabolism, Mutation, Neurofibromin 1 genetics, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
Purpose: Mucosal melanoma represents ~1% of all melanomas, frequently having a poor prognosis due to diagnosis at a late stage of disease. Mucosal melanoma differs from cutaneous melanoma not only in terms of poorer clinical outcome but also on the molecular level having e.g. less BRAF and more frequent KIT mutations than cutaneous melanomas. For the majority of mucosal melanomas oncogenic driver mutations remain unknown., Experimental Design and Results: In our study, 75 tumor tissues from patients diagnosed with mucosal melanoma were analyzed, applying a targeted next generation sequencing panel covering 29 known recurrently mutated genes in melanoma. NF1 and RAS mutations were identified as the most frequently mutated genes occurring in 18.3% and 16.9% of samples, respectively. Mutations in BRAF were identified in 8.4% and KIT in 7.0% of tumor samples., Conclusions: Our study identifies NF1 as the most frequently occurring driver mutation in mucosal melanoma. RAS alterations, consisting of NRAS and KRAS mutations, were the second most frequent mutation type. BRAF and KIT mutations were rare with frequencies below 10% each. Our data indicate that in mucosal melanomas RAS/NF1 alterations are frequent, implying a significant pathogenetic role for MAPK and potentially PI3K pathway activation in these tumors.
- Published
- 2017
- Full Text
- View/download PDF
48. Next-Generation Sequencing to Guide Treatment of Advanced Melanoma.
- Author
-
Griewank KG and Schilling B
- Subjects
- High-Throughput Nucleotide Sequencing methods, Humans, Immunotherapy methods, Melanoma genetics, Melanoma pathology, Mutation, Patient Selection, Proto-Oncogene Proteins B-raf genetics, Skin Neoplasms genetics, Skin Neoplasms pathology, Melanoma therapy, Molecular Targeted Therapy, Skin Neoplasms therapy
- Abstract
Next-generation sequencing (NGS) has provided significant insights into the pathogenesis of human malignancies. In advanced melanoma, two therapeutic avenues have appeared and have immediately become the standard of care, i.e. targeted therapy with small molecule inhibitors, and immune checkpoint blockade. Sequencing has always been essential for determining which patients may benefit from targeted therapies (e.g. the presence of BRAF mutations). While sequencing does not currently help recognize which patients might benefit from immune checkpoint blockade, recent data suggest that this may change. Multiple studies have identified tumor mutation profiles associated with patients benefiting from immune checkpoint blockade therapy. These findings suggest comprehensive tumor sequencing may become a critical step for predicting therapy responses to all systemic therapies. In this review, the current and potential future impact of NGS on treatment decisions in advanced melanoma will be summarized and discussed.
- Published
- 2017
- Full Text
- View/download PDF
49. Acquired IFNγ resistance impairs anti-tumor immunity and gives rise to T-cell-resistant melanoma lesions.
- Author
-
Sucker A, Zhao F, Pieper N, Heeke C, Maltaner R, Stadtler N, Real B, Bielefeld N, Howe S, Weide B, Gutzmer R, Utikal J, Loquai C, Gogas H, Klein-Hitpass L, Zeschnigk M, Westendorf AM, Trilling M, Horn S, Schilling B, Schadendorf D, Griewank KG, and Paschen A
- Subjects
- Antigen Presentation genetics, Antigens, Neoplasm immunology, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Biopsy, Cell Line, Tumor, DNA Mutational Analysis, Datasets as Topic, Drug Resistance, Neoplasm immunology, Histocompatibility Antigens Class I immunology, Humans, Immunotherapy methods, Interferon-gamma metabolism, Janus Kinase 1 genetics, Janus Kinase 1 metabolism, Janus Kinase 2 genetics, Janus Kinase 2 metabolism, Melanoma drug therapy, Melanoma immunology, Melanoma pathology, Mutation, Mutation Rate, Patient-Specific Modeling, Precision Medicine methods, Signal Transduction genetics, Signal Transduction immunology, Skin pathology, Skin Neoplasms drug therapy, Skin Neoplasms immunology, Skin Neoplasms pathology, T-Lymphocytes metabolism, Treatment Outcome, Whole Genome Sequencing, Drug Resistance, Neoplasm genetics, Interferon-gamma immunology, Melanoma genetics, Skin Neoplasms genetics, T-Lymphocytes immunology, Tumor Escape genetics
- Abstract
Melanoma treatment has been revolutionized by antibody-based immunotherapies. IFNγ secretion by CD8
+ T cells is critical for therapy efficacy having anti-proliferative and pro-apoptotic effects on tumour cells. Our study demonstrates a genetic evolution of IFNγ resistance in different melanoma patient models. Chromosomal alterations and subsequent inactivating mutations in genes of the IFNγ signalling cascade, most often JAK1 or JAK2, protect melanoma cells from anti-tumour IFNγ activity. JAK1/2 mutants further evolve into T-cell-resistant HLA class I-negative lesions with genes involved in antigen presentation silenced and no longer inducible by IFNγ. Allelic JAK1/2 losses predisposing to IFNγ resistance development are frequent in melanoma. Subclones harbouring inactivating mutations emerge under various immunotherapies but are also detectable in pre-treatment biopsies. Our data demonstrate that JAK1/2 deficiency protects melanoma from anti-tumour IFNγ activity and results in T-cell-resistant HLA class I-negative lesions. Screening for mechanisms of IFNγ resistance should be considered in therapeutic decision-making.- Published
- 2017
- Full Text
- View/download PDF
50. Epidermal growth factor receptor gaining impact in cutaneous squamous cell carcinoma.
- Author
-
Griewank KG
- Subjects
- Epidermal Growth Factor, Humans, Skin Neoplasms, Carcinoma, Squamous Cell, ErbB Receptors
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.