1
|
Luo J, Bishop JA, DuBois SG, Hanna GJ, Sholl LM, Stelow EB, Thompson LDR, Shapiro GI, French CA. Hiding in plain sight: NUT carcinoma is an unrecognized subtype of squamous cell carcinoma of the lungs and head and neck. Nat Rev Clin Oncol 2025; 22:292-306. [PMID: 39900969 DOI: 10.1038/s41571-025-00986-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 02/05/2025]
Abstract
In the past two decades, treatment for non-small-cell lung cancers (NSCLCs) and head and neck squamous cell carcinoma (HNSCC) has advanced considerably, owing largely to the characterization of distinct oncological subtypes, the development of targeted therapies for each subtype and the advent of immunotherapy. Data emerging over the past two decades suggest that NUT carcinoma, a highly aggressive malignancy driven by a NUT fusion oncoprotein and arising in the lungs, head and neck, and rarely in other sites, is a squamous cell carcinoma (SCC) based on transcriptional, histopathological, cell-of-origin and molecular characteristics. NUT carcinoma has an estimated incidence of 1,400 cases per year in the United States, surpassing that of some rare NSCLC and HNSCC subtypes. However, NUT carcinoma is currently not recognized as an SCC of the lungs or head and neck. The orphan classification of NUT carcinoma as a distinct entity leads to a lack of awareness of this malignancy among oncologists and surgeons, despite early diagnosis being crucial for this cancer type with a median survival of only ~6.5 months. Consequently, NUT carcinoma is underdiagnosed and often misdiagnosed, resulting in limited research and progress in developing effective treatments in one of the most aggressive forms of lung and head and neck cancer. With a growing number of targeted agents that can potentially be used to treat NUT carcinoma, improved recognition through reclassification and inclusion of NUT carcinoma as a squamous NSCLC or an HNSCC when arising in these locations will accelerate the development of effective therapies for this disease. Thus, in the Perspective, we propose such a reclassification of NUT carcinoma as an SCC and discuss the supporting evidence.
Collapse
Affiliation(s)
- Jia Luo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Glenn J Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edward B Stelow
- Department of Pathology, University of Virginia Medical Center, Charlottesville, VA, USA
| | | | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher A French
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Sánchez Becerra MV, Escudero Iriarte C, Travert C, Tian TV, Besse B. Promising response to lurbinectedin in NUT carcinoma: a case report and review of emerging therapeutic strategies. Ann Oncol 2025; 36:225-227. [PMID: 39396757 DOI: 10.1016/j.annonc.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024] Open
Affiliation(s)
- M V Sánchez Becerra
- "Rare Circulating Cells" Translational Platform, Gustave Roussy, Université Paris-Saclay, CNRS UMS3655-INSERM US23 AMMICA, Villejuif; "Identification of Molecular Predictors and new Targets for Cancer Treatment". INSERM, U981, Villejuif; Department of Cancer Medicine, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - C Escudero Iriarte
- Upper GI and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - C Travert
- Thoracic Oncology Department, Hôpital Arnaud de Villeneuve-Centre Hospitalier, Universitaire de Montpellier, Montpellier, France
| | - T V Tian
- Upper GI and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - B Besse
- "Rare Circulating Cells" Translational Platform, Gustave Roussy, Université Paris-Saclay, CNRS UMS3655-INSERM US23 AMMICA, Villejuif; "Identification of Molecular Predictors and new Targets for Cancer Treatment". INSERM, U981, Villejuif; Department of Cancer Medicine, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
3
|
Amrutkar RD, Amesar MV, Chavan LB, Baviskar NS, Bhamare VG. Precision Targeting of BET Proteins - Navigating Disease Pathways, Inhibitor Insights, and Shaping Therapeutic Frontiers: A Comprehensive Review. Curr Drug Targets 2025; 26:147-166. [PMID: 39385413 DOI: 10.2174/0113894501304747240823111337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 10/12/2024]
Abstract
The family of proteins known as Bromodomain and Extra-Terminal (BET) proteins has become a key participant in the control of gene expression, having a significant impact on numerous physiological and pathological mechanisms. This review offers a thorough investigation of the BET protein family, clarifying its various roles in essential cellular processes and its connection to a variety of illnesses, from inflammatory disorders to cancer. The article explores the structural and functional features of BET proteins, emphasizing their special bromodomain modules that control chromatin dynamics by identifying acetylated histones. BET proteins' complex roles in the development of cardiovascular, neurodegenerative, and cancer diseases are carefully investigated, providing insight into possible treatment avenues. In addition, the review carefully examines the history and relevance of BET inhibitors, demonstrating their capacity to modify gene expression profiles and specifically target BET proteins. The encouraging outcomes of preclinical and clinical research highlight BET inhibitors' therapeutic potential across a range of disease contexts. The article summarizes the state of BET inhibitors today and makes predictions about the challenges and future directions of the field. This article provides insights into the changing field of BET protein-targeted interventions by discussing the potential of personalized medicine and combination therapies involving BET inhibitors. This thorough analysis combines many aspects of BET proteins, such as their physiological roles and their roles in pathophysiological conditions. As such, it is an invaluable tool for scientists and medical professionals who are trying to figure out how to treat patients by using this fascinating protein family.
Collapse
Affiliation(s)
- Rakesh D Amrutkar
- Department of Pharmaceutical Chemistry, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| | - Mehul V Amesar
- Department of Pharmaceutical Chemistry, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| | - Lokesh B Chavan
- Department of Pharmaceutical Chemistry, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| | - Nilesh S Baviskar
- Department of Pharmaceutical Chemistry, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| | - Vaibhav G Bhamare
- Department of Pharmaceutics, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| |
Collapse
|
4
|
Li R, Zhang Y, Liu Q, Gao A, Dang Q. Targeted combination chemotherapy effective in nuclear protein in testis carcinoma of lung origin: A case report and review of the literature. Medicine (Baltimore) 2024; 103:e38881. [PMID: 39969296 PMCID: PMC11688100 DOI: 10.1097/md.0000000000038881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/20/2024] [Indexed: 02/20/2025] Open
Abstract
RATIONALE Nuclear protein in testis carcinoma (NC) is a rare, aggressive, poorly differentiated squamous cell carcinoma. By reviewing the entire treatment process of the patient, we aim to explore the treatment experience of NC. PATIENT CONCERNS We report the case of a 27-year-old female patient with NC whose initial symptoms were occasional cough and chest tightness with abdominal distension for more than half a month without any other specific discomfort. DIAGNOSES Computed tomography showed right lung hilar and right middle and lower lobe mass, malignant, right hilar and mediastinal lymph node metastasis, and bilateral cystic solid masses in the adnexal region, malignant possibility. Pathological diagnosis showed nuclear protein in the testis (+). INTERVENTIONS After the failure of first-line chemotherapy with immunocombination, second-line chemotherapy was switched to bevacizumab, which resulted in a progression-free survival of 6 months. OUTCOMES The disease then reprogressed, and she died on November 7, 2022. LESSONS The patient achieved survival of nearly 1 year on multiple courses of therapy, well beyond the currently reported median survival. The patient achieved a 6-month progression-free survival, suggesting that combination therapy with antivascular endothelial growth factor class-targeted agents is a potential approach.
Collapse
Affiliation(s)
- Ran Li
- Shandong Second Medical University, Weifang, China
| | - Ye Zhang
- Shandong Second Medical University, Weifang, China
| | - Qian Liu
- Shandong Second Medical University, Weifang, China
| | - Aiqin Gao
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Qi Dang
- Department of Oncology, Phase I Clinical Trial Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
5
|
Dai W, Qiao X, Fang Y, Guo R, Bai P, Liu S, Li T, Jiang Y, Wei S, Na Z, Xiao X, Li D. Epigenetics-targeted drugs: current paradigms and future challenges. Signal Transduct Target Ther 2024; 9:332. [PMID: 39592582 PMCID: PMC11627502 DOI: 10.1038/s41392-024-02039-0] [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: 08/02/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Epigenetics governs a chromatin state regulatory system through five key mechanisms: DNA modification, histone modification, RNA modification, chromatin remodeling, and non-coding RNA regulation. These mechanisms and their associated enzymes convey genetic information independently of DNA base sequences, playing essential roles in organismal development and homeostasis. Conversely, disruptions in epigenetic landscapes critically influence the pathogenesis of various human diseases. This understanding has laid a robust theoretical groundwork for developing drugs that target epigenetics-modifying enzymes in pathological conditions. Over the past two decades, a growing array of small molecule drugs targeting epigenetic enzymes such as DNA methyltransferase, histone deacetylase, isocitrate dehydrogenase, and enhancer of zeste homolog 2, have been thoroughly investigated and implemented as therapeutic options, particularly in oncology. Additionally, numerous epigenetics-targeted drugs are undergoing clinical trials, offering promising prospects for clinical benefits. This review delineates the roles of epigenetics in physiological and pathological contexts and underscores pioneering studies on the discovery and clinical implementation of epigenetics-targeted drugs. These include inhibitors, agonists, degraders, and multitarget agents, aiming to identify practical challenges and promising avenues for future research. Ultimately, this review aims to deepen the understanding of epigenetics-oriented therapeutic strategies and their further application in clinical settings.
Collapse
Affiliation(s)
- Wanlin Dai
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinbo Qiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Fang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Renhao Guo
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Bai
- Department of Forensic Genetics, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Shuang Liu
- Shenyang Maternity and Child Health Hospital, Shenyang, China
| | - Tingting Li
- Department of General Internal Medicine VIP Ward, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yutao Jiang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuang Wei
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhijing Na
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
| | - Xue Xiao
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China.
| | - Da Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China.
| |
Collapse
|
6
|
Kroening G, Luo J, Evans MG, Adeyelu T, Ou SHI, Arter ZL, Wise-Draper TM, Sukari A, Azmi AS, Braxton DR, Elliott A, Bryant DA, Oberley MJ, Kim C, Shapiro GI, French CA, Nagasaka M. Multiomic Characterization and Molecular Profiling of Nuclear Protein in Testis Carcinoma. JCO Precis Oncol 2024; 8:e2400334. [PMID: 39447095 PMCID: PMC11520346 DOI: 10.1200/po.24.00334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/14/2024] [Accepted: 08/06/2024] [Indexed: 10/26/2024] Open
Abstract
PURPOSE Nuclear protein in testis carcinoma (NC) is an underdiagnosed and aggressive squamous/poorly differentiated cancer characterized by rearrangement of the gene NUTM1 on chromosome 15q14. Co-occurring alternations have not been fully characterized. METHODS We analyzed the genomic and immune landscape of 54 cases of NC that underwent DNA- and RNA-based NGS sequencing (Caris). RESULTS While NC is driven by NUTM1 fusion oncoproteins, co-occurring DNA mutations in epigenetic or cell cycle pathways were observed in 26% of cases. There was no significant difference between the fusion partner of NUTM1 and co-occurring gene mutations. RNA sequencing analysis showed increased MYC pathway activity in NC compared with head and neck squamous cell carcinoma (HNSCC) and lung squamous cell carcinoma (LUSC), which is consistent with the known pathophysiology of NC. Characterization of the NC tumor microenvironment using RNA sequencing revealed significantly lower immune cell infiltration compared with HNSCC and LUSC. NC was 10× higher in patients with HNSCC and LUSC younger than 50 years than in those older than 70 years. CONCLUSION To our knowledge, this is the first series of NC profiled broadly at the DNA and RNA level. We observed fewer intratumoral immune cells by RNA sequencing, which may be associated with anecdotal data of lack of immunotherapy benefit in NC. High MYC pathway activity in NC supports ongoing trials targeting MYC suppression. The incidence of NC among patients younger than 50 years with LUSC/HNSCC supports testing for NC in these patients. The prognosis of NCs remains dismal, and future studies should focus on improving the response to immunotherapy and targeting MYC.
Collapse
Affiliation(s)
- Gianna Kroening
- University of California Irvine School of Medicine, Orange, CA
| | - Jia Luo
- Dana-Farber Cancer Institute, Boston, MA
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | | - Chul Kim
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Geoffrey I. Shapiro
- Dana-Farber Cancer Institute, Boston, MA
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | | | - Misako Nagasaka
- University of California Irvine School of Medicine, Orange, CA
| |
Collapse
|
7
|
Goto K, Kukita Y, Hishima T, Matsushita S, Tsuyuki T, Makihara K, Koga K, Mukumoto S, Honma K. Primary Cutaneous NUT Carcinoma: Clinicopathologic and Genetic Study of 4 Cases. Am J Surg Pathol 2024; 48:942-952. [PMID: 38708710 DOI: 10.1097/pas.0000000000002240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
WHO Classification of Skin Tumors, fifth edition (2023) has newly described primary cutaneous NUT carcinoma; however, information on this cancer type remains scarce. Herein, we performed clinicopathologic and genetic analyses of 4 cases. Four elderly women (median age 77 y, range: 68 to 82 y) were included. The median tumor size was 12.5 (10 to 40 mm). Tumors were located on the scalp, temple, thigh, and palm. Two (50%) patients presented with regional lymph node metastases. Neither distant metastasis nor mortality was observed during patient follow-up of 10.5 (3 to 15) months. Sanger, panel DNA and whole-exome RNA sequencing revealed BRD3::NUTM1 (n=2) and BRD4::NUTM1 (n=2) fusions. Histology of BRD3 -rearranged tumors revealed an epidermal connection, relatively small tumor nests, and ductal or intracytoplasmic luminal formation, whereas that of BRD4 -rearranged tumors revealed large solid nests comprising discohesive tumor cells. NUT, cytokeratins, p63, EMA, TRPS1, c-MYB, CD56, and INSM1 were immunoexpressed to varying degrees in all (100%) tumors. Furthermore, diffuse SOX10 expression was common (3/4, 75%). The literature review of five previously described cases revealed women predominance, no recurrence, frequent BRD3::NUTM1 fusions, and histology of ductoglandular structures. Our study findings and literature suggest elderly women predominance, relatively frequent BRD3::NUTM1 fusions, histopathologic ductoglandular differentiation, absence of abrupt keratinisation, and a characteristic immunoprofile in primary cutaneous NUT carcinoma, unlike in that of other organ. No distant metastasis or disease-associated mortality was seen in all cases with limited follow-up.
Collapse
Affiliation(s)
- Keisuke Goto
- Department of Diagnostic Pathology and Cytology
- Laboratory of Genomic Pathology, Research Center, Osaka International Cancer Institute
- Department of Diagnostic Pathology, Osaka National Hospital, Osaka
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital
- Department of Pathology, Itabashi Central Clinical Laboratory
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo
- Department of Dermato-Oncology/Dermatology, National Hospital Organization Kagoshima Medical Center, Kagoshima
- Department of Diagnostic Pathology, Shizuoka Cancer Center Hospital, Sunto
- Department of Diagnostic Pathology, Chutoen General Medical Center, Kakegawa
| | - Yoji Kukita
- Department of Dermatology, Hyogo Cancer Center, Akashi
| | - Tsunekazu Hishima
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital
| | - Shigeto Matsushita
- Department of Dermato-Oncology/Dermatology, National Hospital Organization Kagoshima Medical Center, Kagoshima
| | - Takuji Tsuyuki
- Department of Diagnostic Pathology, Kainan Hospital, Yatomi
| | - Kosuke Makihara
- Department of Surgical Pathology, Kyushu Rosai Hospital, Kitakyushu
| | - Kaori Koga
- Department of Pathology, Fukuoka University Hospital, Fukuoka
| | | | | |
Collapse
|
8
|
王 艺, 赵 建, 周 小, 刘 剑. [NUT cancer of nasal cavity and sinuses: a case report and literature review]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY HEAD AND NECK SURGERY 2024; 38:530-533;540. [PMID: 38858120 PMCID: PMC11480580 DOI: 10.13201/j.issn.2096-7993.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Indexed: 06/12/2024]
Abstract
NUT Carcinoma(NC) is a rare malignant tumor of unknown origin, which is highly aggressive. It is characterized by chromosome rearrangement accompanied by NUTM1 gene. The pathological manifestations were sudden and focal squamous in poorly differentiated or undifferentiated carcinoma. NUTM1gene rearrangement can be used to diagnose NC. The prognosis of NUT cancer is poor. Clinically, there is no established treatment plan. treatment options mainly comprise surgery, radiotherapy and chemotherapy. A 74-year-old patient with NC of the nasal cavity and sinuses was reported. Her clinical presentation was right nasal congestion with facial swelling. Sinus CT and MRI showed soft tissue density in the right nasal cavity and maxillary sinus with bone destruction. After admission, the patient underwent nasal endoscopic biopsy, and the postoperative pathological FISH staining showed BRD4/NUT fusion t(15, 19). The tumor was significantly reduced after two courses of sequential chemoradiotherapy. Two months later, the patient underwent a partial maxillary resection due to the rapid regrowth of sinusoidal mass, invading the hard palate. The patient died 2 months after surgery due to multiple organ failure resulted from tumor metastasis, with a survival time of 11 months. The clinical characteristics, diagnosis and treatment of this case were reported and related literature was reviewed.
Collapse
Affiliation(s)
- 艺贝 王
- 中日友好医院耳鼻咽喉头颈外科(北京,100029)Department of Otolaryngology Head and Neck Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - 建辉 赵
- 中日友好医院耳鼻咽喉头颈外科(北京,100029)Department of Otolaryngology Head and Neck Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - 小林 周
- 中日友好医院耳鼻咽喉头颈外科(北京,100029)Department of Otolaryngology Head and Neck Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - 剑锋 刘
- 中日友好医院耳鼻咽喉头颈外科(北京,100029)Department of Otolaryngology Head and Neck Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| |
Collapse
|
9
|
Kloker LD, Sidiras M, Flaadt T, Brecht IB, Deinzer CKW, Groß T, Benzler K, Zender L, Lauer UM. Clinical management of NUT carcinoma (NC) in Germany: Analysis of survival, therapy response, tumor markers and tumor genome sequencing in 35 adult patients. Lung Cancer 2024; 189:107496. [PMID: 38301600 DOI: 10.1016/j.lungcan.2024.107496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/22/2024] [Accepted: 01/27/2024] [Indexed: 02/03/2024]
Abstract
NUT carcinomas (NC) are very rare and highly aggressive tumors, molecularly defined by an aberrant gene fusion involving the NUTM1 gene. NCs preferentially arise intrathoracically or in the head and neck region, having a highly adverse prognosis with almost no long-term survivors. Here, we report on a cohort of 35 adult NC patients who were evaluated at University Hospital Tuebingen in an eight year time span, i.e. between 2016 and 2023. Primary objectives were overall survival (OS) and influence of primary tumor locations, fusion gene types and staging on OS. Secondary objectives were patient baseline characteristics, risk factors, tumor markers, treatment decisions and responses to therapy comparing thoracic vs non-thoracic origins. Further, data from tumor genome sequencing were analyzed. In this monocentric German cohort, 54 % of patients had thoracic tumors and 65 % harbored a BRD4-NUTM1 fusion gene. Median OS was 7.5 months, being significantly dependent on primary tumor location and nodal status. Initial misdiagnosis was a problem in 31 % of the cases. Surgery was the first treatment in most patients (46 %) and 80 % were treated with polychemotherapies, showing longer progression free survival (PFS) with ifosfamide-based than with platinum-based regimens. Patients treated with an immune checkpoint inhibitor (ICI) in addition to first-line chemotherapy tended to have longer OS. Initial LDH levels could be identified as a prognostic measure for survival prognosis. Sequencing data highlight aberrant NUTM1 fusion genes as unique tumor driver genes. This is the largest adult European cohort of this orphan tumor disease, showing epidemiologic and molecular features as well as relevant clinical data. Awareness to prevent misdiagnosis, fast contact to a specialized nation-wide center and referral to clinical studies are essential as long-term survival is rarely achieved with any of the current therapeutic regimes.
Collapse
Affiliation(s)
- Linus D Kloker
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tuebingen, Germany.
| | - Mirjana Sidiras
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tuebingen, Germany
| | - Tim Flaadt
- Pediatric Hematology/Oncology, Department of Pediatrics, University Hospital, Tuebingen, Germany
| | - Ines B Brecht
- Pediatric Hematology/Oncology, Department of Pediatrics, University Hospital, Tuebingen, Germany
| | - Christoph K W Deinzer
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tuebingen, Germany
| | - Thorben Groß
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tuebingen, Germany
| | - Katrin Benzler
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tuebingen, Germany
| | - Lars Zender
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tuebingen, Germany; DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy', University of Tuebingen, Tuebingen, Germany; National Center for Tumor Diseases (NCT), NCT Tuebingen, a partnership between DKFZ and the University Hospital Tuebingen, Germany
| | - Ulrich M Lauer
- Department of Medical Oncology and Pneumology, Medical University Hospital, Tuebingen, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Tuebingen, Germany; National Center for Tumor Diseases (NCT), NCT Tuebingen, a partnership between DKFZ and the University Hospital Tuebingen, Germany
| |
Collapse
|
10
|
Krupina K, Goginashvili A, Cleveland DW. Scrambling the genome in cancer: causes and consequences of complex chromosome rearrangements. Nat Rev Genet 2024; 25:196-210. [PMID: 37938738 PMCID: PMC10922386 DOI: 10.1038/s41576-023-00663-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 11/09/2023]
Abstract
Complex chromosome rearrangements, known as chromoanagenesis, are widespread in cancer. Based on large-scale DNA sequencing of human tumours, the most frequent type of complex chromosome rearrangement is chromothripsis, a massive, localized and clustered rearrangement of one (or a few) chromosomes seemingly acquired in a single event. Chromothripsis can be initiated by mitotic errors that produce a micronucleus encapsulating a single chromosome or chromosomal fragment. Rupture of the unstable micronuclear envelope exposes its chromatin to cytosolic nucleases and induces chromothriptic shattering. Found in up to half of tumours included in pan-cancer genomic analyses, chromothriptic rearrangements can contribute to tumorigenesis through inactivation of tumour suppressor genes, activation of proto-oncogenes, or gene amplification through the production of self-propagating extrachromosomal circular DNAs encoding oncogenes or genes conferring anticancer drug resistance. Here, we discuss what has been learned about the mechanisms that enable these complex genomic rearrangements and their consequences in cancer.
Collapse
Affiliation(s)
- Ksenia Krupina
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Alexander Goginashvili
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Don W Cleveland
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA.
| |
Collapse
|
11
|
Zhao D, Cao W, Zha S, Wang Y, Pan Z, Zhang J, Hu K. Primary pulmonary nuclear protein of the testis midline carcinoma: case report and systematic review with pooled analysis. Front Oncol 2024; 13:1308432. [PMID: 38264746 PMCID: PMC10803636 DOI: 10.3389/fonc.2023.1308432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Nuclear protein of the testis (NUT) midline carcinoma (NMC) is a rare tumor, with particularly low incidence in the lungs, and a correspondingly poor prognosis. To determine the clinicopathological characteristics, outcomes, and prognostic factors of primary pulmonary NMC, a case was reported and a systematic review was performed. Twenty-nine records, including ours, involving 62 cases, were finally included. The median age at diagnosis was 29.5 years. At presentation, the most common symptoms at presentation were cough (47.50%) and chest/back pain (37.50%). In terms of diagnosis, 32.14% of NMC cases were identified through immunohistochemistry (IHC); However, a greater number of cases may be misdiagnosed initially, and ultimately, the diagnosis of NMC was confirmed through a combination of IHC and fluorescence in situ hybridization (FISH). Despite the clinical application of various chemotherapy-based treatments, the actual effectiveness remains unsatisfactory. Furthermore, Cox regression analysis of multiple factors identified male gender and concurrent presence of pleural effusion as indicators of shorter survival time in patients. These results emphasize the importance of increased diagnostic awareness among clinical and pathology practitioners concerning NMC. While there is currently no established standard for treating NMC, a treatment approach combining multiple methods shows promise for future research. Concurrently, clinical and foundational investigations addressing variables such as gender and the presence of pleural effusion may yield valuable insights into the diagnosis and treatment of NMC.
Collapse
Affiliation(s)
- Dong Zhao
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shiqian Zha
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yixuan Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhou Pan
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingyi Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
12
|
Chen M, Li S, Jiang L. Clinicopathological molecular characterizations of sinonasal NUT carcinoma: a report of two cases and a literature review. Front Oncol 2024; 13:1296862. [PMID: 38239638 PMCID: PMC10794637 DOI: 10.3389/fonc.2023.1296862] [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: 09/19/2023] [Accepted: 12/01/2023] [Indexed: 01/22/2024] Open
Abstract
Background Nuclear protein in testis (NUT) carcinoma (NC) is a rare, aggressive tumor with a typical NUTM1 gene rearrangement. Methods Herein, we report a series of 2 cases of sinonasal NC: one in a 16-year-old woman and one in a 37-year-old man. Immunohistochemistry (IHC) staining for NUT (C52B1), fluorescence in situ hybridization (FISH), and next generation sequencing (NGS) sequencing were performed to investigate the morphological and genetic features of sinonasal NC. Results The two cases presented similar pathological features and IHC markers, and typical morphological changes, including undifferentiated cells and abrupt keratinization, were observed, with numerous mitotic figures and widespread tumor necrosis. Diffuse expression of NUT, CK, p63, and p40 was noted, while the tumors were negative for synaptophysin, chromogranin A, S-100, EBV-ISH, and PD-L1. Both tumors harbored a NUTM1 rearrangement. Subsequent sequencing revealed a rare BRD3::NUTM1 fusion and a classic BRD4::NUTM1 fusion. In addition, MCL1 copy number gain (2.1), low tumor mutation burden and stable microsatellites, were also confirmed. Case 1 received surgery and chemoradiotherapy but died 13 months after local recurrence and subsequent lung and bone metastasis. Case 2 underwent chemoradiotherapy and unfortunately died from the disease 6 months later. A review of all previously reported cases of sinonasal NCs (n=55) revealed that these tumors occur more frequently in female pediatric patients (n=11, male: female =3:8), whereas this sex difference is not observed in adult patients (n=44, male: female =23:21). The median survival times of pediatric and adult patients were 17 and 13.8 months, respectively. Conclusion Sinonasal NC presents typical undifferentiated or poorly differentiated cells, abrupt keratinization features and heterogeneous genotypes, including BRD4::NUTM1 and BRD3::NUTM1 fusions, with low tumor mutation burden and stable microsatellites.
Collapse
Affiliation(s)
| | | | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Yoshida A. NUT carcinoma and thoracic SMARCA4-deficient undifferentiated tumour: facts and controversies. Histopathology 2024; 84:86-101. [PMID: 37873676 DOI: 10.1111/his.15063] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/09/2023] [Accepted: 09/24/2023] [Indexed: 10/25/2023]
Abstract
NUT carcinoma and thoracic SMARCA4-deficient undifferentiated tumour are unique entities in the 5th edition of the World Health Organisation (WHO) Classification of Thoracic Tumours, whose definitions include molecular genetic abnormalities. These aggressive tumours require rapid work-ups on biopsies, but a broad list of differential diagnoses poses challenges for practising pathologists. This review provides an update on their key clinicopathological and molecular characteristics, as well as controversies regarding tumour classification and diagnostic strategy. Phenotypical assessment plays a substantial role in diagnosis because recurrent and predictable clinicopathological findings exist, including robust immunohistochemical phenotypes. Accurate diagnosis is crucial for appropriate management and a clearer understanding of the disease.
Collapse
Affiliation(s)
- Akihiko Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
- Rare Cancer Center, National Cancer Center, Tokyo, Japan
| |
Collapse
|
14
|
Hu R, Hou H, Li Y, Zhang M, Li X, Chen Y, Guo Y, Sun H, Zhao S, Liao M, Cao D, Yan Q, Chen X, Yin M. Combined BET and MEK Inhibition synergistically suppresses melanoma by targeting YAP1. Theranostics 2024; 14:593-607. [PMID: 38169595 PMCID: PMC10758063 DOI: 10.7150/thno.85437] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024] Open
Abstract
Rationale: The response rate to the MEK inhibitor trametinib in BRAF-mutated melanoma patients is less than 30%, and drug resistance develops rapidly, but the mechanism is still unclear. Yes1-associated transcriptional regulator (YAP1) is highly expressed in melanoma and may be related to MEK inhibitor resistance. The purpose of this study was to investigate the mechanism of YAP1 in MEK inhibitor resistance in melanoma and to screen YAP1 inhibitors to further determine whether YAP1 inhibition reverses MEK inhibitor resistance. Methods: On the one hand, we analyzed paired melanoma and adjacent tissue samples using RNA-seq and found that the Hippo-YAP1 signaling pathway was the top upregulated pathway. On the other hand, we evaluated the transcriptomes of melanoma samples from patients before and after trametinib treatment and investigated the correlation between YAP1 expression and trametinib resistance. Then, we screened for inhibitors that repress YAP1 expression and investigated the mechanisms. Finally, we investigated the antitumor effect of YAP1 inhibition combined with MEK inhibition both in vitro and in vivo. Results: We found that YAP1 expression levels upon trametinib treatment in melanoma patients were correlated with resistance to trametinib. YAP1 was translocated into the nucleus after trametinib treatment in melanoma cells, which could render resistance to MEK inhibition. Thus, we screened for inhibitors that repress YAP1 expression and identified multiple bromodomain and extra-terminal (BET) inhibitors, including NHWD-870, as hits. BET inhibition repressed YAP1 expression by decreasing BRD4 binding to the YAP1 promoter. Consistently, YAP1 overexpression was sufficient to reverse the proliferation defect caused by BRD4 depletion. In addition, the BET inhibitor NHWD-870 acted synergistically with trametinib to suppress melanoma growth in vitro and in vivo. Conclusions: We identified a new vulnerability for MEK inhibitor-resistant melanomas, which activated Hippo pathway due to elevated YAP1 activity. Inhibition of BRD4 using BET inhibitors suppressed YAP1 expression and led to blunted melanoma growth when combined with treatment with the MEK inhibitor trametinib.
Collapse
Affiliation(s)
- Rui Hu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huihui Hou
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yao Li
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yanzhong Chen
- The first clinical college of Chongqing Medical University, Chongqing, China
| | - Ying Guo
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hongyin Sun
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shuang Zhao
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Mengting Liao
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Health Management of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Dongsheng Cao
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Qin Yan
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Mingzhu Yin
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Cancer Early Detection and Treatment Center (CEDTC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
- Translational Medicine Research Center (TMRC), School of Medicine Chongqing University, Shapingba, Chongqing, China
| |
Collapse
|
15
|
Durall RT, Huang J, Wojenski L, Huang Y, Gokhale PC, Leeper BA, Nash JO, Ballester PL, Davidson S, Shlien A, Sotirakis E, Bertaux F, Dubus V, Luo J, Wu CJ, Keskin DB, Eagen KP, Shapiro GI, French CA. The BRD4-NUT Fusion Alone Drives Malignant Transformation of NUT Carcinoma. Cancer Res 2023; 83:3846-3860. [PMID: 37819236 PMCID: PMC10690098 DOI: 10.1158/0008-5472.can-23-2545] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/26/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
NUT carcinoma (NC) is an aggressive squamous carcinoma defined by the BRD4-NUT fusion oncoprotein. Routinely effective systemic treatments are unavailable for most NC patients. The lack of an adequate animal model precludes identifying and leveraging cell-extrinsic factors therapeutically in NC. Here, we created a genetically engineered mouse model (GEMM) of NC that forms a Brd4::NUTM1 fusion gene upon tamoxifen induction of Sox2-driven Cre. The model displayed complete disease penetrance, with tumors arising from the squamous epithelium weeks after induction and all mice succumbing to the disease shortly thereafter. Closely resembling human NC (hNC), GEMM tumors (mNC) were poorly differentiated squamous carcinomas with high expression of MYC that metastasized to solid organs and regional lymph nodes. Two GEMM-derived cell lines were developed whose transcriptomic and epigenetic landscapes harbored key features of primary GEMM tumors. Importantly, GEMM tumor and cell line transcriptomes co-classified with those of human NC. BRD4-NUT also blocked differentiation and maintained the growth of mNC as in hNC. Mechanistically, GEMM primary tumors and cell lines formed large histone H3K27ac-enriched domains, termed megadomains, that were invariably associated with the expression of key NC-defining proto-oncogenes, Myc and Trp63. Small-molecule BET bromodomain inhibition (BETi) of mNC induced differentiation and growth arrest and prolonged survival of NC GEMMs, as it does in hNC models. Overall, tumor formation in the NC GEMM is definitive evidence that BRD4-NUT alone can potently drive the malignant transformation of squamous progenitor cells into NC. SIGNIFICANCE The development of an immunocompetent model of NUT carcinoma that closely mimics the human disease provides a valuable global resource for mechanistic and preclinical studies to improve treatment of this incurable disease.
Collapse
Affiliation(s)
- R. Taylor Durall
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Julianna Huang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Yeying Huang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Prafulla C. Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Brittaney A. Leeper
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joshua O. Nash
- Program in Genetics and Genome Biology, The Hospital for Sick Children (SickKids), University of Toronto, Toronto, Ontario, Canada
- Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Pedro L. Ballester
- Program in Genetics and Genome Biology, The Hospital for Sick Children (SickKids), University of Toronto, Toronto, Ontario, Canada
| | - Scott Davidson
- Program in Genetics and Genome Biology, The Hospital for Sick Children (SickKids), University of Toronto, Toronto, Ontario, Canada
| | - Adam Shlien
- Program in Genetics and Genome Biology, The Hospital for Sick Children (SickKids), University of Toronto, Toronto, Ontario, Canada
- Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Jia Luo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Derin B. Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kyle P. Eagen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christopher A. French
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Huang Y, Durall RT, Luong NM, Hertzler HJ, Huang J, Gokhale PC, Leeper BA, Persky NS, Root DE, Anekal PV, Montero Llopis PD, David CN, Kutok JL, Raimondi A, Saluja K, Luo J, Zahnow CA, Adane B, Stegmaier K, Hawkins CE, Ponne C, Le Q, Shapiro GI, Lemieux ME, Eagen KP, French CA. EZH2 Cooperates with BRD4-NUT to Drive NUT Carcinoma Growth by Silencing Key Tumor Suppressor Genes. Cancer Res 2023; 83:3956-3973. [PMID: 37747726 PMCID: PMC10843040 DOI: 10.1158/0008-5472.can-23-1475] [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: 05/17/2023] [Revised: 07/31/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
NUT carcinoma is an aggressive carcinoma driven by the BRD4-NUT fusion oncoprotein, which activates chromatin to promote expression of progrowth genes. BET bromodomain inhibitors (BETi) are a promising treatment for NUT carcinoma that can impede BRD4-NUT's ability to activate genes, but the efficacy of BETi as monotherapy is limited. Here, we demonstrated that enhancer of zeste homolog 2 (EZH2), which silences genes through establishment of repressive chromatin, is a dependency in NUT carcinoma. Inhibition of EZH2 with the clinical compound tazemetostat potently blocked growth of NUT carcinoma cells. Epigenetic and transcriptomic analysis revealed that tazemetostat reversed the EZH2-specific H3K27me3 silencing mark and restored expression of multiple tumor suppressor genes while having no effect on key oncogenic BRD4-NUT-regulated genes. Indeed, H3K27me3 and H3K27ac domains were found to be mutually exclusive in NUT carcinoma cells. CDKN2A was identified as the only gene among all tazemetostat-derepressed genes to confer resistance to tazemetostat in a CRISPR-Cas9 screen. Combined inhibition of EZH2 and BET synergized to downregulate cell proliferation genes, resulting in more pronounced growth arrest and differentiation than either inhibitor alone. In preclinical models, combined tazemetostat and BETi synergistically blocked tumor growth and prolonged survival of NUT carcinoma-xenografted mice, with complete remission without relapse in one cohort. Identification of EZH2 as a dependency in NUT carcinoma substantiates the reliance of NUT carcinoma tumor cells on epigenetic dysregulation of functionally opposite, yet highly complementary, chromatin regulatory pathways to maintain NUT carcinoma growth. SIGNIFICANCE Repression of tumor suppressor genes, including CDKN2A, by EZH2 provides a mechanistic rationale for combining EZH2 and BET inhibitors for the clinical treatment of NUT carcinoma. See related commentary by Kazansky and Kentsis, p. 3827.
Collapse
Affiliation(s)
- Yeying Huang
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - R. Taylor Durall
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nhi M. Luong
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hans J. Hertzler
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Julianna Huang
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Prafulla C. Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brittaney A. Leeper
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - David E. Root
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Praju V. Anekal
- MicRoN, Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Karan Saluja
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston, TX, USA
| | - Jia Luo
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Cynthia A. Zahnow
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Biniam Adane
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA
| | - Catherine E. Hawkins
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Christopher Ponne
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Quan Le
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Kyle P. Eagen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. French
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Nishimura Y, Ryo E, Inoue S, Kawazu M, Ueno T, Namikawa K, Takahashi A, Ogata D, Yoshida A, Yamazaki N, Mano H, Yatabe Y, Mori T. Strategic Approach to Heterogeneity Analysis of Cutaneous Adnexal Carcinomas Using Computational Pathology and Genomics. JID INNOVATIONS 2023; 3:100229. [PMID: 37965425 PMCID: PMC10641284 DOI: 10.1016/j.xjidi.2023.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 07/30/2023] [Accepted: 08/07/2023] [Indexed: 11/16/2023] Open
Abstract
Cutaneous adnexal tumors are neoplasms that arise from skin appendages. Their morphologic diversity and phenotypic variability with rare progression to malignancy make them difficult to diagnose and classify, and there is currently no established treatment strategy. To overcome these difficulties, this study investigated the transcription factor SOX9 expression, morphology, and genetics of skin adnexal tumors for understanding their biology, especially their histogenesis. We showed that cutaneous adnexal tumors and their nontumor counterparts of skin and appendages exhibit expression patterns similar to that of SOX9. Its expression intensity and pattern, as well as histopathologic evaluation of tumors, were analyzed using digital images of 69 normal skin adnexal 9-type organs and 185 skin adnexal 29-type tumors as references. It was possible to distinguish basal cell carcinoma from squamous cell carcinoma, sebaceous carcinoma, and pilomatrixoma with significant differences, along with porocarcinoma from squamous cell carcinoma. Furthermore, unsupervised machine learning "computational pathology" was used to derive a multiregion whole-exome sequencing fusion method termed "genocomputed pathology." The genocomputed pathology of three representable adnexal carcinomas (porocarcinoma, hidradenocarcinoma, and spiradenocarcinoma) was evaluated for total nine cases. We showed that there was more heterogeneity than expected within the tumors as well as the coexistence of components lacking driver fusion genes. The presence or absence of potential driver genes, such as PIK3CA, YAP1, and PTEN, in each region was identified, highlighting a therapeutic strategy for cutaneous adnexal carcinoma encompassing heterogeneous tumors.
Collapse
Affiliation(s)
- Yuuki Nishimura
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Eijitsu Ryo
- Division of Molecular Pathology, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Satoshi Inoue
- Division of Cellular Signaling, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Masahito Kawazu
- Division of Cellular Signaling, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Kenjiro Namikawa
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akira Takahashi
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Dai Ogata
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akihiko Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Naoya Yamazaki
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Division of Molecular Pathology, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Taisuke Mori
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
- Division of Molecular Pathology, National Cancer Center Reserch Institute, Tokyo, Japan
| |
Collapse
|
18
|
Huang Y, Durall RT, Luong NM, Hertzler HJ, Huang J, Gokhale PC, Leeper BA, Persky NS, Root DE, Anekal PV, Montero Llopis PD, David CN, Kutok JL, Raimondi A, Saluja K, Luo J, Zahnow CA, Adane B, Stegmaier K, Hawkins CE, Ponne C, Le Q, Shapiro GI, Lemieux ME, Eagen KP, French CA. EZH2 synergizes with BRD4-NUT to drive NUT carcinoma growth through silencing of key tumor suppressor genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.15.553204. [PMID: 37645799 PMCID: PMC10461970 DOI: 10.1101/2023.08.15.553204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
NUT carcinoma (NC) is an aggressive carcinoma driven by the BRD4-NUT fusion oncoprotein, which activates chromatin to promote expression of pro-growth genes. BET bromodomain inhibitors (BETi) impede BRD4-NUT's ability to activate genes and are thus a promising treatment but limited as monotherapy. The role of gene repression in NC is unknown. Here, we demonstrate that EZH2, which silences genes through establishment of repressive chromatin, is a dependency in NC. Inhibition of EZH2 with the clinical compound tazemetostat (taz) potently blocked growth of NC cells. Epigenetic and transcriptomic analysis revealed that taz reversed the EZH2-specific H3K27me3 silencing mark, and restored expression of multiple tumor suppressor genes while having no effect on key oncogenic BRD4- NUT-regulated genes. CDKN2A was identified as the only gene amongst all taz-derepressed genes to confer resistance to taz in a CRISPR-Cas9 screen. Combined EZH2 inhibition and BET inhibition synergized to downregulate cell proliferation genes resulting in more pronounced growth arrest and differentiation than either inhibitor alone. In pre-clinical models, combined taz and BETi synergistically blocked growth and prolonged survival of NC-xenografted mice, with all mice cured in one cohort. STATEMENT OF SIGNIFICANCE Identification of EZH2 as a dependency in NC substantiates the reliance of NC tumor cells on epigenetic dysregulation of functionally opposite, yet highly complementary chromatin regulatory pathways to maintain NC growth. In particular, repression of CDKN2A expression by EZH2 provides a mechanistic rationale for combining EZH2i with BETi for the clinical treatment of NC.
Collapse
Affiliation(s)
- Yeying Huang
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - R. Taylor Durall
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nhi M. Luong
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hans J. Hertzler
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Julianna Huang
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Prafulla C. Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brittaney A. Leeper
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - David E. Root
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Praju V. Anekal
- MicRoN, Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Karan Saluja
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston, TX, USA
| | - Jia Luo
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Cynthia A. Zahnow
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Biniam Adane
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA
| | - Catherine E. Hawkins
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Christopher Ponne
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Quan Le
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Kyle P. Eagen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. French
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Bornhorst M, Eze A, Bhattacharya S, Putnam E, Almira-Suarez MI, Rossi C, Kambhampati M, Almalvez M, Barseghyan M, Del Risco N, Dotson D, Turner J, Myseros JS, Vilain E, Packer RJ, Nazarian J, Rood B, Barseghyan H. Optical genome mapping identifies a novel pediatric embryonal tumor with a ZNF532::NUTM1 fusion. J Pathol 2023; 260:329-338. [PMID: 37203791 PMCID: PMC10330119 DOI: 10.1002/path.6085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 03/06/2023] [Accepted: 03/31/2023] [Indexed: 05/20/2023]
Abstract
The molecular characteristics of pediatric brain tumors have not only allowed for tumor subgrouping but have led to the introduction of novel treatment options for patients with specific tumor alterations. Therefore, an accurate histologic and molecular diagnosis is critical for optimized management of all pediatric patients with brain tumors, including central nervous system embryonal tumors. We present a case where optical genome mapping identified a ZNF532::NUTM1 fusion in a patient with a unique tumor best characterized histologically as a central nervous system embryonal tumor with rhabdoid features. Additional analyses including immunohistochemistry for NUT protein, methylation array, whole genome, and RNA-sequencing was done to confirm the presence of the fusion in the tumor. This is the first description of a pediatric patient with a ZNF532::NUTM1 fusion, yet the histology of this tumor is similar to that of adult cancers with ZNF::NUTM1 fusions reported in the literature. Although rare, the distinct pathology and underlying molecular characteristics of the ZNF532::NUTM1 tumor separates this from other embryonal tumors. Therefore, screening for this or similar NUTM1 rearrangements should be considered for all patients with unclassified central nervous system tumors with rhabdoid features to ensure accurate diagnosis. Ultimately, with additional cases, we may be able to better inform therapeutic management for these patients. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Miriam Bornhorst
- Division of Hematology/Oncology, Children’s National Hospital, Washington, DC 20010, USA
- Brain Tumor Institute, Children’s National Hospital, Washington, DC 20010, USA
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
- Department of Pediatrics, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20037, USA
- Center for Neuroscience and Behavioral Medicine, Children’s National Hospital, Washington, DC, 20010, USA
| | - Augustine Eze
- Brain Tumor Institute, Children’s National Hospital, Washington, DC 20010, USA
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
| | - Surajit Bhattacharya
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
| | - Ethan Putnam
- Brain Tumor Institute, Children’s National Hospital, Washington, DC 20010, USA
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
| | | | - Christopher Rossi
- Divison of Pathology, Children’s National Hospital, Washington, DC, 20010, USA
| | - Madhuri Kambhampati
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
| | - Miguel Almalvez
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
| | - Mariam Barseghyan
- Department of Obstetrics and Gynecology, MedStar Georgetown University Hospital, Washington, DC, 20007, USA
| | - Nicole Del Risco
- School of Medicine and Health Sciences, George Washington University, Washington, DC, 20037, USA
| | | | - Joyce Turner
- Division of Genetics and Metabolism, Children’s National Hospital, Washington, DC, 20010, USA
| | - John S. Myseros
- Division of Neurosurgery, Children’s National Hospital, Washington, DC, 20010, USA
| | - Eric Vilain
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
- Center for Genomics and Precision Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Roger J. Packer
- Brain Tumor Institute, Children’s National Hospital, Washington, DC 20010, USA
- Center for Neuroscience and Behavioral Medicine, Children’s National Hospital, Washington, DC, 20010, USA
| | - Javad Nazarian
- Brain Tumor Institute, Children’s National Hospital, Washington, DC 20010, USA
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
- Center for Genomics and Precision Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Brian Rood
- Division of Hematology/Oncology, Children’s National Hospital, Washington, DC 20010, USA
- Brain Tumor Institute, Children’s National Hospital, Washington, DC 20010, USA
- Department of Pediatrics, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20037, USA
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC, 20010, USA
| | - Hayk Barseghyan
- Center for Genetics Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
- Center for Genomics and Precision Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| |
Collapse
|
20
|
Guo J, Zheng Q, Peng Y. BET proteins: Biological functions and therapeutic interventions. Pharmacol Ther 2023; 243:108354. [PMID: 36739915 DOI: 10.1016/j.pharmthera.2023.108354] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Bromodomain and extra-terminal (BET) family member proteins (BRD2, BRD3, BRD4 and BRDT) play a pivotal role in interpreting the epigenetic information of histone Kac modification, thus controlling gene expression, remodeling chromatin structures and avoid replicative stress-induced DNA damages. Abnormal activation of BET proteins is tightly correlated to various human diseases, including cancer. Therefore, BET bromodomain inhibitors (BBIs) were considered as promising therapeutics to treat BET-related diseases, raising >70 clinical trials in the past decades. Despite preliminary effects achieved, drug resistance and adverse events represent two major challenges for current BBIs development. In this review, we will introduce the biological functions of BET proteins in both physiological and pathological conditions; and summarize the progress in current BBI drug development. Moreover, we will also discuss the major challenges in the front of BET inhibitor development and provide rational strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Jiawei Guo
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingquan Zheng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
21
|
Chen M, Chen X, Zhang Y, Wang W, Jiang L. Clinical and molecular features of pulmonary NUT carcinoma characterizes diverse responses to immunotherapy, with a pathologic complete response case. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04621-5. [PMID: 36752907 DOI: 10.1007/s00432-023-04621-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/27/2023] [Indexed: 02/09/2023]
Abstract
PURPOSE Nuclear protein in testis (NUT) carcinoma is an uncommon malignant cancer characterized by NUTM1 rearrangement. We aimed to investigate the clinicopathological and molecular features and immunotherapy of pulmonary NUT carcinoma. METHODS Immunohistochemistry (IHC) for NUT (C52B1) and programmed cell death ligand 1 (PD-L1: 22C3) and fluorescence in situ hybridization (FISH) for NUTM1 break and BRD4-NUTM1 fusion were performed on six pulmonary NUT carcinoma samples. RESULTS The 6 pulmonary NUT carcinoma samples were obtained from 5 males and 1 female, with ages ranging from 31 to 73 years (average, 46 years). Five tumors occurred in the lobes, with one in the trachea. Pathologically, all cases showed primitive-appearing round to epithelioid cells growing in nests and sheets. Squamous differentiation and abrupt keratinization were observed. All tumors expressed the NUT protein and p63, and 4 tumors showed focal synaptophysin, but PD-L1 expression was not observed. All cases displayed NUTM1 rearrangement, 5 had BRD4-NUTM1 fusion, and one had an unknown partner. Three patients presented regional lymph node involvement at diagnosis. Five patients underwent intensive radiation and/or chemotherapy. Furthermore, 2 patients (1 and 2) received a combination of PD-L1 inhibitor and chemotherapy. Patient 1 exhibited a poor response and soon showed tumor progression and metastasis; however, patient 2 responded remarkably and achieved pathologic complete response (pCR) without uncontrollable adverse events. The overall survival time was 2.9 months. CONCLUSIONS Pulmonary NUT carcinoma exhibits poorly differentiated morphological features with diffuse NUT staining, low PD-L1 expression, and NUTM1 rearrangement. Despite its poor prognosis, it presents a diverse response to immunotherapy. Immune checkpoint inhibitors (ICIs) need to be further explored in NUT carcinoma.
Collapse
Affiliation(s)
- Min Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaohua Chen
- Department of Pathology, The Second People's Hospital of Yibin, Yibin, 644000, China
| | - Ying Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weiya Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
22
|
Kuhn E, Pescia C, Mendogni P, Nosotti M, Ferrero S. Thymic Epithelial Tumors: An Evolving Field. Life (Basel) 2023; 13:314. [PMID: 36836670 PMCID: PMC9964105 DOI: 10.3390/life13020314] [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: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Despite their rarity, thymic epithelial tumors (TETs) have attracted much interest over the years, leading to an impressive number of histological and staging classifications. At present, TETs are divided by the WHO classification into four main subtypes: type A, type AB, and type B thymomas (subdivided into B1, B2, and B3), and thymic carcinomas, going from the more indolent to the most aggressive ones. Among many debated staging proposals, the TNM and the Masaoka-Koga staging systems have been widely accepted and used in routine practice. The four-tiered histological classification is symmetrically mirrored by the molecular subgrouping of TETs, which identifies an A-like and an AB-like cluster, with frequent GTF2I and HRAS mutations; an intermediate B-like cluster, with a T-cell signaling profile; and a carcinoma-like cluster comprising thymic carcinomas with frequent CDKN2A and TP53 alterations and a high tumor molecular burden. Molecular investigations have opened the way to tailored therapies, such as tyrosine kinase inhibitors targeting KIT, mTOR, and VEGFR, and immune-checkpoints that have been adopted as second-line systemic treatments. In this review, we discuss the crucial events that led to the current understanding of TETs, while disclosing the next steps in this intriguing field.
Collapse
Affiliation(s)
- Elisabetta Kuhn
- S.C. Anatomia Patologica, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20122 Milano, Italy
| | - Carlo Pescia
- S.C. Anatomia Patologica, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20122 Milano, Italy
| | - Paolo Mendogni
- S.C. Chirurgia Toracica e Trapianti di Polmone, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Mario Nosotti
- S.C. Chirurgia Toracica e Trapianti di Polmone, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
- Dipartimento di Patofisiologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20122 Milano, Italy
| | - Stefano Ferrero
- S.C. Anatomia Patologica, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20122 Milano, Italy
| |
Collapse
|
23
|
Barletta JA, Gilday SD, Afkhami M, Bell D, Bocklage T, Boisselier P, Chau NG, Cipriani NA, Costes-Martineau V, Ghossein RA, Hertzler HJ, Kramer AM, Limaye S, Lopez CA, Ng TL, Weissferdt A, Xu B, Zhang S, French CA. NUTM1 -rearranged Carcinoma of the Thyroid : A Distinct Subset of NUT Carcinoma Characterized by Frequent NSD3 - NUTM1 Fusions. Am J Surg Pathol 2022; 46:1706-1715. [PMID: 36040068 PMCID: PMC9669222 DOI: 10.1097/pas.0000000000001967] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
NUT carcinoma (NC) is a rare subtype of squamous cell carcinoma defined by NUTM1 rearrangements encoding NUT fusion oncoproteins (the most frequent fusion partner being BRD4 ) that carries a very poor prognosis, with most patients dying in under 1 year. Only rare primary thyroid NCs have been reported. Here, we evaluated a series of 14 cases. The median patient age at diagnosis was 38 years (range: 17 to 72 y). Eight of 13 cases with slides available for review (62%) showed a morphology typical of NC, whereas 5 (38%) had a non-NC-like morphology, some of which had areas of cribriform or fused follicular architecture resembling a follicular cell-derived thyroid carcinoma. For cases with immunohistochemistry results, 85% (11/13) were positive for NUT on biopsy or resection, though staining was significantly decreased on resection specimens due to fixation; 55% (6/11) were positive for PAX8, and 54% (7/13) for TTF-1. Tumors with a non-NC-like morphology were all positive for PAX8 and TTF-1. The fusion partner was known in 12 cases: 9 (75%) cases had a NSD3-NUTM1 fusion, and 3 (25%) had a BRD4-NUTM1 fusion. For our cohort, the 2-year overall survival (OS) was 69%, and the 5-year OS was 58%. Patients with NC-like tumors had a significantly worse OS compared with that of patients with tumors with a non-NC-like morphology ( P =0.0462). Our study shows that NC of the thyroid can mimic other thyroid primaries, has a high rate of NSD3 - NUTM1 fusions, and an overall more protracted clinical course compared with nonthyroid primary NC.
Collapse
Affiliation(s)
- Justine A. Barletta
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven D. Gilday
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michelle Afkhami
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Diana Bell
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Theresa Bocklage
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Pierre Boisselier
- Department of Radiation Oncology, ICM, Montpellier Cancer Institute, Montpellier, France
| | - Nicole G. Chau
- Department of Medical Oncology, BC Cancer, Vancouver BC, Canada
| | | | | | - Ronald A. Ghossein
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hans J. Hertzler
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Sewanti Limaye
- Kokilaben Dhirubhai Ambani Hospital and Medical Research Institute, Mumbai, India
| | - Carlos A. Lopez
- Division of Medical Oncology & Hematology, Northwell Health Cancer Institute, Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY 11042, USA
| | - Tony L. Ng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Annikka Weissferdt
- Department of Pathology and Laboratory Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Bin Xu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Songlin Zhang
- Department of Pathology and Laboratory Medicine, McGovern Medical School, Houston, TX
| | - Christopher A. French
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Lauer UM, Hinterleitner M, Horger M, Ohnesorge PV, Zender L. NUT Carcinoma—An Underdiagnosed Malignancy. Front Oncol 2022; 12:914031. [PMID: 35957893 PMCID: PMC9360329 DOI: 10.3389/fonc.2022.914031] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
NUT carcinoma (NC) is a rare and highly aggressive malignancy with a dismal prognosis and a median survival of 6–9 months only. Although very few cases of NC are reported each year, the true prevalence is estimated to be much higher, with NC potentially widely underdiagnosed due to the lack of awareness. NC primarily occurs in midline structures including thorax, head, and neck; however, other sites such as pancreas and kidney are also affected, albeit at lower frequencies. NC is characterized by a single translocation involving the NUTM1 (NUT midline carcinoma family member 1) gene and different partner genes. The resulting fusion proteins initiate tumorigenesis through a mechanism involving BET (bromo-domain and extra-terminal motif) proteins such as Bromodomain-containing protein 4 (BRD4) and inordinate acetylation of chromatin, leading to the dysregulation of growth and differentiation genes. While no clinical characteristics are specific for NC, some histologic features can be indicative; therefore, patients with these tumor characteristics should be routinely tested for NUTM1. The diagnosis of NC using immunohistochemistry with a highly specific antibody is straightforward. There are currently no standard-of-care treatment options for patients with NC. However, novel therapies specifically addressing the unique tumorigenic mechanism are under investigation, including BET inhibitors. This review aims to raise awareness of this underdiagnosed cancer entity and provide all patients the opportunity to be properly diagnosed and referred to a clinical study.
Collapse
Affiliation(s)
- Ulrich M. Lauer
- Medical Oncology and Pneumology, Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- *Correspondence: Ulrich M. Lauer,
| | - Martina Hinterleitner
- Medical Oncology and Pneumology, Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Marius Horger
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Tübingen, Germany
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tübingen, Germany
| | - Paul V. Ohnesorge
- Medical Oncology and Pneumology, Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Lars Zender
- Medical Oncology and Pneumology, Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| |
Collapse
|
25
|
French CA, Cheng ML, Hanna GJ, DuBois SG, Chau NG, Hann CL, Storck S, Salgia R, Trucco M, Tseng J, Stathis A, Piekarz R, Lauer UM, Massard C, Bennett K, Coker S, Tontsch-Grunt U, Sos ML, Liao S, Wu CJ, Polyak K, Piha-Paul SA, Shapiro GI. Report of the First International Symposium on NUT Carcinoma. Clin Cancer Res 2022; 28:2493-2505. [PMID: 35417004 PMCID: PMC9197941 DOI: 10.1158/1078-0432.ccr-22-0591] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/28/2022] [Accepted: 04/08/2022] [Indexed: 12/15/2022]
Abstract
NUT carcinoma is a rare, aggressive cancer defined by rearrangements of the NUTM1 gene. No routinely effective treatments of NUT carcinoma exist, despite harboring a targetable oncoprotein, most commonly BRD4-NUT. The vast majority of cases are fatal. Poor awareness of the disease is a major obstacle to progress in the treatment of NUT carcinoma. While the incidence likely exceeds that of Ewing sarcoma, and BRD4-NUT heralded the bromodomain and extra-terminal domain (BET) inhibitor class of selective epigenetic modulators, NUT carcinoma is incorrectly perceived as "impossibly rare," and therefore receives comparatively little private or governmental funding or prioritization by pharma. To raise awareness, propagate scientific knowledge, and initiate a consensus on standard and targeted treatment of NUT carcinoma, we held the First International Symposium on NUT Carcinoma on March 3, 2021. This virtual event had more than eighty attendees from the Americas, Europe, Asia, and Australia. Patients with NUT carcinoma and family members were represented and shared perspectives. Broadly, the four areas discussed by experts in the field included (1) the biology of NUT carcinoma; (2) standard approaches to the treatment of NUT carcinoma; (3) results of clinical trials using BET inhibitors; and (4) future directions, including novel BET bromodomain inhibitors, combinatorial approaches, and immunotherapy. It was concluded that standard chemotherapeutic approaches and first-generation BET bromodomain inhibitors, the latter complicated by a narrow therapeutic window, are only modestly effective in a minority of cases. Nonetheless, emerging second-generation targeted inhibitors, novel rational synergistic combinations, and the incorporation of immuno-oncology approaches hold promise to improve the prognosis of this disease.
Collapse
Affiliation(s)
| | | | | | - Steven G. DuBois
- Dana-Farber Cancer Institute, Boston, MA, USA,Boston Children’s Hospital, Boston, MA, USA
| | - Nicole G. Chau
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | | | - Simone Storck
- Swabian Children’s Cancer Center, Paediatric and Adolescent Medicine, University Medical Center Augsburg, Augsburg, Germany
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | | | | | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland and Faculty of Biomedical Sciences, Universita della Svizzera Italiana, Lugano, Switzerland
| | - Richard Piekarz
- Investigational Drug Branch, Cancer Therapy Evaluation Program (CTEP), Bethesda, MD
| | | | - Christophe Massard
- Gustave Roussy-Molecular Radiotherapy INSERM U1030, Faculty of Medicine Kremlin-Bicêtre and Paris-Saclay University , France
| | | | - Shodeinde Coker
- Bristol-Myers Squibb Company, Lawrenceville, New Jersey, USA
| | | | - Martin L. Sos
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Pathology, Molecular Pathology University of Cologne, Cologne, Germany and Department of Translational Genomics and Center for Molecular Medicine Cologne, Cologne, Germany
| | - Sida Liao
- TScan Therapeutics, Waltham, MA, USA
| | | | | | - Sarina A. Piha-Paul
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
26
|
Abreu RF, Oliveira TBD, Hertzler H, Toledo RN, D'Almeida Costa F, Lopes Pinto CA, Nunes WA, Nascimento AF, French CA, Nascimento AG. NUT Carcinoma, an under-recognized malignancy: a clinicopathologic and molecular series of 6 cases showing a subset of patients with better prognosis and a rare ZNF532::NUTM1 fusion. Hum Pathol 2022; 126:87-99. [PMID: 35623465 DOI: 10.1016/j.humpath.2022.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 12/21/2022]
Abstract
NUT carcinoma (NC) is a rare malignancy with aggressive clinical behavior, defined by rearrangements involving the NUTM1 gene locus. This entity is often under recognized and its diagnosis may be challenging. In this study, we describe a subset of patients that, despite the molecularly proven diagnosis of NC, show improved outcome. In addition, we describe one case with the novel ZNF532::NUTM1 fusion. All cases of NC diagnosed from 2013 to 2022 in our department were retrieved. FISH using dual color bring-together probes and next-generation sequencing assay were performed to characterize the fusions involving NUTM1. Amongst 6 patients identified, 5 were male with a median age of 35.6 years. Four patients had primary tumor in the head and neck region (2 ethmoid sinus, 1 parotid gland, 1 lacrimal gland); 1 in the mediastinum, and another presented with femoral bone tumor. In all cases, the initial diagnoses were not NC. The cases showed different morphological patterns, including monomorphic, rhabdoid and pleomorphic appearance. One case showed pseudopapillary pattern. By immunohistochemistry, all tumors showed squamous differentiation and ≥ 50% of neoplastic cells with nuclear positivity for NUT antibody. One case expressed WT1 (C-terminus) and other showed chromogranin positivity. Genetic study revealed a BRD4::NUTM1 fusion in all head and neck cases, BRD3::NUTM1 in mediastinum case, and ZNF532::NUTM1 fusion in the femur bone case. They were treated with surgical resection plus chemo and radiotherapy. The median overall survival was 23.11 months (1.6-83.3 mo) and the median disease free survival was 14.86 months (0-54.4 mo). The patients with longer overall survival were one with a lacrimal gland primary (83.3 months) and other with a parotid lesion (31.9 months). Both patients were primarily treated with complete surgical resection. Anatomic location may be directly related to the overall survival in NC cases. Resectability of the lesion is also an important factor related to survival. Pathologists should include NC in the differential diagnosis of any poorly differentiated and undifferentiated monomorphic malignancy, regardless of its anatomic location.
Collapse
Affiliation(s)
| | | | - Hans Hertzler
- Clinical Research Coordinator for NUT Carcinoma Patients, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, USA
| | - Ronaldo Nunes Toledo
- Department of Head and Neck Surgery, A.C.Camargo Cancer Center, Sao Paulo, 01509-010, Brazil
| | | | | | - Warley Abreu Nunes
- Department of Pathology, A.C.Camargo Cancer Center, Sao Paulo, 01509-010, Brazil
| | - Alessandra F Nascimento
- Department of Pathology, University Hospitals Cleveland Medical Center, Associate Professor of Pathology, Case Western Reserve University, Cleveland, 44106, USA
| | | | | |
Collapse
|
27
|
Abstract
INTRODUCTION Radical surgery is the best therapeutic option for thymic malignancies. However, patients with advanced or recurrent thymic malignancies often require chemotherapy or radiotherapy. Since thymic malignancies are rare cancers, the efficacy and safety of treatments have been verified based on small Phase 2 trials or retrospective studies. AREA COVERED We comprehensively reviewed the treatment strategies for thymic malignancies, including surgery, radiotherapy, and pharmacotherapy, including cytotoxic chemotherapy, molecular-targeted therapy, and immunotherapy. Additionally, we reviewed specific situations, such as pleural dissemination, central nervous system metastasis, and paraneoplastic syndrome. EXPERT OPINION Cytotoxic chemotherapy remains the standard option in pharmacotherapy. However, multikinase inhibitors, such as sunitinib and lenvatinib, and immune checkpoint inhibitors including pembrolizumab have been developed to treat thymic carcinomas. Now, a Phase 2 study is evaluating whether lenvatinib plus pembrolizumab benefits patients with type B3 thymoma or thymic carcinoma. Phosphatidylinositol 3-kinase/AKT/ mammalian target of rapamycin inhibitors may contribute to disease control and octreotide scan is only applicable to somatostatin analogues. Although the genomic characteristics of thymic malignancies have been analyzed, few actionable mutations have been detected in general. The development of a treatment strategy using combination pharmacotherapy is anticipated.
Collapse
Affiliation(s)
- Yutaka Muto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| |
Collapse
|
28
|
Zhou J, Duan M, Jiao Q, Chen C, Xing A, Su P, Tang J, Zhang H, Liu Z. Primary Thyroid NUT Carcinoma With High PD-L1 Expression and Novel Massive IGKV Gene Fusions: A Case Report With Treatment Implications and Literature Review. Front Oncol 2022; 11:778296. [PMID: 35127482 PMCID: PMC8807656 DOI: 10.3389/fonc.2021.778296] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/23/2021] [Indexed: 12/17/2022] Open
Abstract
Background Nuclear protein in testis (NUT) carcinoma (NC) is a rare and aggressive undifferentiated carcinoma that typically arises from midline supradiaphragmatic structures. It is uniquely driven by a NUT gene rearrangement on chromosome 15q14. Few thyroid NCs have been reported and there are no established treatment guidelines for NUT carcinoma. Method Ultrasound-guided fine needle aspiration smear was performed for the preoperative diagnosis of thyroid lesions. Cytopathology, histology, and immunochemical staining all indicated NC. Fluorescence in situ hybridization (FISH), qRT-PCR, and next-generation sequencing (NGS) were used to analyze the genetic characteristics of NC. Results We describe a rare case of thyrogenic NC in a 38-year-old male with cytological, histological, immunohistochemical, and genetic features. Cytological smears and histopathological specimens showed typical features of NC. Immunohistochemistry confirmed strong immunoreactivity with NUT, EMA, P63, TTF-1, and c-myc. CK19 was positive exclusively in sudden keratosis. No immunoreactivity was found for neuroendocrine markers. FISH was applied to isolate the NUT gene on chromosome 15q14. The NGS results revealed a BRD4-NUT gene fusion, which was further confirmed by RT-qPCR. Structural variation (SV) of NUTM1 occurred in the exon region, and the mutation site was 15q14. Moreover, BRD4 single-nucleotide variation (SNV) occurs in the 3′ UTR at mutation site 19p13.12. The PD-L1 combined predictive score was over 30%. The patient received chemotherapy, followed by programmed cell death 1 (PD-1) inhibition with camrelizumab, and died 10 months after surgery. Conclusion Thyroid NC is an extremely rare and fatal malignant tumor. It is necessary to consider NC when squamous differentiation is observed cytologically or histologically. NGS is an effective tool for obtaining the final diagnosis and obtaining a better understanding of tumor pathogenesis. A large number of IGKV gene fusions in addition to the BRD4-NUT fusion may play a role in the pathogenesis and immunotherapy response of NC. Immunotherapy for NC remains to be explored due to the rarity of this aggressive malignancy.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Miao Duan
- Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Chunyan Chen
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Aiyan Xing
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China
| | - Peng Su
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China
| | - Juan Tang
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Hui Zhang
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Hui Zhang, ; Zhiyan Liu,
| | - Zhiyan Liu
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Hui Zhang, ; Zhiyan Liu,
| |
Collapse
|
29
|
NUTM1-Rearranged Neoplasms-A Heterogeneous Group of Primitive Tumors with Expanding Spectrum of Histology and Molecular Alterations-An Updated Review. Curr Oncol 2021; 28:4485-4503. [PMID: 34898574 PMCID: PMC8628659 DOI: 10.3390/curroncol28060381] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022] Open
Abstract
Nuclear protein of testis (NUT), a protein product of the NUTM1 gene (located on the long arm of chromosome 15) with highly restricted physiologic expression in post-meiotic spermatids, is the oncogenic driver of a group of emerging neoplasms when fused with genes involved in transcription regulation. Although initially identified in a group of lethal midline carcinomas in which NUT forms fusion proteins with bromodomain proteins, NUTM1-rearrangement has since been identified in tumors at non-midline locations, with non-bromodomain partners and with varied morphology. The histologic features of these tumors have also expanded to include sarcoma, skin adnexal tumors, and hematologic malignancies that harbor various fusion partners and are associated with markedly different clinical courses varying from benign to malignant. Most of these tumors have nondescript primitive morphology and therefore should be routinely considered in any undifferentiated neoplasm. The diagnosis is facilitated by the immunohistochemical use of the monoclonal C52 antibody, fluorescence in situ hybridization (FISH), and, recently, RNA-sequencing. The pathogenesis is believed to be altered expression of oncogenes or tumor suppressor genes by NUT-mediated genome-wide histone modification. NUTM1-rearranged neoplasms respond poorly to classical chemotherapy and radiation therapy. Targeted therapies such as bromodomain and extraterminal domain inhibitor (BETi) therapy are being developed. This current review provides an update on NUTM1-rearranged neoplasms, focusing on the correlation between basic sciences and clinical aspects.
Collapse
|
30
|
Marx A, Chan JKC, Chalabreysse L, Dacic S, Detterbeck F, French CA, Hornick JL, Inagaki H, Jain D, Lazar AJ, Marino M, Marom EM, Moreira AL, Nicholson AG, Noguchi M, Nonaka D, Papotti MG, Porubsky S, Sholl LM, Tateyama H, Thomas de Montpréville V, Travis WD, Rajan A, Roden AC, Ströbel P. The 2021 WHO Classification of Tumors of the Thymus and Mediastinum: What Is New in Thymic Epithelial, Germ Cell, and Mesenchymal Tumors? J Thorac Oncol 2021; 17:200-213. [PMID: 34695605 DOI: 10.1016/j.jtho.2021.10.010] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 01/12/2023]
Abstract
This overview of the fifth edition of the WHO classification of thymic epithelial tumors (including thymomas, thymic carcinomas, and thymic neuroendocrine tumors [NETs]), mediastinal germ cell tumors, and mesenchymal neoplasms aims to (1) list established and new tumor entities and subtypes and (2) focus on diagnostic, molecular, and conceptual advances since publication of the fourth edition in 2015. Diagnostic advances are best exemplified by the immunohistochemical characterization of adenocarcinomas and the recognition of genetic translocations in metaplastic thymomas, rare B2 and B3 thymomas, and hyalinizing clear cell carcinomas. Advancements at the molecular and tumor biological levels of utmost oncological relevance are the findings that thymomas and most thymic carcinomas lack currently targetable mutations, have an extraordinarily low tumor mutational burden, but typically have a programmed death-ligand 1high phenotype. Finally, data underpinning a conceptual advance are illustrated for the future classification of thymic NETs that may fit into the classification scheme of extrathoracic NETs. Endowed with updated clinical information and state-of-the-art positron emission tomography and computed tomography images, the fifth edition of the WHO classification of thymic epithelial tumors, germ cell tumors, and mesenchymal neoplasms with its wealth of new diagnostic and molecular insights will be a valuable source for pathologists, radiologists, surgeons, and oncologists alike. Therapeutic perspectives and research challenges will be addressed as well.
Collapse
Affiliation(s)
- Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany.
| | - John K C Chan
- Department of Pathology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, People's Republic of China
| | - Lara Chalabreysse
- Department of Pathology, Groupement Hospitalier Est, Bron Cedex Lyon, France
| | - Sanja Dacic
- Department of Pathology, University Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Frank Detterbeck
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Christopher A French
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hiroshi Inagaki
- Department of Pathology and Molecular Diagnostics, Nagoya City University, Nagoya, Japan
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Alexander J Lazar
- Department of Pathology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mirella Marino
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico Regina Elena National Cancer Institute, Rome, Italy
| | - Edith M Marom
- Department of Diagnostic Imaging, Chaim Sheba Medical Center, affiliated with Tel Aviv University, Ramat Gan, Israel
| | - Andre L Moreira
- Department of Pathology, New York University Langone Health, New York, New York
| | - Andrew G Nicholson
- Department of Histopathology, Royal Brompton and Harefield Hospitals and National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Masayuki Noguchi
- Department of Diagnostic Pathology, University of Tsukuba, Tsukuba-shi, Japan
| | - Daisuke Nonaka
- Department of Cellular Pathology, Guy's and St Thomas' NHS Foundation Trust, St Thomas' Hospital, London, United Kingdom
| | | | - Stefan Porubsky
- Department of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hisashi Tateyama
- Department of Pathology, Kasugai Municipal Hospital, Kasugai, Japan
| | | | - William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Arun Rajan
- Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
31
|
Li X, Shi H, Zhang W, Bai C, He M, Ta N, Huang H, Ning Y, Fang C, Qin H, Dong Y. Immunotherapy and Targeting the Tumor Microenvironment: Current Place and New Insights in Primary Pulmonary NUT Carcinoma. Front Oncol 2021; 11:690115. [PMID: 34660264 PMCID: PMC8515126 DOI: 10.3389/fonc.2021.690115] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Primary pulmonary nuclear protein of testis carcinoma is a rare and highly aggressive malignant tumor. It accounts for approximately 0.22% of primary thoracic tumors and is little known, so it is often misdiagnosed as pulmonary squamous cell carcinoma. No effective treatment has been formed yet, and the prognosis is extremely poor. This review aims to summarize the etiology, pathogenesis, diagnosis, treatment, and prognosis of primary pulmonary nuclear protein of testis carcinoma in order to better recognize it and discuss the current and innovative strategies to overcome it. With the increasing importance of cancer immunotherapy and tumor microenvironment, the review also discusses whether immunotherapy and targeting the tumor microenvironment can improve the prognosis of primary pulmonary nuclear protein of testis carcinoma and possible treatment strategies. We reviewed and summarized the clinicopathological features of all patients with primary pulmonary nuclear protein of testis carcinoma who received immunotherapy, including initial misdiagnosis, disease stage, immunohistochemical markers related to tumor neovascularization, and biomarkers related to immunotherapy, such as PD-L1 (programmed death-ligand 1) and TMB (tumor mutational burden). In the meanwhile, we summarized and analyzed the progression-free survival (PFS) and the overall survival (OS) of patients with primary pulmonary nuclear protein of testis carcinoma treated with PD-1 (programmed cell death protein 1)/PD-L1 inhibitors and explored potential population that may benefit from immunotherapy. To the best of our knowledge, this is the first review on the exploration of the tumor microenvironment and immunotherapy effectiveness in primary pulmonary nuclear protein of testis carcinoma.
Collapse
Affiliation(s)
- Xiang Li
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hui Shi
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chong Bai
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Miaoxia He
- Department of Pathology, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Na Ta
- Department of Pathology, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Haidong Huang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yunye Ning
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chen Fang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hao Qin
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yuchao Dong
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
32
|
BET Proteins as Attractive Targets for Cancer Therapeutics. Int J Mol Sci 2021; 22:ijms222011102. [PMID: 34681760 PMCID: PMC8538173 DOI: 10.3390/ijms222011102] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Transcriptional dysregulation is a hallmark of cancer and can be an essential driver of cancer initiation and progression. Loss of transcriptional control can cause cancer cells to become dependent on certain regulators of gene expression. Bromodomain and extraterminal domain (BET) proteins are epigenetic readers that regulate the expression of multiple genes involved in carcinogenesis. BET inhibitors (BETis) disrupt BET protein binding to acetylated lysine residues of chromatin and suppress the transcription of various genes, including oncogenic transcription factors. Phase I and II clinical trials demonstrated BETis’ potential as anticancer drugs against solid tumours and haematological malignancies; however, their clinical success was limited as monotherapies. Emerging treatment-associated toxicities, drug resistance and a lack of predictive biomarkers limited BETis’ clinical progress. The preclinical evaluation demonstrated that BETis synergised with different classes of compounds, including DNA repair inhibitors, thus supporting further clinical development of BETis. The combination of BET and PARP inhibitors triggered synthetic lethality in cells with proficient homologous recombination. Mechanistic studies revealed that BETis targeted multiple essential homologous recombination pathway proteins, including RAD51, BRCA1 and CtIP. The exact mechanism of BETis’ anticancer action remains poorly understood; nevertheless, these agents provide a novel approach to epigenome and transcriptome anticancer therapy.
Collapse
|
33
|
Wang X, Wang J, Luo X, Lu J, Wang L, Li Q, Wang EH. Diagnosis of NUT Carcinoma Despite False-Negative Next-Generation Sequencing Results: A Case Report and Literature Review. Onco Targets Ther 2021; 14:4621-4633. [PMID: 34475768 PMCID: PMC8407784 DOI: 10.2147/ott.s327722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/18/2021] [Indexed: 12/23/2022] Open
Abstract
Nuclear protein in testis (NUT) carcinoma (NC) is a poorly differentiated malignant tumor with a poor prognosis, which is caused by the NUTM1 gene rearrangement. Positive staining of NUT using immunohistochemistry (IHC) or gene rearrangement of NUTM1 revealed by genetic analysis, such as fluorescence in situ hybridization (FISH) or next-generation sequencing (NGS), are important strategies used for accurate diagnosis. In the current study, we present a case of NC in an 18-year-old man who had a chief complaint of nasal congestion, nasal bleeding, and anosmia. Magnetic resonance imaging revealed a mass in the nasal cavity and nasal septum. The initial pathological diagnosis was basaloid squamous cell carcinoma. Based on the tumor location and abrupt keratinization, further genetic tests were performed, and NC was diagnosed using FISH, which was further verified by IHC. However, neither DNA-based NGS nor RNA-based NGS revealed the NUTM1 gene rearrangement. Using this case as a basis, we have reviewed the related literature, compared the common diagnostic methods of NC, and discussed the advantages and limitations of current tools employed for molecular analysis of the gene fusion.
Collapse
Affiliation(s)
- Xi Wang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, 110001, People's Republic of China
| | - Jinping Wang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, 110001, People's Republic of China
| | - Xue Luo
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, 110001, People's Republic of China
| | - Jinxi Lu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, 110001, People's Republic of China
| | - Liang Wang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, 110001, People's Republic of China
| | - Qingchang Li
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, 110001, People's Republic of China
| | - En-Hua Wang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, 110001, People's Republic of China
| |
Collapse
|
34
|
Leeman R, Pinkney K, Bradley J, Ruiz R, DuBois SG, French C, Trucco M. NUT Carcinoma Without Upfront Surgical Resection: A Case Report. J Pediatr Hematol Oncol 2021; 43:e707-e710. [PMID: 32555033 PMCID: PMC8887700 DOI: 10.1097/mph.0000000000001865] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/19/2020] [Indexed: 01/26/2023]
Abstract
Nuclear protein in testis carcinoma is a rare and highly aggressive carcinoma associated with a 70% mortality rate 1 year from diagnosis and a median survival of only 6.5 months. No established treatment protocol exists, although some success has been achieved using a multimodal approach including early surgical resection and adjuvant chemotherapy and radiation. Prior studies have not demonstrated successful treatment in the absence of upfront surgical resection. We describe the first reported case of a patient with unresectable nuclear protein in testis carcinoma treated successfully with definitive chemotherapy using the Scandinavian Sarcoma Group IX Protocol and concurrent radiation therapy, but without surgical resection.
Collapse
Affiliation(s)
- Rachel Leeman
- Department of Pediatrics, University of Miami Miller School of Medicine/Jackson Health System, Miami, FL, USA
| | - Kerice Pinkney
- Division of Pediatric Hematology/Oncology, Joe DiMaggio Children’s Hospital, Hollywood, FL, USA
| | - Julie Bradley
- Department of Radiation Oncology, University of Florida College of Medicine, Jacksonville, FL USA
| | - Robert Ruiz
- Department of Pathology, Memorial Health System, Hollywood, FL, USA
| | - Steven G. DuBois
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Christopher French
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA, Dana-Farber Cancer Institute
| | - Matteo Trucco
- Department of Pediatric Hematology Oncology and BMT, Cleveland Clinic Children’s, Cleveland, Ohio, USA
| |
Collapse
|
35
|
Lantuejoul S, Pissaloux D, Ferretti GR, McLeer A. NUT carcinoma of the lung. Semin Diagn Pathol 2021; 38:72-82. [PMID: 34176698 DOI: 10.1053/j.semdp.2021.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 11/11/2022]
Abstract
NUT carcinoma of the thorax is a rare and very aggressive tumor, whose definition is based on the demonstration of a nuclear protein in testis (NUTM1; also known as NUT) gene fusion on 15q14 with different partners from the bromodomain-containing proteins gene family. This fusion results in an activation of MYC oncoprotein responsible for the tumor's aggressivity. NUT carcinoma arises preferentially in young adults, presenting a large thoracic mass frequently associated with lymph nodes, bone or pleural metastases. At histology, this tumor is often poorly differentiated, mainly composed of sheets of small cells with scant cytoplasm, a round nucleus with a central nucleolus. Focal areas of squamous differentiation can be observed. Mitoses and necrosis are frequent, as well as neutrophilic infiltrate. The diagnosis is based on the detection of NUT protein expression by immunohistochemistry using the rabbit monoclonal antibody C52B1 in more than 50% of the tumor nuclei. This technique offers 87% sensitivity and nearly 100% specificity with reference to FISH or RT-PCR, which confirm the NUTM1 rearrangement. The differential diagnoses include basaloid carcinoma of the lung, small cell carcinoma, thymic carcinoma (basaloid variant), SMARCA4_deficient thoracic sarcoma, other NUTM1 rearranged undifferentiated tumors, small round cell tumors, non-Hodgkin lymphoma/leukemia, and melanoma. The prognosis of NUT carcinoma remains very poor, with a median survival of 6.7 months, and 1- and 2-year overall survival rates of 30% and 19%, respectively. NUT carcinoma is often refractory to conventional chemotherapy, but ifosfamide-based regimens or BET inhibitors could represent promising therapies.
Collapse
Affiliation(s)
- Sylvie Lantuejoul
- Centre Léon Bérard, Department of biopathology, 28 rue Laënnec, 69373 Lyon, France; University of Grenoble Alpes, F-38000, Grenoble, France; Cancer Research Center of Lyon, Equipe Labellisée Ligue contre le Cancer, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France.
| | - Daniel Pissaloux
- Centre Léon Bérard, Department of biopathology, 28 rue Laënnec, 69373 Lyon, France; Cancer Research Center of Lyon, Equipe Labellisée Ligue contre le Cancer, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Gilbert R Ferretti
- University of Grenoble Alpes, F-38000, Grenoble, France; CHU Grenoble-Alpes, Department of radiology, 38043 Grenoble, France; Institute for Advanced Biosciences, CNRS UMR5309, Inserm U1209, Grenoble Alpes, University F-38000, Grenoble, France
| | - Anne McLeer
- University of Grenoble Alpes, F-38000, Grenoble, France; CHU Grenoble-Alpes, Department of pathology, 38043 Grenoble, France; Institute for Advanced Biosciences, CNRS UMR5309, Inserm U1209, Grenoble Alpes, University F-38000, Grenoble, France
| |
Collapse
|
36
|
What Is New in Biomarker Testing at Diagnosis of Advanced Non-Squamous Non-Small Cell Lung Carcinoma? Implications for Cytology and Liquid Biopsy. JOURNAL OF MOLECULAR PATHOLOGY 2021. [DOI: 10.3390/jmp2020015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The discovery and clinical validation of biomarkers predictive of the response of non-squamous non-small-cell lung carcinomas (NS-NSCLC) to therapeutic strategies continue to provide new data. The evaluation of novel treatments is based on molecular analyses aimed at determining their efficacy. These tests are increasing in number, but the tissue specimens are smaller and smaller and/or can have few tumor cells. Indeed, in addition to tissue samples, complementary cytological and/or blood samples can also give access to these biomarkers. To date, it is recommended and necessary to look for the status of five genomic molecular biomarkers (EGFR, ALK, ROS1, BRAFV600, NTRK) and of a protein biomarker (PD-L1). However, the short- and more or less long-term emergence of new targeted treatments of genomic alterations on RET and MET, but also on others’ genomic alteration, notably on KRAS, HER2, NRG1, SMARCA4, and NUT, have made cellular and blood samples essential for molecular testing. The aim of this review is to present the interest in using cytological and/or liquid biopsies as complementary biological material, or as an alternative to tissue specimens, for detection at diagnosis of new predictive biomarkers of NS-NSCLC.
Collapse
|
37
|
Shorstova T, Foulkes WD, Witcher M. Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer 2021; 124:1478-1490. [PMID: 33723398 PMCID: PMC8076232 DOI: 10.1038/s41416-021-01321-0] [Citation(s) in RCA: 220] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/12/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
The transcriptional upregulation of oncogenes is a driving force behind the progression of many tumours. However, until a decade ago, the concept of 'switching off' these oncogenic pathways represented a formidable challenge. Research has revealed that members of the bromo- and extra-terminal domain (BET) motif family are key activators of oncogenic networks in a spectrum of cancers; their function depends on their recruitment to chromatin through two bromodomains (BD1 and BD2). The advent of potent inhibitors of BET proteins (BETi), which target either one or both bromodomains, represents an important step towards the goal of suppressing oncogenic networks within tumours. Here, we discuss the biology of BET proteins, advances in BETi design and highlight potential biomarkers predicting their activity. We also outline the logic of incorporating BETi into combination therapies to enhance its efficacy. We suggest that understanding mechanisms of activity, defining predictive biomarkers and identifying potent synergies represents a roadmap for clinical success using BETi.
Collapse
Affiliation(s)
- Tatiana Shorstova
- grid.414980.00000 0000 9401 2774Departments of Oncology and Experimental Medicine, McGill University, Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, Montreal, QC Canada
| | - William D. Foulkes
- grid.414980.00000 0000 9401 2774Departments of Oncology and Human Genetics, McGill University, Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, Montreal, QC Canada
| | - Michael Witcher
- grid.414980.00000 0000 9401 2774Departments of Oncology and Experimental Medicine, McGill University, Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, Montreal, QC Canada
| |
Collapse
|
38
|
Chen M, Yang J, Lv L, Li Y, Tang Y, Liu W, Wang W, Jiang L. Comprehensive genetic profiling of six pulmonary nuclear protein in testis carcinomas with a novel micropapillary histological subtype in two cases. Hum Pathol 2021; 115:56-66. [PMID: 33713695 DOI: 10.1016/j.humpath.2021.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/11/2021] [Indexed: 02/05/2023]
Abstract
Nuclear protein in testis (NUT) carcinoma (NC) is a rare and aggressive neoplasm associated with a rearrangement of the NUT gene on chromosome 15q14. To date, genomic alterations of NCs, especially those in the lung, are poorly understood. In this study, immunohistochemistry staining, fluorescence in situ hybridization, and two next-generation sequencing (NGS) panels of 56 and 701 genes were used to explore the clinical, pathological, and genetic profiling of pulmonary NCs. Six pulmonary NC cases were confirmed, with a mean age of 41 years (range: 22-69 years) and a median survival time of 6.5 months (range: 2-19 months). Morphologically, typical abrupt keratinization was observed in four of six cases (67%), and two patients presented a mixed pattern of classical squamous component and micropapillary adenocarcinoma morphology. We also identified a case with NUT gene amplification instead of rearrangement. Furthermore, NGS analysis demonstrated the following fusions: BRD4-NUTM1 (2/4 cases) and NSD3-NUTM1 (2/4 cases), and the analysis highlighted 53 gene mutations, including 50 (94.3%, 50/53) single-nucleotide variations (SNVs) and three (5.7%, 3/53) long insertions/deletions. SNVs of MUC16 were the most common and occurred in three cases (75%). Moreover, SNVs of EPHA8, FANCA, TRIO, and USP6 were detected in two of four cases (50%). These 53 mutated genes were involved in 13 functional pathways based on enrichment analysis, especially in the PI3K-Akt signaling pathway. Finally, none of the cases showed obvious copy number variations and had low tumor mutational burden and stable microsatellite sites.
Collapse
Affiliation(s)
- Min Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jieliang Yang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Lixia Lv
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Yuli Li
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Yuan Tang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Weiya Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
39
|
Gu B, Hakun MC. Challenges and Opportunities in NUT Carcinoma Research. Genes (Basel) 2021; 12:genes12020235. [PMID: 33562801 PMCID: PMC7915910 DOI: 10.3390/genes12020235] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
NUT carcinoma (NC) is a type of aggressive cancer driven by chromosome translocations. Fusion genes between a DNA-binding protein, such as bromodomain and extraterminal domain (BET) proteins, and the testis-specific protein NUTM1 generated by these translocations drive the formation of NC. NC can develop in very young children without significant accumulation of somatic mutations, presenting a relatively clean model to study the genetic etiology of oncogenesis. However, after 20 years of research, a few challenging questions still remain for understanding the mechanism and developing therapeutics for NC. In this short review, we first briefly summarize the current knowledge regarding the molecular mechanism and targeted therapy development of NC. We then raise three challenging questions: (1) What is the cell of origin of NC? (2) How does the germline analogous epigenetic reprogramming process driven by the BET-NUTM1 fusion proteins cause NC? and (3) How will BET-NUTM1 targeted therapies be developed? We propose that with the unprecedented technological advancements in genome editing, animal models, stem cell biology, organoids, and chemical biology, we have unique opportunities to address these challenges.
Collapse
Affiliation(s)
- Bin Gu
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University East Lansing, MI 48824, USA
- Department of Biomedical Engineering; Michigan State University East Lansing, MI 48824, USA;
- Correspondence:
| | - Maxwell C. Hakun
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University East Lansing, MI 48824, USA
- Department of Biomedical Engineering; Michigan State University East Lansing, MI 48824, USA;
| |
Collapse
|
40
|
Supercharging BRD4 with NUT in carcinoma. Oncogene 2021; 40:1396-1408. [PMID: 33452461 PMCID: PMC7914217 DOI: 10.1038/s41388-020-01625-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023]
Abstract
NUT carcinoma (NC) is an extremely aggressive squamous cancer with no effective therapy. NC is driven, most commonly, by the BRD4-NUT fusion oncoprotein. BRD4-NUT combines the chromatin-binding bromo- and extraterminal domain-containing (BET) protein, BRD4, with an unstructured, poorly understood protein, NUT, which recruits and activates the histone acetyltransferase p300. Recruitment of p300 to chromatin by BRD4 is believed to lead to the formation of hyperacetylated nuclear foci, as seen by immunofluorescence. BRD4-NUT nuclear foci correspond with massive contiguous regions of chromatin co-enriched with BRD4-NUT, p300, and acetylated histones, termed "megadomains" (MD). Megadomains stretch for as long as 2 MB. Proteomics has defined a BRD4-NUT chromatin complex in which members that associate with BRD4 also exist as rare NUT-fusion partners. This suggests that the common pathogenic denominator is the presence of both BRD4 and NUT, and that the function of BRD4-NUT may mimic that of wild-type BRD4. If so, then MDs may function as massive super-enhancers, activating transcription in a BET-dependent manner. Common targets of MDs across multiple NCs and tissues are three stem cell-related transcription factors frequently implicated in cancer: MYC, SOX2, and TP63. Recently, MDs were found to form a novel nuclear sub-compartment, called subcompartment M (subM), where MD-MD interactions occur both intra- and inter-chromosomally. Included in subM are MYC, SOX2, and TP63. Here we explore the possibility that if MDs are simply large super-enhancers, subM may exist in other cell systems, with broad implications for how 3D organization of the genome may function in gene regulation and maintenance of cell identity. Finally, we discuss how our knowledge of BRD4-NUT function has been leveraged for the therapeutic development of first-in-class BET inhibitors and other targeted strategies.
Collapse
|
41
|
Chatzopoulos K, Boland JM. Update on genetically defined lung neoplasms: NUT carcinoma and thoracic SMARCA4-deficient undifferentiated tumors. Virchows Arch 2021; 478:21-30. [PMID: 33409598 DOI: 10.1007/s00428-020-03011-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/07/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022]
Abstract
NUT carcinoma, also known as NUT midline carcinoma, is an aggressive malignancy mainly affecting the midline structures of younger patients and almost invariably leading to death within a few months of the diagnosis. Morphologically, NUT carcinoma consists of sheets of monomorphous small or medium size cells with scant cytoplasm, commonly featuring areas of abrupt squamous differentiation with keratinization. Immunohistochemistry for NUT protein is sensitive and specific, typically showing a speckled nuclear reactivity, assisting in diagnosis. The molecular background of NUT carcinoma includes the reciprocal translocation t(15;19) leading to expression of the BRD4-NUT fusion transcript with oncogenic properties. Other less common genes may occasionally be fused with NUT not only in NUT carcinoma but also in other soft tissue tumors, highlighting the fact that NUT-rearranged tumors may represent a larger and more diverse family of neoplasms. Thoracic SMARCA4-deficient undifferentiated tumors are aggressive malignancies diagnosed more often in young male smokers, which often lead to death within a few months. SMARCA4-deficient tumors show undifferentiated morphology with occasional hepatoid and rhabdoid features. Immunohistochemically, the hallmark of diagnosis is loss of expression of SMARCA4 (BRG1). Concurrent loss of SMARCA2 expression, as well as expression of one or more stem cell markers SOX2, CD34, or SALL4 is common. Truncating mutations in SMARCA4, a catalytic subunit of the mammalian BAF (SWI/SNF) complex, are the dominant oncogenic molecular event underlying the pathogenesis of these tumors. SMARCA4 deficiency can also be seen as a passenger somatic event in multiple solid neoplasms manifesting as focal dedifferentiation and rhabdoid morphology.
Collapse
Affiliation(s)
- Kyriakos Chatzopoulos
- Division of Anatomic Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jennifer M Boland
- Division of Anatomic Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
42
|
Wang S, Li J, Tong W, Li H, Feng Q, Teng B. Advances in the pathogenesis and treatment of nut carcinoma: a narrative review. Transl Cancer Res 2020; 9:6505-6515. [PMID: 35117258 PMCID: PMC8798738 DOI: 10.21037/tcr-20-1884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 09/12/2020] [Indexed: 11/06/2022]
Abstract
NUT carcinoma (NC) is a rare, highly invasive and fatal tumor and often misdiagnosed. It typically arises from the mediastinum and midline organs and has complicated pathogenesis and poor outcome. Genetically, its pathogenesis is related to a chromosomal rearrangement involving the NUTM1 gene. In most cases, the main oncoprotein is BRD4-NUT with a translocation between NUTM1 and BRD4 genes, but in a few cases, the oncoprotein is BRD3-NUT, or NSD3-NUT. Studies have shown that the histone hyperacetylation and BRD4 hyperphosphorylation may lead to the activation of cancer circuits. Abnormal production of microRNA, inactivation of tumor suppressor genes and abnormal activation of several signaling pathways are proposed as potential mechanisms underlying the pathogenesis of NC. Currently, there is no consensus on its standard treatment for NC. Extent of surgical resection with negative margins, initial radiotherapy and part of chemotherapy regimens may significantly associated with the improvement of progression-free survival (PFS) rate and overall survival (OS) rate. Some bromodomain and extraterminal inhibitors (BETis) have shown encouraging results in the clinical trials on NC, but delayed drug resistance is still an important issue that needs to be resolved. Histone deacetylase inhibitors are also found to possess the potential in the treatment of NC. Herein, we summarize recent advances in the pathogenesis and treatment of NC.
Collapse
Affiliation(s)
- Sanchun Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jinqiu Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Weifang Tong
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Hejie Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Qingjie Feng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Bo Teng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
McLean-Holden AC, Moore SA, Gagan J, French CA, Sher D, Truelson JM, Bishop JA. NUT Carcinoma in a Patient with Unusually Long Survival and False Negative FISH Results. Head Neck Pathol 2020; 15:698-703. [PMID: 32918711 PMCID: PMC8134642 DOI: 10.1007/s12105-020-01220-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022]
Abstract
Nuclear protein in testis (NUT) carcinoma is a rare and highly aggressive epithelial malignancy defined by rearrangement of the NUTM1 gene on chromosome 15q14. Histologically, NUT carcinoma is an undifferentiated carcinoma formed by sheets and nests of primitive and monotonous "round blue cells" with foci of abrupt keratinization in a subset. NUT carcinoma runs a fulminant clinical course and is almost always quickly lethal, with a median overall survival of only 6.7 months. There is no consensus regarding treatment for this disease, and most patients respond poorly to conventional chemotherapy and radiation. We report a case of NUT carcinoma in an African-American man who initially presented in 2009 with a tracheal mass at age 28. Although fluorescence in situ hybridization (FISH) assays for NUTM1 and BRD4 rearrangements were negative, he was diagnosed based on diffusely positive NUT immunostaining and BRD4-NUTM1 on RNA sequencing. Since his initial presentation, he has undergone multiple surgical procedures and radiation therapy. His tumor has recurred twice, but he has survived for 129 months and is currently alive without disease. Long-term survival of patients with NUT carcinoma is incredibly unusual, especially in patients with tumors that exhibit a BRD4 rearrangement. False negative FISH is a pitfall in diagnosing NUT carcinoma; NUT immunostaining and RNA sequencing are more sensitive diagnostic methods.
Collapse
Affiliation(s)
- Anne C McLean-Holden
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Samantha A Moore
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey Gagan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher A French
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Sher
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John M Truelson
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- UT Southwestern Medical Center, Clements University Hospital, UH04.250 6201 Harry Hines Blvd, Dallas, TX, 75390, USA.
| |
Collapse
|
44
|
Morrison-Smith CD, Knox TM, Filic I, Soroko KM, Eschle BK, Wilkens MK, Gokhale PC, Giles F, Griffin A, Brown B, Shapiro GI, Zucconi BE, Cole PA, Lemieux ME, French CA. Combined Targeting of the BRD4-NUT-p300 Axis in NUT Midline Carcinoma by Dual Selective Bromodomain Inhibitor, NEO2734. Mol Cancer Ther 2020; 19:1406-1414. [PMID: 32371576 DOI: 10.1158/1535-7163.mct-20-0087] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/12/2020] [Accepted: 04/13/2020] [Indexed: 11/16/2022]
Abstract
NUT midline carcinoma (NMC) is a rare, aggressive subtype of squamous carcinoma that is driven by the BRD4-NUT fusion oncoprotein. BRD4, a BET protein, binds to chromatin through its two bromodomains, and NUT recruits the p300 histone acetyltransferse (HAT) to activate transcription of oncogenic target genes. BET-selective bromodomain inhibitors have demonstrated on-target activity in patients with NMC, but with limited efficacy. P300, like BRD4, contains a bromodomain. We show that combining selective p300/CBP and BET bromodomain inhibitors, GNE-781 and OTX015, respectively, induces cooperative depletion of MYC and synergistic inhibition of NMC growth. Treatment of NMC cells with the novel dual p300/CBP and BET bromodomain-selective inhibitor, NEO2734, potently inhibits growth and induces differentiation of NMC cells in vitro; findings that correspond with potentiated transcriptional effects from combined BET and p300 bromodomain inhibition. In three disseminated NMC xenograft models, NEO2734 provided greater growth inhibition, with tumor regression and significant survival benefit seen in two of three models, compared with a lead clinical BET inhibitor or "standard" chemotherapy. Our findings provide a strong rationale for clinical study of NEO2734 in patients with NMC. Moreover, the synergistic inhibition of NMC growth by CBP/p300 and BET bromodomain inhibition lays the groundwork for greater mechanistic understanding of the interplay between p300 and BRD4-NUT that drives this cancer.
Collapse
Affiliation(s)
- Chevaun D Morrison-Smith
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tatiana M Knox
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ivona Filic
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kara M Soroko
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Benjamin K Eschle
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Margaret K Wilkens
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Prafulla C Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Francis Giles
- Developmental Therapeutics Consortium, Chicago, Ilinois
| | | | - Bill Brown
- Paraza Pharma Inc., Montreal, Quebec, Canada
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Beth E Zucconi
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Philip A Cole
- Department of Medicine, Division of Genetics, Brigham and Women's Hospital and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | | | - Christopher A French
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
45
|
Chau NG, Ma C, Danga K, Al-Sayegh H, Nardi V, Barrette R, Lathan CS, DuBois SG, Haddad RI, Shapiro GI, Sallan SE, Dhar A, Nelson JJ, French CA. An Anatomical Site and Genetic-Based Prognostic Model for Patients With Nuclear Protein in Testis (NUT) Midline Carcinoma: Analysis of 124 Patients. JNCI Cancer Spectr 2020; 4:pkz094. [PMID: 32328562 PMCID: PMC7165803 DOI: 10.1093/jncics/pkz094] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/02/2019] [Accepted: 10/31/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND NUT midline carcinoma, renamed NUT carcinoma (NC), is an aggressive squamous cancer defined by rearrangement of the NUTM1 gene. Although a subset of patients can be cured, for the majority of patients the prognosis is grim. We sought to classify patients into risk groups based on molecular and clinicopathologic factors at the time of diagnosis. METHODS Clinicopathologic variables and survival outcomes were extracted for a total of 141 NC patients from the NUT midline carcinoma Registry using questionnaires and medical records. Translocation type was identified by molecular analyses. Survival tree regression analysis was performed to determine risk factors associated with overall survival (OS). RESULTS For 141 patients, the median age at diagnosis was 23.6 years. Fifty-one percent had thoracic origin compared with 49% nonthoracic sites (41% head and neck, 6% bone or soft tissue, 1% other). The median OS was 6.5 months (95% confidence interval [CI] = 5.8 to 9.1 months). Most patients had the BRD4-NUTM1 fusion (78%), followed by BRD3-NUTM1 (15%) and NSD3-NUTM1 (6%). Survival tree regression identified three statistically distinct risk groups among 124 patients classified by anatomical site and genetics: group A is nonthoracic primary, BRD3-, or NSD3-NUT (n = 12, median OS = 36.5 months, 95% CI = 12.5 to not reported months); group B is nonthoracic primary, BRD4-NUT (n = 45, median OS = 10 months, 95% CI = 7 to 14.6 months); and group C is thoracic primary (n = 67, median OS = 4.4 months, 95% CI = 3.5 to 5.6 months). Only groups A and B had long-term (≥3 years, n = 12) survivors. CONCLUSIONS We identify three risk groups defined by anatomic site and NUT fusion type. Nonthoracic primary with non-BRD4-NUT fusion confers the best prognosis, followed by nonthoracic primary with BRD4-NUT. Thoracic NC patients, regardless of the NUT fusion, have the worst survival.
Collapse
Affiliation(s)
- Nicole G Chau
- Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Clement Ma
- Harvard Medical School, Boston, MA
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | | | - Hasan Al-Sayegh
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Valentina Nardi
- Harvard Medical School, Boston, MA
- Massachusetts General Hospital, Boston, MA
| | | | | | - Steven G DuBois
- Harvard Medical School, Boston, MA
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Robert I Haddad
- Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Geoffrey I Shapiro
- Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Stephen E Sallan
- Harvard Medical School, Boston, MA
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | | | | | - Christopher A French
- Harvard Medical School, Boston, MA
- Brigham and Women’s Hospital, Boston, MA
- GlaxoSmithKline, Collegeville, PA
| |
Collapse
|
46
|
McEvoy CR, Fox SB, Prall OWJ. Emerging entities in NUTM1-rearranged neoplasms. Genes Chromosomes Cancer 2020; 59:375-385. [PMID: 32060986 DOI: 10.1002/gcc.22838] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 12/11/2022] Open
Abstract
Structural alterations of NUTM1 were originally thought to be restricted to poorly differentiated carcinomas with variable squamous differentiation originating in the midline organs of children and adolescents. Termed NUT carcinomas (NCs), they were defined by a t(15;19) chromosomal rearrangement that was found to result in a BRD4-NUTM1 gene fusion. However, the use of DNA and RNA-based next-generation sequencing has recently revealed a multitude of new NUTM1 fusion partners in a diverse array of neoplasms including sarcoma-like tumors, poromas, and acute lymphoblastic leukemias (ALLs) that we propose to call NUTM1-rearranged neoplasms (NRNs). Intriguingly, the nosology of NRNs often correlates with the functional classification of the fusion partner, suggesting different oncogenic mechanisms within each NRN division. Indeed, whereas NCs are characterized by their aggressiveness and intransigence to standard therapeutic measures, the more positive clinical outcomes seen in some sarcoma and ALL NRNs may reflect these mechanistic differences. Here we provide a broad overview of the molecular, nosological, and clinical features in these newly discovered neoplastic entities. We describe how aberrant expression of NUTM1 due to fusion with an N-terminal DNA/chromatin-binding protein can generate a potentially powerful chromatin modifier that can give rise to oncogenic transformation in numerous cellular contexts. We also conclude that classification, clinical behavior, and therapeutic options may be best defined by the NUTM1 fusion partner rather than by tumor morphology or immunohistochemical profile.
Collapse
Affiliation(s)
- Christopher R McEvoy
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Owen W J Prall
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
47
|
Zhang H, Liu M, Zhang J, Luo S, Wang C, Zhao Z, Ge Y, Zhang J, Geng F. Successful treatment of a case with NUT midline carcinoma in the larynx and review of the literature. Clin Case Rep 2020; 8:176-181. [PMID: 31998511 PMCID: PMC6982469 DOI: 10.1002/ccr3.2568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/28/2019] [Accepted: 11/08/2019] [Indexed: 11/20/2022] Open
Abstract
In this report, we gave the first case of successful treatment for laryngeal NMC, which is exceedingly rare with dismal prognosis. intensity-modulated radiation therapy accompanied by traditional Chinese medicine was administrated for the young woman, instead of radical resection, and she got continuous remission for more than 2 years, with no recurrence detected.
Collapse
Affiliation(s)
- Hai Zhang
- Department of OncologyThe Hospital of 81st Group Army PLAZhang Jia KouChina
| | - Ming‐Hua Liu
- Department of PharmacyThe Hospital of 81st Group Army PLAZhang Jia KouChina
| | - Jie Zhang
- Department of Radiation MedicineFaculty of Preventive MedicineAir Force Medical UniversityXi'anChina
| | - Shu‐Peng Luo
- Department of OncologyThe Hospital of 81st Group Army PLAZhang Jia KouChina
| | - Chao‐Bo Wang
- Department of OncologyThe Hospital of 81st Group Army PLAZhang Jia KouChina
- Outpatient Department of Dongcheng Fourth Cadres'SanatoriumBeijingChina
| | - Zeng‐Hu Zhao
- Department of OncologyThe Hospital of 81st Group Army PLAZhang Jia KouChina
| | - Yan‐Li Ge
- Department of OncologyThe Hospital of 81st Group Army PLAZhang Jia KouChina
| | - Jian‐Yu Zhang
- Department of OncologyThe Hospital of 81st Group Army PLAZhang Jia KouChina
| | - Feng‐Hao Geng
- Department of OncologyThe Hospital of 81st Group Army PLAZhang Jia KouChina
- Department of Radiation MedicineFaculty of Preventive MedicineAir Force Medical UniversityXi'anChina
| |
Collapse
|
48
|
NUT midline carcinoma: Current concepts and future perspectives of a novel tumour entity. Crit Rev Oncol Hematol 2019; 144:102826. [DOI: 10.1016/j.critrevonc.2019.102826] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 01/22/2023] Open
|
49
|
Hermsen MA, Riobello C, García-Marín R, Cabal VN, Suárez-Fernández L, López F, Llorente JL. Translational genomics of sinonasal cancers. Semin Cancer Biol 2019; 61:101-109. [PMID: 31560943 DOI: 10.1016/j.semcancer.2019.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023]
Abstract
The sinonasal cavities harbor a wide variety of histologically distinct cancers, the majority very aggressive with 5-year survival rates between 30-60% and local recurrence as the main cause of death. This is a complex anatomic area, close to structures such the eyes and the brain, which is of special relevance for surgery and postoperative radiotherapy. The low incidence of these rare tumors hampers accumulation of experience with diagnosis and clinical managment as well as knowledge on recurrent genetic aberrations or testing of new treatment strategies. However, recent years have seen a growing number of publications on genetic aberrations providing data that can aid or fine-tune classification and provide molecular targets for treatment with specific inhibitors. In addition, new sinonasal cancer models are created that enable preclinical testing of candidate inhibitor drugs. With more and more novel targeted therapies being developed, options for personalized treatment of sinonasal cancer patients are now opening up.
Collapse
Affiliation(s)
- Mario A Hermsen
- Dept. Head and Neck Oncology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.
| | - Cristina Riobello
- Dept. Head and Neck Oncology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Rocío García-Marín
- Dept. Head and Neck Oncology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Virginia N Cabal
- Dept. Head and Neck Oncology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Laura Suárez-Fernández
- Dept. Head and Neck Oncology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Fernando López
- Dept. Otolaryngology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - José L Llorente
- Dept. Otolaryngology, Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
50
|
Lee JJK, Park S, Park H, Kim S, Lee J, Lee J, Youk J, Yi K, An Y, Park IK, Kang CH, Chung DH, Kim TM, Jeon YK, Hong D, Park PJ, Ju YS, Kim YT. Tracing Oncogene Rearrangements in the Mutational History of Lung Adenocarcinoma. Cell 2019; 177:1842-1857.e21. [PMID: 31155235 DOI: 10.1016/j.cell.2019.05.013] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/18/2019] [Accepted: 05/03/2019] [Indexed: 01/13/2023]
Abstract
Mutational processes giving rise to lung adenocarcinomas (LADCs) in non-smokers remain elusive. We analyzed 138 LADC whole genomes, including 83 cases with minimal contribution of smoking-associated mutational signature. Genomic rearrangements were not correlated with smoking-associated mutations and frequently served as driver events of smoking-signature-low LADCs. Complex genomic rearrangements, including chromothripsis and chromoplexy, generated 74% of known fusion oncogenes, including EML4-ALK, CD74-ROS1, and KIF5B-RET. Unlike other collateral rearrangements, these fusion-oncogene-associated rearrangements were frequently copy-number-balanced, representing a genomic signature of early oncogenesis. Analysis of mutation timing revealed that fusions and point mutations of canonical oncogenes were often acquired in the early decades of life. During a long latency, cancer-related genes were disrupted or amplified by complex rearrangements. The genomic landscape was different between subgroups-EGFR-mutant LADCs had frequent whole-genome duplications with p53 mutations, whereas fusion-oncogene-driven LADCs had frequent SETD2 mutations. Our study highlights LADC oncogenesis driven by endogenous mutational processes.
Collapse
Affiliation(s)
- Jake June-Koo Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States; Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, United States
| | - Seongyeol Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Hansol Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Sehui Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jongkeun Lee
- Clinical Genomics Analysis Branch, National Cancer Center, Goyang 10408, Korea
| | - Junehawk Lee
- Korea Institute of Science and Technology Information, Daejeon 34141, Korea
| | - Jeonghwan Youk
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Kijong Yi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Yohan An
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - In Kyu Park
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul 03080, Korea
| | - Chang Hyun Kang
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul 03080, Korea
| | - Doo Hyun Chung
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Tae Min Kim
- Seoul National University Cancer Research Institute, Seoul 03080, Korea; Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea; Seoul National University Cancer Research Institute, Seoul 03080, Korea
| | - Dongwan Hong
- Clinical Genomics Analysis Branch, National Cancer Center, Goyang 10408, Korea
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States; Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, United States
| | - Young Seok Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
| | - Young Tae Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul 03080, Korea; Seoul National University Cancer Research Institute, Seoul 03080, Korea.
| |
Collapse
|