1
|
Newman JA, Gavard AE, Imprachim N, Aitkenhead H, Sheppard HE, Te Poele R, Clarke PA, Hossain MA, Temme L, Oh HJ, Wells CI, Davis-Gilbert ZW, Workman P, Gileadi O, Drewry DH. Structural insights into human brachyury DNA recognition and discovery of progressible binders for cancer therapy. Nat Commun 2025; 16:1596. [PMID: 39952925 PMCID: PMC11828899 DOI: 10.1038/s41467-025-56213-1] [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: 05/09/2024] [Accepted: 01/10/2025] [Indexed: 02/17/2025] Open
Abstract
Brachyury is a transcription factor that plays an essential role in tumour growth of the rare bone cancer chordoma and is implicated in other solid tumours. Brachyury is minimally expressed in healthy tissues, making it a potential therapeutic target. Unfortunately, as a ligandless transcription factor, brachyury has historically been considered undruggable. To investigate direct targeting of brachyury by small molecules, we determine the structure of human brachyury both alone and in complex with DNA. The structures provide insights into DNA binding and the context of the chordoma associated G177D variant. We use crystallographic fragment screening to identify hotspots on numerous pockets on the brachyury surface. Finally, we perform follow-up chemistry on fragment hits and describe the progression of a thiazole chemical series into binders with low µM potency. Thus we show that brachyury is ligandable and provide an example of how crystallographic fragment screening may be used to target protein classes that are difficult to address using other approaches.
Collapse
Affiliation(s)
- Joseph A Newman
- Centre for Medicines Discovery, University of Oxford, Oxford, UK.
| | - Angeline E Gavard
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Exscientia, Oxford, UK
| | - Nergis Imprachim
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Hazel Aitkenhead
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Diamond Light Source Ltd, Didcot, UK
| | - Hadley E Sheppard
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
- Sano Genetics Ltd, Cambridge, UK
| | - Robert Te Poele
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
| | - Paul A Clarke
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
| | - Mohammad Anwar Hossain
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Louisa Temme
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Hans J Oh
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carrow I Wells
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- GlaxoSmithKline, Collegeville, PA, USA
| | - Zachary W Davis-Gilbert
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paul Workman
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK.
| | - Opher Gileadi
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- SGC Karolinska, Centre for Molecular Medicine, Stockholm, Sweden
| | - David H Drewry
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
2
|
Zheng BW, Guo W. Multi-omics analysis unveils the role of inflammatory cancer-associated fibroblasts in chordoma progression. J Pathol 2025; 265:69-83. [PMID: 39611243 DOI: 10.1002/path.6369] [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: 07/17/2024] [Revised: 09/25/2024] [Accepted: 10/13/2024] [Indexed: 11/30/2024]
Abstract
Cancer-associated fibroblasts (CAFs) constitute the primary cellular component of the stroma in chordomas, characterized by an abundance of mucinous stromal elements, potentially facilitating their initiation and progression; however, this inference has yet to be fully confirmed. In this study, single-cell RNA sequencing (scRNA-seq), spatial transcriptomics (ST), bulk RNA-seq, multiplexed quantitative immunofluorescence (QIF), and in vivo and in vitro experiments were performed to determine the heterogeneity, spatial distribution, and clinical significance of CAFs in chordoma. ScRNA-seq was performed on 87,693 single cells derived from seven tumor samples and four control nucleus pulposus samples. A distinct CAF cluster distinguished by the upregulated expression of inflammatory genes and enriched functionality in activating inflammation-associated cells was identified. Pseudotime trajectory and cell communication analyses suggested that this inflammatory CAF (iCAF) subset originated from normal fibroblasts and interacted extensively with tumors and various other cell types. By integrating the scRNA-seq results with ST, the presence of iCAF in chordoma tissue was further confirmed, indicating their positioning at a distance from the tumor cells. Bulk RNA-seq data analysis from 126 patients revealed a correlation between iCAF signature scores, chordoma invasiveness, and poor prognosis. QIF validation involving an additional 116 patients found that although iCAFs were not in close proximity to tumor cells compared with other CAF subsets, their density correlated with malignant tumor phenotypes and adverse outcomes. In vivo and in vitro experiments further confirmed that iCAFs accelerate the malignant progression of chordomas. These findings could provide insights into the development of novel therapeutic strategies. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bo-Wen Zheng
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Peking University, Beijing, PR China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, PR China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Peking University, Beijing, PR China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, PR China
| |
Collapse
|
3
|
Aydemir E, Yılmaz NZ, Bayrak ÖF, Sahin F. Investigating the Effects of Chordoma Cell-Derived Exosomes on the Tumorigenicity of Nucleus Pulposus Cells. J Neurol Surg B Skull Base 2024; 85:161-167. [PMID: 38449582 PMCID: PMC10914466 DOI: 10.1055/a-2018-4627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Objective Interaction of tumor cells with the surrounding environment is essential for tumor growth and progression that eventually leads to metastasis. Growing evidence shows that extracellular vesicles also known as exosomes play a crucial role in signaling between the tumor and its microenvironment. Tumor-derived exosomes have generally protumorigenic effects such as metastasis, hypoxia, angiogenesis, and epithelial-mesenchymal transition. Methods In this study, exosomes were isolated from a chordoma cell line, MUG-Chor1, and characterized subsequently. The number of exosomes was determined and introduced into the healthy nucleus pulposus (NP) cells for 140 days. The protumorigenic effects of a chordoma cell line-derived exosomes that initiate the tumorigenesis on NP cells were investigated. The impact of tumor-derived exosomes on various cellular events including cell cycle, migration, proliferation, apoptosis, and viability has been studied by treating NP cells with chordoma cell-line-derived exosomes cells. Results Upon treatment with exosomes, the NP cells not only gained a chordoma-like morphology but also molecular characteristics such as alterations in the levels of certain gene expressions. The migratory and angiogenic capabilities of NP cells increased after treatment with chordoma-derived exosomes. Conclusion Based on our findings, we can conclude that exosomes carry information from tumor cells and may exert tumorigenic effects on nontumorous cells.
Collapse
Affiliation(s)
- Esra Aydemir
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, Türkiye
| | - Nur Zübeyda Yılmaz
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ömer Faruk Bayrak
- Department of Medical Genetics, Yeditepe University Medical School and Yeditepe University Hospital, Istanbul, Türkiye
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| |
Collapse
|
4
|
Li H, Tang Y, Ruan X, Zhang J, Liu H, Yu S, Chen H, Yang H, Zhang K, Chen K. N6-methyladenosine-modified circTEAD1 stabilizes Yap1 mRNA to promote chordoma tumorigenesis. Clin Transl Med 2024; 14:e1658. [PMID: 38659080 PMCID: PMC11043093 DOI: 10.1002/ctm2.1658] [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: 10/21/2023] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Chordoma, a rare bone tumour with aggressive local invasion and high recurrence rate with limited understanding of its molecular mechanisms. Circular RNAs (circRNAs) have been extensively implicated in tumorigenesis, yet their involvement in chordoma remains largely unexplored. N6-methyladenosine (m6A) modification holds a crucial function in regulating protein translation, RNA degradation and transcription. METHODS Initially, screening and validation of circTEAD1 in chordoma were conducted by high-throughput sequencing. Subsequently, sh-circTEAD1 and an overexpression plasmid were constructed. Colony formation assays, cell counting kit-8, Transwell and wound healing assays were utilized to validate the function of circTEAD1 in vitro. RNA pull-down assays identified the binding proteins of circTEAD1, which underwent verification through RNA immunoprecipitation (RIP). Methylated RIP assays were conducted to detect the m6A binding sites. Following this, luciferase assay, RT-qPCR, RIP and Western blotting analyses were conducted, revealing that Yap1 was the direct target of circTEAD1. Afterwards, the same methods were utilized for the validation of the function of Yap1 in chordoma in vitro. Finally, the regulatory relationship between circTEAD1 and Yap1 in chordoma was verified by an in vivo tumour formation assay. RESULTS CircTEAD1 was identified as an upregulated circRNA in chordoma specimens, with heightened circTEAD1 expression emerging as a prognostic indicator. In vitro experiments convincingly demonstrated that circTEAD1 significantly promoted chordoma cell invasion, migration and aggressiveness. Furthermore, the analysis revealed that methyltransferase-like 3-mediated m6A modification facilitated the cytoplasmic export of circTEAD1. The circTEAD1/IGF2BP3/Yap1 mRNA RNA-protein ternary complex not only bolstered the stability of Yap1 mRNA but also exerted a pivotal role in driving chordoma tumorigenesis. CONCLUSIONS In this study, the role of m6A-modified circTEAD1 in chordoma was identified. The findings offer novel insights into the potential molecular targets for chordoma therapy, shedding light on the intricate interplay between circRNAs, m6A modification and Yap1 mRNA in chordoma pathogenesis.
Collapse
Affiliation(s)
- Hanwen Li
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Yingchuang Tang
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Xingbang Ruan
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Junxin Zhang
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Hao Liu
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Shiyu Yu
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Hao Chen
- Institute of Translational Medicine, Medical CollegeYangzhou UniversityYangzhouPeople's Republic of China
| | - Huilin Yang
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Kai Zhang
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Kangwu Chen
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| |
Collapse
|
5
|
Jin L, Jin A, Wang L, Qi X, Jin Y, Zhang C, Niu M. NRP1 Induces Enhanced Stemness and Chemoresistance in Glioma Cells via YAP. Biol Pharm Bull 2024; 47:166-174. [PMID: 38220212 DOI: 10.1248/bpb.b23-00630] [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] [Indexed: 01/16/2024]
Abstract
Neuropilin-1 (NRP1), a transmembrane glycoprotein, plays an important role in the malignant progression of gliomas; however, its role in chemoresistance is not fully understood. In this study, we observed the effects of NRP1 on the stemness and chemoresistance of glioma cells and the mediating role of Yes-associated protein (YAP). We constructed NRP1 overexpressing LN-229 glioma cells. Cells were treated with recombinant NRP1 protein (rNRP1) and the YAP inhibitor Super-TDU when necessary. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to detect the sensitivity of cells to temozolomide (TMZ). Sphere and clone formation assays were performed to detect the sphere- and clone-forming abilities of cells. Western blotting was performed to detect cellular CD133, CD44, p-LATS1, and p-YAP protein expression. Immunofluorescence and flow cytometry were used to detect the subcellular localization of YAP and apoptosis, respectively. We found that both NRP1 overexpression and rNRP1 treatment enhanced self-renewal, TMZ resistance, and CD133 and CD44 protein expression in LN-229 cells. NRP1 overexpression and rNRP1 treatment also induced LATS1 and YAP dephosphorylation and YAP nuclear translocation. Super-TDU inhibits NRP1 overexpression-induced enhanced self-renewal and TMZ resistance in LN-229 cells. Our study suggests that NRP1 induces increased stemness in glioma cells, resulting in chemoresistance, and that this effect is associated with YAP activation.
Collapse
Affiliation(s)
| | - Ai Jin
- Cangzhou People's Hospital
| | | | | | | | | | | |
Collapse
|
6
|
Perez-Vega C, Akinduro OO, Ruiz-Garcia HJ, Ghaith AKA, Almeida JP, Jentoft ME, Mahajan A, Janus JR, Bendok BR, Choby GW, Middlebrooks EH, Trifiletti DM, Chaichana KL, Laack NN, Quinones-Hinojosa A, Van Gompel JJ. Extent of Surgical Resection as a Predictor of Tumor Progression in Skull Base Chordomas: A Multicenter Volumetric Analysis. World Neurosurg 2024; 181:e620-e627. [PMID: 37898264 DOI: 10.1016/j.wneu.2023.10.101] [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: 08/07/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/30/2023]
Abstract
INTRODUCTION Skull-base chordomas are aggressive tumors with a propensity for recurrence/progression. Even with standard of care (SoC), 5-year recurrence rates are variable (19%-54%). This high recurrence/progression rate correlates with increased morbidity and mortality. We sought to analyze a multicenter cohort of skull base chordomas to identify predictors of progression in patients receiving SoC. METHODS The [Blinded]-Neurosurgery data registry was queried for skull base chordomas treated from 2008-2020. Patients with the histopathologic diagnosis of chordoma were included. The cohort was composed of patients with preoperative and postoperative magnetic resonance imaging. Tumor volume and radiologic characteristics were obtained from axial T2 sequences using a Digital Imaging and Communications in Medicine viewer. Survival analysis was performed using Kaplan-Meier method, and time-to-event multivariate regression was performed to identify independent predictors of progression. RESULTS The cohort included 195 patients, of which 66 patients met inclusion criteria; median age was 44, and 28 (42%) were females. Fifty-four (82%) received SoC, 7 (11%) resection only, and 5 (8%) radiotherapy only. Median preoperative and postoperative tumor volumes were 11.55 cm3 (0.33-54.89) and 0.34 cm3 (0-42.52), respectively. Recurrence rate with SoC was 37%. Postoperative tumor volume (P = 0.010) correlated with progression. A postoperative volume of >4.9 cm3 (P = 0.044), ≤81.3% of tumor resection (P = 0.02), and lower-clivus location (P < 0.005) correlated with decreased time to progression. CONCLUSIONS Skull base chordomas can be challenging to resect. Even though maximal resection and radiotherapy improve rate of tumor progression, many of these lesions eventually recur. We have identified a postoperative tumor volume of ≥4.9 cm3 and extent of resection of ≤81.3% in this cohort as predictors of progression in patients receiving SoC.
Collapse
Affiliation(s)
- Carlos Perez-Vega
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | | | - Joao P Almeida
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, Florida, USA
| | - Mark E Jentoft
- Department of Lab Medicine and Pathology, Jacksonville, Florida, USA
| | - Anita Mahajan
- Department of Radiation Oncology, Rochester, Minnesota, USA
| | | | | | - Garret W Choby
- Department of Otolaryngology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | - Nadia N Laack
- Department of Radiation Oncology, Rochester, Minnesota, USA
| | | | - Jamie J Van Gompel
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
7
|
Şişli HB, Hayal TB, Şenkal S, Bulut E, Kıratlı B, Asutay AB, Şahin F, Bayrak ÖF, Doğan A. Activation of Wnt Pathway Suppresses Growth of MUG-Chor1 Chordoma Cell Line. Cell Biochem Biophys 2023; 81:823-837. [PMID: 37751039 DOI: 10.1007/s12013-023-01178-5] [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] [Accepted: 09/15/2023] [Indexed: 09/27/2023]
Abstract
Chordoma as a malignant bone tumor, occurs along the axial skeleton and does not have an effective therapy. Brachyury, which is a crucial player for the formation of early embryonic notochord, is abundantly found in both sporadic and familial chordoma. During embryonic development, Brachyury expression was reported to be regulated by the Wnt pathway. The objective of the study is to investigate the role of Wnt signaling in a human chordoma cell line in terms of proliferation, survival, and invasiveness. We tried to elucidate the signaling events that regulate Chordoma cancer. In this regard, Wnt pathway was activated or inhibited using various strategies including small molecules, siRNA-based knockdown and overexpression applications. The results indicated the negative regulatory effect of Wnt signaling activity on proliferation and migration capacity of the chordoma cells. It was revealed that when GSK3β was inhibited, the Wnt pathway was activated and negatively regulated T/Bra expression. Activity of the Wnt pathway caused cell cycle arrest, reduced migration potential of the cells, and led to cell death. Therefore, the present study suggests that the Wnt pathway plays a key role in suppressing the proliferation and invasive characteristics of human chordoma cells and has a great potential as a therapeutic target in further clinical studies.
Collapse
Affiliation(s)
- Hatice Burcu Şişli
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Taha Bartu Hayal
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Selinay Şenkal
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Ezgi Bulut
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Binnur Kıratlı
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Ayla Burçin Asutay
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Fikrettin Şahin
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Ömer Faruk Bayrak
- Department of Medical Genetics, School of Medicine, Yeditepe University, İstanbul, 34755, Turkey
| | - Ayşegül Doğan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey.
| |
Collapse
|
8
|
Ma T, Bai J, Zhang Y. Current understanding of brachyury in chordoma. Biochim Biophys Acta Rev Cancer 2023; 1878:189010. [PMID: 39492486 DOI: 10.1016/j.bbcan.2023.189010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/07/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
Chordomas are rare malignant tumors that pose significant challenges in terms of effective treatment options. Surgical resection remains the only established approach that has proven useful in the treatment of chordoma. However, recent evidence has shed light on the role of brachyury, also known as the T-gene and TBXT, as both a diagnostic marker and a potential therapeutic target in chordoma. Considering these developments, this review aims to provide a comprehensive summary of the current knowledge and understanding of brachyury in chordomas.
Collapse
Affiliation(s)
- Tianshun Ma
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiwei Bai
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Xu J, Shi Q, Wang B, Ji T, Guo W, Ren T, Tang X. The role of tumor immune microenvironment in chordoma: promising immunotherapy strategies. Front Immunol 2023; 14:1257254. [PMID: 37720221 PMCID: PMC10502727 DOI: 10.3389/fimmu.2023.1257254] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Chordoma is a rare malignant bone tumor with limited therapeutic options, which is resistant to conventional chemotherapy and radiotherapy, and targeted therapy is also shown with little efficacy. The long-standing delay in researching its mechanisms of occurrence and development has resulted in the dilemma of no effective treatment targets and no available drugs in clinical practice. In recent years, the role of the tumor immune microenvironment in driving tumor growth has become a hot and challenging topic in the field of cancer research. Immunotherapy has shown promising results in the treatment of various tumors. However, the study of the immune microenvironment of chordoma is still in its infancy. In this review, we aim to present a comprehensive reveal of previous exploration on the chordoma immune microenvironment and propose promising immunotherapy strategies for chordoma based on these characteristics.
Collapse
Affiliation(s)
- Jiuhui Xu
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Qianyu Shi
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Boyang Wang
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Tao Ji
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
10
|
Lin WH, Feathers RW, Cooper LM, Lewis-Tuffin LJ, Chen J, Sarkaria JN, Anastasiadis PZ. A Syx-RhoA-Dia1 signaling axis regulates cell cycle progression, DNA damage, and therapy resistance in glioblastoma. JCI Insight 2023; 8:e157491. [PMID: 37427593 PMCID: PMC10371349 DOI: 10.1172/jci.insight.157491] [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: 12/13/2021] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Glioblastomas (GBM) are aggressive tumors that lack effective treatments. Here, we show that the Rho family guanine nucleotide exchange factor Syx promotes GBM cell growth both in vitro and in orthotopic xenografts derived from patients with GBM. Growth defects upon Syx depletion are attributed to prolonged mitosis, increased DNA damage, G2/M cell cycle arrest, and cell apoptosis, mediated by altered mRNA and protein expression of various cell cycle regulators. These effects are phenocopied by depletion of the Rho downstream effector Dia1 and are due, at least in part, to increased phosphorylation, cytoplasmic retention, and reduced activity of the YAP/TAZ transcriptional coactivators. Furthermore, targeting Syx signaling cooperates with radiation treatment and temozolomide (TMZ) to decrease viability in GBM cells, irrespective of their inherent response to TMZ. The data indicate that a Syx-RhoA-Dia1-YAP/TAZ signaling axis regulates cell cycle progression, DNA damage, and therapy resistance in GBM and argue for its targeting for cancer treatment.
Collapse
Affiliation(s)
- Wan-Hsin Lin
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ryan W. Feathers
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Lisa M. Cooper
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Jiaxiang Chen
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
11
|
Hu Y, Lu B, Deng Z, Xing F, Hsu W. Virus-like particle-based delivery of Cas9/guide RNA ribonucleoprotein efficiently edits the brachyury gene and inhibits chordoma growth in vivo. Discov Oncol 2023; 14:70. [PMID: 37198417 DOI: 10.1007/s12672-023-00680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023] Open
Abstract
PURPOSE Chordoma is a rare and aggressive bone cancer driven by the developmental transcription factor brachyury. Efforts to target brachyury are hampered by the absence of ligand-accessible small-molecule binding pockets. Genome editing with CRISPR systems provides an unprecedented opportunity to modulate undruggable transcription factor targets. However, delivery of CRISPR remains a bottleneck for in vivo therapy development. The aim was to investigate the in vivo therapeutic efficiency of Cas9/guide RNA (gRNA) ribonucleoprotein (RNP) delivery through a novel virus-like particle (VLP) by fusing an aptamer-binding protein to the lentiviral nucleocapsid protein. METHODS The p24 based ELISA and transmission electron microscopy were used to determine the characterization of engineered VLP-packaged Cas9/gRNA RNP. The deletion efficiency of brachyury gene in chordoma cells and tissues was measured by genome cleavage detection assay. RT-PCR, Western blot, immunofluorescence staining, and IHC were employed to test the function of brachyury deletion. Cell growth and tumor volume were measured to evaluate the therapeutic efficiency of brachyury deletion by VLP-packaged Cas9/gRNA RNP. RESULTS Our "all-in-one" VLP-based Cas9/gRNA RNP system allows for transient expression of Cas9 in chordoma cells, but maintains efficient editing capacity leading to approximately 85% knockdown of brachyury with subsequent inhibition of chordoma cell proliferation and tumor progression. In addition, this VLP-packaged brachyury-targeting Cas9 RNP avoids systemic toxicities in vivo. CONCLUSION Our preclinical studies demonstrate the potential of VLP-based Cas9/gRNA RNP gene therapy for the treatment of brachyury-dependent chordoma.
Collapse
Affiliation(s)
- Yunping Hu
- Department of Neurological Surgery, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| | - Baisong Lu
- Medical Center Boulevard, Wake Forest University Institute for Regenerative Medicine, Winston-Salem, NC, 27157, USA
| | - Zhiyong Deng
- Department of Physiology and Pharmacology, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Fei Xing
- Department of Cancer Biology, Medical Center Boulevard, Wake Forest University School of Medicine, Winston- Salem, NC, 27157, USA
| | - Wesley Hsu
- Department of Neurological Surgery, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
12
|
Saeui CT, Shah SR, Fernandez-Gil BI, Zhang C, Agatemor C, Dammen-Brower K, Mathew MP, Buettner M, Gowda P, Khare P, Otamendi-Lopez A, Yang S, Zhang H, Le A, Quinoñes-Hinojosa A, Yarema KJ. Anticancer Properties of Hexosamine Analogs Designed to Attenuate Metabolic Flux through the Hexosamine Biosynthetic Pathway. ACS Chem Biol 2023; 18:151-165. [PMID: 36626752 DOI: 10.1021/acschembio.2c00784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Altered cellular metabolism is a hallmark of cancer pathogenesis and progression; for example, a near-universal feature of cancer is increased metabolic flux through the hexosamine biosynthetic pathway (HBP). This pathway produces uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), a potent oncometabolite that drives multiple facets of cancer progression. In this study, we synthesized and evaluated peracetylated hexosamine analogs designed to reduce flux through the HBP. By screening a panel of analogs in pancreatic cancer and glioblastoma multiform (GBM) cells, we identified Ac4Glc2Bz─a benzyl-modified GlcNAc mimetic─as an antiproliferative cancer drug candidate that down-regulated oncogenic metabolites and reduced GBM cell motility at concentrations non-toxic to non-neoplastic cells. More specifically, the growth inhibitory effects of Ac4Glc2Bz were linked to reduced levels of UDP-GlcNAc and concomitant decreases in protein O-GlcNAc modification in both pancreatic cancer and GBM cells. Targeted metabolomics analysis in GBM cells showed that Ac4Glc2Bz disturbed glucose metabolism, amino acid pools, and nucleotide precursor biosynthesis, consistent with reduced proliferation and other anti-oncogenic properties of this analog. Furthermore, Ac4Glc2Bz reduced the invasion, migration, and stemness of GBM cells. Importantly, normal metabolic functions mediated by UDP-GlcNAc were not disrupted in non-neoplastic cells, including maintenance of endogenous levels of O-GlcNAcylation with no global disruption of N-glycan production. Finally, a pilot in vivo study showed that a potential therapeutic window exists where animals tolerated 5- to 10-fold higher levels of Ac4Glc2Bz than projected for in vivo efficacy. Together, these results establish GlcNAc analogs targeting the HBP through salvage mechanisms as a new therapeutic approach to safely normalize an important facet of aberrant glucose metabolism associated with cancer.
Collapse
Affiliation(s)
- Christopher T Saeui
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Sagar R Shah
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | | | - Cissy Zhang
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Christian Agatemor
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Kris Dammen-Brower
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Mohit P Mathew
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Matthew Buettner
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Prateek Gowda
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Pratik Khare
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | | | - Shuang Yang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Hui Zhang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Anne Le
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | | | - Kevin J Yarema
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| |
Collapse
|
13
|
Lu ZG, Shen J, Yang J, Wang JW, Zhao RC, Zhang TL, Guo J, Zhang X. Nucleic acid drug vectors for diagnosis and treatment of brain diseases. Signal Transduct Target Ther 2023; 8:39. [PMID: 36650130 PMCID: PMC9844208 DOI: 10.1038/s41392-022-01298-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/08/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
Nucleic acid drugs have the advantages of rich target selection, simple in design, good and enduring effect. They have been demonstrated to have irreplaceable superiority in brain disease treatment, while vectors are a decisive factor in therapeutic efficacy. Strict physiological barriers, such as degradation and clearance in circulation, blood-brain barrier, cellular uptake, endosome/lysosome barriers, release, obstruct the delivery of nucleic acid drugs to the brain by the vectors. Nucleic acid drugs against a single target are inefficient in treating brain diseases of complex pathogenesis. Differences between individual patients lead to severe uncertainties in brain disease treatment with nucleic acid drugs. In this Review, we briefly summarize the classification of nucleic acid drugs. Next, we discuss physiological barriers during drug delivery and universal coping strategies and introduce the application methods of these universal strategies to nucleic acid drug vectors. Subsequently, we explore nucleic acid drug-based multidrug regimens for the combination treatment of brain diseases and the construction of the corresponding vectors. In the following, we address the feasibility of patient stratification and personalized therapy through diagnostic information from medical imaging and the manner of introducing contrast agents into vectors. Finally, we take a perspective on the future feasibility and remaining challenges of vector-based integrated diagnosis and gene therapy for brain diseases.
Collapse
Affiliation(s)
- Zhi-Guo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| | - Jie Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jun Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jing-Wen Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Rui-Chen Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Tian-Lu Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Jing Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| |
Collapse
|
14
|
Cancer Vaccines for Triple-Negative Breast Cancer: A Systematic Review. Vaccines (Basel) 2023; 11:vaccines11010146. [PMID: 36679991 PMCID: PMC9866612 DOI: 10.3390/vaccines11010146] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the subtype of breast cancer with the poorest outcomes, and is associated with a high risk of relapse and metastasis. The treatment choices for this malignancy have been confined to conventional chemotherapeutic agents, due to a lack of expression of the canonical molecular targets. Immunotherapy has been recently changing the treatment paradigm for many types of tumors, and the approach of evoking active immune responses in the milieu of breast tumors through cancer vaccines has been introduced as one of the most novel immunotherapeutic approaches. Accordingly, a number of vaccines for the treatment or prevention of recurrence have been developed and are currently being studied in TNBC patients, while none have yet received any approvals. To elucidate the efficacy and safety of these vaccines, we performed a systematic review of the available literature on the topic. After searching the PubMed, Scopus, Web of Science, Embase, Cochrane CENTRAL, and Google Scholar databases, a total of 5701 results were obtained, from which 42 clinical studies were eventually included based on the predefined criteria. The overall quality of the included studies was acceptable. However, due to a lack of reporting outcomes of survival or progression in some studies (which were presented as conference abstracts) as well as the heterogeneity of the reported outcomes and study designs, we were not able to carry out a meta-analysis. A total of 32 different vaccines have so far been evaluated in TNBC patients, with the majority belonging to the peptide-based vaccine type. The other vaccines were in the cell or nucleic acid (RNA/DNA)-based categories. Most vaccines proved to be safe with low-grade, local adverse events and could efficiently evoke cellular immune responses; however, most trials were not able to demonstrate significant improvements in clinical indices of efficacy. This is in part due to the limited number of randomized studies, as well as the limited TNBC population of each trial. However, due to the encouraging results of the currently published trials, we anticipate that this strategy could show its potential through larger, phase III randomized studies in the near future.
Collapse
|
15
|
Ervin EH, French R, Chang CH, Pauklin S. Inside the stemness engine: Mechanistic links between deregulated transcription factors and stemness in cancer. Semin Cancer Biol 2022; 87:48-83. [PMID: 36347438 DOI: 10.1016/j.semcancer.2022.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/22/2022] [Accepted: 11/03/2022] [Indexed: 11/07/2022]
Abstract
Cell identity is largely determined by its transcriptional profile. In tumour, deregulation of transcription factor expression and/or activity enables cancer cell to acquire a stem-like state characterised by capacity to self-renew, differentiate and form tumours in vivo. These stem-like cancer cells are highly metastatic and therapy resistant, thus warranting a more complete understanding of the molecular mechanisms downstream of the transcription factors that mediate the establishment of stemness state. Here, we review recent research findings that provide a mechanistic link between the commonly deregulated transcription factors and stemness in cancer. In particular, we describe the role of master transcription factors (SOX, OCT4, NANOG, KLF, BRACHYURY, SALL, HOX, FOX and RUNX), signalling-regulated transcription factors (SMAD, β-catenin, YAP, TAZ, AP-1, NOTCH, STAT, GLI, ETS and NF-κB) and unclassified transcription factors (c-MYC, HIF, EMT transcription factors and P53) across diverse tumour types, thereby yielding a comprehensive overview identifying shared downstream targets, highlighting unique mechanisms and discussing complexities.
Collapse
Affiliation(s)
- Egle-Helene Ervin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford, OX3 7LD, United Kingdom.
| | - Rhiannon French
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford, OX3 7LD, United Kingdom.
| | - Chao-Hui Chang
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford, OX3 7LD, United Kingdom.
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford, OX3 7LD, United Kingdom.
| |
Collapse
|
16
|
Freed DM, Sommer J, Punturi N. Emerging target discovery and drug repurposing opportunities in chordoma. Front Oncol 2022; 12:1009193. [PMID: 36387127 PMCID: PMC9647139 DOI: 10.3389/fonc.2022.1009193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/11/2022] [Indexed: 09/01/2023] Open
Abstract
The development of effective and personalized treatment options for patients with rare cancers like chordoma is hampered by numerous challenges. Biomarker-guided repurposing of therapies approved in other indications remains the fastest path to redefining the treatment paradigm, but chordoma's low mutation burden limits the impact of genomics in target discovery and precision oncology efforts. As our knowledge of oncogenic mechanisms across various malignancies has matured, it's become increasingly clear that numerous properties of tumors transcend their genomes - leading to new and uncharted frontiers of therapeutic opportunity. In this review, we discuss how the implementation of cutting-edge tools and approaches is opening new windows into chordoma's vulnerabilities. We also note how a convergence of emerging observations in chordoma and other cancers is leading to the identification and evaluation of new therapeutic hypotheses for this rare cancer.
Collapse
|
17
|
Hu Y, Lu Y, Xing F, Hsu W. FGFR1/MAPK-directed brachyury activation drives PD-L1-mediated immune evasion to promote lung cancer progression. Cancer Lett 2022; 547:215867. [PMID: 35985510 DOI: 10.1016/j.canlet.2022.215867] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/02/2022]
Abstract
Immune checkpoint inhibitors provide promising benefits for patients with cancer. However, efficacy has been encumbered by high resistance rates. It is critical to understand the basic mechanisms of tumor-mediated resistance to this treatment modality. Previous studies have found that the transcription factor brachyury is highly expressed in lung cancer. Here, we show that brachyury activation induces the upregulation of PD-L1 leading to inactivation of T cell proliferation in vitro and inhibited infiltration of CD8+ and CD3+ T cells into tumor in an immunocompetent mouse model. We further demonstrate that FGFR1/MAPK activation regulates brachyury and PD-L1 expressions and promotes immunosuppression. Blocking FGFR1/MAPK suppresses brachyury and PD-L1 expressions, revives immune activity, and reverses the resistance to anti-PD-1 treatment to produce a durable therapeutic response. We also find that lung cancer patients with high activation of the FGFR1-MAPK-brachyury-PD-L1 signature and low expression of CD8A, CD3D, or PDCD1 have worse survival outcomes. These findings elucidate a novel mechanism of immune escape from immune checkpoint therapy and provide an opportunity to enhance its therapeutic efficacy in the treatment of a subset of FGFR1/MAPK/brachyury/PD-L1-driven lung cancer.
Collapse
Affiliation(s)
- Yunping Hu
- Department of Neurological Surgery, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Yong Lu
- The Methodist Hospital Research Institute, 6670 Bertner Avenue, Houston, Houston, TX, 77030, USA
| | - Fei Xing
- Department of Cancer Biology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Wesley Hsu
- Department of Neurological Surgery, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
18
|
Bozsodi A, Scholtz B, Papp G, Sapi Z, Biczo A, Varga PP, Lazary A. Potential molecular mechanism in self-renewal is associated with miRNA dysregulation in sacral chordoma - A next-generation RNA sequencing study. Heliyon 2022; 8:e10227. [PMID: 36033338 PMCID: PMC9404356 DOI: 10.1016/j.heliyon.2022.e10227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/24/2022] [Accepted: 08/04/2022] [Indexed: 12/04/2022] Open
Abstract
Background Chordoma, the most frequent malignant primary spinal neoplasm, characterized by a high rate of recurrence, is an orphan disease where the clarification of the molecular oncogenesis would be crucial to developing new, effective therapies. Dysregulated expression of non-coding RNAs, especially microRNAs (miRNA) has a significant role in cancer development. Methods Next-generation RNA sequencing (NGS) was used for the combinatorial analysis of mRNA-miRNA gene expression profiles in sacral chordoma and nucleus pulposus samples. Advanced bioinformatics workflow was applied to the data to predict miRNA-mRNA regulatory networks with altered activity in chordoma. Results A large set of significantly dysregulated miRNAs in chordoma and their differentially expressed target genes have been identified. Several molecular pathways related to tumorigenesis and the modulation of the immune system are predicted to be dysregulated due to aberrant miRNA expression in chordoma. We identified a gene set including key regulators of the Hippo pathway, which is targeted by differently expressed miRNAs, and validated their altered expression by RT-qPCR. These newly identified miRNA/RNA interactions are predicted to have a role in the self-renewal process of chordoma stem cells, which might sustain the high rate of recurrence for this tumor. Conclusions Our results can significantly contribute to the designation of possible targets for the development of anti-chordoma therapies.
Collapse
Affiliation(s)
- Arpad Bozsodi
- National Center for Spinal Disorders, Buda Health Center, Királyhágó u. 1-3, Budapest, H-1126, Hungary
- School of PhD Studies, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Beata Scholtz
- Genomic Medicine and Bioinformatic Core Facility, Dept. of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Gergo Papp
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Zoltan Sapi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Adam Biczo
- National Center for Spinal Disorders, Buda Health Center, Királyhágó u. 1-3, Budapest, H-1126, Hungary
| | - Peter Pal Varga
- National Center for Spinal Disorders, Buda Health Center, Királyhágó u. 1-3, Budapest, H-1126, Hungary
| | - Aron Lazary
- National Center for Spinal Disorders, Buda Health Center, Királyhágó u. 1-3, Budapest, H-1126, Hungary
- Department of Spine Surgery, Department of Orthopaedics, Semmelweis University, Királyhágó u. 1-3, Budapest, H-1126, Hungary
- Corresponding author.
| |
Collapse
|
19
|
Perftoran improves Visudyne-photodynamic therapy via suppressing hypoxia pathway in murine lung cancer. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2022. [DOI: 10.1016/j.jrras.2022.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Niu G, Hao J, Sheng S, Wen F. Role of T-box genes in cancer, epithelial-mesenchymal transition, and cancer stem cells. J Cell Biochem 2021; 123:215-230. [PMID: 34897787 DOI: 10.1002/jcb.30188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/13/2022]
Abstract
Sharing a common DNA binding motif called T-box, transcription factor T-box gene family controls embryonic development and is also involved in cancer progression and metastasis. Cancer metastasis shows therapy resistance and involves complex processes. Among them, epithelial-mesenchymal transition (EMT) triggers cancer cell invasiveness and the acquisition of stemness of cancer cells, called cancer stem cells (CSCs). CSCs are a small fraction of tumor bulk and are capable of self-renewal and tumorsphere formation. Recent progress has highlighted the critical roles of T-box genes in cancer progression, EMT, and CSC function, and such regulatory functions of T-box genes have emerged as potential therapeutic candidates for cancer. Herein we summarize the current understanding of the regulatory mechanisms of T-box genes in cancer, EMT, and CSCs, and discuss the implications of targeting T-box genes as anticancer therapeutics.
Collapse
Affiliation(s)
- Gengle Niu
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Jin Hao
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Surui Sheng
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyuan Wen
- Department of Outpatient, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
21
|
Jalessi M, Gholami MS, Razmara E, Hassanzadeh S, Sadeghipour A, Jahanbakhshi A, Tabibkhooei A, Bahrami E, Falah M. Association between TBXT rs2305089 polymorphism and chordoma in Iranian patients identified by a developed T-ARMS-PCR assay. J Clin Lab Anal 2021; 36:e24150. [PMID: 34837714 PMCID: PMC8761424 DOI: 10.1002/jcla.24150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/05/2021] [Accepted: 11/18/2021] [Indexed: 11/20/2022] Open
Abstract
Background Chordoma is a locally aggressive bone tumor with a high capability of recurrence. Because chordoma often occurs at critical locations next to neurovascular structures, there is an urgent need to introduce validated biomarkers. T‐box transcription factor T (TBXT; OMIM: 601397) plays an important role in the pathogenesis and survival of chordoma cells. Methods Herein, we aimed to show whether rs2305089 polymorphism is correlated with chordoma in the Iranian population. In order to detect rs2305089, tetra‐primer amplification refractory mutation system‐polymerase chain reaction (T‐ARMS‐PCR) was used. In total, 19 chordoma patients and 108 normal healthy individuals were recruited and screened using T‐ARMS‐PCR. The results were subsequently validated by Sanger sequencing. Results The genotype distributions and allele frequencies were significantly different among the patient and healthy groups (p‐value <0.05). The A allele of rs2305089 showed a significant positive association with chordoma risk (p‐value <0.05). DNA sequencing verified the T‐ARMS‐PCR results as well. This study demonstrated the association between TBXT rs2305089 and chordoma in an Iranian population using a simple, accurate, and cost‐effective T‐ARMS‐PCR assay. Conclusions Our results were in line with those of previous studies showing that TBXT rs2305089 is associated with chordoma development. We also developed an efficient T‐ARMS‐PCR assay to determine the genotype of rs2305089.
Collapse
Affiliation(s)
- Maryam Jalessi
- Skull Base Research Center, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.,ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Saeed Gholami
- Skull Base Research Center, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.,Department of Hematology and Blood Transfusion, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ehsan Razmara
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Sajad Hassanzadeh
- Skull Base Research Center, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Sadeghipour
- Pathology Department, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Amin Jahanbakhshi
- Skull Base Research Center, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Tabibkhooei
- Department of Neurosurgery, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Eshagh Bahrami
- Skull Base Research Center, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.,Department of Neurosurgery, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Falah
- Skull Base Research Center, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.,ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Chen X, Lo SFL, Bettegowda C, Ryan DM, Gross JM, Hu C, Kleinberg L, Sciubba DM, Redmond KJ. High-dose hypofractionated stereotactic body radiotherapy for spinal chordoma. J Neurosurg Spine 2021; 35:674-683. [PMID: 34388713 DOI: 10.3171/2021.2.spine202199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/02/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Spinal chordoma is locally aggressive and has a high rate of recurrence, even after en bloc resection. Conventionally fractionated adjuvant radiation leads to suboptimal tumor control, and data regarding hypofractionated regimens are limited. The authors hypothesized that neoadjuvant stereotactic body radiotherapy (SBRT) may overcome its intrinsic radioresistance, improve surgical margins, and allow preservation of critical structures during surgery. The purpose of this study is to review the feasibility and early outcomes of high-dose hypofractionated SBRT, with a focus on neoadjuvant SBRT. METHODS Electronic medical records of patients with spinal chordoma treated using image-guided SBRT between 2009 and 2019 at a single institution were retrospectively reviewed. RESULTS Twenty-eight patients with 30 discrete lesions (24 in the mobile spine) were included. The median follow-up duration was 20.8 months (range 2.3-126.3 months). The median SBRT dose was 40 Gy (range 15-50 Gy) in 5 fractions (range 1-5 fractions). Seventeen patients (74% of those with newly diagnosed lesions) received neoadjuvant SBRT, of whom 15 (88%) underwent planned en bloc resection, all with negative margins. Two patients (12%) developed surgical wound-related complications after neoadjuvant SBRT and surgery, and 4 (two grade 3 and two grade 2) experienced postoperative complications unrelated to the surgical site. Of the remaining patients with newly diagnosed lesions, 5 received adjuvant SBRT for positive or close surgical margins, and 1 received SBRT alone. Seven recurrent lesions were treated with SBRT alone, including 2 after failure of prior conventional radiation. The 2-year overall survival rate was 92% (95% confidence interval [CI] 71%-98%). Patients with newly diagnosed chordoma had longer median survival (not reached) than those with recurrent lesions (27.7 months, p = 0.006). The 2-year local control rate was 96% (95% CI 74%-99%). Among patients with radiotherapy-naïve lesions, no local recurrence was observed with a biologically effective dose ≥ 140 Gy, maximum dose of the planning target volume (PTV) ≥ 47 Gy, mean dose of the PTV ≥ 39 Gy, or minimum dose to 80% of the PTV ≥ 36 Gy (5-fraction equivalent doses). All acute toxicities from SBRT were grade 1-2, and no myelopathy was observed. CONCLUSIONS Neoadjuvant high-dose, hypofractionated SBRT for spinal chordoma is safe and does not increase surgical morbidities. Early outcomes at 2 years are promising, although long-term follow-up is pending.
Collapse
Affiliation(s)
- Xuguang Chen
- Departments of1Radiation Oncology and Molecular Radiation Sciences
| | | | | | | | - John M Gross
- 4Pathology, Johns Hopkins University School of Medicine; and
| | - Chen Hu
- 5Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | | |
Collapse
|
23
|
Sumransub N, Murugan P, Marette S, Clohisy DR, Skubitz KM. Multiple malignant tumors in a patient with familial chordoma, a case report. BMC Med Genomics 2021; 14:213. [PMID: 34465320 PMCID: PMC8406958 DOI: 10.1186/s12920-021-01064-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Chordoma is a rare bone tumor that is typically resistant to chemotherapy and is associated with genetic abnormalities of the T-box transcription factor T (TBXT) gene, which encodes the transcription factor brachyury. Brachyury is felt to be a major contributor to the development of chordomas. CASE PRESENTATION We describe a 67-year-old woman who developed an undifferentiated pleomorphic sarcoma in her thigh. Despite treatment with standard chemotherapy regimens, she had a rapidly progressive course of disease with pulmonary metastases and passed away 8 months from diagnosis with pulmonary complications. Her medical history was remarkable in that she had a spheno-occipital chordoma at age 39 and later developed multiple other tumors throughout her life including Hodgkin lymphoma and squamous cell carcinoma and basal cell carcinoma of the skin. She had a family history of chordoma and her family underwent extensive genetic study in the past and were found to have a duplication of the TBXT gene. CONCLUSIONS Brachyury has been found to associate with tumor progression, treatment resistance, and metastasis in various epithelial cancers, and it might play roles in tumorigenesis and aggressiveness in this patient with multiple rare tumors and germ line duplication of the TBXT gene. Targeting this molecule may be useful for some malignancies.
Collapse
Affiliation(s)
- Nuttavut Sumransub
- Department of Medicine, University of Minnesota, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | - Paari Murugan
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN, 55455, USA
- The Masonic Cancer Center, 425 E River Pkwy, Minneapolis, MN, 55455, USA
| | - Shelly Marette
- The Masonic Cancer Center, 425 E River Pkwy, Minneapolis, MN, 55455, USA
- Department of Radiology, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | - Denis R Clohisy
- The Masonic Cancer Center, 425 E River Pkwy, Minneapolis, MN, 55455, USA
- Department of Orthopaedic Surgery, 2450 Riverside Ave Suite R200, Minneapolis, MN, 55454, USA
| | - Keith M Skubitz
- Department of Medicine, University of Minnesota, 420 Delaware St SE, Minneapolis, MN, 55455, USA.
- The Masonic Cancer Center, 425 E River Pkwy, Minneapolis, MN, 55455, USA.
- Department of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, 420 Delaware St. SE MMC 480, Minneapolis, MN, 55455, USA.
| |
Collapse
|
24
|
Mathsyaraja H, Catchpole J, Freie B, Eastwood E, Babaeva E, Geuenich M, Cheng PF, Ayers J, Yu M, Wu N, Moorthi S, Poudel KR, Koehne A, Grady W, Houghton AM, Berger AH, Shiio Y, MacPherson D, Eisenman RN. Loss of MGA repression mediated by an atypical polycomb complex promotes tumor progression and invasiveness. eLife 2021; 10:e64212. [PMID: 34236315 PMCID: PMC8266391 DOI: 10.7554/elife.64212] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 06/24/2021] [Indexed: 12/19/2022] Open
Abstract
MGA, a transcription factor and member of the MYC network, is mutated or deleted in a broad spectrum of malignancies. As a critical test of a tumor suppressive role, we inactivated Mga in two mouse models of non-small cell lung cancer using a CRISPR-based approach. MGA loss significantly accelerated tumor growth in both models and led to de-repression of non-canonical Polycomb ncPRC1.6 targets, including genes involved in metastasis and meiosis. Moreover, MGA deletion in human lung adenocarcinoma lines augmented invasive capabilities. We further show that MGA-MAX, E2F6, and L3MBTL2 co-occupy thousands of promoters and that MGA stabilizes these ncPRC1.6 subunits. Lastly, we report that MGA loss also induces a pro-growth effect in human colon organoids. Our studies establish MGA as a bona fide tumor suppressor in vivo and suggest a tumor suppressive mechanism in adenocarcinomas resulting from widespread transcriptional attenuation of MYC and E2F target genes mediated by MGA-MAX associated with a non-canonical Polycomb complex.
Collapse
Affiliation(s)
- Haritha Mathsyaraja
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Jonathen Catchpole
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Brian Freie
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Emily Eastwood
- Human Biology and Public Health Sciences Divisions, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Ekaterina Babaeva
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Michael Geuenich
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Pei Feng Cheng
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Jessica Ayers
- Clinical Research Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Ming Yu
- Clinical Research Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Nan Wu
- Human Biology and Public Health Sciences Divisions, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Sitapriya Moorthi
- Human Biology and Public Health Sciences Divisions, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Kumud R Poudel
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Amanda Koehne
- Comparative Pathology, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - William Grady
- Clinical Research Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Department of Medicine, University of Washington School of MedicineSeattleUnited States
| | - A McGarry Houghton
- Human Biology and Public Health Sciences Divisions, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Clinical Research Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Alice H Berger
- Human Biology and Public Health Sciences Divisions, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Yuzuru Shiio
- Greehey Children's Cancer Research Institute, The University of Texas Health Science CenterSan AntonioUnited States
| | - David MacPherson
- Human Biology and Public Health Sciences Divisions, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Robert N Eisenman
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| |
Collapse
|
25
|
The novel FAT4 activator jujuboside A suppresses NSCLC tumorigenesis by activating HIPPO signaling and inhibiting YAP nuclear translocation. Pharmacol Res 2021; 170:105723. [PMID: 34116210 DOI: 10.1016/j.phrs.2021.105723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/29/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022]
Abstract
FAT atypical cadherin 4 (FAT4) has been identified as a tumor suppressor in lung cancers. However, no agent for lung cancer treatment targeting FAT4 has been used in the clinic. Jujuboside A (JUA) is a major active compound in Semen Ziziphi Spinosae. Semen Ziziphi Spinosae is a traditional Chinese herbal medicine used clinically for tumor treatment to improve patients' quality of life. However, the anti-lung cancer activity and the underlying mechanisms of JUA are not yet fully understood. Here, we demonstrated the anti-lung cancer activity of JUA in two lung cancer mice models and three non-small cell lung cancer (NSCLC) cell lines, and further illustrated its underlying mechanisms. JUA suppressed the occurrence and development of lung cancer and extended mice survival in vivo, and suppressed NSCLC cell activities through cell cycle arrest, proliferation suppression, stemness inhibition and senescence promotion. Moreover, JUA directly bound with and activated FAT4, subsequently activating FAT4-HIPPO signaling and inhibiting YAP nuclear translocation. Knockdown of FAT4 diminished JUA's effects on HIPPO signaling, YAP nuclear translocation, cell proliferation and cellular senescence. In conclusion, JUA significantly suppressed NSCLC tumorigenesis by regulating FAT4-HIPPO-YAP signaling. Our findings suggest that JUA is a novel FAT4 activator that can be developed as a promising NSCLC therapeutic agent targeting the FAT4-HIPPO-YAP pathway.
Collapse
|
26
|
Yepes S, Shah NN, Bai J, Koka H, Li C, Gui S, McMaster ML, Xiao Y, Jones K, Wang M, Vogt A, Zhu B, Zhu B, Hutchinson A, Yeager M, Hicks B, Carter B, Freedman ND, Beane-Freeman L, Chanock SJ, Zhang Y, Parry DM, Yang XR, Goldstein AM. Rare Germline Variants in Chordoma-Related Genes and Chordoma Susceptibility. Cancers (Basel) 2021; 13:cancers13112704. [PMID: 34070849 PMCID: PMC8197919 DOI: 10.3390/cancers13112704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Chordoma is an extremely rare bone cancer that has not been fully characterized and few risk factors have been identified, highlighting the need for improving our understanding of the disease biology. Our study aims to identify chordoma susceptibility genes by investigating 265 genes involved in chordoma-related signaling pathways and other biological processes on germline DNA of 138 chordoma patients of European ancestry compared to internal control datasets and general population databases. Results were intersected with whole genome sequencing data from 80 skull-base chordoma patients of Chinese ancestry. Several rare loss-of-function and predicted deleterious missense variants were enriched in chordoma cases in both datasets, suggesting a complex model of pathways potentially involved in chordoma development and susceptibility, warranting further investigation in larger studies. Abstract Background: Chordoma is a rare bone cancer with an unknown etiology. TBXT is the only chordoma susceptibility gene identified to date; germline single nucleotide variants and copy number variants in TBXT have been associated with chordoma susceptibility in familial and sporadic chordoma. However, the genetic susceptibility of chordoma remains largely unknown. In this study, we investigated rare germline genetic variants in genes involved in TBXT/chordoma-related signaling pathways and other biological processes in chordoma patients from North America and China. Methods: We identified variants that were very rare in general population and internal control datasets and showed evidence for pathogenicity in 265 genes in a whole exome sequencing (WES) dataset of 138 chordoma patients of European ancestry and in a whole genome sequencing (WGS) dataset of 80 Chinese patients with skull base chordoma. Results: Rare and likely pathogenic variants were identified in 32 of 138 European ancestry patients (23%), including genes that are part of notochord development, PI3K/AKT/mTOR, Sonic Hedgehog, SWI/SNF complex and mesoderm development pathways. Rare pathogenic variants in COL2A1, EXT1, PDK1, LRP2, TBXT and TSC2, among others, were also observed in Chinese patients. Conclusion: We identified several rare loss-of-function and predicted deleterious missense variants in germline DNA from patients with chordoma, which may influence chordoma predisposition and reflect a complex susceptibility, warranting further investigation in large studies.
Collapse
Affiliation(s)
- Sally Yepes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Correspondence: (S.Y.); (A.M.G.)
| | - Nirav N. Shah
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Jiwei Bai
- Beijing Tiantan Hospital, Beijing 100070, China; (J.B.); (C.L.); (S.G.); (Y.Z.)
| | - Hela Koka
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Chuzhong Li
- Beijing Tiantan Hospital, Beijing 100070, China; (J.B.); (C.L.); (S.G.); (Y.Z.)
| | - Songbai Gui
- Beijing Tiantan Hospital, Beijing 100070, China; (J.B.); (C.L.); (S.G.); (Y.Z.)
| | - Mary Lou McMaster
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Yanzi Xiao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201, USA
| | - Mingyi Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201, USA
| | - Aurelie Vogt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201, USA
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201, USA
| | - Brian Carter
- American Cancer Society, Inc, Atlanta, GA 30303, USA;
| | - Neal D. Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Laura Beane-Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Yazhuo Zhang
- Beijing Tiantan Hospital, Beijing 100070, China; (J.B.); (C.L.); (S.G.); (Y.Z.)
| | - Dilys M. Parry
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Xiaohong R. Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
| | - Alisa M. Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (N.N.S.); (H.K.); (M.L.M.); (Y.X.); (K.J.); (M.W.); (A.V.); (B.Z.); (B.Z.); (A.H.); (M.Y.); (B.H.); (N.D.F.); (L.B.-F.); (S.J.C.); (D.M.P.); (X.R.Y.)
- Correspondence: (S.Y.); (A.M.G.)
| |
Collapse
|
27
|
Akinduro OO, Suarez-Meade P, Garcia D, Brown DA, Sarabia-Estrada R, Attia S, Gokaslan ZL, Quiñones-Hinojosa A. Targeted Therapy for Chordoma: Key Molecular Signaling Pathways and the Role of Multimodal Therapy. Target Oncol 2021; 16:325-337. [PMID: 33893940 DOI: 10.1007/s11523-021-00814-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chordoma is a rare but devastating tumor that arises in the cranial skull base or spine. There are currently no US Food and Drug Administration-approved targeted therapies for chordoma, and little understanding of whether using more than one therapy has benefit over monotherapy. OBJECTIVE The objective of this study was to systematically review the current status of clinical trials completed for patients with chordoma to determine if multimodal therapy offers a benefit in progression-free survival over monomodal therapy. METHODS We performed a systematic review of the literature according to Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines to review the available clinical trials of targeted therapy for chordoma. We compiled the clinical data to determine if there is a benefit of multimodal therapy over monotherapy. RESULTS Our search resulted in 11 clinical trials including 270 patients with advanced chordoma who were treated with targeted therapies. The most commonly employed targeted therapies acted within the following pathways: platelet-derived growth factor receptor (187 patients), vascular endothelial growth factor (66 patients), and mammalian target of rapamycin (43 patients). Reported progression-free survival for included studies ranged from 2.5 to 58 months, with the longest progression-free survival in a trial that included a platelet-derived growth factor receptor inhibitor, nilotinib, and concurrent radiotherapy (58.2 months). There was a higher range of progression-free survival for trials treating patients with multimodal therapy (10.2-14 months vs 2.5-9.2 months, except for a monotherapy trial published in 2020 with a progression-free survival of 18 months), and those published in 2018 or later (14-58.2 months vs 2.5-10.2 months). Only 23% of patients with chordoma in published clinical trials have been treated with multimodal therapy. CONCLUSIONS Progression-free survival may be enhanced by the use of targeted therapy with concurrent radiotherapy, use of multimodal therapy, and use of newer targeted therapy. Future clinical trials should consider use of concurrent radiotherapy and multimodal therapy for patients with advanced chordoma.
Collapse
Affiliation(s)
- Oluwaseun O Akinduro
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA
| | - Paola Suarez-Meade
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA
| | - Diogo Garcia
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA
| | | | - Rachel Sarabia-Estrada
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA
| | - Steven Attia
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Ziya L Gokaslan
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Alfredo Quiñones-Hinojosa
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA.
| |
Collapse
|
28
|
Wang L, Guan X, Hu Q, Wu Z, Chen W, Song L, Wang K, Tian K, Cao C, Zhang D, Ma J, Tong X, Zhang B, Zhang J, Zeng C. TGFB3 downregulation causing chordomagenesis and its tumor suppression role maintained by Smad7. Carcinogenesis 2021; 42:913-923. [PMID: 34057989 DOI: 10.1093/carcin/bgab022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/02/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Chordoma is a rare bone tumor arising from notochordal remnants, but the underlying mechanism remains elusive. By integrated mRNA and microRNA analyses, we found significant downregulation of TGFB3 along with upregulation of its inhibitor, miR-29 family in chordoma comparing with notochord. Somatic copy number gains of miR-29 loci in chordoma highlighted a mechanism of inactivation of TGFB3 signaling in tumor formation. In zebrafish, knockout and knockdown homologous tgfb3 resulted in a chordoma-like neoplasm. On the other hand, Smad7 negative feedback regulation of transforming growth factor-β (TGF-β) signaling is retentive in chordoma cell UM-Chor1 despite its disruption in most cancer cells (e.g. A549). Therefore, contrary to other cancers, exogenous TGF-β activated Smad7 by downregulating miR-182 and inhibited cell migration and invasion in UM-Chor1. Meanwhile, TGF-β decreased chordoma characteristic protein Brachyury. Altogether, downregulation of TGFB3 causes chordomagenesis, showing a feasible target for therapies. The retention of Smad7 negative regulation may maintain the suppressor role of TGF-β in chordoma.
Collapse
Affiliation(s)
- Liang Wang
- Neurosurgery Department, Beijing Tiantan Hospital, Capital Medical University, Tiantan Xili, Dongcheng District, Beijing, China.,China National Clinical Research Center for Neurological Diseases, NCRC-ND, Tiantan Xili, Dongcheng District, Beijing, China
| | - Xiaonan Guan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qingtao Hu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhen Wu
- Neurosurgery Department, Beijing Tiantan Hospital, Capital Medical University, Tiantan Xili, Dongcheng District, Beijing, China.,China National Clinical Research Center for Neurological Diseases, NCRC-ND, Tiantan Xili, Dongcheng District, Beijing, China
| | - Wei Chen
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lairong Song
- Neurosurgery Department, Beijing Tiantan Hospital, Capital Medical University, Tiantan Xili, Dongcheng District, Beijing, China.,China National Clinical Research Center for Neurological Diseases, NCRC-ND, Tiantan Xili, Dongcheng District, Beijing, China
| | - Ke Wang
- Neurosurgery Department, Beijing Tiantan Hospital, Capital Medical University, Tiantan Xili, Dongcheng District, Beijing, China.,China National Clinical Research Center for Neurological Diseases, NCRC-ND, Tiantan Xili, Dongcheng District, Beijing, China
| | - Kaibing Tian
- Neurosurgery Department, Beijing Tiantan Hospital, Capital Medical University, Tiantan Xili, Dongcheng District, Beijing, China.,China National Clinical Research Center for Neurological Diseases, NCRC-ND, Tiantan Xili, Dongcheng District, Beijing, China
| | - Chunwei Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Dake Zhang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Junpeng Ma
- Neurosurgery Department, Beijing Tiantan Hospital, Capital Medical University, Tiantan Xili, Dongcheng District, Beijing, China.,China National Clinical Research Center for Neurological Diseases, NCRC-ND, Tiantan Xili, Dongcheng District, Beijing, China
| | - Xiangjun Tong
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Bo Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Junting Zhang
- Neurosurgery Department, Beijing Tiantan Hospital, Capital Medical University, Tiantan Xili, Dongcheng District, Beijing, China.,China National Clinical Research Center for Neurological Diseases, NCRC-ND, Tiantan Xili, Dongcheng District, Beijing, China
| | - Changqing Zeng
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
29
|
Locquet MA, Dechaume AL, Berchard P, Abbes L, Pissaloux D, Tirode F, Ramos I, Bedoucha J, Valantin J, Karanian M, Perret R, Gille O, Blay JY, Dutour A. Aldehyde Dehydrogenase, a Therapeutic Target in Chordoma: Analysis in 3D Cellular Models. Cells 2021; 10:cells10020399. [PMID: 33672032 PMCID: PMC7919493 DOI: 10.3390/cells10020399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/29/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Chordomas are rare, slow-growing tumors of the axial skeleton. These tumors are locally aggressive and refractory to conventional therapies. Radical surgery and radiation remain the first-line treatments. Despite these aggressive treatments, chordomas often recur and second-line treatment options are limited. The mechanisms underlying chordoma radioresistance remain unknown, although several radioresistant cancer cells have been shown to respond favorably to aldehyde dehydrogenase (ALDH) inhibition. The study of chordoma has been delayed by small patient cohorts and few available models due to the scarcity of these tumors. We thus created cellular 3D models of chordoma by using low-adherence culture systems. Then, we evaluated their radiosensitivity using colony-forming and spheroid size assays. Finally, we determined whether pharmacologically inhibiting ALDH increased their radiosensitivity. We found that 3D cellular models of chordoma (derived from primary, relapse, and metastatic tumors) reproduce the histological and gene expression features of the disease. The metastatic, relapse, and primary spheroids displayed high, medium, and low radioresistance, respectively. Moreover, inhibiting ALDH decreased the radioresistance in all three models.
Collapse
Affiliation(s)
- Marie-Anaïs Locquet
- Team Cell Death and Pediatric Cancer, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008 Lyon, France; (M.-A.L.); (A.-L.D.); (P.B.); (L.A.); (I.R.); (J.B.); (J.-Y.B.)
| | - Anne-Lise Dechaume
- Team Cell Death and Pediatric Cancer, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008 Lyon, France; (M.-A.L.); (A.-L.D.); (P.B.); (L.A.); (I.R.); (J.B.); (J.-Y.B.)
| | - Paul Berchard
- Team Cell Death and Pediatric Cancer, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008 Lyon, France; (M.-A.L.); (A.-L.D.); (P.B.); (L.A.); (I.R.); (J.B.); (J.-Y.B.)
| | - Lhorra Abbes
- Team Cell Death and Pediatric Cancer, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008 Lyon, France; (M.-A.L.); (A.-L.D.); (P.B.); (L.A.); (I.R.); (J.B.); (J.-Y.B.)
| | - Daniel Pissaloux
- Department of Biopathology, Centre Leon Berard, F-69008 Lyon, France;
- Team Genetics, Epigenetics and Biology of Sarcomas, Univ Lyon, Université Claude Bernard Lyon 1, INSERM1052, CNRS5286, Cancer Research Center of Lyon, Centre Leon Berard, F-69008 Lyon, France; (F.T.); (M.K.)
| | - Franck Tirode
- Team Genetics, Epigenetics and Biology of Sarcomas, Univ Lyon, Université Claude Bernard Lyon 1, INSERM1052, CNRS5286, Cancer Research Center of Lyon, Centre Leon Berard, F-69008 Lyon, France; (F.T.); (M.K.)
| | - Inès Ramos
- Team Cell Death and Pediatric Cancer, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008 Lyon, France; (M.-A.L.); (A.-L.D.); (P.B.); (L.A.); (I.R.); (J.B.); (J.-Y.B.)
| | - Julie Bedoucha
- Team Cell Death and Pediatric Cancer, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008 Lyon, France; (M.-A.L.); (A.-L.D.); (P.B.); (L.A.); (I.R.); (J.B.); (J.-Y.B.)
| | - Julie Valantin
- Research Pathology Platform, Department of Translational Research and Innovation, Centre Leon Berard, F-69008 Lyon, France;
- Fondation Synergie Lyon Cancer, F-69008 Lyon, France
| | - Marie Karanian
- Department of Biopathology, Centre Leon Berard, F-69008 Lyon, France;
- Team Genetics, Epigenetics and Biology of Sarcomas, Univ Lyon, Université Claude Bernard Lyon 1, INSERM1052, CNRS5286, Cancer Research Center of Lyon, Centre Leon Berard, F-69008 Lyon, France; (F.T.); (M.K.)
| | - Raul Perret
- Department of Biopathology, Institut Bergonié, F-33000 Bordeaux, France;
| | - Olivier Gille
- Orthopedic Spinal Surgery Unit 1, Bordeaux University Hospital, F-33000 Bordeaux, France;
| | - Jean-Yves Blay
- Team Cell Death and Pediatric Cancer, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008 Lyon, France; (M.-A.L.); (A.-L.D.); (P.B.); (L.A.); (I.R.); (J.B.); (J.-Y.B.)
- Medical Oncology Department, Centre Leon Berard, F-69008 Lyon, France
| | - Aurélie Dutour
- Team Cell Death and Pediatric Cancer, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008 Lyon, France; (M.-A.L.); (A.-L.D.); (P.B.); (L.A.); (I.R.); (J.B.); (J.-Y.B.)
- Correspondence:
| |
Collapse
|
30
|
Sheppard HE, Dall’Agnese A, Park WD, Shamim MH, Dubrulle J, Johnson HL, Stossi F, Cogswell P, Sommer J, Levy J, Sharifnia T, Wawer MJ, Nabet B, Gray NS, Clemons PA, Schreiber SL, Workman P, Young RA, Lin CY. Targeted brachyury degradation disrupts a highly specific autoregulatory program controlling chordoma cell identity. CELL REPORTS MEDICINE 2021; 2:100188. [PMID: 33521702 PMCID: PMC7817874 DOI: 10.1016/j.xcrm.2020.100188] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 08/14/2020] [Accepted: 12/17/2020] [Indexed: 12/31/2022]
Abstract
Chordomas are rare spinal tumors addicted to expression of the developmental transcription factor brachyury. In chordomas, brachyury is super-enhancer associated and preferentially downregulated by pharmacologic transcriptional CDK inhibition, leading to cell death. To understand the underlying basis of this sensitivity, we dissect the brachyury transcription regulatory network and compare the consequences of brachyury degradation with transcriptional CDK inhibition. Brachyury defines the chordoma super-enhancer landscape and autoregulates through binding its super-enhancer, and its locus forms a transcriptional condensate. Transcriptional CDK inhibition and brachyury degradation disrupt brachyury autoregulation, leading to loss of its transcriptional condensate and transcriptional program. Compared with transcriptional CDK inhibition, which globally downregulates transcription, leading to cell death, brachyury degradation is much more selective, inducing senescence and sensitizing cells to anti-apoptotic inhibition. These data suggest that brachyury downregulation is a core tenet of transcriptional CDK inhibition and motivates developing strategies to target brachyury and its autoregulatory feedback loop. Brachyury defines the chordoma super-enhancer landscape Brachyury autoregulates through a transcriptional condensate CDK7/12/13i and brachyury degradation target the brachyury transcriptional condensate Brachyury degradation inhibits chordoma identity genes and induces senescence
Collapse
Affiliation(s)
- Hadley E. Sheppard
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Woojun D. Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - M. Hamza Shamim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Julien Dubrulle
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hannah L. Johnson
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fabio Stossi
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Joan Levy
- Chordoma Foundation, Durham, NC 27713, USA
| | - Tanaz Sharifnia
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | | | - Behnam Nabet
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nathanael S. Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Paul A. Clemons
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Stuart L. Schreiber
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| | - Richard A. Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charles Y. Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Therapeutic Innovation Center, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Corresponding author
| |
Collapse
|
31
|
Wong BS, Shah SR, Yankaskas CL, Bajpai VK, Wu PH, Chin D, Ifemembi B, ReFaey K, Schiapparelli P, Zheng X, Martin SS, Fan CM, Quiñones-Hinojosa A, Konstantopoulos K. A microfluidic cell-migration assay for the prediction of progression-free survival and recurrence time of patients with glioblastoma. Nat Biomed Eng 2020; 5:26-40. [PMID: 32989283 PMCID: PMC7855796 DOI: 10.1038/s41551-020-00621-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 09/01/2020] [Indexed: 01/22/2023]
Abstract
Clinical scores, molecular markers and cellular phenotypes have been used to predict the clinical outcomes of patients with glioblastoma. However, their clinical use has been hampered by confounders such as patient co-morbidities, by the tumoral heterogeneity of molecular and cellular markers, and by the complexity and cost of high-throughput single-cell analysis. Here, we show that a microfluidic assay for the quantification of cell migration and proliferation can categorize patients with glioblastoma according to progression-free survival. We quantified with a composite score the ability of primary glioblastoma cells to proliferate (via the protein biomarker Ki-67) and to squeeze through microfluidic channels, mimicking aspects of the tight perivascular conduits and white-matter tracts in brain parenchyma. The assay retrospectively categorized 28 patients according to progression-free survival (short-term or long-term) with an accuracy of 86%, predicted time to recurrence and correctly categorized five additional patients on the basis of survival prospectively. RNA sequencing of the highly motile cells revealed differentially expressed genes that correlated with poor prognosis. Our findings suggest that cell-migration and proliferation levels can predict patient-specific clinical outcomes.
Collapse
Affiliation(s)
- Bin Sheng Wong
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Sagar R Shah
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, USA
| | - Christopher L Yankaskas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Vivek K Bajpai
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Deborah Chin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Brent Ifemembi
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Karim ReFaey
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, USA
| | | | - Xiaobin Zheng
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Stuart S Martin
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | | | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA. .,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA. .,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
32
|
Gill CM, Fowkes M, Shrivastava RK. Emerging Therapeutic Targets in Chordomas: A Review of the Literature in the Genomic Era. Neurosurgery 2020; 86:E118-E123. [PMID: 31504814 DOI: 10.1093/neuros/nyz342] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/13/2019] [Indexed: 12/18/2022] Open
Abstract
Chordomas are rare primary malignant tumors of the bones that occur along the skull base, spine, and sacrum. Long-term survival and neurological outcome continue to be challenging with continued low percentages of long-term survival. Recent studies have used genome, exome, transcriptome, and proteome sequencing to assess the mutational profile of chordomas. Most notably, Brachyury, or T-protein, has been shown to be an early mutational event in chordoma evolution. Clinically actionable mutations, including in the PI3K pathway, were identified. Preliminary evidence suggests that there may be mutational differences associated with primary tumor location. In this study, we review the therapeutic landscape of chordomas and discuss emerging targets in the genomic era.
Collapse
Affiliation(s)
- Corey M Gill
- Department of Neurosurgery, Mount Sinai Medical Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mary Fowkes
- Department of Pathology, Mount Sinai Medical Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Raj K Shrivastava
- Department of Neurosurgery, Mount Sinai Medical Center, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
33
|
Bhargav AG, Mondal SK, Garcia CA, Green JJ, Quiñones‐Hinojosa A. Nanomedicine Revisited: Next Generation Therapies for Brain Cancer. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Adip G. Bhargav
- Mayo Clinic College of Medicine and Science Mayo Clinic 200 First Street SW Rochester MN 55905 USA
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| | - Sujan K. Mondal
- Department of Pathology University of Pittsburgh School of Medicine 200 Lothrop Street Pittsburgh PA 15213 USA
| | - Cesar A. Garcia
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| | - Jordan J. Green
- Departments of Biomedical Engineering, Neurosurgery, Oncology, Ophthalmology, Materials Science and Engineering, and Chemical and Biomolecular Engineering, Translational Tissue Engineering Center, Bloomberg‐Kimmel Institute for Cancer Immunotherapy, Institute for Nanobiotechnology Johns Hopkins University School of Medicine 400 N. Broadway, Smith 5017 Baltimore MD 21231 USA
| | - Alfredo Quiñones‐Hinojosa
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
- Departments of Otolaryngology‐Head and Neck Surgery/Audiology Neuroscience, Cancer Biology, and Anatomy Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| |
Collapse
|
34
|
Chen M, Wu Y, Zhang H, Li S, Zhou J, Shen J. The Roles of Embryonic Transcription Factor BRACHYURY in Tumorigenesis and Progression. Front Oncol 2020; 10:961. [PMID: 32695672 PMCID: PMC7338565 DOI: 10.3389/fonc.2020.00961] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/15/2020] [Indexed: 12/16/2022] Open
Abstract
Transcription factor brachyury, with a DNA-binding T-domain, regulates posterior mesoderm formation and notochord development through binding with highly conserved palindromic consensus sequence in a variety of organisms. The absence of brachyury expression in majority of adult normal tissues and exclusive tumor-specific expression provides the potential to be developed into a novel and promising diagnostic and therapeutic target in cancer. As a sensitive and specific marker in the diagnosis of chordoma, brachyury protein has been verified to involve in the process of carcinogenesis and progression of chordoma and several epithelial carcinomas in various studies, but the mechanism by which brachyury promotes tumor cells migrate, invade and metastasis still remains less clear. To this end, we attempt to summarize the literature on the upstream regulatory pathway of brachyury transcription and downstream controlling network by brachyury activation, all of which involve in both the embryonic development and tumor progression. We present the respective correlation of brachyury expression with tumor progression, distant metastasis, survival rate and prognosis in several types of tumor samples (including chordoma, lung cancer, breast carcinoma, and prostate cancer), and various brachyury gain-of-function and loss-of-function experiments are summarized to explore its specific role in respective tumor cell line in vitro. In addition, we also discuss another two programs relating to brachyury function: epithelial-to-mesenchymal transition (EMT) and cell cycle control, both of which implicate in the regulation of brachyury on biological behavior of tumor cells. This review will provide an overview of the function of master transcriptional factor brachyury, compare the similarities and differences of its role between embryonic development and carcinogenesis, and list the evidence on which brachyury-target therapies have the potential to help control advanced cancer populations.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of Orthopeadic Surgery, Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi, China
| | - Yinghui Wu
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| | - Hong Zhang
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| | - Suoyuan Li
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jun Shen
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
35
|
Lv GH, Zou MX, Liu FS, Zhang Y, Huang W, Ye A, Zhang QS, Wang XB, Li J, Jiang Y, She XL. Clinicopathological and Prognostic Characteristics in Extra-Axial Chordomas: An Integrative Analysis of 86 Cases and Comparison With Axial Chordomas. Neurosurgery 2020; 85:E527-E542. [PMID: 30892619 DOI: 10.1093/neuros/nyz073] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 02/14/2019] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Currently, clinical characteristics and prognostic factors of extra-axial chordoma (EAC) remain poorly understood. OBJECTIVE To characterize clinicopathological characteristics in a large EAC cohort and investigate their correlation with survival. We also attempted to compare these outcomes with axial chordoma (AC). METHODS Medline and Embase searches (from inception to February 28, 2018) were conducted to identify eligible studies as per predefined criteria. The local database at our center was also retrospectively reviewed to include additional patients. RESULTS Forty-three studies from the literature and 86 patients from our local institute were identified, resulting in a total of 86 EAC patients and 75 AC patients for analysis. Overall, EAC had similar characteristics to AC, except for having higher CAM5.2 expression, common lobular growth pattern, and better prognosis. Whereas wide surgical resection was consistently associated with favorable survival in both EAC and AC cohorts on univariate analyses, most parameters showed differential prognostic implications between the 2 groups. Significant prognostic factors for local recurrence-free survival on multivariate analysis included type of surgery in both cohorts and tumor Brachyury expression and adjuvant radiotherapy in AC cohort. Multivariate analysis of overall survival demonstrated that type of surgery, tumor Brachyury expression, and duration of symptoms were significant predictors in the AC cohort, whereas none of the analyzed parameters were predictive of overall survival for the EAC group. CONCLUSION These data suggest potentially distinct biological behaviors between EAC and AC and may provide useful information to better understand the prognostic characteristics and improve the outcome prediction of EAC patients.
Collapse
Affiliation(s)
- Guo-Hua Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Xiang Zou
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fu-Sheng Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Zhang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Huang
- Institute of Precision Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - An Ye
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qian-Shi Zhang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Bin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Ling She
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
36
|
Pinto F, Costa ÂM, Santos GC, Matsushita MM, Costa S, Silva VA, Miranda-Gonçalves V, Lopes CM, Clara CA, Becker AP, Neder L, Hajj GN, da Cunha IW, Jones C, Andrade RP, Reis RM. The T-box transcription factor brachyury behaves as a tumor suppressor in gliomas. J Pathol 2020; 251:87-99. [PMID: 32154590 DOI: 10.1002/path.5419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 12/19/2022]
Abstract
The oncogene brachyury (TBXT) is a T-box transcription factor that is overexpressed in multiple solid tumors and is associated with tumor aggressiveness and poor patient prognosis. Gliomas comprise the most common and aggressive group of brain tumors, and at the present time the functional and clinical impact of brachyury expression has not been investigated previously in these neoplasms. Brachyury expression (mRNA and protein) was assessed in normal brain (n = 67), glioma tissues (n = 716) and cell lines (n = 42), and further in silico studies were undertaken using genomic databases totaling 3115 samples. Our glioma samples were analyzed for copy number (n = 372), promoter methylation status (n = 170), and mutation status (n = 1569 tissues and n = 52 cell lines) of the brachyury gene. The prognostic impact of brachyury expression was studied in 1524 glioma patient tumors. The functional impact of brachyury on glioma proliferation, viability, and cell death was evaluated both in vitro and in vivo. Brachyury was expressed in the normal brain, and significantly downregulated in glioma tissues. Loss of brachyury was associated with tumor aggressiveness and poor survival in glioma patients. Downregulation of brachyury was not associated with gene deletion, promoter methylation, or inactivating point mutations. Brachyury re-expression in glioma cells was found to decrease glioma tumorigenesis by induction of autophagy. These data strongly suggest that brachyury behaves as a tumor suppressor gene in gliomas by modulating autophagy. It is important to note that brachyury constitutes an independent positive biomarker of patient prognosis. Our findings indicate that the role of brachyury in tumorigenesis may be tissue-dependent and demands additional investigation to guide rational interventions. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Filipe Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal.,I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
| | - Ângela M Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal.,I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Gisele C Santos
- Department of Pathology, Barretos Cancer Hospital, São Paulo, Brazil
| | | | - Sandra Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal
| | - Viviane Ao Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
| | - Vera Miranda-Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal
| | - Celeste M Lopes
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Carlos A Clara
- Neurosurgery Department, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Aline P Becker
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
| | - Luciano Neder
- Department of Pathology and Forensic Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), São Paulo, Brazil
| | - Glaucia Nm Hajj
- International Research Center, AC Camargo Cancer Center, São Paulo, Brazil
| | - Isabela W da Cunha
- Department of Molecular Diagnosis, Anatomic Pathology Department, AC Camargo Cancer Center, São Paulo, Brazil
| | - Chris Jones
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research (ICR), Sutton, UK
| | - Raquel P Andrade
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal.,Regenerative Medicine Program; Department of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal.,CBMR, Centre for Biomedical Research, Universidade do Algarve, Faro, Portugal
| | - Rui M Reis
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal.,Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
| |
Collapse
|
37
|
Brachyury: Strategies for Drugging an Intractable Cancer Therapeutic Target. Trends Cancer 2020; 6:271-273. [PMID: 32209441 DOI: 10.1016/j.trecan.2020.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 11/23/2022]
Abstract
New approaches to drug discovery are unlocking enormous therapeutic potential residing in cancer-specific molecules. Brachyury is emerging as an exciting new drug target for the rare bone cancer chordoma. Here, recent advances targeting Brachyury in chordoma are discussed and how these might open doors to the targeting of other, more common cancer types.
Collapse
|
38
|
Ozair MZ, Shah PP, Mathios D, Lim M, Moss NS. New Prospects for Molecular Targets for Chordomas. Neurosurg Clin N Am 2020; 31:289-300. [PMID: 32147018 DOI: 10.1016/j.nec.2019.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chordomas are malignant, highly recurrent tumors of the midline skeleton that arise from the remnants of the notochord. The development of systemic therapy is critically important to ultimately managing this tumor. Several ongoing trials are attempting to use molecular targeted therapies for mutated pathways in recurrent and advanced chordomas and have shown promise. In addition, immunotherapies, including brachyury-directed vaccination and checkpoint inhibition, have also been attempted with encouraging results. This article discusses the major pathways that have been implicated in the pathogenesis of chordoma with an emphasis on molecular vulnerabilities that future therapies are attempting to exploit.
Collapse
Affiliation(s)
- Mohammad Zeeshan Ozair
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Pavan Pinkesh Shah
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21287, USA
| | - Dimitrios Mathios
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21287, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21287, USA
| | - Nelson S Moss
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
39
|
Shamul JG, Shah SR, Kim J, Schiapparelli P, Vazquez-Ramos CA, Lee BJ, Patel KK, Shin A, Quinones-Hinojosa A, Green JJ. Verteporfin-Loaded Anisotropic Poly(Beta-Amino Ester)-Based Micelles Demonstrate Brain Cancer-Selective Cytotoxicity and Enhanced Pharmacokinetics. Int J Nanomedicine 2019; 14:10047-10060. [PMID: 31920302 PMCID: PMC6935022 DOI: 10.2147/ijn.s231167] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/13/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Nanomedicine can improve traditional therapies by enhancing the controlled release of drugs at targeted tissues in the body. However, there still exists disease- and therapy-specific barriers that limit the efficacy of such treatments. A major challenge in developing effective therapies for one of the most aggressive brain tumors, glioblastoma (GBM), is affecting brain cancer cells while avoiding damage to the surrounding healthy brain parenchyma. Here, we developed poly(ethylene glycol) (PEG)-poly(beta-amino ester) (PBAE) (PEG-PBAE)-based micelles encapsulating verteporfin (VP) to increase tumor-specific targeting. METHODS Biodegradable, pH-sensitive micelles of different shapes were synthesized via nanoprecipitation using two different triblock PEG-PBAE-PEG copolymers varying in their relative hydrophobicity. The anti-tumor efficacy of verteporfin loaded in these anisotropic and spherical micelles was evaluated in vitro using patient-derived primary GBM cells. RESULTS For anisotropic micelles, uptake efficiency was ~100% in GBM cells (GBM1A and JHGBM612) while only 46% in normal human astrocytes (NHA) at 15.6 nM VP (p ≤ 0.0001). Cell killing of GBM1A and JHGBM612 vs NHA was 52% and 77% vs 29%, respectively, at 24 hrs post-treatment of 125 nM VP-encapsulated in anisotropic micelles (p ≤ 0.0001), demonstrating the tumor cell-specific selectivity of VP. Moreover, anisotropic micelles showed an approximately fivefold longer half-life in blood circulation than the analogous spherical micelles in a GBM xenograft model in mice. In this model, micelle accumulation to tumors was significantly greater for anisotropic micelle-treated mice compared to spherical micelle-treated mice at both 8 hrs (~1.8-fold greater, p ≤ 0.001) and 24 hrs (~2.1-fold greater, p ≤ 0.0001). CONCLUSION Overall, this work highlights the promise of a biodegradable anisotropic micelle system to overcome multiple drug delivery challenges and enhance efficacy and safety for the treatment of brain cancer.
Collapse
Affiliation(s)
- James G Shamul
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
| | - Sagar R Shah
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL32224, USA
| | - Jayoung Kim
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
| | | | | | - Ben J Lee
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
| | - Kisha K Patel
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
| | - Alyssa Shin
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
| | | | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD21231, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer, and The Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD21231, USA
- Department of Ophthalmology, Department of Materials Science and Engineering, and Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21231, USA
| |
Collapse
|
40
|
Yan X, Li Z, Li H, Liu P, Zhao Z, Cheng S, Wang Z, Zhang Q. Inhibition Of Glycogen Synthase Kinase 3 Beta Suppresses The Growth And Survival Of Skull Base Chordoma Cells By Downregulating Brachyury Expression. Onco Targets Ther 2019; 12:9783-9791. [PMID: 31819479 PMCID: PMC6874116 DOI: 10.2147/ott.s218930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/31/2019] [Indexed: 01/06/2023] Open
Abstract
Purpose Chordomas are locally aggressive tumors arising from notochordal remnants. Brachyury, a protein coded by T-gene, is crucial for chordoma cell proliferation. The aim of this study was to evaluate the effects of glycogen synthase kinase 3 beta (GSK3β) activity on brachyury expression and on the growth and survival of skull base chordoma cells. Patients and methods In this study, 16 paraffin-embedded specimens of primary skull base chordomas were analyzed for the expression of phosphorylated GSK3β and brachyury using immunohistochemistry. The UM-Chor1 cell line derived from a clival chordoma was treated with AR-A014418 (AR), an inhibitor of GSK3β, and brachyury expression was analyzed by qRT-PCR and Western blotting. The possible mechanism by which brachyury regulates the Wnt/β-catenin signaling pathway was investigated by immunocytochemistry. The effects of AR on cell proliferation as well as sensitivity to chemotherapeutic drugs were also examined. Results The results suggested that phosphorylated GSK3β and brachyury were upregulated in chordoma tissues. The GSK3β inhibitor (AR) decreased brachyury expression and suppressed the growth and survival of the chordoma cells, possibly via regulation of the Wnt/β-catenin signaling pathway. Moreover, AR increased the sensitivity of chordoma cells to chemotherapeutic drugs in vitro. Conclusion This study provides evidence for the clinical development of the GSK3β inhibitor (AR-A014418) as a potential chemotherapeutic adjuvant for the treatment of chordoma.
Collapse
Affiliation(s)
- Xudong Yan
- Department of Otolaryngology-Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zhiyuan Li
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Hong Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Pei Liu
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Zehang Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Shan Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Zhenlin Wang
- Department of Otolaryngology-Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Qiuhang Zhang
- Department of Otolaryngology-Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
41
|
Liu L, Wang T, Yang X, Xu C, Liao Z, Wang X, Su D, Li Y, Zhou H, Qiu X, Chen Y, Huang D, Lian C, Su P. MTNR1B loss promotes chordoma recurrence by abrogating melatonin-mediated β-catenin signaling repression. J Pineal Res 2019; 67:e12588. [PMID: 31140197 DOI: 10.1111/jpi.12588] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/16/2019] [Accepted: 05/22/2019] [Indexed: 01/03/2023]
Abstract
Chordoma is an extremely rare malignant bone tumor with a high rate of relapse. While cancer stem cells (CSCs) are closely associated with tumor recurrence, which depend on its capacity to self-renew and induce chemo-/radioresistance, whether and how CSCs participate in chordoma recurrence remains unclear. The current study found that tumor cells in recurrent chordoma displayed more dedifferentiated CSC-like properties than those in corresponding primary tumor tissues. Meanwhile, MTNR1B deletion along with melatonin receptor 1B (MTNR1B) down-regulation was observed in recurrent chordoma. Further investigation revealed that activation of Gαi2 by MTNR1B upon melatonin stimulation could inhibit SRC kinase activity via recruiting CSK and SRC, increasing SRC Y530 phosphorylation, and decreasing SRC Y419 phosphorylation. This subsequently suppressed β-catenin signaling and stemness via decreasing β-catenin p-Y86/Y333/Y654. However, MTNR1B loss in chordoma mediated increased CSC properties, chemoresistance, and tumor progression by releasing melatonin's repression of β-catenin signaling. Clinically, MTNR1B deletion was found to correlate with patients' survival. Together, our study establishes a novel convergence between melatonin and β-catenin signaling pathways and reveals the significance of this cross talk in chordoma recurrence. Besides, we propose that MTNR1B is a potential biomarker for prediction of chordoma prognosis and selection of treatment options, and chordoma patients might benefit from targeting MTNR1B/Gαi2/SRC/β-catenin axis.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/deficiency
- Biomarkers, Tumor/genetics
- Bone Neoplasms/drug therapy
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Bone Neoplasms/pathology
- Cell Line, Tumor
- Chondroma/drug therapy
- Chondroma/genetics
- Chondroma/metabolism
- Chondroma/pathology
- Female
- Humans
- Melatonin/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Receptor, Melatonin, MT2/deficiency
- Receptor, Melatonin, MT2/metabolism
- Signal Transduction/drug effects
- Xenograft Model Antitumor Assays
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Lei Liu
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Center for Peripheral Nerve Tissue Engineering and Technology Research, Guangzhou, China
- Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tingting Wang
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Yang
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Caixia Xu
- Research Centre for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiheng Liao
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xudong Wang
- Department of Spine Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Deying Su
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongyong Li
- Research Centre for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hang Zhou
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xianjian Qiu
- Department of Spine Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuyu Chen
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dongsheng Huang
- Department of Spine Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chengjie Lian
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peiqiang Su
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Center for Peripheral Nerve Tissue Engineering and Technology Research, Guangzhou, China
- Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China
| |
Collapse
|
42
|
Kozielski KL, Ruiz-Valls A, Tzeng SY, Guerrero-Cázares H, Rui Y, Li Y, Vaughan HJ, Gionet-Gonzales M, Vantucci C, Kim J, Schiapparelli P, Al-Kharboosh R, Quiñones-Hinojosa A, Green JJ. Cancer-selective nanoparticles for combinatorial siRNA delivery to primary human GBM in vitro and in vivo. Biomaterials 2019; 209:79-87. [PMID: 31026613 PMCID: PMC7122460 DOI: 10.1016/j.biomaterials.2019.04.020] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/24/2019] [Accepted: 04/11/2019] [Indexed: 01/15/2023]
Abstract
Novel treatments for glioblastoma (GBM) are urgently needed, particularly those which can simultaneously target GBM cells' ability to grow and migrate. Herein, we describe a synthetic, bioreducible, biodegradable polymer that can package and deliver hundreds of siRNA molecules into a single nanoparticle, facilitating combination therapy against multiple GBM-promoting targets. We demonstrate that siRNA delivery with these polymeric nanoparticles is cancer-selective, thereby avoiding potential side effects in healthy cells. We show that we can deliver siRNAs targeting several anti-GBM genes (Robo1, YAP1, NKCC1, EGFR, and survivin) simultaneously and within the same nanoparticles. Robo1 (roundabout homolog 1) siRNA delivery by biodegradable particles was found to trigger GBM cell death, as did non-viral delivery of NKCC1, EGFR, and survivin siRNA. Most importantly, combining several anti-GBM siRNAs into a nanoparticle formulation leads to high GBM cell death, reduces GBM migration in vitro, and reduces tumor burden over time following intratumoral administration. We show that certain genes, like survivin and EGFR, are important for GBM survival, while NKCC1, is more crucial for cancer cell migration. This represents a powerful platform technology with the potential to serve as a multimodal therapeutic for cancer.
Collapse
Affiliation(s)
- Kristen L Kozielski
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA; Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, 70569, Germany
| | - Alejandro Ruiz-Valls
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Hugo Guerrero-Cázares
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yuan Rui
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Yuxin Li
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Hannah J Vaughan
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Marissa Gionet-Gonzales
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Casey Vantucci
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Jayoung Kim
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Paula Schiapparelli
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Rawan Al-Kharboosh
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Alfredo Quiñones-Hinojosa
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA; Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Materials Science and Engineering, Department of Chemical and Biomolecular Engineering, Department of Ophthalmology, The Sidney Kimmel Comprehensive Cancer, And the Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA.
| |
Collapse
|
43
|
Shah SR, Kim J, Schiapparelli P, Vazquez-Ramos CA, Martinez-Gutierrez JC, Ruiz-Valls A, Inman K, Shamul JG, Green JJ, Quinones-Hinojosa A. Verteporfin-Loaded Polymeric Microparticles for Intratumoral Treatment of Brain Cancer. Mol Pharm 2019; 16:1433-1443. [PMID: 30803231 PMCID: PMC7337228 DOI: 10.1021/acs.molpharmaceut.8b00959] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Glioblastoma (GBMs) is the most common and aggressive type of primary brain tumor in adults with dismal prognosis despite radical surgical resection coupled with chemo- and radiotherapy. Recent studies have proposed the use of small-molecule inhibitors, including verteporfin (VP), to target oncogenic networks in cancers. Here we report efficient encapsulation of water-insoluble VP in poly(lactic- co-glycolic acid) microparticles (PLGA MP) of ∼1.5 μm in diameter that allows tunable, sustained release. Treatment with naked VP and released VP from PLGA MP decreased cell viability of patient-derived primary GBM cells in vitro by ∼70%. Moreover, naked VP treatment significantly increased radiosensitivity of GBM cells, thereby enhancing overall tumor cell killing ability by nearly 85%. Our in vivo study demonstrated that two intratumoral administrations of sustained slow-releasing VP-loaded PLGA MPs separated by two weeks significantly attenuated tumor growth by ∼67% in tumor volume in a subcutaneous patient-derived GBM xenograft model over 26 d. Additionally, our in vitro data indicate broader utility of VP for treatment for other solid cancers, including chordoma, malignant meningioma, and various noncentral nervous system-derived carcinomas. Collectively, our work suggests that the use of VP-loaded PLGA MP may be an effective local therapeutic strategy for a variety of solid cancers, including unresectable and orphan tumors, which may decrease tumor burden and ultimately improve patient prognosis.
Collapse
Affiliation(s)
- Sagar R. Shah
- Department of Neurosurgery, Mayo Clinic, Jacksonville, Florida 32224, United States
- Department of Biomedical Engineering, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
- Translational Tissue Engineering Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Jayoung Kim
- Department of Biomedical Engineering, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
- Translational Tissue Engineering Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Paula Schiapparelli
- Department of Neurosurgery, Mayo Clinic, Jacksonville, Florida 32224, United States
| | | | | | - Alejandro Ruiz-Valls
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, Maryland 21231, United States
| | - Kyle Inman
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, Maryland 21231, United States
| | - James G. Shamul
- Department of Biomedical Engineering, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
- Translational Tissue Engineering Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Jordan J. Green
- Department of Biomedical Engineering, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
- Translational Tissue Engineering Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
- Department of Oncology, the Sidney Kimmel Comprehensive Cancer, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, Maryland 21231, United States
- Department of Ophthalmology, Department of Materials Science and Engineering, and Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | | |
Collapse
|
44
|
Meng T, Jin J, Jiang C, Huang R, Yin H, Song D, Cheng L. Molecular Targeted Therapy in the Treatment of Chordoma: A Systematic Review. Front Oncol 2019; 9:30. [PMID: 30775316 PMCID: PMC6367227 DOI: 10.3389/fonc.2019.00030] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/10/2019] [Indexed: 12/21/2022] Open
Abstract
Objectives: Chordoma is a rare bone malignancy that affects the spine and skull base. Treatment dilemma leads to a high rate of local relapse and distant metastases. Molecular targeted therapy (MTT) is an option for advanced chordoma, but its therapeutic efficacy and safety have not been investigated systematically. Therefore, a systematic review was conducted on studies reporting MTT regimens for chordoma. Methods: Clinical trials, case series and case reports on chordoma MTT were identified using MEDLINE, Cochrane library and EMBASE, and systematically reviewed. Data on clinical outcomes, such as median overall survival, progression-free survival, response rate and adverse events (AEs) were extracted and analyzed. Results: Thirty-three eligible studies were selected for the systematic review, which indicated that imatinib and erlotinib were the most frequently used molecular targeted inhibitors (MTIs) for chordoma. For PDGFR-positive and/or EGFR-positive chordoma, clinical benefits were achieved with acceptable AEs. Monotherapy is preferred as the first-line of treatment, and combined drug therapy as the second-line treatment. In addition, the brachyury vaccine has shown promising results. Conclusions: The selection of MTIs for patients with advanced or relapsed chordoma should be based on gene mutation screening and immunohistochemistry (IHC). Monotherapy of TKIs is recommended as the first-line management, and combination therapy (two TKIs or TKI plus mTOR inhibitor) may be the choice for drug-resistant chordoma. Brachyury vaccine is a promising therapeutic strategy and requires more clinical trials to evaluate its safety and efficacy.
Collapse
Affiliation(s)
- Tong Meng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Shanghai Bone Tumor Institution, Shanghai, China.,Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiali Jin
- Department of Central Laboratory, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Cong Jiang
- Beth Israel Deaconess Medical Center, BIDMC Cancer Center, Harvard Medical School, Cancer Research Institute, Boston, MA, United States
| | - Runzhi Huang
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Huabin Yin
- Shanghai Bone Tumor Institution, Shanghai, China.,Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dianwen Song
- Shanghai Bone Tumor Institution, Shanghai, China.,Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Tongji University, Shanghai, China
| |
Collapse
|
45
|
Sharifnia T, Wawer MJ, Chen T, Huang QY, Weir BA, Sizemore A, Lawlor MA, Goodale A, Cowley GS, Vazquez F, Ott CJ, Francis JM, Sassi S, Cogswell P, Sheppard HE, Zhang T, Gray NS, Clarke PA, Blagg J, Workman P, Sommer J, Hornicek F, Root DE, Hahn WC, Bradner JE, Wong KK, Clemons PA, Lin CY, Kotz JD, Schreiber SL. Small-molecule targeting of brachyury transcription factor addiction in chordoma. Nat Med 2019; 25:292-300. [PMID: 30664779 PMCID: PMC6633917 DOI: 10.1038/s41591-018-0312-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022]
Abstract
Chordoma is a primary bone cancer with no approved therapy1. The identification of therapeutic targets in this disease has been challenging due to the infrequent occurrence of clinically actionable somatic mutations in chordoma tumors2,3. Here we describe the discovery of therapeutically targetable chordoma dependencies via genome-scale CRISPR-Cas9 screening and focused small-molecule sensitivity profiling. These systematic approaches reveal that the developmental transcription factor T (brachyury; TBXT) is the top selectively essential gene in chordoma, and that transcriptional cyclin-dependent kinase (CDK) inhibitors targeting CDK7/12/13 and CDK9 potently suppress chordoma cell proliferation. In other cancer types, transcriptional CDK inhibitors have been observed to downregulate highly expressed, enhancer-associated oncogenic transcription factors4,5. In chordoma, we find that T is associated with a 1.5-Mb region containing 'super-enhancers' and is the most highly expressed super-enhancer-associated transcription factor. Notably, transcriptional CDK inhibition leads to preferential and concentration-dependent downregulation of cellular brachyury protein levels in all models tested. In vivo, CDK7/12/13-inhibitor treatment substantially reduces tumor growth. Together, these data demonstrate small-molecule targeting of brachyury transcription factor addiction in chordoma, identify a mechanism of T gene regulation that underlies this therapeutic strategy, and provide a blueprint for applying systematic genetic and chemical screening approaches to discover vulnerabilities in genomically quiet cancers.
Collapse
Affiliation(s)
| | | | - Ting Chen
- New York University Langone Medical Center, New York, NY, USA
| | - Qing-Yuan Huang
- New York University Langone Medical Center, New York, NY, USA
- Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Barbara A Weir
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Janssen R&D, Cambridge, MA, USA
| | - Ann Sizemore
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew A Lawlor
- Dana-Farber Cancer Institute, Boston, MA, USA
- Massachusetts General Hospital, Charlestown, MA, USA
| | - Amy Goodale
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Glenn S Cowley
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Janssen R&D, Spring House, PA, USA
| | | | - Christopher J Ott
- Dana-Farber Cancer Institute, Boston, MA, USA
- Massachusetts General Hospital, Charlestown, MA, USA
| | - Joshua M Francis
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Gritstone Oncology, Cambridge, MA, USA
| | - Slim Sassi
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Paul A Clarke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | | | - Francis Hornicek
- Massachusetts General Hospital, Boston, MA, USA
- UCLA Medical Center, Santa Monica, CA, USA
| | - David E Root
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - James E Bradner
- Dana-Farber Cancer Institute, Boston, MA, USA
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kwok K Wong
- New York University Langone Medical Center, New York, NY, USA
| | | | | | - Joanne D Kotz
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Jnana Therapeutics, Boston, MA, USA.
| | - Stuart L Schreiber
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
46
|
Characterization of a Clival Chordoma Xenograft Model Reveals Tumor Genomic Instability. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2902-2911. [DOI: 10.1016/j.ajpath.2018.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 07/28/2018] [Accepted: 08/02/2018] [Indexed: 01/24/2023]
|
47
|
Shah SR, Quinones-Hinojosa A, Xia S. Advances in Brain Cancer: Creating Monoallelic Single Point Mutation in IDH1 by Single Base Editing. JOURNAL OF ONCOLOGY RESEARCH AND THERAPY 2018; 5:166. [PMID: 31328182 PMCID: PMC6641564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mutations in the Isocitrate Dehydrogenase 1 (IDH1) gene occur in 70% of grade II and grade III gliomas, 10% of acute myeloid leukemia, as well as cholangiocarcinomas, melanomas, and chondrosarcomas. Numerous mechanisms have been proposed to illustrate the biological function of mutant IDH1. Most functional studies of mutant IDH1 have been conducted in exogenous overexpression systems with the IDH1 wild type background. This mini-review comments on recent publication by Wei et al, in which a highly efficient "single base editing" approach was employed to generate monoallelic IDH1 R132H mutation without the induction of a double strand break in the IDH1 gene.
Collapse
Affiliation(s)
- Sagar R. Shah
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Shuli Xia
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
48
|
Gao L, Cheng XL, Cao H. LncRNA THOR attenuates cisplatin sensitivity of nasopharyngeal carcinoma cells via enhancing cells stemness. Biochimie 2018; 152:63-72. [DOI: 10.1016/j.biochi.2018.06.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/20/2018] [Indexed: 12/20/2022]
|
49
|
Mechanoregulation and pathology of YAP/TAZ via Hippo and non-Hippo mechanisms. Clin Transl Med 2018; 7:23. [PMID: 30101371 PMCID: PMC6087706 DOI: 10.1186/s40169-018-0202-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023] Open
Abstract
Yes-associated protein (YAP) and its paralog WW domain containing transcription regulator 1 (TAZ) are important regulators of multiple cellular functions such as proliferation, differentiation, and survival. On the tissue level, YAP/TAZ are essential for embryonic development, organ size control and regeneration, while their deregulation leads to carcinogenesis or other diseases. As an underlying principle for YAP/TAZ-mediated regulation of biological functions, a growing body of research reveals that YAP/TAZ play a central role in delivering information of mechanical environments surrounding cells to the nucleus transcriptional machinery. In this review, we discuss mechanical cue-dependent regulatory mechanisms for YAP/TAZ functions, as well as their clinical significance in cancer progression and treatment.
Collapse
|
50
|
Kim J, Shamul JG, Shah SR, Shin A, Lee BJ, Quinones-Hinojosa A, Green JJ. Verteporfin-Loaded Poly(ethylene glycol)-Poly(beta-amino ester)-Poly(ethylene glycol) Triblock Micelles for Cancer Therapy. Biomacromolecules 2018; 19:3361-3370. [PMID: 29940101 DOI: 10.1021/acs.biomac.8b00640] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Amphiphilic polymers can be used to form micelles to deliver water-insoluble drugs. A biodegradable poly(ethylene glycol) (PEG)-poly(beta-amino ester) (PBAE)-PEG triblock copolymer was developed that is useful for drug delivery. It was shown to successfully encapsulate and pH-dependently release a water-insoluble, small molecule anticancer drug, verteporfin. PEG-PBAE-PEG micelle morphology was also controlled through variations to the hydrophobicity of the central PBAE block of the copolymer in order to evade macrophage uptake. Spherical micelles were 50 nm in diameter, while filamentous micelles were 31 nm in width with an average aspect ratio of 20. When delivered to RAW 264.7 mouse macrophages, filamentous micelles exhibited a 89% drop in cellular uptake percentage and a 5.6-fold drop in normalized geometric mean cellular uptake compared to spherical micelles. This demonstrates the potential of high-aspect-ratio, anisotropically shaped PEG-PBAE-PEG micelles to evade macrophage-mediated clearance. Both spherical and filamentous micelles also showed therapeutic efficacy in human triple-negative breast cancer and small cell lung cancer cells without requiring photodynamic therapy to achieve an anticancer effect. Both spherical and filamentous micelles were more effective in killing lung cancer cells than breast cancer cells at equivalent verteporfin concentrations, while spherical micelles were shown to be more effective than filamentous micelles against both cancer cells. Spherical and filamentous micelles at 5 and 10 μM respective verteporfin concentration resulted in 100% cell killing of lung cancer cells, but both micelles required a higher verteporfin concentration of 20 μM to kill breast cancer cells at the levels of 80% and 50% respectively. This work demonstrates the potential of PEG-PBAE-PEG as a biodegradable, anisotropic drug delivery system as well as the in vitro use of verteporfin-loaded micelles for cancer therapy.
Collapse
Affiliation(s)
| | | | - Sagar R Shah
- Department of Neurosurgery , Mayo Clinic , Jacksonville , Florida 32224 , United States
| | | | | | | | - Jordan J Green
- Department of Ophthalmology, Department of Neurosurgery, Department of Materials Science and Engineering, and Department of Chemical and Biomolecular Engineering , Johns Hopkins University , Baltimore , Maryland 21218 , United States
| |
Collapse
|