1
|
Carter ME, Benegiamo-Chilla A, Kloker LD, Paulsen N, Potkrajcic V, Paulsen F, Nemeth A, Steger V, Schulze M, Biskup S, Benzler K, Singer S, Lauer UM, Zender L, Deinzer CKW. Case report: Pulmonary Ewing sarcoma disguised as non-small cell lung cancer. Front Oncol 2024; 14:1449119. [PMID: 39575427 PMCID: PMC11578818 DOI: 10.3389/fonc.2024.1449119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/11/2024] [Indexed: 11/24/2024] Open
Abstract
Ewing sarcoma is the second most common primary malignant bone cancer in children and adolescents. This rare type of cancer is characterized by its high malignancy and therefore high risk of metastases. Typically, Ewing sarcomas originate from bones. However, extraosseous Ewing sarcoma such as pulmonary Ewing sarcoma can also be found. In this case report, we present a 55-year old male patient who was initially diagnosed with non-small cell lung cancer at his local district hospital. However, the diagnosis was changed to one of pulmonary Ewing sarcoma after subsequent histopathological and molecular pathological analysis performed in a reference pathology laboratory. After patient referral to a certified (according to the German Cancer Society) high-volume sarcoma center, multimodal chemotherapy was initiated based on recently published clinical data as opposed to the more commonly used treatment regimen in Europe. The patient responded well to treatment and underwent a complete surgical tumor resection followed by radiotherapy. In summary, this case report highlights the importance of a rigorous and timely histopathological examination of biopsy samples by a specialized cancer center to enable a correct diagnosis of the cancer type. Additionally, molecular pathology plays a crucial part in this analysis and allows the necessary differentiation between cancer types. Up to now, there is no international treatment guideline available for the treatment of Ewing sarcoma. Patients should be referred to specialist centers to allow the best possible treatment of the cancer type in view of current published clinical data. In the case of Ewing sarcoma, and in accordance with the most recent research, patients should be treated with vincristine, doxorubicin and cyclophosphamide plus ifosfamide and etoposide in combination with local treatment such as surgery and/or radiotherapy because this has been demonstrated to be the more effective therapy.
Collapse
Affiliation(s)
- Mary E. Carter
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tübingen, Germany
| | | | - Linus D. Kloker
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tübingen, Germany
| | - Nikolas Paulsen
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tübingen, Germany
| | - Vlatko Potkrajcic
- Department of Radiation Oncology, University Hospital, Tübingen, Germany
| | - Frank Paulsen
- Department of Radiation Oncology, University Hospital, Tübingen, Germany
| | - Attila Nemeth
- Department of Thoracic and Cardiovascular Surgery, University Hospital, Tübingen, Germany
| | - Volker Steger
- Department of Thoracic and Cardiovascular Surgery, University Hospital, Tübingen, Germany
| | | | - Saskia Biskup
- Zentrum für Humangenetik Tübingen, Tübingen, Germany
- CeGaT GmbH, Center for Genomics and Transcriptomics, Tübingen, Germany
| | - Katrin Benzler
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tübingen, Germany
| | - Stephan Singer
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Ulrich M. Lauer
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tübingen, Germany
- University of Tübingen, iFIT Cluster of Excellence (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Zender
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tübingen, Germany
- University of Tübingen, iFIT Cluster of Excellence (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph K. W. Deinzer
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tübingen, Germany
| |
Collapse
|
2
|
He Z, Gu Y, Yang H, Fu Q, Zhao M, Xie Y, Liu Y, Du W. Identification and verification of a novel anoikis-related gene signature with prognostic significance in clear cell renal cell carcinoma. J Cancer Res Clin Oncol 2023; 149:11661-11678. [PMID: 37402968 DOI: 10.1007/s00432-023-05012-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/19/2023] [Indexed: 07/06/2023]
Abstract
PURPOSE Clear cell renal cell carcinomas (ccRCCs) are the most common form of renal cancer in the world. The loss of extracellular matrix (ECM) stimulates cell apoptosis, known as anoikis. A resistance to anoikis in cancer cells is believed to contribute to tumor malignancy, particularly metastasis; however, the potential influence of anoikis on the prognosis of ccRCC patients is not fully understood. METHODS In this study, anoikis-related genes (ARGs) with discrepant expression were selected from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The anoikis-related gene signature (ARS) was built using a combination of the univariate Cox and least absolute shrinkage and selection operator (LASSO) analyses. ARS was also evaluated for their prognostic value. We explored the tumor microenvironment and enrichment pathways between different clusters of ccRCC. We also examined differences in clinical characteristics, immune cell infiltration and drug sensitivity between the high- and low-risk sets. In addition, we utilized three external databases and quantitative real-time polymerase chain reaction (qRT-PCR) to validate the expression and prognosis of ARGs. RESULTS Eight ARGs (PLAUR, HMCN1, CDKN2A, BID, GLI2, PLG, PRKCQ and IRF6) were identified as anoikis-related prognostic factors. According to Kaplan-Meier (KM) analysis, ccRCC patients with high-risk ARGs have a worse prognosis. The risk score was found to be a significant independent prognostic indicator. According to tumor microenvironment (TME) scores, stromal score, immune score, and estimated score of the high-risk group were superior to those of the low-risk group. There were significant differences between the two groups regarding the amount of infiltrated immune cells, immune checkpoint expression as well as drug sensitivity. A nomogram was constructed using ccRCC clinical features and risk scores. The signature and the nomogram both performed well in predicting overall survival (OS) for ccRCC patients. According to a decision curve analysis (DCA), clinical treatment options for patients with ccRCC could be improved using this model. CONCLUSION The results of validation from external databases and qRT-PCR were basically agreement with findings in TCGA and GEO databases. The ARS serving as biomarkers may provide an important reference for individual therapy of ccRCC patients.
Collapse
Affiliation(s)
- Zhiqiang He
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Yufan Gu
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Huan Yang
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Qian Fu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Maofang Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Yuhan Xie
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Yi Liu
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| | - Wenlong Du
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
3
|
Wang X, Luo Z, Chen J, Chen Y, Ji D, Fan L, Chen L, Zhao Q, Hu P, Sun P, Jia Z, Guo J, Si L. First-in-human phase I dose-escalation and dose-expansion trial of the selective MEK inhibitor HL-085 in patients with advanced melanoma harboring NRAS mutations. BMC Med 2023; 21:2. [PMID: 36600247 PMCID: PMC9814429 DOI: 10.1186/s12916-022-02669-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 11/18/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND HL-085 is a selective, orally administered MEK1/2 inhibitor. We aimed to evaluate the safety and efficacy of HL-085 in patients with advanced melanoma harboring NRAS mutations. METHODS This was a multicenter phase 1 study. HL-085 was administered twice daily in a standard 3 + 3 dose-escalation design (10 dose cohorts; 0.5-18 mg twice daily), followed by dose expansion at the recommended phase II dose (RP2D). The primary endpoints included tolerability, dose-limiting toxicity (DLT), maximum tolerated dose (MTD) and RP2D. RESULTS Between September 13, 2017, and January 18, 2021, 42 patients were enrolled (dose escalation phase: n = 30; dose expansion phase: n = 12). No DLT was reported during dose escalation and MTD was not reached with HL-085 doses up to 18 mg twice daily. The RP2D was 12 mg twice daily. The most common all-grade drug-related adverse events (AEs) across all dose levels were rash (61.9%), increased creatine phosphokinase (CK, 59.5%), face edema (50.0%), increased aspartate aminotransferase (47.6%), peripheral edema (40.5%), diarrhea (33.3%), alanine aminotransferase (33.3%), and paronychia (19.0%), most of which were grade 1 and 2. Most frequency of grade ≥ 3 AEs were CK (14.2%), asthenia (7.1%), peripheral edema (4.8%), and acneiform dermatitis (4.8%). In the cohort of 12 mg twice daily dose (15 patients), confirmed objective response rate was 26.7%; disease control rate was 86.7%; median duration of response was 2.9 months; median progression-free survival was 3.6 months. CONCLUSIONS The HL-085 showed acceptable tolerability and substantial clinical activity in patients with advanced melanoma harboring NRAS mutations. TRIAL REGISTRATION Trial registration ClinicalTrials.gov number: NCT03973151.
Collapse
Affiliation(s)
- Xuan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Zhiguo Luo
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing Chen
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Chen
- Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Dongmei Ji
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Li Fan
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Chen
- Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Qian Zhao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Peng Sun
- Department of Clinical Research and Development, Shanghai Kechow Pharma, Inc., Shanghai, China
| | - Zhongwei Jia
- Department of Clinical Research and Development, Shanghai Kechow Pharma, Inc., Shanghai, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Research Institute, Beijing, China.
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Research Institute, Beijing, China.
| |
Collapse
|
4
|
Crescitelli R, Filges S, Karimi N, Urzì O, Alonso-Agudo T, Ståhlberg A, Lötvall J, Lässer C, Olofsson Bagge R. Extracellular vesicle DNA from human melanoma tissues contains cancer-specific mutations. Front Cell Dev Biol 2022; 10:1028854. [PMID: 36531960 PMCID: PMC9751452 DOI: 10.3389/fcell.2022.1028854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/18/2022] [Indexed: 10/19/2023] Open
Abstract
Liquid biopsies are promising tools for early diagnosis and residual disease monitoring in patients with cancer, and circulating tumor DNA isolated from plasma has been extensively studied as it has been shown to contain tumor-specific mutations. Extracellular vesicles (EVs) present in tumor tissues carry tumor-derived molecules such as proteins and nucleic acids, and thus EVs can potentially represent a source of cancer-specific DNA. Here we identified the presence of tumor-specific DNA mutations in EVs isolated from six human melanoma metastatic tissues and compared the results with tumor tissue DNA and plasma DNA. Tumor tissue EVs were isolated using enzymatic treatment followed by ultracentrifugation and iodixanol density cushion isolation. A panel of 34 melanoma-related genes was investigated using ultra-sensitive sequencing (SiMSen-seq). We detected mutations in six genes in the EVs (BRAF, NRAS, CDKN2A, STK19, PPP6C, and RAC), and at least one mutation was detected in all melanoma EV samples. Interestingly, the mutant allele frequency was higher in DNA isolated from tumor-derived EVs compared to total DNA extracted directly from plasma DNA, supporting the potential role of tumor EVs as future biomarkers in melanoma.
Collapse
Affiliation(s)
- Rossella Crescitelli
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stefan Filges
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Nasibeh Karimi
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ornella Urzì
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D), University of Palermo, Gothenburg, Italy
| | - Tamara Alonso-Agudo
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
5
|
Dobre EG, Constantin C, Neagu M. Skin Cancer Research Goes Digital: Looking for Biomarkers within the Droplets. J Pers Med 2022; 12:jpm12071136. [PMID: 35887633 PMCID: PMC9323323 DOI: 10.3390/jpm12071136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 12/24/2022] Open
Abstract
Skin cancer, which includes the most frequent malignant non-melanoma carcinomas (basal cell carcinoma, BCC, and squamous cell carcinoma, SCC), along with the difficult to treat cutaneous melanoma (CM), pose important worldwide issues for the health care system. Despite the improved anti-cancer armamentarium and the latest scientific achievements, many skin cancer patients fail to respond to therapies, due to the remarkable heterogeneity of cutaneous tumors, calling for even more sophisticated biomarker discovery and patient monitoring approaches. Droplet digital polymerase chain reaction (ddPCR), a robust method for detecting and quantifying low-abundance nucleic acids, has recently emerged as a powerful technology for skin cancer analysis in tissue and liquid biopsies (LBs). The ddPCR method, being capable of analyzing various biological samples, has proved to be efficient in studying variations in gene sequences, including copy number variations (CNVs) and point mutations, DNA methylation, circulatory miRNome, and transcriptome dynamics. Moreover, ddPCR can be designed as a dynamic platform for individualized cancer detection and monitoring therapy efficacy. Here, we present the latest scientific studies applying ddPCR in dermato-oncology, highlighting the potential of this technology for skin cancer biomarker discovery and validation in the context of personalized medicine. The benefits and challenges associated with ddPCR implementation in the clinical setting, mainly when analyzing LBs, are also discussed.
Collapse
Affiliation(s)
- Elena-Georgiana Dobre
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91–95, 050095 Bucharest, Romania;
- Correspondence:
| | - Carolina Constantin
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Monica Neagu
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91–95, 050095 Bucharest, Romania;
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
6
|
Gracie L, Pan Y, Atenafu EG, Ward DG, Teng M, Pallan L, Stevens NM, Khoja L. Circulating tumour DNA (ctDNA) in metastatic melanoma, a systematic review and meta-analysis. Eur J Cancer 2021; 158:191-207. [PMID: 34757258 DOI: 10.1016/j.ejca.2021.09.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/31/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Circulating tumour DNA (ctDNA) is an emerging biomarker in melanoma. We performed a systematic review and meta-analysis to explore its clinical utility as a prognostic, pharmacodynamic (PD) and predictive biomarker. METHODS A systematic search was conducted from Jan 2015 to April 2021, of the electronic databases PubMed, Cochrane Library and Ovid MEDLINE to identify studies. Studies were restricted to those published in English within the last 5 years, evaluating ctDNA in humans in ≥10 patients. Survival data were extracted for meta-analysis using pooled treatment effect (TE), i.e. log hazard ratios (HRs) and corresponding standard error of TE for progression-free survival or overall survival differences in patients who were ctDNA positive or negative. PRISMA statement guidelines were followed. RESULTS A meta-analysis of 19 studies grouped according to methodology of ctDNA detection, revealed a combined estimate for HR of progression-free survival (13 studies using droplet digital Polymerase Chain Reaction (ddPCR) methodology (N = 1002) of 2.10 (95% CI: 1.71-2.59) revealing a poorer prognosis when ctDNA was detected. This result was confirmed in the smaller analysis of (non-ddPCR, N = 347) five studies: HR = 2.45 (95% CI: 1.29-4.63). Similar findings were found in the overall survival analysis of nine studies (ddPCR methodology, N = 841) where the combined HR was 2.78 (95% CI: 2.21-3.49) and of the five studies (non-ddPCR methodology, N = 326) where the combined HR was 2.58 (95% CI: 1.74-3.84). Serial ctDNA levels on treatment showed a pharmacodynamic role reflecting response or resistance earlier than radiological assessment. CONCLUSIONS Circulating tumour DNA is a predictive, prognostic and PD biomarker in melanoma. Technical standardisation of assays is required before clinical adoption.
Collapse
Affiliation(s)
- Lara Gracie
- College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Yi Pan
- Centre for Computational Biology and Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Eshetu G Atenafu
- Biostatistics Department, Princess Margaret Cancer Center, University Health Network, University Way, Toronto, Ontario, Canada
| | - Douglas G Ward
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Mabel Teng
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Department of Oncology, Birmingham B15 2TH, UK
| | - Lallit Pallan
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Department of Oncology, Birmingham B15 2TH, UK
| | - Neil M Stevens
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Leila Khoja
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
7
|
Jones J, Cain S, Pesic-Smith J, Choong PFM, Morokoff AP, Drummond KJ, Dabscheck G. Circulating tumor DNA for malignant peripheral nerve sheath tumors in neurofibromatosis type 1. J Neurooncol 2021; 154:265-274. [PMID: 34529228 DOI: 10.1007/s11060-021-03846-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/14/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE The leading cause of early death in patients with neurofibromatosis type 1 (NF1) is malignant peripheral nerve sheath tumor (MPNST). The principles of management include early diagnosis, surgical clearance and close monitoring for tumor recurrence. Current methods for diagnosis, detection of residual disease and monitoring tumor burden are inadequate, as clinical and radiological features are non-specific for malignancy in patients with multiple tumors and lack the sensitivity to identify early evidence of malignant transformation or tumor recurrence. Circulating tumor DNA (ctDNA) is a promising tool in cancer management and has the potential to improve the care of patients with NF1. In the following article we summarise the current understanding of the genomic landscape of MPNST, report on the previous literature of ctDNA in MPNST and outline the potential clinical applications for ctDNA in NF1 associated MPNST. Finally, we describe our prospective cohort study protocol investigating the utility of using ctDNA as an early diagnostic tool for MPNSTs in NF1 patients.
Collapse
Affiliation(s)
- Jordan Jones
- Department of Surgery, University of Melbourne, Melbourne, VIC, Australia. .,Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, VIC, 3050, Australia.
| | - Sarah Cain
- Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, VIC, 3050, Australia
| | - Jonathan Pesic-Smith
- Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, VIC, 3050, Australia
| | - Peter F M Choong
- Department of Surgery, University of Melbourne, Melbourne, VIC, Australia.,Department of Orthopaedics, St Vincent's Hospital, Melbourne, VIC, Australia.,Bone and Soft Tissue Sarcoma Service, Perter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Andrew P Morokoff
- Department of Surgery, University of Melbourne, Melbourne, VIC, Australia.,Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, VIC, 3050, Australia
| | - Kate J Drummond
- Department of Surgery, University of Melbourne, Melbourne, VIC, Australia.,Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, VIC, 3050, Australia
| | - Gabriel Dabscheck
- Department of Neurology, Royal Children's Hospital, Melbourne, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Tolmeijer SH, Koornstra RHT, de Groot JWB, Geerlings MJ, van Rens DH, Boers-Sonderen MJ, Schalken JA, Gerritsen WR, Ligtenberg MJL, Mehra N. Plasma BRAF Mutation Detection for the Diagnostic and Monitoring Trajectory of Patients with LDH-High Stage IV Melanoma. Cancers (Basel) 2021; 13:3913. [PMID: 34359813 PMCID: PMC8345527 DOI: 10.3390/cancers13153913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 01/16/2023] Open
Abstract
For patients with newly diagnosed metastatic melanoma, rapid BRAF mutation (mBRAF) assessment is vital to promptly initiate systemic therapy. Additionally, blood-based biomarkers are desired to monitor and predict treatment response. Circulating tumor DNA (ctDNA) has shown great promise for minimally invasive mBRAF assessment and treatment monitoring, but validation studies are needed. This prospective study utilized longitudinal plasma samples at regular timepoints (0, 6, 12, 18 weeks) to address the clinical validity of ctDNA measurements in stage IV melanoma patients with elevated serum lactate dehydrogenase (LDH > 250U/L) starting first-line systemic treatment. Using droplet digital PCR, the plasma mBRAF abundance was assessed in 53 patients with a BRAFV600 tissue mutation. Plasma mBRAF was detected in 50/51 patients at baseline (98% sensitivity; median fraction abundance of 19.5%) and 0/17 controls (100% specificity). Patients in whom plasma mBRAF became undetectable during the first 12-18 weeks of treatment had a longer progression-free survival (30.2 vs. 4.0 months; p < 0.001) and cancer-specific survival (not reached vs. 10.2 months; p < 0.001) compared to patients with detectable mBRAF. The ctDNA dynamics outperformed LDH and S100 dynamics. These results confirm the clinical validity of ctDNA measurements as a minimally invasive biomarker for the diagnostic and monitoring trajectory of patients with LDH-high stage IV melanoma.
Collapse
Affiliation(s)
- Sofie H. Tolmeijer
- Department of Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Rutger H. T. Koornstra
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
- Department of Medical Oncology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | | | - Maartje J. Geerlings
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (M.J.G.); (M.J.L.L.)
| | - Dirk H. van Rens
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
| | - Marye J. Boers-Sonderen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
| | - Jack A. Schalken
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Winald R. Gerritsen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
| | - Marjolijn J. L. Ligtenberg
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (M.J.G.); (M.J.L.L.)
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
| |
Collapse
|
9
|
Forschner A, Sinnberg T, Mroz G, Schroeder C, Reinert CP, Gatidis S, Bitzer M, Eigentler T, Garbe C, Niessner H, Röcken M, Roggia C, Armeanu-Ebinger S, Riess O, Mattern S, Nann D, Bonzheim I. Case Report: Combined CDK4/6 and MEK Inhibition in Refractory CDKN2A and NRAS Mutant Melanoma. Front Oncol 2021; 11:643156. [PMID: 33732653 PMCID: PMC7959243 DOI: 10.3389/fonc.2021.643156] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/25/2021] [Indexed: 01/14/2023] Open
Abstract
There are only limited treatment options for metastatic NRAS mutant melanoma patients with resistance to immune checkpoint inhibitors. Besides activation of the mitogen-activated protein (MAP) kinase pathway, they often have additional disturbances in cell cycle regulation. However, unlike BRAF mutant melanoma, no targeted therapy has yet been approved for NRAS mutant melanoma so far. Here we present a NRAS mutant melanoma patient with response to combined binimetinib and ribociclib therapy following characterization of the molecular defects of the tumor by panel sequencing. Next generation sequencing (708 cancer genes) of a soft tissue metastasis revealed a homozygous deletion of CDKN2A in addition to the previously known NRAS mutation, as well as amplification of CCNE1 and CDK6. Immunohistochemical staining of the altered cell cycle genes confirmed loss of p16, reduced expression of p21 and high expression of CDK6 and cyclin D1. As the patient had been progressive on combined immunotherapy, targeted therapy with combined MEK and CDK4/6 inhibition was initiated as recommended by the molecular tumor board. Response to treatment was monitored with PET/CT and liquid biopsy, serum LDH, and S100. In addition, a patient-derived xenograft (PDX) was used to prove the efficacy of the two drugs in combination. Furthermore, senescence-associated beta-galactosidase staining showed that more cells were senescent under the combination treatment of binimetinib and ribociclib. Our case demonstrates how an individualized, molecular-based therapeutic approach could be found based on next-generation sequencing results. Furthermore our report highlights the fruitful and efficient collaboration of dermatooncologists, human geneticists, molecular pathologists, biochemists, radiologists, and nuclear physicians. Further studies are urgently needed to expand the very limited therapeutic landscape of NRAS mutated melanoma.
Collapse
Affiliation(s)
- Andrea Forschner
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Tobias Sinnberg
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany.,iFIT Cluster of Excellence (EXC 2180), University of Tübingen, Tübingen, Germany
| | - Gabi Mroz
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Christian Philipp Reinert
- Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Michael Bitzer
- Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Eigentler
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Heike Niessner
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Martin Röcken
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany.,iFIT Cluster of Excellence (EXC 2180), University of Tübingen, Tübingen, Germany
| | - Cristiana Roggia
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Sorin Armeanu-Ebinger
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Olaf Riess
- iFIT Cluster of Excellence (EXC 2180), University of Tübingen, Tübingen, Germany.,German Deutsche Forschungsgemeinschaft (DFG) Next Generation Sequencing (NGS) Competence Center, NGS Competence Center Tübingen (NCCT), Tübingen, Germany
| | - Sven Mattern
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Dominik Nann
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Irina Bonzheim
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
10
|
Circulating tumour DNA in patients with melanoma receiving targeted therapy. Lancet Oncol 2021; 22:291-293. [PMID: 33587892 DOI: 10.1016/s1470-2045(20)30758-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/21/2022]
|