1
|
Hu X, Chai J, Zhang B, Hu C. Primary pulmonary alveolar soft part sarcoma with ASPSCR1-TFE3 gene fusion: Case report and literature review. Medicine (Baltimore) 2024; 103:e40249. [PMID: 39495972 PMCID: PMC11537591 DOI: 10.1097/md.0000000000040249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/08/2024] [Indexed: 11/06/2024] Open
Abstract
RATIONALE Primary pulmonary alveolar soft part sarcoma (ASPS) is an extremely rare disease characterized by a specific genetic abnormality - the ASPSCR1-TFE3 gene fusion. PATIENT CONCERNS This study presented a 27-year-old male patient who experienced persistent chest tightness for over 6 months. DIAGNOSES The computed tomography (CT) scan and enhanced CT scan revealed a mass in the medial segment of the right middle lobe of his lung. The patients then underwent further diagnosis. Pathological examination showed the tumor to be consisting of polygonal cells with abundant eosinophilic or transparent cytoplasm arranged in nests. Next-generation sequencing reported ASPSCR1-TFE3 gene fusion, confirming the final diagnosis of primary pulmonary ASPS. Regular follow-ups of 12 months showed no signs of tumor recurrence. INTERVENTIONS The patients underwent the medial segment resection of the right middle lobe for treatment. OUTCOMES A CT examination 3 months after the operation showed that the patient had improved. The last review showed no recurrence or metastasis. LESSONS This case report highlights the importance of detailed diagnosis, prompt treatment, and close monitoring of patients with ASPS.
Collapse
Affiliation(s)
- Xijian Hu
- Graduate School, Shanxi Medical University, Taiyuan, China
| | - Jing Chai
- Graduate School, Shanxi Medical University, Taiyuan, China
| | - Bin Zhang
- Graduate School, Shanxi Medical University, Taiyuan, China
| | - Chengguang Hu
- Department of Thoracic Surgery, Shanxi Cancer Hospital, Taiyuan, China
| |
Collapse
|
2
|
Ishiguro N, Nakagawa M. ASPSCR1::TFE3-mediated upregulation of insulin receptor substrate 2 (IRS-2) activates PI3K/AKT signaling and promotes malignant phenotype. Int J Biochem Cell Biol 2024; 176:106676. [PMID: 39419345 DOI: 10.1016/j.biocel.2024.106676] [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: 05/24/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024]
Abstract
The ASPSCR1::TFE3 fusion gene, resulting from chromosomal translocation, is detected in alveolar soft part sarcoma (ASPS) and a subset of renal cell carcinomas (RCC). The ASPSCR1::TFE3 oncoprotein, functioning as an aberrant transcription factor, contributes to tumor development and progression by inappropriately upregulating target genes. Here, we identified insulin receptor substrate 2 (IRS-2), a cytoplasmic adaptor protein, as a novel transcriptional target of ASPSCR1::TFE3. Ectopic expression of ASPSCR1::TFE3 led to increased IRS-2 mRNA and protein levels. Chromatin immunoprecipitation and luciferase assays demonstrated that ASPSCR1::TFE3 bound to the IRS-2 promoter region and enhanced its transcription. Moreover, IRS-2 was highly expressed in the ASPSCR1::TFE3-positive RCC cell line FU-UR1, while small interfering RNA-mediated depletion of ASPSCR1::TFE3 markedly decreased IRS-2 mRNA and protein levels. Functionally, IRS-2 knockdown attenuated activation of the PI3K/AKT pathway and reduced proliferation, migration, invasion, adhesion, and clonogenicity in FU-UR1 cells. Pharmacological inhibition of IRS-2 also reduced AKT activation as well as cell viability, clonogenicity, migration, invasion, and adhesion. These findings suggest that IRS-2, regulated by ASPSCR1::TFE3, promotes tumor progression by activating PI3K/AKT signaling and enhancing the malignant phenotype.
Collapse
MESH Headings
- Humans
- Insulin Receptor Substrate Proteins/metabolism
- Insulin Receptor Substrate Proteins/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Signal Transduction
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Cell Line, Tumor
- Up-Regulation
- Gene Expression Regulation, Neoplastic
- Cell Movement/genetics
- Cell Proliferation
- Phenotype
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/metabolism
- Kidney Neoplasms/pathology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Sarcoma, Alveolar Soft Part/pathology
- Sarcoma, Alveolar Soft Part/genetics
- Sarcoma, Alveolar Soft Part/metabolism
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Naoko Ishiguro
- Department of Pathobiological Science and Technology, Faculty of Medicine, Tottori University, Yonago, Japan.
| | - Mayumi Nakagawa
- Department of Pathobiological Science and Technology, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
3
|
Li J, Huang K, McBride F, Sadagopan A, Gallant DS, Thakur M, Khanna P, Li B, Ge M, Weiss CN, Achom M, Xu Q, Huang K, Ryback BA, Gui M, Bar-Peled L, Viswanathan SR. TFE3 fusions direct an oncogenic transcriptional program that drives OXPHOS and unveils vulnerabilities in translocation renal cell carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607311. [PMID: 39149323 PMCID: PMC11326252 DOI: 10.1101/2024.08.09.607311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Translocation renal cell carcinoma (tRCC) is an aggressive subtype of kidney cancer driven by TFE3 gene fusions, which act via poorly characterized downstream mechanisms. Here we report that TFE3 fusions transcriptionally rewire tRCCs toward oxidative phosphorylation (OXPHOS), contrasting with the highly glycolytic metabolism of most other renal cancers. This TFE3 fusion-driven OXPHOS program, together with heightened glutathione levels found in renal cancers, renders tRCCs sensitive to reductive stress - a metabolic stress state induced by an imbalance of reducing equivalents. Genome-scale CRISPR screening identifies tRCC-selective vulnerabilities linked to this metabolic state, including EGLN1, which hydroxylates HIF-1α and targets it for proteolysis. Inhibition of EGLN1 compromises tRCC cell growth by stabilizing HIF-1a and promoting metabolic reprogramming away from OXPHOS, thus representing a vulnerability to OXPHOS-dependent tRCC cells. Our study defines a distinctive tRCC-essential metabolic program driven by TFE3 fusions and nominates EGLN1 inhibition as a therapeutic strategy to counteract fusion-induced metabolic rewiring.
Collapse
Affiliation(s)
- Jiao Li
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
- Department of Medicine, Harvard Medical School; Boston, MA, USA
| | - Kaimeng Huang
- Department of Medicine, Harvard Medical School; Boston, MA, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fiona McBride
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Ananthan Sadagopan
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Daniel. S Gallant
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Meha Thakur
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Prateek Khanna
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Bingchen Li
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
- Department of Medicine, Harvard Medical School; Boston, MA, USA
| | - Maolin Ge
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Cary N. Weiss
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mingkee Achom
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
- Department of Medicine, Harvard Medical School; Boston, MA, USA
| | - Qingru Xu
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
| | - Kun Huang
- Molecular Imaging Core and Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Birgitta A. Ryback
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Miao Gui
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, Zhejiang, China; Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Liron Bar-Peled
- Department of Medicine, Harvard Medical School; Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Srinivas R. Viswanathan
- Department of Medical Oncology, Dana-Farber Cancer Institute; Boston, MA, USA
- Department of Medicine, Harvard Medical School; Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Department of Medicine, Brigham and Women’s Hospital; Boston, MA, USA
| |
Collapse
|
4
|
Sicinska E, Kola VSR, Kerfoot JA, Taddei ML, Al-Ibraheemi A, Hsieh YH, Church AJ, Landesman-Bollag E, Landesman Y, Hemming ML. ASPSCR1::TFE3 Drives Alveolar Soft Part Sarcoma by Inducing Targetable Transcriptional Programs. Cancer Res 2024; 84:2247-2264. [PMID: 38657118 PMCID: PMC11250573 DOI: 10.1158/0008-5472.can-23-2115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 02/09/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Alveolar soft part sarcoma (ASPS) is a rare mesenchymal malignancy driven by the ASPSCR1::TFE3 fusion. A better understanding of the mechanisms by which this oncogenic transcriptional regulator drives cancer growth is needed to help identify potential therapeutic targets. In this study, we characterized the transcriptional and chromatin landscapes of ASPS tumors and preclinical models, identifying the essential role of ASPSCR1::TFE3 in tumor cell viability by regulating core transcriptional programs involved in cell proliferation, angiogenesis, and mitochondrial biology. ASPSCR1::TFE3 directly interacted with key epigenetic regulators at enhancers and promoters to support ASPS-associated transcription. Among the effector programs driven by ASPSCR1::TFE3, cell proliferation was driven by high levels of cyclin D1 expression. Disruption of cyclin D1/CDK4 signaling led to a loss of ASPS proliferative capacity, and combined inhibition of CDK4/6 and angiogenesis halted tumor growth in xenografts. These results define the ASPS oncogenic program, reveal mechanisms by which ASPSCR1::TFE3 controls tumor biology, and identify a strategy for therapeutically targeting tumor cell-intrinsic vulnerabilities. Significance: The ASPSCR1::TFE3 fusion propels the growth of alveolar soft part sarcoma by activating transcriptional programs that regulate proliferation, angiogenesis, mitochondrial biogenesis, and differentiation and can be therapeutically targeted to improve treatment.
Collapse
MESH Headings
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Sarcoma, Alveolar Soft Part/genetics
- Sarcoma, Alveolar Soft Part/pathology
- Sarcoma, Alveolar Soft Part/metabolism
- Humans
- Animals
- Mice
- Cell Proliferation/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Gene Expression Regulation, Neoplastic
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cyclin-Dependent Kinase 4/genetics
- Cyclin-Dependent Kinase 4/metabolism
- Cyclin-Dependent Kinase 4/antagonists & inhibitors
- Female
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Ewa Sicinska
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Vijaya S R Kola
- Division of Hematology and Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts
| | - Joseph A Kerfoot
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Madeleine L Taddei
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alyaa Al-Ibraheemi
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yi-Hsuan Hsieh
- Division of Hematology and Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts
| | - Alanna J Church
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Esther Landesman-Bollag
- Department of Medicine, Section of Hematology and Oncology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts
| | - Yosef Landesman
- Cure Alveolar Soft Part Sarcoma International, Brookline, Massachusetts
| | - Matthew L Hemming
- Division of Hematology and Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts
| |
Collapse
|
5
|
Stanton BZ, Pomella S. Epigenetic determinants of fusion-driven sarcomas: paradigms and challenges. Front Cell Dev Biol 2024; 12:1416946. [PMID: 38946804 PMCID: PMC11211607 DOI: 10.3389/fcell.2024.1416946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/14/2024] [Indexed: 07/02/2024] Open
Abstract
We describe exciting recent advances in fusion-driven sarcoma etiology, from an epigenetics perspective. By exploring the current state of the field, we identify and describe the central mechanisms that determine sarcomagenesis. Further, we discuss seminal studies in translational genomics, which enabled epigenetic characterization of fusion-driven sarcomas. Important context for epigenetic mechanisms include, but are not limited to, cell cycle and metabolism, core regulatory circuitry, 3-dimensional chromatin architectural dysregulation, integration with ATP-dependent chromatin remodeling, and translational animal modeling. Paradoxically, while the genetic requirements for oncogenic transformation are highly specific for the fusion partners, the epigenetic mechanisms we as a community have uncovered are categorically very broad. This dichotomy prompts the question of whether the investigation of rare disease epigenomics should prioritize studying individual cell populations, thereby examining whether the mechanisms of chromatin dysregulation are specific to a particular tumor. We review recent advances focusing on rhabdomyosarcoma, synovial sarcoma, alveolar soft part sarcoma, clear cell sarcoma, undifferentiated round cell sarcoma, Ewing sarcoma, myxoid/round liposarcoma, epithelioid hemangioendothelioma and desmoplastic round cell tumor. The growing number of groundbreaking discoveries in the field, motivated us to anticipate further exciting advances in the area of mechanistic epigenomics and direct targeting of fusion transcription factors in the years ahead.
Collapse
Affiliation(s)
- Benjamin Z. Stanton
- Nationwide Children’s Hospital, Center for Childhood Cancer Research, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
- Department of Biological Chemistry and Pharmacology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Silvia Pomella
- Department of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
6
|
Kucinski JP, Calderon D, Kendall GC. Biological and therapeutic insights from animal modeling of fusion-driven pediatric soft tissue sarcomas. Dis Model Mech 2024; 17:dmm050704. [PMID: 38916046 PMCID: PMC11225592 DOI: 10.1242/dmm.050704] [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] [Indexed: 06/26/2024] Open
Abstract
Survival for children with cancer has primarily improved over the past decades due to refinements in surgery, radiation and chemotherapy. Although these general therapies are sometimes curative, the cancer often recurs, resulting in poor outcomes for patients. Fusion-driven pediatric soft tissue sarcomas are genetically defined by chromosomal translocations that create a chimeric oncogene. This distinctive, almost 'monogenic', genetic feature supports the generation of animal models to study the respective diseases in vivo. This Review focuses on a subset of fusion-driven pediatric soft tissue sarcomas that have transgenic animal tumor models, which includes fusion-positive and infantile rhabdomyosarcoma, synovial sarcoma, undifferentiated small round cell sarcoma, alveolar soft part sarcoma and clear cell sarcoma. Studies using the animal models of these sarcomas have highlighted that pediatric cancers require a specific cellular state or developmental stage to drive tumorigenesis, as the fusion oncogenes cause different outcomes depending on their lineage and timing of expression. Therefore, understanding these context-specific activities could identify targetable activities and mechanisms critical for tumorigenesis. Broadly, these cancers show dependencies on chromatin regulators to support oncogenic gene expression and co-opting of developmental pathways. Comparative analyses across lineages and tumor models will further provide biological and therapeutic insights to improve outcomes for these children.
Collapse
Affiliation(s)
- Jack P. Kucinski
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology PhD Program, The Ohio State University, Columbus, OH 43210, USA
| | - Delia Calderon
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology PhD Program, The Ohio State University, Columbus, OH 43210, USA
| | - Genevieve C. Kendall
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology PhD Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43215, USA
| |
Collapse
|
7
|
Wachtel M, Surdez D, Grünewald TGP, Schäfer BW. Functional Classification of Fusion Proteins in Sarcoma. Cancers (Basel) 2024; 16:1355. [PMID: 38611033 PMCID: PMC11010897 DOI: 10.3390/cancers16071355] [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: 02/29/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Sarcomas comprise a heterogeneous group of malignant tumors of mesenchymal origin. More than 80 entities are associated with different mesenchymal lineages. Sarcomas with fibroblastic, muscle, bone, vascular, adipocytic, and other characteristics are distinguished. Nearly half of all entities contain specific chromosomal translocations that give rise to fusion proteins. These are mostly pathognomonic, and their detection by various molecular techniques supports histopathologic classification. Moreover, the fusion proteins act as oncogenic drivers, and their blockade represents a promising therapeutic approach. This review summarizes the current knowledge on fusion proteins in sarcoma. We categorize the different fusion proteins into functional classes, including kinases, epigenetic regulators, and transcription factors, and describe their mechanisms of action. Interestingly, while fusion proteins acting as transcription factors are found in all mesenchymal lineages, the others have a more restricted pattern. Most kinase-driven sarcomas belong to the fibroblastic/myofibroblastic lineage. Fusion proteins with an epigenetic function are mainly associated with sarcomas of unclear differentiation, suggesting that epigenetic dysregulation leads to a major change in cell identity. Comparison of mechanisms of action reveals recurrent functional modes, including antagonism of Polycomb activity by fusion proteins with epigenetic activity and recruitment of histone acetyltransferases by fusion transcription factors of the myogenic lineage. Finally, based on their biology, we describe potential approaches to block the activity of fusion proteins for therapeutic intervention. Overall, our work highlights differences as well as similarities in the biology of fusion proteins from different sarcomas and provides the basis for a functional classification.
Collapse
Affiliation(s)
- Marco Wachtel
- Department of Oncology and Children’s Research Center, University Children’s Hospital, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland
| | - Didier Surdez
- Balgrist University Hospital, Faculty of Medicine, University of Zurich (UZH), CH-8008 Zurich, Switzerland
| | - Thomas G. P. Grünewald
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Hopp-Children’s Cancer Center (KiTZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Beat W. Schäfer
- Department of Oncology and Children’s Research Center, University Children’s Hospital, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland
| |
Collapse
|
8
|
Pozner A, Li L, Verma SP, Wang S, Barrott JJ, Nelson ML, Yu JSE, Negri GL, Colborne S, Hughes CS, Zhu JF, Lambert SL, Carroll LS, Smith-Fry K, Stewart MG, Kannan S, Jensen B, John CM, Sikdar S, Liu H, Dang NH, Bourdage J, Li J, Vahrenkamp JM, Mortenson KL, Groundland JS, Wustrack R, Senger DL, Zemp FJ, Mahoney DJ, Gertz J, Zhang X, Lazar AJ, Hirst M, Morin GB, Nielsen TO, Shen PS, Jones KB. ASPSCR1-TFE3 reprograms transcription by organizing enhancer loops around hexameric VCP/p97. Nat Commun 2024; 15:1165. [PMID: 38326311 PMCID: PMC10850509 DOI: 10.1038/s41467-024-45280-5] [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: 02/21/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024] Open
Abstract
The t(X,17) chromosomal translocation, generating the ASPSCR1::TFE3 fusion oncoprotein, is the singular genetic driver of alveolar soft part sarcoma (ASPS) and some Xp11-rearranged renal cell carcinomas (RCCs), frustrating efforts to identify therapeutic targets for these rare cancers. Here, proteomic analysis identifies VCP/p97, an AAA+ ATPase with known segregase function, as strongly enriched in co-immunoprecipitated nuclear complexes with ASPSCR1::TFE3. We demonstrate that VCP is a likely obligate co-factor of ASPSCR1::TFE3, one of the only such fusion oncoprotein co-factors identified in cancer biology. Specifically, VCP co-distributes with ASPSCR1::TFE3 across chromatin in association with enhancers genome-wide. VCP presence, its hexameric assembly, and its enzymatic function orchestrate the oncogenic transcriptional signature of ASPSCR1::TFE3, by facilitating assembly of higher-order chromatin conformation structures demonstrated by HiChIP. Finally, ASPSCR1::TFE3 and VCP demonstrate co-dependence for cancer cell proliferation and tumorigenesis in vitro and in ASPS and RCC mouse models, underscoring VCP's potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Amir Pozner
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Li Li
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Shiv Prakash Verma
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Shuxin Wang
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Jared J Barrott
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Mary L Nelson
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jamie S E Yu
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Gian Luca Negri
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Shane Colborne
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | | | - Ju-Fen Zhu
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Sydney L Lambert
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Lara S Carroll
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kyllie Smith-Fry
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Michael G Stewart
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Sarmishta Kannan
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Bodrie Jensen
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Cini M John
- Department of Microbiology, Immunology and Infectious Disease, University of Calgary, Calgary, AB, Canada
| | - Saif Sikdar
- Department of Microbiology, Immunology and Infectious Disease, University of Calgary, Calgary, AB, Canada
| | - Hongrui Liu
- Department of Microbiology, Immunology and Infectious Disease, University of Calgary, Calgary, AB, Canada
| | - Ngoc Ha Dang
- Department of Oncology, Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer Bourdage
- Department of Oncology, Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jinxiu Li
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jeffery M Vahrenkamp
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Katelyn L Mortenson
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - John S Groundland
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Rosanna Wustrack
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Donna L Senger
- Department of Oncology, Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Oncology, McGill University and Lady Davis Institute for Medical Research, Montreal, QC, Canada
| | - Franz J Zemp
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Douglas J Mahoney
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jason Gertz
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Xiaoyang Zhang
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Alexander J Lazar
- Departments of Anatomic Pathology, Translational Molecular Pathology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martin Hirst
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
- Department of Microbiology and Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Gregg B Morin
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Torsten O Nielsen
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Peter S Shen
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Kevin B Jones
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
9
|
Spinnato P, Papalexis N, Colangeli M, Miceli M, Crombé A, Parmeggiani A, Palmerini E, Righi A, Bianchi G. Imaging Features of Alveolar Soft Part Sarcoma: Single Institution Experience and Literature Review. Clin Pract 2023; 13:1369-1382. [PMID: 37987424 PMCID: PMC10660714 DOI: 10.3390/clinpract13060123] [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: 08/31/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023] Open
Abstract
Alveolar soft part sarcoma (ASPS) is an extremely rare and aggressive soft-tissue sarcoma (STS) subtype with poor prognosis and limited response to radiation therapy and chemotherapy. Prompt recognition and referral to sarcoma centers for appropriate management are crucial for patients' survival. The purpose of this study was to report ASPS pre-treatment imaging features and to examine the existing literature on this topic. Twelve patients (7 women, 5 men-mean age 27.1 ± 10.7 years) were included from our single-center experience. Ultrasonography (US), computed tomography (CT), and magnetic resonance imaging (MRI) available were reviewed according to an analysis grid incorporating features from the latest research on STS. Clinical, histological, and outcome data were collected. MRI was available in 10 patients (83.3%), US in 7 patients (58.3%), and CT in 3 patients (25%). Mean longest tumor diameter was 7.6 ± 2.9 cm, and all tumors were deeply seated. Large peritumoral feeding vessels were systematically found and identified on ultrasonography (7/7), MRI (10/10), and CT (3/3). US revealed a well-defined heterogeneous hypoechoic pattern, with abundant flow signals in all patients (7/7). In all patients, MRI showed mildly high signal intensity (SI) on T1-WI and high SI on T2-WI and peritumoral edema. Moreover, flow-voids (due to arteriosus high-flow) into the peritumoral/intratumoral feeding vessels were detected in the MRI fluid-sensitive sequences of all patients. At baseline, whole-body contrast-enhanced CT revealed metastases in 8/12 (66.7%) patients. A pre-treatment longest diameter > 5 cm was significantly associated with distant metastases at diagnosis (p = 0.01). A maximum diameter > 5 cm represents a risk of metastatic disease at diagnosis (odds ratio = 45.0000 (95% CI: 1.4908-1358.3585), p = 0.0285). In the comprehensive literature review, we found 14 articles (case series or original research) focusing on ASPS imaging, with a total of 151 patients included. Merging our experience with the data from the existing literature, we conclude that the hallmark of ASPS imaging at presentation are the following characteristics: deep location, a slight hyperintense MRI SI on T1-WI and a hyperintense SI on T2-WI, numerous MRI flow voids, high internal vascularization, and large peritumoral feeding vessels.
Collapse
Affiliation(s)
- Paolo Spinnato
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Nicolas Papalexis
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Marco Colangeli
- Department of Orthopaedic Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Marco Miceli
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Amandine Crombé
- Department of Musculoskeletal Imaging, Pellegrin University Hospital, University of Bordeaux, 33000 Bordeaux, France
| | - Anna Parmeggiani
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Emanuela Palmerini
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Alberto Righi
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Giuseppe Bianchi
- Orthopaedic Oncology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| |
Collapse
|
10
|
Pozner A, Verma SP, Li L, Wang S, Barrott JJ, Nelson ML, Yu JSE, Negri GL, Colborne S, Hughes CS, Zhu JF, Lambert SL, Carroll LS, Smith-Fry K, Stewart MG, Kannan S, Jensen B, Mortenson KL, John C, Sikdar S, Liu H, Dang NH, Bourdage J, Li J, Vahrenkamp JM, Groundland JS, Wustrack R, Senger DL, Zemp FJ, Mahoney DJ, Gertz J, Zhang X, Lazar AJ, Hirst M, Morin GB, Nielsen TO, Shen PS, Jones KB. ASPSCR1-TFE3 reprograms transcription by organizing enhancer loops around hexameric VCP/p97. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560242. [PMID: 37873234 PMCID: PMC10592841 DOI: 10.1101/2023.09.29.560242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The t(X,17) chromosomal translocation, generating the ASPSCR1-TFE3 fusion oncoprotein, is the singular genetic driver of alveolar soft part sarcoma (ASPS) and some Xp11-rearranged renal cell carcinomas (RCC), frustrating efforts to identify therapeutic targets for these rare cancers. Proteomic analysis showed that VCP/p97, an AAA+ ATPase with known segregase function, was strongly enriched in co-immunoprecipitated nuclear complexes with ASPSCR1-TFE3. We demonstrate that VCP is a likely obligate co-factor of ASPSCR1-TFE3, one of the only such fusion oncoprotein co-factors identified in cancer biology. Specifically, VCP co-distributed with ASPSCR1-TFE3 across chromatin in association with enhancers genome-wide. VCP presence, its hexameric assembly, and its enzymatic function orchestrated the oncogenic transcriptional signature of ASPSCR1-TFE3, by facilitating assembly of higher-order chromatin conformation structures as demonstrated by HiChIP. Finally, ASPSCR1-TFE3 and VCP demonstrated co-dependence for cancer cell proliferation and tumorigenesis in vitro and in ASPS and RCC mouse models, underscoring VCP's potential as a novel therapeutic target.
Collapse
|
11
|
Chen AP, Sharon E, O'Sullivan-Coyne G, Moore N, Foster JC, Hu JS, Van Tine BA, Conley AP, Read WL, Riedel RF, Burgess MA, Glod J, Davis EJ, Merriam P, Naqash AR, Fino KK, Miller BL, Wilsker DF, Begum A, Ferry-Galow KV, Deshpande HA, Schwartz GK, Ladle BH, Okuno SH, Beck JC, Chen JL, Takebe N, Fogli LK, Rosenberger CL, Parchment RE, Doroshow JH. Atezolizumab for Advanced Alveolar Soft Part Sarcoma. N Engl J Med 2023; 389:911-921. [PMID: 37672694 PMCID: PMC10729808 DOI: 10.1056/nejmoa2303383] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
BACKGROUND Alveolar soft part sarcoma (ASPS) is a rare soft-tissue sarcoma with a poor prognosis and no established therapy. Recently, encouraging responses to immune checkpoint inhibitors have been reported. METHODS We conducted an investigator-initiated, multicenter, single-group, phase 2 study of the anti-programmed death ligand 1 (PD-L1) agent atezolizumab in adult and pediatric patients with advanced ASPS. Atezolizumab was administered intravenously at a dose of 1200 mg (in patients ≥18 years of age) or 15 mg per kilogram of body weight with a 1200-mg cap (in patients <18 years of age) once every 21 days. Study end points included objective response, duration of response, and progression-free survival according to Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1, as well as pharmacodynamic biomarkers of multistep drug action. RESULTS A total of 52 patients were evaluated. An objective response was observed in 19 of 52 patients (37%), with 1 complete response and 18 partial responses. The median time to response was 3.6 months (range, 2.1 to 19.1), the median duration of response was 24.7 months (range, 4.1 to 55.8), and the median progression-free survival was 20.8 months. Seven patients took a treatment break after 2 years of treatment, and their responses were maintained through the data-cutoff date. No treatment-related grade 4 or 5 adverse events were recorded. Responses were noted despite variable baseline expression of programmed death 1 and PD-L1. CONCLUSIONS Atezolizumab was effective at inducing sustained responses in approximately one third of patients with advanced ASPS. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT03141684.).
Collapse
Affiliation(s)
- Alice P Chen
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Elad Sharon
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Geraldine O'Sullivan-Coyne
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Nancy Moore
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Jared C Foster
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James S Hu
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brian A Van Tine
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Anthony P Conley
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - William L Read
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Richard F Riedel
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Melissa A Burgess
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - John Glod
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Elizabeth J Davis
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Priscilla Merriam
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Abdul R Naqash
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Kristin K Fino
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brandon L Miller
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Deborah F Wilsker
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Asma Begum
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Katherine V Ferry-Galow
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Hari A Deshpande
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Gary K Schwartz
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brian H Ladle
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Scott H Okuno
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Jill C Beck
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James L Chen
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Naoko Takebe
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Laura K Fogli
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Christina L Rosenberger
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Ralph E Parchment
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James H Doroshow
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| |
Collapse
|
12
|
Tanaka M, Chuaychob S, Homme M, Yamazaki Y, Lyu R, Yamashita K, Ae K, Matsumoto S, Kumegawa K, Maruyama R, Qu W, Miyagi Y, Yokokawa R, Nakamura T. ASPSCR1::TFE3 orchestrates the angiogenic program of alveolar soft part sarcoma. Nat Commun 2023; 14:1957. [PMID: 37029109 PMCID: PMC10082046 DOI: 10.1038/s41467-023-37049-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/01/2023] [Indexed: 04/09/2023] Open
Abstract
Alveolar soft part sarcoma (ASPS) is a soft part malignancy affecting adolescents and young adults. ASPS is characterized by a highly integrated vascular network, and its high metastatic potential indicates the importance of ASPS's prominent angiogenic activity. Here, we find that the expression of ASPSCR1::TFE3, the fusion transcription factor causatively associated with ASPS, is dispensable for in vitro tumor maintenance; however, it is required for in vivo tumor development via angiogenesis. ASPSCR1::TFE3 is frequently associated with super-enhancers (SEs) upon its DNA binding, and the loss of its expression induces SE-distribution dynamic modification related to genes belonging to the angiogenesis pathway. Using epigenomic CRISPR/dCas9 screening, we identify Pdgfb, Rab27a, Sytl2, and Vwf as critical targets associated with reduced enhancer activities due to the ASPSCR1::TFE3 loss. Upregulation of Rab27a and Sytl2 promotes angiogenic factor-trafficking to facilitate ASPS vascular network construction. ASPSCR1::TFE3 thus orchestrates higher ordered angiogenesis via modulating the SE activity.
Collapse
Affiliation(s)
- Miwa Tanaka
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan.
- Project for Cancer Epigenomics, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Surachada Chuaychob
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Mizuki Homme
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Division of Cell Biology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yukari Yamazaki
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Ruyin Lyu
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kyoko Yamashita
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Keisuke Ae
- Department of Orthopedic Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Seiichi Matsumoto
- Department of Orthopedic Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kohei Kumegawa
- Project for Cancer Epigenomics, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Reo Maruyama
- Project for Cancer Epigenomics, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Wei Qu
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Takuro Nakamura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
- Department of Experimental Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
13
|
Choi JH, Ro JY. The Recent Advances in Molecular Diagnosis of Soft Tissue Tumors. Int J Mol Sci 2023; 24:ijms24065934. [PMID: 36983010 PMCID: PMC10051446 DOI: 10.3390/ijms24065934] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Soft tissue tumors are rare mesenchymal tumors with divergent differentiation. The diagnosis of soft tissue tumors is challenging for pathologists owing to the diversity of tumor types and histological overlap among the tumor entities. Present-day understanding of the molecular pathogenesis of soft tissue tumors has rapidly increased with the development of molecular genetic techniques (e.g., next-generation sequencing). Additionally, immunohistochemical markers that serve as surrogate markers for recurrent translocations in soft tissue tumors have been developed. This review aims to provide an update on recently described molecular findings and relevant novel immunohistochemical markers in selected soft tissue tumors.
Collapse
Affiliation(s)
- Joon Hyuk Choi
- Department of Pathology, Yeungnam University College of Medicine, 170 Hyeonchung-ro, Namgu, Daegu 42415, Republic of Korea
| | - Jae Y Ro
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Medical College, Cornell University, Houston, TX 77030, USA
| |
Collapse
|
14
|
Karapetyan L, Gooding W, Li A, Yang X, Knight A, Abushukair HM, Vargas De Stefano D, Sander C, Karunamurthy A, Panelli M, Storkus WJ, Tarhini AA, Kirkwood JM. Sentinel Lymph Node Gene Expression Signature Predicts Recurrence-Free Survival in Cutaneous Melanoma. Cancers (Basel) 2022; 14:4973. [PMID: 36291758 PMCID: PMC9599365 DOI: 10.3390/cancers14204973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/29/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022] Open
Abstract
We sought to develop a sentinel lymph node gene expression signature score predictive of disease recurrence in patients with cutaneous melanoma. Gene expression profiling was performed on SLN biopsies using U133A 2.0 Affymetrix gene chips. The top 25 genes associated with recurrence-free survival (RFS) were selected and a penalized regression function was used to select 12 genes with a non-zero coefficient. A proportional hazards regression model was used to evaluate the association between clinical covariates, gene signature score, and RFS. Among the 45 patients evaluated, 23 (51%) had a positive SLN. Twenty-one (46.7%) patients developed disease recurrence. For the top 25 differentially expressed genes (DEG), 12 non-zero penalized coefficients were estimated (CLGN, C1QTNF3, ADORA3, ARHGAP8, DCTN1, ASPSCR1, CHRFAM7A, ZNF223, PDE6G, CXCL3, HEXIM1, HLA-DRB). This 12-gene signature score was significantly associated with RFS (p < 0.0001) and produced a bootstrap C index of 0.888. In univariate analysis, Breslow thickness, presence of primary tumor ulceration, SLN positivity were each significantly associated with RFS. After simultaneously adjusting for these prognostic factors in relation to the gene signature, the 12-gene score remained a significant independent predictor for RFS (p < 0.0001). This SLN 12-gene signature risk score is associated with melanoma recurrence regardless of SLN status and may be used as a prognostic factor for RFS.
Collapse
Affiliation(s)
- Lilit Karapetyan
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - William Gooding
- Hillman Cancer Center, Biostatistics Facility, Pittsburgh, PA 15213, USA
| | - Aofei Li
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Xi Yang
- Department of Medicine, Brigham and Women’s Hospital and Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Andrew Knight
- Department of Medicine, Division of General Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Hassan M. Abushukair
- Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Danielle Vargas De Stefano
- Department of Pathology, Division of Pediatric Pathology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Cindy Sander
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Arivarasan Karunamurthy
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Departments of Dermatology and Pathology, Divisions of Dermatopathology and Molecular Genetic Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | - Walter J. Storkus
- Departments of Dermatology, Immunology, Pathology and Bioengineering, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Ahmad A. Tarhini
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John M. Kirkwood
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Medicine, Division of Hematology/Oncology; University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
15
|
A Rag GTPase dimer code defines the regulation of mTORC1 by amino acids. Nat Cell Biol 2022; 24:1394-1406. [PMID: 36097072 PMCID: PMC9481461 DOI: 10.1038/s41556-022-00976-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 07/13/2022] [Indexed: 12/14/2022]
Abstract
Amino acid availability controls mTORC1 activity via a heterodimeric Rag GTPase complex that functions as a scaffold at the lysosomal surface, bringing together mTORC1 with its activators and effectors. Mammalian cells express four Rag proteins (RagA–D) that form dimers composed of RagA/B bound to RagC/D. Traditionally, the Rag paralogue pairs (RagA/B and RagC/D) are referred to as functionally redundant, with the four dimer combinations used interchangeably in most studies. Here, by using genetically modified cell lines that express single Rag heterodimers, we uncover a Rag dimer code that determines how amino acids regulate mTORC1. First, RagC/D differentially define the substrate specificity downstream of mTORC1, with RagD promoting phosphorylation of its lysosomal substrates TFEB/TFE3, while both Rags are involved in the phosphorylation of non-lysosomal substrates such as S6K. Mechanistically, RagD recruits mTORC1 more potently to lysosomes through increased affinity to the anchoring LAMTOR complex. Furthermore, RagA/B specify the signalling response to amino acid removal, with RagB-expressing cells maintaining lysosomal and active mTORC1 even upon starvation. Overall, our findings reveal key qualitative differences between Rag paralogues in the regulation of mTORC1, and underscore Rag gene duplication and diversification as a potentially impactful event in mammalian evolution. Gollwitzer, Grützmacher et al. and Figlia et al. establish that the various Rag GTPase genes and isoforms differentially regulate mTORC1 activity and distinctly modulate the responsiveness of mammalian cells to amino acid availability.
Collapse
|
16
|
Ogura K, Elkrief A, Bowman AS, Koche RP, de Stanchina E, Benayed R, Mauguen A, Mattar MS, Khodos I, Meyers PA, Healey JH, Tap WD, Hameed M, Zehir A, Shukla N, Sawyers C, Bose R, Slotkin E, Ladanyi M. Prospective Clinical Genomic Profiling of Ewing Sarcoma: ERF and FGFR1 Mutations as Recurrent Secondary Alterations of Potential Biologic and Therapeutic Relevance. JCO Precis Oncol 2022; 6:e2200048. [PMID: 35952322 PMCID: PMC9384944 DOI: 10.1200/po.22.00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ewing sarcoma (ES) is a primitive sarcoma defined by EWSR1-ETS fusions as the primary driver alteration. To better define the landscape of cooperating secondary genetic alterations in ES, we analyzed clinical genomic profiling data of 113 patients with ES, a cohort including more adult patients (> 18 years) and more patients with advanced stage at presentation than previous genomic cohorts.
Collapse
Affiliation(s)
- Koichi Ogura
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Arielle Elkrief
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anita S Bowman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elisa de Stanchina
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,AstraZeneca Pharmaceuticals, Wilmington, DE
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marissa S Mattar
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Inna Khodos
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Paul A Meyers
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John H Healey
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Surgery, Orthopaedic Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - William D Tap
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Meera Hameed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,AstraZeneca Pharmaceuticals, Wilmington, DE
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Charles Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY.,HHMI, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rohit Bose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA.,Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA.,Department of Urology, University of California, San Francisco, San Francisco, CA.,Benioff Initiative for Prostate Cancer Research, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - Emily Slotkin
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
17
|
Tretiakova MS. Chameleon TFE3-translocation RCC and How Gene Partners Can Change Morphology: Accurate Diagnosis Using Contemporary Modalities. Adv Anat Pathol 2022; 29:131-140. [PMID: 35180736 DOI: 10.1097/pap.0000000000000332] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Translocation renal cell carcinoma (tRCC) with TFE3 gene rearrangements has been born as a distinct entity 20 years ago. These relatively rare tumors were notable among other RCC subtypes because of their disproportionally high incidence among children and young adults. Initial reports were focused on describing unifying morphologic criteria and typical clinical presentation. Follow-up studies of ancillary immunohistochemical and hybridization techniques provided additional diagnostic tools allowing recognition of tRCC tumors in practice. However, a growing body of literature also expanded the clinicomorphologic spectrum of tRCCs, to include a significant morphologic overlap with other RCC variants thus blurring the diagnostic clarity of this entity. More recent molecular studies utilizing next-generation sequencing technology accelerated recognition of numerous novel gene partners fusing at different breakpoints with the TFE3 gene. Accumulating data indicates that morphologic and clinical heterogeneity of tRCC could be explained by fusion subtypes, and knowledge of TFE3 partnering genes may be important in predicting tumor behavior. Herein we provided a comprehensive analysis of ∼400 tRCC cases with known TFE3 fusion partners, estimated their relative incidence and summarized clinicomorphologic features associated with most common fusion subtypes. Our data was based on an extensive literature review and had a special focus on comparing immunohistochemistry, fluorescent in situ hybridization and contemporary molecular studies for the accurate diagnosis of tRCC.
Collapse
Affiliation(s)
- Maria S Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| |
Collapse
|
18
|
CIC-mediated modulation of MAPK signaling opposes receptor tyrosine kinase inhibitor response in kinase-addicted sarcoma. Cancer Res 2022; 82:1110-1127. [DOI: 10.1158/0008-5472.can-21-1397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/15/2021] [Accepted: 01/20/2022] [Indexed: 11/16/2022]
|
19
|
Smith RS, Odintsov I, Liu Z, Lui AJW, Hayashi T, Vojnic M, Suehara Y, Delasos L, Mattar MS, Hmeljak J, Ramirez HA, Shaw M, Bui G, Hartono AB, Gladstone E, Kunte S, Magnan H, Khodos I, De Stanchina E, La Quaglia MP, Yao J, Laé M, Lee SB, Spraggon L, Pratilas CA, Ladanyi M, Somwar R. Novel patient-derived models of DSRCT enable validation of ERBB signaling as a potential therapeutic vulnerability. Dis Model Mech 2021; 15:273569. [PMID: 34841430 PMCID: PMC8807576 DOI: 10.1242/dmm.047621] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/12/2021] [Indexed: 11/20/2022] Open
Abstract
Desmoplastic small round cell tumor (DSRCT) is characterized by the t(11;22)(p13;q12) translocation, which fuses the transcriptional regulatory domain of EWSR1 with the DNA-binding domain of WT1, resulting in the oncogenic EWSR1-WT1 fusion protein. The paucity of DSRCT disease models has hampered preclinical therapeutic studies on this aggressive cancer. Here, we developed preclinical disease models and mined DSRCT expression profiles to identify genetic vulnerabilities that could be leveraged for new therapies. We describe four DSRCT cell lines and one patient-derived xenograft model. Transcriptomic, proteomic and biochemical profiling showed evidence of activation of the ERBB pathway. Ectopic expression of EWSR1-WT1 resulted in upregulation of ERRB family ligands. Treatment of DSRCT cell lines with ERBB ligands resulted in activation of EGFR, ERBB2, ERK1/2 and AKT, and stimulation of cell growth. Antagonizing EGFR function with shRNAs, small-molecule inhibitors (afatinib, neratinib) or an anti-EGFR antibody (cetuximab) inhibited proliferation of DSRCT cells. Finally, treatment of mice bearing DSRCT xenografts with a combination of cetuximab and afatinib significantly reduced tumor growth. These data provide a rationale for evaluating EGFR antagonists in patients with DSRCT. This article has an associated First Person interview with the joint first authors of the paper. Summary: Novel models of desmoplastic small round cell tumor (DSRCT) reveal a role for the ERBB pathway in regulating growth of this sarcoma and provide a rationale for evaluating EGFR antagonists in patients with DSRCT.
Collapse
Affiliation(s)
- Roger S Smith
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Igor Odintsov
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zebing Liu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allan Jo-Weng Lui
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Takuo Hayashi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Morana Vojnic
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yoshiyuki Suehara
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lukas Delasos
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marissa S Mattar
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julija Hmeljak
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hillary A Ramirez
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melissa Shaw
- Gerstner School of Graduate Studies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gabrielle Bui
- Gerstner School of Graduate Studies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Eric Gladstone
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Siddharth Kunte
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heather Magnan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Inna Khodos
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa De Stanchina
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael P La Quaglia
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinjuan Yao
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marick Laé
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean B Lee
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Lee Spraggon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christine A Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Romel Somwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
20
|
Wang L, Liu X, Yue M, Liu Z, Zhang Y, Ma Y, Luo J, Li W, Bai J, Yao H, Chen Y, Li X, Feng D, Song X. Identification of hub genes in bladder cancer based on weighted gene co-expression network analysis from TCGA database. Cancer Rep (Hoboken) 2021; 5:e1557. [PMID: 34541834 PMCID: PMC9458504 DOI: 10.1002/cnr2.1557] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
Background Muscular invasive bladder cancer (MIBC) is a common malignant tumor in the world. Because of their heterogeneity in prognosis and response to treatment, biomarkers that can predict survival or help make treatment decisions in patients with MIBC are essential for individualized treatment. Aim We aimed to integrate bioinformatics research methods to identify a set of effective biomarkers capable of predicting, diagnosing, and treating MIBC. To provide a new theoretical basis for the diagnosis and treatment of bladder cancer. Methods and results Gene expression profiles and clinical data of MIBC were obtained by downloading from the Cancer Genome Atlas database. A dataset of 129 MIBC cases and controls was included. 2084 up‐regulated genes and 2961 down‐regulated genes were identified by differentially expressed gene (DEG) analysis. Then, gene ontology analysis was performed to explore the biological functions of DEGs, respectively. The up‐regulated DEGs are mainly enriched in epidermal cell differentiation, mitotic nuclear division, and so forth. They are also involved in the cell cycle, p53 signaling pathway, PPAR signaling pathway, and so forth. The weighted gene co‐expression network analysis yielded five modules related to pathological stages and grading, of which blue and turquoise were the most relevant modules for MIBC. Next, Using Kaplan–Meier survival analysis to identify further hub genes, the screening criteria at p ≤ .05, we found CNKSR1, HIP1R, CFL2, TPM1, CSRP1, SYNM, POPDC2, PJA2, and RBBP8NL genes associated with the progression and prognosis of MIBC patients. Finally, immunohistochemistry experiments further confirmed that CNKSR1 plays a vital role in the tumorigenic context of MIBC. Conclusion The research suggests that CNKSR1, POPDC2, and PJA2 may be novel biomarkers as therapeutic targets for MIBC, especially we used immunohistochemical further to validate CNKSR1 as a therapeutic target for MIBC which may help to improve the prognosis for MIBC.
Collapse
Affiliation(s)
- Lei Wang
- College of Life Sciences, Xinyang Normal University, Xinyang, China.,College of Life Medicine, Xinyang Normal University, Xinyang, China
| | - Xudong Liu
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Miao Yue
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Zhe Liu
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Yu Zhang
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Ying Ma
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Jia Luo
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Wuling Li
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Jiangshan Bai
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Hongmei Yao
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Yuxuan Chen
- Department of Recovery Medicine, People's Liberation Army 990 Hospital, Xinyang, China
| | - Xiaofeng Li
- Department of Pathology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinqiang Song
- College of Life Sciences, Xinyang Normal University, Xinyang, China.,College of Life Medicine, Xinyang Normal University, Xinyang, China
| |
Collapse
|
21
|
O'Sullivan Coyne G, Naqash AR, Sankaran H, Chen AP. Advances in the management of alveolar soft part sarcoma. Curr Probl Cancer 2021; 45:100775. [PMID: 34284873 DOI: 10.1016/j.currproblcancer.2021.100775] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/21/2022]
Abstract
Alveolar Soft Part Sarcoma is one of the less commonly diagnosed soft tissue sarcoma subtypes, an infrequent subtype within the already rare category of human malignancy of sarcoma. In this article we will summarize the histopathological features, natural history and distinct molecular and biological features that have become increasingly appreciated with newer technologies and precision oncology. We will discuss the contemporary management of this disease as well as emerging treatment options.
Collapse
Affiliation(s)
- Geraldine O'Sullivan Coyne
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Abdul Rafeh Naqash
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Hari Sankaran
- Biometric Research Program, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alice P Chen
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
22
|
Kannan S, Lock I, Ozenberger BB, Jones KB. Genetic drivers and cells of origin in sarcomagenesis. J Pathol 2021; 254:474-493. [DOI: 10.1002/path.5617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/01/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Sarmishta Kannan
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Ian Lock
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Benjamin B Ozenberger
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| | - Kevin B Jones
- Departments of Orthopaedics and Oncological Sciences Huntsman Cancer Institute, University of Utah School of Medicine Salt Lake City UT USA
| |
Collapse
|
23
|
Ogura K, Somwar R, Hmeljak J, Magnan H, Benayed R, Momeni Boroujeni A, Bowman AS, Mattar MS, Khodos I, de Stanchina E, Jungbluth A, Asher M, Odintsov I, Hartono AB, LaQuaglia MP, Slotkin E, Pratilas CA, Lee SB, Spraggon L, Ladanyi M. Therapeutic Potential of NTRK3 Inhibition in Desmoplastic Small Round Cell Tumor. Clin Cancer Res 2020; 27:1184-1194. [PMID: 33229458 DOI: 10.1158/1078-0432.ccr-20-2585] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/27/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Desmoplastic small round cell tumor (DSRCT) is a highly lethal intra-abdominal sarcoma of adolescents and young adults. DSRCT harbors a t(11;22)(p13:q12) that generates the EWSR1-WT1 chimeric transcription factor, the key oncogenic driver of DSRCT. EWSR1-WT1 rewires global gene expression networks and activates aberrant expression of targets that together mediate oncogenesis. EWSR1-WT1 also activates a neural gene expression program. EXPERIMENTAL DESIGN Among these neural markers, we found prominent expression of neurotrophic tyrosine kinase receptor 3 (NTRK3), a druggable receptor tyrosine kinase. We investigated the regulation of NTRK3 by EWSR1-WT1 and its potential as a therapeutic target in vitro and in vivo, the latter using novel patient-derived models of DSRCT. RESULTS We found that EWSR1-WT1 binds upstream of NTRK3 and activates its transcription. NTRK3 mRNA is highly expressed in DSRCT compared with other major chimeric transcription factor-driven sarcomas and most DSRCTs are strongly immunoreactive for NTRK3 protein. Remarkably, expression of NTRK3 kinase domain mRNA in DSRCT is also higher than in cancers with NTRK3 fusions. Abrogation of NTRK3 expression by RNAi silencing reduces growth of DSRCT cells and pharmacologic targeting of NTRK3 with entrectinib is effective in both in vitro and in vivo models of DSRCT. CONCLUSIONS Our results indicate that EWSR1-WT1 directly activates NTRK3 expression in DSRCT cells, which are dependent on its expression and activity for growth. Pharmacologic inhibition of NTRK3 by entrectinib significantly reduces growth of DSRCT cells both in vitro and in vivo, providing a rationale for clinical evaluation of NTRK3 as a therapeutic target in DSRCT.
Collapse
Affiliation(s)
- Koichi Ogura
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Romel Somwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julija Hmeljak
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heather Magnan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Anita S Bowman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marissa S Mattar
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Inna Khodos
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Anti-tumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Achim Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marina Asher
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Igor Odintsov
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alifiani B Hartono
- Department of Pathology & Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Michael P LaQuaglia
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily Slotkin
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christine A Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Sean Bong Lee
- Department of Pathology & Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Lee Spraggon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. .,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
24
|
Stockwin LH. Alveolar soft-part sarcoma (ASPS) resembles a mesenchymal stromal progenitor: evidence from meta-analysis of transcriptomic data. PeerJ 2020; 8:e9394. [PMID: 32596059 PMCID: PMC7307565 DOI: 10.7717/peerj.9394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022] Open
Abstract
Alveolar soft-part sarcoma (ASPS) is an extremely rare malignancy characterized by the unbalanced translocation der(17)t(X;17)(p11;q25). This translocation generates a fusion protein, ASPL-TFE3, that drives pathogenesis through aberrant transcriptional activity. Although considerable progress has been made in identifying ASPS therapeutic vulnerabilities (e.g., MET inhibitors), basic research efforts are hampered by the lack of appropriate in vitro reagents with which to study the disease. In this report, previously unmined microarray data for the ASPS cell line, ASPS-1, was analyzed relative to the NCI sarcoma cell line panel. These data were combined with meta-analysis of pre-existing ASPS patient microarray and RNA-seq data to derive a platform-independent ASPS transcriptome. Results demonstrated that ASPS-1, in the context of the NCI sarcoma cell panel, had some similarities to normal mesenchymal cells and connective tissue sarcomas. The cell line was characterized by high relative expression of transcripts such as CRYAB, MT1G, GCSAML, and SV2B. Notably, ASPS-1 lacked mRNA expression of myogenesis-related factors MYF5, MYF6, MYOD1, MYOG, PAX3, and PAX7. Furthermore, ASPS-1 had a predicted mRNA surfaceome resembling an undifferentiated mesenchymal stromal cell through expression of GPNMB, CD9 (TSPAN29), CD26 (DPP4), CD49C (ITGA3), CD54 (ICAM1), CD63 (TSPAN30), CD68 (SCARD1), CD130 (IL6ST), CD146 (MCAM), CD147 (BSG), CD151 (SFA-1), CD166 (ALCAM), CD222 (IGF2R), CD230 (PRP), CD236 (GPC), CD243 (ABCB1), and CD325 (CDHN). Subsequent re-analysis of ASPS patient data generated a consensus expression profile with considerable overlap between studies. In common with ASPS-1, elevated expression was noted for CTSK, DPP4, GPNMB, INHBE, LOXL4, PSG9, SLC20A1, STS, SULT1C2, SV2B, and UPP1. Transcripts over-expressed only in ASPS patient samples included ABCB5, CYP17A1, HIF1A, MDK, P4HB, PRL, and PSAP. These observations are consistent with that expected for a mesenchymal progenitor cell with adipogenic, osteogenic, or chondrogenic potential. In summary, the consensus data generated in this study highlight the unique and highly conserved nature of the ASPS transcriptome. Although the ability of the ASPL-TFE3 fusion to perturb mRNA expression must be acknowledged, the prevailing ASPS transcriptome resembles that of a mesenchymal stromal progenitor.
Collapse
|
25
|
Dickson BC, T-S Chung C, Hurlbut DJ, Marrano P, Shago M, Sung YS, Swanson D, Zhang L, Antonescu CR. Genetic diversity in alveolar soft part sarcoma: A subset contain variant fusion genes, highlighting broader molecular kinship with other MiT family tumors. Genes Chromosomes Cancer 2020; 59:23-29. [PMID: 31433528 PMCID: PMC7057290 DOI: 10.1002/gcc.22803] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/14/2019] [Accepted: 08/17/2019] [Indexed: 01/25/2023] Open
Abstract
Alveolar soft part sarcoma (ASPS) is a rare malignancy that, since its initial description, remains a neoplasm of uncertain histogenesis. The disease-defining molecular event characterizing the diagnosis of ASPS is the ASPSCR1-TFE3 fusion gene. Following identification of an index case of ASPS with a novel TFE3 fusion partner, we performed a retrospective review to determine whether this represents an isolated event. We identified two additional cases, for a total of three cases lacking ASPSCR1 partners. The average patient age was 46 years (range, 17-65); two patients were female. The sites of origin included the transverse colon, foot, and dura. Each case exhibited a histomorphology typical of ASPS, and immunohistochemistry was positive for TFE3 in all cases. Routine molecular testing of the index patient demonstrated a HNRNPH3-TFE3 gene fusion; the remaining cases were found to have DVL2-TFE3 or PRCC-TFE3 fusion products. The latter two fusions have previously been identified in renal cell carcinoma; to our knowledge, this is the first report of a HNRNPH3-TFE3 gene fusion. These findings highlight a heretofore underrecognized genetic diversity in ASPS, which appears to more broadly molecularly overlap with that of translocation-associated renal cell carcinoma and PEComa. These results have immediate implications in the diagnosis of ASPS since assays reliant upon ASPSCR1 may yield a false negative result. While these findings further understanding of the molecular pathogenesis of ASPS, issues related to the histogenesis of this unusual neoplasm remain unresolved.
Collapse
Affiliation(s)
- Brendan C. Dickson
- Department of Pathology and Laboratory Medicine, Mount Sinai Health System, Toronto, ON, Canada,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada,Corresponding Authors: Brendan C. Dickson, Pathology & Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5, , Cristina R. Antonescu, Department of Pathology, Memorial Sloan Kettering Cancer Center New York, NY, USA 10021,
| | - Catherine T-S Chung
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada,Department of Paediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - David J. Hurlbut
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Paula Marrano
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada,Department of Paediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Mary Shago
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada,Department of Paediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Yun-Shao Sung
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Swanson
- Department of Pathology and Laboratory Medicine, Mount Sinai Health System, Toronto, ON, Canada
| | - Lei Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
26
|
Baba M, Furuya M, Motoshima T, Lang M, Funasaki S, Ma W, Sun HW, Hasumi H, Huang Y, Kato I, Kadomatsu T, Satou Y, Morris N, Karim BO, Ileva L, Kalen JD, Wilan Krisna LA, Hasumi Y, Sugiyama A, Kurahashi R, Nishimoto K, Oyama M, Nagashima Y, Kuroda N, Araki K, Eto M, Yao M, Kamba T, Suda T, Oike Y, Schmidt LS, Linehan WM. TFE3 Xp11.2 Translocation Renal Cell Carcinoma Mouse Model Reveals Novel Therapeutic Targets and Identifies GPNMB as a Diagnostic Marker for Human Disease. Mol Cancer Res 2019; 17:1613-1626. [PMID: 31043488 PMCID: PMC6679785 DOI: 10.1158/1541-7786.mcr-18-1235] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/12/2019] [Accepted: 04/26/2019] [Indexed: 12/16/2022]
Abstract
Renal cell carcinoma (RCC) associated with Xp11.2 translocation (TFE3-RCC) has been recently defined as a distinct subset of RCC classified by characteristic morphology and clinical presentation. The Xp11 translocations involve the TFE3 transcription factor and produce chimeric TFE3 proteins retaining the basic helix-loop-helix leucine zipper structure for dimerization and DNA binding suggesting that chimeric TFE3 proteins function as oncogenic transcription factors. Diagnostic biomarkers and effective forms of therapy for advanced cases of TFE3-RCC are as yet unavailable. To facilitate the development of molecular based diagnostic tools and targeted therapies for this aggressive kidney cancer, we generated a translocation RCC mouse model, in which the PRCC-TFE3 transgene is expressed specifically in kidneys leading to the development of RCC with characteristic histology. Expression of the receptor tyrosine kinase Ret was elevated in the kidneys of the TFE3-RCC mice, and treatment with RET inhibitor, vandetanib, significantly suppressed RCC growth. Moreover, we found that Gpnmb (Glycoprotein nonmetastatic B) expression was notably elevated in the TFE3-RCC mouse kidneys as seen in human TFE3-RCC tumors, and confirmed that GPNMB is the direct transcriptional target of TFE3 fusions. While GPNMB IHC staining was positive in 9/9 cases of TFE3-RCC, Cathepsin K, a conventional marker for TFE3-RCC, was positive in only 67% of cases. These data support RET as a potential target and GPNMB as a diagnostic marker for TFE3-RCC. The TFE3-RCC mouse provides a preclinical in vivo model for the development of new biomarkers and targeted therapeutics for patients affected with this aggressive form of RCC. IMPLICATIONS: Key findings from studies with this preclinical mouse model of TFE3-RCC underscore the potential for RET as a therapeutic target for treatment of patients with TFE3-RCC, and suggest that GPNMB may serve as diagnostic biomarker for TFE3 fusion RCC.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Animals
- Apoptosis
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Cycle Proteins/genetics
- Cell Proliferation
- Child
- Chromosomes, Human, X
- Disease Models, Animal
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Middle Aged
- Neoplasm Proteins/genetics
- Oncogene Proteins, Fusion
- Prognosis
- Survival Rate
- Translocation, Genetic
- Tumor Cells, Cultured
- Young Adult
Collapse
Affiliation(s)
- Masaya Baba
- Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mitsuko Furuya
- Department of Molecular Pathology, Yokohama City University, Yokohama, Japan
| | | | - Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Shintaro Funasaki
- Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Wenjuan Ma
- Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, Maryland
| | - Hisashi Hasumi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Department of Urology, Yokohama City University, Yokohama, Japan
| | - Ying Huang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ikuma Kato
- Department of Molecular Pathology, Yokohama City University, Yokohama, Japan
| | | | - Yorifumi Satou
- Laboratory of Retroviral Genomics and Transcriptomics, International Research Center for Medical Sciences (IRCMS), Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Nicole Morris
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Baktiar O Karim
- Pathology/Histotechnology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Lilia Ileva
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Luh Ade Wilan Krisna
- Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Yukiko Hasumi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Aiko Sugiyama
- DSK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Ryoma Kurahashi
- Department of Urology, Kumamoto University, Kumamoto, Japan
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Koshiro Nishimoto
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Masafumi Oyama
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Naoto Kuroda
- Department of Pathology, Kochi Red Cross Hospital, Kochi, Japan
| | - Kimi Araki
- Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Masatoshi Eto
- Department of Urology, Kyushyu University, Fukuoka, Japan
| | - Masahiro Yao
- Department of Urology, Yokohama City University, Yokohama, Japan
| | - Tomomi Kamba
- Department of Urology, Kumamoto University, Kumamoto, Japan
| | - Toshio Suda
- Laboratory of Stem Cell Regulation, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore
| | - Yuichi Oike
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
27
|
Kurahashi R, Kadomatsu T, Baba M, Hara C, Itoh H, Miyata K, Endo M, Morinaga J, Terada K, Araki K, Eto M, Schmidt LS, Kamba T, Linehan WM, Oike Y. MicroRNA-204-5p: A novel candidate urinary biomarker of Xp11.2 translocation renal cell carcinoma. Cancer Sci 2019; 110:1897-1908. [PMID: 31006167 PMCID: PMC6549932 DOI: 10.1111/cas.14026] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/02/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022] Open
Abstract
Xp11.2 translocation renal cell carcinoma (Xp11 tRCC) is a rare sporadic pediatric kidney cancer caused by constitutively active TFE3 fusion proteins. Tumors in patients with Xp11 tRCC tend to recur and undergo frequent metastasis, in part due to lack of methods available to detect early‐stage disease. Here we generated transgenic (Tg) mice overexpressing the human PRCC‐TFE3 fusion gene in renal tubular epithelial cells, as an Xp11 tRCC mouse model. At 20 weeks of age, mice showed no histological abnormalities in kidney but by 40 weeks showed Xp11 tRCC development and related morphological and histological changes. MicroRNA (miR)‐204‐5p levels in urinary exosomes of 40‐week‐old Tg mice showing tRCC were significantly elevated compared with levels in control mice. MicroRNA‐204‐5p expression also significantly increased in primary renal cell carcinoma cell lines established both from Tg mouse tumors and from tumor tissue from 2 Xp11 tRCC patients. All of these lines secreted miR‐204‐5p‐containing exosomes. Notably, we also observed increased miR‐204‐5p levels in urinary exosomes in 20‐week‐old renal PRCC‐TFE3 Tg mice prior to tRCC development, and those levels were equivalent to those in 40‐week‐old Tg mice, suggesting that miR‐204‐5p increases follow expression of constitutively active TFE3 fusion proteins in renal tubular epithelial cells prior to overt tRCC development. Finally, we confirmed that miR‐204‐5p expression significantly increases in noncancerous human kidney cells after overexpression of a PRCC‐TFE3 fusion gene. These findings suggest that miR‐204‐5p in urinary exosomes could be a useful biomarker for early diagnosis of patients with Xp11 tRCC.
Collapse
Affiliation(s)
- Ryoma Kurahashi
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaya Baba
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Chiaki Hara
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hitoshi Itoh
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Motoyoshi Endo
- Department of Molecular Biology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Clinical Research, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Nephrology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazutoyo Terada
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kimi Araki
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Laura S Schmidt
- Basic Science Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA.,Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tomomi Kamba
- Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| |
Collapse
|
28
|
Dufresne A, Brahmi M, Karanian M, Blay JY. Using biology to guide the treatment of sarcomas and aggressive connective-tissue tumours. Nat Rev Clin Oncol 2019; 15:443-458. [PMID: 29666441 DOI: 10.1038/s41571-018-0012-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sarcomas are a heterogeneous group of malignancies that arise from cells of a mesenchymal origin. Surgery forms the mainstay of the treatment of most patients with localized sarcoma and might be followed or preceded by chemotherapy and/or radiotherapy. In the metastatic setting, systemic treatments tend to improve survival and control symptoms. However, the adverse events and sometimes disappointing outcomes associated with these empirical approaches to treatment indicate a need for new approaches. The advent of next-generation sequencing (NGS) has enabled more targeted treatment of many malignancies based on the presence of specific alterations. NGS analyses of sarcomas have revealed the presence of many alterations that can be targeted using therapies that are already used in patients with other forms of cancer. In this Review, we describe the genomic alterations considered to define specific nosological subgroups of sarcoma and whose contribution to oncogenesis provides a biological rationale for the use of a specific targeted therapy. We also report several less successful examples that should guide researchers and clinicians to better define the extent to which the identification of driver molecular alterations should influence the development of novel treatments.
Collapse
Affiliation(s)
- Armelle Dufresne
- Department of Medical Oncology, Centre Leon Berard, Lyon, France.
| | - Mehdi Brahmi
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Marie Karanian
- Department of Pathology, Centre Leon Berard, Lyon, France
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| |
Collapse
|
29
|
NEAT1-TFE3 and KAT6A-TFE3 renal cell carcinomas, new members of MiT family translocation renal cell carcinoma. Mod Pathol 2019; 32:710-716. [PMID: 30622287 PMCID: PMC6486435 DOI: 10.1038/s41379-018-0191-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/31/2022]
Abstract
Microphthalmia-associated transcription factor (MiT) family translocation renal cell carcinoma harbors variable gene fusions involving either TFE3 or TFEB genes. Multiple 5' fusion partners for TFE3 have been reported, including ASPSCR1, CLTC, DVL2, LUC7L3, KHSRP, PRCC, PARP14, NONO, SFPQ1, MED15, and RBM10. Each of these fusion genes activates TFE3 transcription which can be detected by immunostaining. Using targeted RNA-sequencing, TFE3 fusion gene partners were identified in 5 cases of TFE3 immunohistochemistry positive translocation renal cell carcinoma. Three cases demonstrated known fusions: ASPSCR1-TFE3, MED15-TFE3 and RBM10-TFE3. However, two cases showed unreported NEAT1-TFE3 and KAT6A-TFE3 fusion transcripts. The NEAT1-TFE3 RCC arose in a 59-year-old male; which demonstrated overlapping morphological features seen in NEAT2(MALAT1)-TFEB t(6;11) renal cell carcinoma, including biphasic alveolar/nested tumor cells with eosinophilic cytoplasm. The KAT6A-TFE3 renal cell carcinoma demonstrated typical morphological features of TFE3/Xp11 renal cell carcinoma including papillae, eosinophilic cytoplasm with focal clearing and abundant psammoma bodies. KAT6A gene fusion was reported in some cases of acute myeloid leukemia, which has not been previously reported in solid tumors. This report highlights the genetic complexity of TFE3 translocation renal cell carcinoma; and RNA-sequencing is a powerful approach for elucidating the underlying genetic alterations.
Collapse
|
30
|
Kato S, Hayashi T, Suehara Y, Hamanoue H, Yamanaka S, Ichikawa Y, Higurashi T, Ohashi K, Yamaguchi S, Nozaki Y, Terao Y, Saito T. Multicenter experience with large panel next-generation sequencing in patients with advanced solid cancers in Japan. Jpn J Clin Oncol 2019; 49:174-182. [PMID: 30541038 DOI: 10.1093/jjco/hyy173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/31/2018] [Indexed: 11/14/2022] Open
Abstract
Background Application of next-generation DNA sequencing (NGS) has recently become increasingly common in the field of clinical oncology in several countries around the world. In Japan also, a system for applying NGS to routine clinical practice is gradually being established. During this process, we introduced in Japan the tumor-profiling MSK-IMPACT (Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets) assay. Methods We present here our initial experience with the use of MSK-IMPACT in 68 patients selected from two institutions in Japan between June 2016 and October 2017. Results MSK-IMPACT sequencing was successful and yielded results in specimens obtained from 64 of the 68 patients, representing an overall assay success rate of 94.1%. The top three cancer types tested were endometrial cancer (17.2%), pancreatic cancer (15.6%) and colorectal cancer (12.5%). Evaluation of the clinical actionability of the genetic alterations revealed that 25.0% of patients (n = 16) harbored at least one actionable alteration. However, enrolling the patients in a genomically matched clinical trial was difficult, mainly because most clinical trials are limited to tumors arising from a specific organ/site. One patient with microsatellite instability-high status, as determined by MSK-IMPACT, was treated with pembrolizumab and showed partial response. Conclusions Although tumor profiling by NGS and administration of genomically matched therapy is a promising strategy, because of its high cost, we need to consider how we can fit it into the Japanese medical system. Towards this end, we believe that it is important to share our initial experience for furthering precision medicine in Japan.
Collapse
Affiliation(s)
- Shingo Kato
- Department of Clinical Cancer Genomics, Yokohama City University Hospital, Yokohama, Japan
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suehara
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Haruka Hamanoue
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama, Japan
| | - Shoji Yamanaka
- Department of Pathology, Yokohama City University Hospital, Yokohama, Japan
| | - Yasushi Ichikawa
- Department of Oncology, Yokohama City University Hospital, Yokohama, Japan
| | - Takuma Higurashi
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Kenichi Ohashi
- Department of Pathology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Shigeo Yamaguchi
- Department of Clinical Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yumi Nozaki
- Department of Clinical Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yasuhisa Terao
- Department of Gynecology and Obstetrics, Juntendo University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
31
|
Perry JA, Seong BKA, Stegmaier K. Biology and Therapy of Dominant Fusion Oncoproteins Involving Transcription Factor and Chromatin Regulators in Sarcomas. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2019. [DOI: 10.1146/annurev-cancerbio-030518-055710] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A third of soft tissue sarcomas have been shown to carry recurrent, characteristic chromosomal translocations, many of which generate fusion proteins that act as dominant transcription factors or as chromatin regulators. With routine use of massively parallel sequencing and advances in technology for the study of epigenetics and protein complexes, the last decade has seen a marked advancement in the identification of novel fusions and in our understanding of the mechanisms by which they contribute to the malignant state. Moreover, with new approaches in chemistry, such as the strategy of targeted protein degradation, we are now better poised to address these previously intractable targets. In this review, we describe three of the most common fusion-driven sarcomas (Ewing sarcoma, alveolar rhabdomyosarcoma, and synovial sarcoma), mechanistic themes emerging across these diseases, and novel approaches to their targeting.
Collapse
Affiliation(s)
- Jennifer A. Perry
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Bo Kyung Alex Seong
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
32
|
Perera RM, Di Malta C, Ballabio A. MiT/TFE Family of Transcription Factors, Lysosomes, and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2019; 3:203-222. [PMID: 31650096 PMCID: PMC6812561 DOI: 10.1146/annurev-cancerbio-030518-055835] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer cells have an increased demand for energy sources to support accelerated rates of growth. When nutrients become limiting, cancer cells may switch to nonconventional energy sources that are mobilized through nutrient scavenging pathways involving autophagy and the lysosome. Thus, several cancers are highly reliant on constitutive activation of these pathways to degrade and recycle cellular materials. Here, we focus on the MiT/TFE family of transcription factors, which control transcriptional programs for autophagy and lysosome biogenesis and have emerged as regulators of energy metabolism in cancer. These new findings complement earlier reports that chromosomal translocations and amplifications involving the MiT/TFE genes contribute to the etiology and pathophysiology of renal cell carcinoma, melanoma, and sarcoma, suggesting pleiotropic roles for these factors in a wider array of cancers. Understanding the interplay between the oncogenic and stress-adaptive roles of MiT/TFE factors could shed light on fundamental mechanisms of cellular homeostasis and point to new strategies for cancer treatment.
Collapse
Affiliation(s)
- Rushika M Perera
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143, USA
| | - Chiara Di Malta
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
- Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
33
|
Drug sensitivity testing on patient-derived sarcoma cells predicts patient response to treatment and identifies c-Sarc inhibitors as active drugs for translocation sarcomas. Br J Cancer 2019; 120:435-443. [PMID: 30745580 PMCID: PMC6462037 DOI: 10.1038/s41416-018-0359-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 10/26/2018] [Accepted: 11/22/2018] [Indexed: 11/17/2022] Open
Abstract
Background Heterogeneity and low incidence comprise the biggest challenge in sarcoma diagnosis and treatment. Chemotherapy, although efficient for some sarcoma subtypes, generally results in poor clinical responses and is mostly recommended for advanced disease. Specific genomic aberrations have been identified in some sarcoma subtypes but few of them can be targeted with approved drugs. Methods We cultured and characterised patient-derived sarcoma cells and evaluated their sensitivity to 525 anti-cancer agents including both approved and non-approved drugs. In total, 14 sarcomas and 5 healthy mesenchymal primary cell cultures were studied. The sarcoma biopsies and derived cells were characterised by gene panel sequencing, cancer driver gene expression and by detecting specific fusion oncoproteins in situ in sarcomas with translocations. Results Soft tissue sarcoma cultures were established from patient biopsies with a success rate of 58%. The genomic profile and drug sensitivity testing on these samples helped to identify targeted inhibitors active on sarcomas. The cSrc inhibitor Dasatinib was identified as an active drug in sarcomas carrying chromosomal translocations. The drug sensitivity of the patient sarcoma cells ex vivo correlated with the response to the former treatment of the patient. Conclusions Our results show that patient-derived sarcoma cells cultured in vitro are relevant and practical models for genotypic and phenotypic screens aiming to identify efficient drugs to treat sarcoma patients with poor treatment options.
Collapse
|
34
|
Verma SP, Das P. Monensin induces cell death by autophagy and inhibits matrix metalloproteinase 7 (MMP7) in UOK146 renal cell carcinoma cell line. In Vitro Cell Dev Biol Anim 2018; 54:736-742. [DOI: 10.1007/s11626-018-0298-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/24/2018] [Indexed: 02/02/2023]
|
35
|
Molecular Landscape in Alveolar Soft Part Sarcoma: Implications for Molecular Targeted Therapy. Biomed Pharmacother 2018; 103:889-896. [DOI: 10.1016/j.biopha.2018.04.117] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/09/2018] [Accepted: 04/16/2018] [Indexed: 12/20/2022] Open
|
36
|
Mukaihara K, Tanabe Y, Kubota D, Akaike K, Hayashi T, Mogushi K, Hosoya M, Sato S, Kobayashi E, Okubo T, Kim Y, Kohsaka S, Saito T, Kaneko K, Suehara Y. Cabozantinib and dastinib exert anti-tumor activity in alveolar soft part sarcoma. PLoS One 2017; 12:e0185321. [PMID: 28945796 PMCID: PMC5612696 DOI: 10.1371/journal.pone.0185321] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022] Open
Abstract
Background Alveolar soft part sarcoma (ASPS) is an extremely rare metastatic soft tissue tumor with a poor prognosis for which no effective systemic therapies have yet been established. Therefore, the development of novel effective treatment approaches is required. Tyrosine kinases (TKs) are being increasingly used as therapeutic targets in a variety of cancers. The purpose of this study was to identify novel therapeutic target TKs and to clarify the efficacy of TK inhibitors (TKIs) in the treatment of ASPS. Experimental design To identify novel therapeutic target TKs in ASPS, we evaluated the antitumor effects and kinase activity of three TKIs (pazopanib, dasatinib, and cabozantinib) against ASPS cells using an in vitro assay. Based on these results, we then investigated the phosphorylation activities of the identified targets using western blotting, in addition to examining antitumor activity through in vivo assays of several TKIs to determine both the efficacy of these substances and accurate targets. Results In cell proliferation and invasion assays using pazopanib, cabozantinib, and dasatinib, all three TKIs inhibited the cell growth in ASPS cells. Statistical analyses of the cell proliferation and invasion assays revealed that dasatinib had a significant inhibitory effect in cell proliferation assays, and cabozantinib exhibited marked inhibitory effects on cellular functions in both assays. Through western blotting, we also confirmed that cabozantinib inhibited c-MET phosphorylation and dasatinib inhibited SRC phosphorylation in dose-dependent fashion. Mice that received cabozantinib and dasatinib had significantly smaller tumor volumes than control animals, demonstrating the in vivo antitumor activity of, these substances. Conclusions Our findings suggest that cabozantinib and dasatinib may be more effective than pazopanib against ASPS cells. These in vitro and in vivo data suggest that c-MET may be a potential therapeutic target in ASPS, and cabozantinib may be a particularly useful therapeutic option for patients with ASPS, including those with pazopanib-resistant ASPS.
Collapse
Affiliation(s)
- Kenta Mukaihara
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yu Tanabe
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Daisuke Kubota
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Keisuke Akaike
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kaoru Mogushi
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Masaki Hosoya
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Shingo Sato
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University Graduate School and Faculty of Medicine, Tokyo, Japan
| | - Eisuke Kobayashi
- Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Taketo Okubo
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Youngji Kim
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Shinji Kohsaka
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuo Kaneko
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suehara
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
37
|
Novel chimeric transcript RRM2-c2orf48 promotes metastasis in nasopharyngeal carcinoma. Cell Death Dis 2017; 8:e3047. [PMID: 28906488 PMCID: PMC5636969 DOI: 10.1038/cddis.2017.402] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 06/29/2017] [Accepted: 07/14/2017] [Indexed: 12/13/2022]
Abstract
Recently, chimeric transcripts have been found to be associated with the pathogenesis and poor prognosis of malignant tumors. Through our preliminary experiment, a novel chimeric transcript called chimeric transcript RRM2-c2orf48 was detected in C666-1, a classical cell line of human nasopharyngeal carcinoma (NPC). Therefore, the objective of this study was to demonstrate the existence and expression of novel chimeric transcript RRM2-c2orf48 and to explore the main functions and mechanisms of RRM2-c2orf48 in NPC. In this study, the expression of RRM2-c2orf48 was evaluated in NPC cells and specimens. Effects of RRM2-c2orf48 on migration and invasive capacities were detected invivo and vitro. Moreover, ways in which RRM2-c2orf48 increases the invasive capacities of NPC were explored. As a result, the presence of novel chimeric transcript RRM2-c2orf48 was confirmed in C666-1 by RT-PCR and sequencing, and it was a read-through between RRM2 and c2orf48 through the transcription of interchromosome. Higher expressions of novel RRM2-c2orf48 were detected in NPC cell lines and NPC tissue specimens relative to the controls and its expression was be statistically relevant to TNM staging. High level of RRM2-c2orf48 could increase the migration and invasive capacities of NPC cells, potentially as a result of NPC cell epithelial–mesenchymal transition. RRM2-c2orf48 could also enhance resistance of chemotherapy. In vivo, RRM2-c2orf48 could enhance lung and lymph node metastasis in nude mice. These results demonstrate that high levels of RRM2-c2orf48 expression may be a useful predictor of NPC patients of metastatic potency, presenting potential implications for NPC diagnosis and therapy.
Collapse
|
38
|
Ju X, Sun K, Liu R, Li S, Abulajiang G, Zou H, Lan J, Ren Y, Jiang J, Liang W, Pang L, Li F. Exploring the Histogenesis and Diagnostic Strategy Using Immunoassay and RT-PCR in Alveolar Soft Part Sarcoma. Pathol Oncol Res 2017; 24:593-600. [DOI: 10.1007/s12253-017-0280-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 07/25/2017] [Indexed: 12/14/2022]
|
39
|
Dupain C, Harttrampf AC, Urbinati G, Geoerger B, Massaad-Massade L. Relevance of Fusion Genes in Pediatric Cancers: Toward Precision Medicine. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 6:315-326. [PMID: 28325298 PMCID: PMC5363511 DOI: 10.1016/j.omtn.2017.01.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 12/19/2022]
Abstract
Pediatric cancers differ from adult tumors, especially by their very low mutational rate. Therefore, their etiology could be explained in part by other oncogenic mechanisms such as chromosomal rearrangements, supporting the possible implication of fusion genes in the development of pediatric cancers. Fusion genes result from chromosomal rearrangements leading to the juxtaposition of two genes. Consequently, an abnormal activation of one or both genes is observed. The detection of fusion genes has generated great interest in basic cancer research and in the clinical setting, since these genes can lead to better comprehension of the biological mechanisms of tumorigenesis and they can also be used as therapeutic targets and diagnostic or prognostic biomarkers. In this review, we discuss the molecular mechanisms of fusion genes and their particularities in pediatric cancers, as well as their relevance in murine models and in the clinical setting. We also point out the difficulties encountered in the discovery of fusion genes. Finally, we discuss future perspectives and priorities for finding new innovative therapies in childhood cancer.
Collapse
Affiliation(s)
- Célia Dupain
- Vectorology and Anticancer Therapies, UMR 8203 CNRS, University Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Anne Catherine Harttrampf
- Vectorology and Anticancer Therapies, UMR 8203 CNRS, University Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Giorgia Urbinati
- Vectorology and Anticancer Therapies, UMR 8203 CNRS, University Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Birgit Geoerger
- Vectorology and Anticancer Therapies, UMR 8203 CNRS, University Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Liliane Massaad-Massade
- Vectorology and Anticancer Therapies, UMR 8203 CNRS, University Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France.
| |
Collapse
|
40
|
Abstract
The MiT family of translocation-associated renal cell carcinomas comprise approximately 40 % of renal cell carcinomas in young patients but only up to 4 % of renal cell carcinomas in adult patients. The Xp11.2 translocation-associated tumors are the most frequent and were included in the 2004 World Health Organization (WHO) classification. They contain a fusion of the TFE3 gene with ASPSCR1, PRCC, NONO, SPFQ or CLTC resulting in an immunohistochemically detectable nuclear overexpression of TFE3. The Xp11.2 translocation-associated renal cell carcinomas are characterized by ample clear cytoplasm, papillary architecture and abundant psammoma bodies. The TFEB translocation-associated renal cell carcinomas are much rarer and show a biphasic architecture. Fluorescence in situ hybridization permits the detection of a translocation by means of a break apart probe for the TFE3 and TFEB genes and is recommended for the diagnosis of renal cell carcinomas in patients under 30 years of age. The TFE3 and TFEB translocation-associated tumors are classified as MiT family translocation carcinomas in the new WHO classification.The rare renal cell carcinomas harboring an ALK rearrangement with fusion to VCL in young patients with sickle cell trait show a characteristic morphology and are listed in the new WHO classification as a provisional entity.
Collapse
|
41
|
Ishiguro N, Yoshida H. ASPL-TFE3 Oncoprotein Regulates Cell Cycle Progression and Induces Cellular Senescence by Up-Regulating p21. Neoplasia 2016; 18:626-635. [PMID: 27673450 PMCID: PMC5037204 DOI: 10.1016/j.neo.2016.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/05/2016] [Accepted: 08/11/2016] [Indexed: 01/30/2023] Open
Abstract
Alveolar soft part sarcoma is an extremely rare soft tissue sarcoma with poor prognosis. It is characterized by the unbalanced recurrent chromosomal translocation der(17)t(X;17)(p11;q25), resulting in the generation of an ASPL-TFE3 fusion gene. ASPL-TFE3 oncoprotein functions as an aberrant transcriptional factor and is considered to play a crucial role in the tumorigenesis of alveolar soft part sarcoma. However, the underlying molecular mechanisms are poorly understood. In this study, we identified p21 (p21WAF1/CIP1) as a direct transcriptional target of ASPL-TFE3. Ectopic ASPL-TFE3 expression in 293 cells resulted in cell cycle arrest and significant increases in protein and mRNA levels of p21. ASPL-TFE3 activated p21 expression in a p53-independent manner through direct transcriptional interactions with the p21 promoter region. When ASPL-TFE3 was expressed in human bone marrow–derived mesenchymal stem cells in a tetracycline-inducible manner, we observed the up-regulation of p21 expression and the induction of senescence-associated β-galactosidase activity. Suppression of p21 significantly decreased the induction of ASPL-TFE3-mediated cellular senescence. Furthermore, ASPL-TFE3 expression in mesenchymal stem cells resulted in a significant up-regulation of proinflammatory cytokines associated with senescence-associated secretory phenotype (SASP). These results show that ASPL-TFE3 regulates cell cycle progression and induces cellular senescence by up-regulating p21 expression. In addition, our data suggest a potential mechanism by which ASPL-TFE3-induced senescence may play a role in tumorigenesis by inducing SASP, which could promote the protumorigenic microenvironment.
Collapse
Affiliation(s)
- Naoko Ishiguro
- Department of Pathobiological Science and Technology, Faculty of Medicine, Tottori University, 86 Nishimachi, Yonago, Tottori 683-8503, Japan.
| | - Haruhiko Yoshida
- Department of Pathology, Yonago Medical Center, 4-17-1 Kuzumo, Yonago, Tottori 683-0006, Japan
| |
Collapse
|
42
|
Harwood JL, Alexander JH, Mayerson JL, Scharschmidt TJ. Targeted Chemotherapy in Bone and Soft-Tissue Sarcoma. Orthop Clin North Am 2015; 46:587-608. [PMID: 26410647 DOI: 10.1016/j.ocl.2015.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Historically surgical intervention has been the mainstay of therapy for bone and soft-tissue sarcomas, augmented with adjuvant radiation for local control. Although cytotoxic chemotherapy revolutionized the treatment of many sarcomas, classic treatment regimens are fraught with side effects while outcomes have plateaued. However, since the approval of imatinib in 2002, research into targeted chemotherapy has increased exponentially. With targeted therapies comes the potential for decreased side effects and more potent, personalized treatment options. This article reviews the evolution of medical knowledge regarding sarcoma, the basic science of sarcomatogenesis, and the major targets and pathways now being studied.
Collapse
Affiliation(s)
- Jared L Harwood
- Department of Orthopaedics, The Ohio State University, 725 Prior Hall, 376 West 10 Avenue, Columbus, OH 43210, USA
| | - John H Alexander
- Department of Orthopaedics, The Ohio State University, 725 Prior Hall, 376 West 10 Avenue, Columbus, OH 43210, USA
| | - Joel L Mayerson
- Department of Orthopaedics, The Ohio State University, 725 Prior Hall, 376 West 10 Avenue, Columbus, OH 43210, USA.
| | - Thomas J Scharschmidt
- Department of Orthopaedics, The Ohio State University, 725 Prior Hall, 376 West 10 Avenue, Columbus, OH 43210, USA
| |
Collapse
|
43
|
Paillard C, Coulomb A, Helfre S, Orbach D. [Alveolar soft part sarcoma in pediatric patients]. Bull Cancer 2015; 102:792-8. [PMID: 26235420 DOI: 10.1016/j.bulcan.2015.05.004] [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: 01/12/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
Abstract
Alveolar soft part sarcoma, ASPS, is a rare malignant tumor, with preferential primary localization in limbs, usually occurring in adolescents and young adults. This sarcoma, well defined histologically and at molecular level, has an indolent course, but a high potential metastatic pulmonary and cerebral evolution, sometimes late. ASPS is characterized by an almost specific translocation t(X, 17)(p11;25) which creates a fusion protein, APSL-TFE3, acting as an aberrant transcription factor. An in-bloc resection of the primary tumor is the treatment of choice in cases of localized disease. Conventional chemotherapy is generally ineffective. The role of radiotherapy is discussed in case of micro- or macroscopical incomplete residue. It seems to reduce local recurrence, but did not influence overall survival. The 5 years survival rate in children, adolescents and young adults is close to 80% in case of localized disease but poorer in presence of metastases. Recently, systemic anti-tumoral treatments have been focused on the use of targeted therapies. Anti-angiogenic drugs and tyrosine kinase inhibitors are the most promising approaches, but require further study. Prognostic risk factors in the literature are age (>10Y), tumor size (>5cm) and presence of metastases. This article reviews the clinical manifestations, diagnosis modalities, radiographic characteristics and therapeutic strategy of this disease in the pediatric population.
Collapse
Affiliation(s)
- Catherine Paillard
- Hôpital Hautepierre, service d'hématologie et d'oncologie pédiatrique, avenue Molière, 67098 Strasbourg, France; Inserm UMR-S1109, université de Strasbourg, laboratoire d'immuno-rhumatologie moléculaire, 67098 Strasbourg, France.
| | - Aurore Coulomb
- Hôpital Armand-Trousseau-Assistance publique, service d'anatomie pathologique, 26, avenue du Dr-Arnold-Netter, 75012 Paris, France
| | - Sylvie Helfre
- Institut Curie, service de radiothérapie, 26, rue d'Ulm, 75005 Paris, France
| | - Daniel Orbach
- Institut Curie, département d'oncologie pédiatrique adolescent jeune adulte, 26, rue d'Ulm, 75005 Paris, France
| |
Collapse
|
44
|
Diaz-Cano SJ. Pathological bases for a robust application of cancer molecular classification. Int J Mol Sci 2015; 16:8655-75. [PMID: 25898411 PMCID: PMC4425102 DOI: 10.3390/ijms16048655] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/07/2015] [Indexed: 12/12/2022] Open
Abstract
Any robust classification system depends on its purpose and must refer to accepted standards, its strength relying on predictive values and a careful consideration of known factors that can affect its reliability. In this context, a molecular classification of human cancer must refer to the current gold standard (histological classification) and try to improve it with key prognosticators for metastatic potential, staging and grading. Although organ-specific examples have been published based on proteomics, transcriptomics and genomics evaluations, the most popular approach uses gene expression analysis as a direct correlate of cellular differentiation, which represents the key feature of the histological classification. RNA is a labile molecule that varies significantly according with the preservation protocol, its transcription reflect the adaptation of the tumor cells to the microenvironment, it can be passed through mechanisms of intercellular transference of genetic information (exosomes), and it is exposed to epigenetic modifications. More robust classifications should be based on stable molecules, at the genetic level represented by DNA to improve reliability, and its analysis must deal with the concept of intratumoral heterogeneity, which is at the origin of tumor progression and is the byproduct of the selection process during the clonal expansion and progression of neoplasms. The simultaneous analysis of multiple DNA targets and next generation sequencing offer the best practical approach for an analytical genomic classification of tumors.
Collapse
Affiliation(s)
- Salvador J Diaz-Cano
- King's Health Partners, Cancer Studies, King's College Hospital-Viapath, Denmark Hill, London SE5-9RS, UK.
| |
Collapse
|
45
|
Alveolar soft part sarcoma of the oral and maxillofacial region: clinical analysis in a series of 18 patients. Oral Surg Oral Med Oral Pathol Oral Radiol 2015; 119:396-401. [DOI: 10.1016/j.oooo.2014.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/03/2014] [Accepted: 12/12/2014] [Indexed: 11/17/2022]
|
46
|
Goldberg JM, Fisher DE, Demetri GD, Neuberg D, Allsop SA, Fonseca C, Nakazaki Y, Nemer D, Raut CP, George S, Morgan JA, Wagner AJ, Freeman GJ, Ritz J, Lezcano C, Mihm M, Canning C, Hodi FS, Dranoff G. Biologic Activity of Autologous, Granulocyte-Macrophage Colony-Stimulating Factor Secreting Alveolar Soft-Part Sarcoma and Clear Cell Sarcoma Vaccines. Clin Cancer Res 2015; 21:3178-86. [PMID: 25805798 DOI: 10.1158/1078-0432.ccr-14-2932] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/20/2015] [Indexed: 12/17/2022]
Abstract
PURPOSE Alveolar soft-part sarcoma (ASPS) and clear cell sarcoma (CCS) are rare mesenchymal malignancies driven by chromosomal translocations that activate members of the microphthalmia transcription factor (MITF) family. However, in contrast to malignant melanoma, little is known about their immunogenicity. To learn more about the host response to ASPS and CCS, we conducted a phase I clinical trial of vaccination with irradiated, autologous sarcoma cells engineered by adenoviral-mediated gene transfer to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF). EXPERIMENTAL DESIGN Metastatic tumors from ASPS and CCS patients were resected, processed to single-cell suspensions, transduced with a replication-defective adenoviral vector encoding GM-CSF, and irradiated. Immunizations were administered subcutaneously and intradermally weekly three times and then every other week. RESULTS Vaccines were successfully manufactured for 11 of the 12 enrolled patients. Eleven subjects received from three to 13 immunizations. Toxicities were restricted to grade 1-2 skin reactions at inoculation sites. Vaccination elicited local dendritic cell infiltrates and stimulated T cell-mediated delayed-type hypersensitivity reactions to irradiated, autologous tumor cells. Antibody responses to tissue-type plasminogen activator (tTPA) and angiopoietins-1/2 were detected. Tumor biopsies showed programmed death-1 (PD-1)-positive CD8(+) T cells in association with PD ligand-1 (PD-L1)-expressing sarcoma cells. No tumor regressions were observed. CONCLUSIONS Vaccination with irradiated, GM-CSF-secreting autologous sarcoma cell vaccines is feasible, safe, and biologically active. Concurrent targeting of angiogenic cytokines and antagonism of the PD-1-negative regulatory pathway might intensify immune-mediated tumor destruction.
Collapse
Affiliation(s)
- John M Goldberg
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Pediatrics, Children's Hospital, Harvard Medical School, Boston, Massachusetts. Department of Pediatrics, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - David E Fisher
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Pediatrics, Children's Hospital, Harvard Medical School, Boston, Massachusetts. Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - George D Demetri
- Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Donna Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts
| | - Stephen A Allsop
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Catia Fonseca
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yukoh Nakazaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Nemer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Chandrajit P Raut
- Department of Surgical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Suzanne George
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey A Morgan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrew J Wagner
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Pediatrics, Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Cecilia Lezcano
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Martin Mihm
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christine Canning
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - F Stephen Hodi
- Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Glenn Dranoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Cancer Vaccine Center, Center for Immuno-oncology, and Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
47
|
Puls F, Niblett A, Clarke J, Kindblom LG, McCulloch T. YAP1-TFE3 epithelioid hemangioendothelioma: a case without vasoformation and a new transcript variant. Virchows Arch 2015; 466:473-8. [PMID: 25680571 DOI: 10.1007/s00428-015-1730-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/14/2015] [Accepted: 01/29/2015] [Indexed: 12/11/2022]
Abstract
Epithelioid hemangioendothelioma is a rare vascular tumor of borderline malignancy characterized by recurrent WWTR1-CAMTA1 gene fusions in approximately 90 % of cases. In addition, a recurrent YAP1-TFE3 gene fusion has been identified in WWTR1-CAMTA1 negative epithelioid hemangioendotheliomas. This subset has been reported as having a distinct morphology with more obvious vasoformation, voluminous eosinophilic cytoplasm, and TFE3 positivity on immunohistochemistry. We report a case of a YAP1-TFE3 translocated epithelioid hemangioendothelioma arising in a groin lymph node in a 29-year-old male. Plump spindle cell morphology and absence of vasoformation made correct diagnosis particularly difficult. Immunohistochemistry showed nuclear positivity for both ERG and TFE3, fluorescence in situ hybridization showed break apart for TFE3 and RT-PCR identified a YAP1 exon1 to TFE3 exon 6 transcript, a previously unreported fusion variant. Awareness of this solid morphology and variant fusion will aid in identification of future cases of this rare vascular tumor.
Collapse
Affiliation(s)
- Florian Puls
- Department of Musculoskeletal Pathology, Royal Orthopaedic Hospital NHS Foundation Trust, Birmingham, UK,
| | | | | | | | | |
Collapse
|
48
|
Abstract
The MiT subfamily of transcription factors includes TFE3, TFEB, TFC, and MiTF. Gene fusions involving two of these transcription factors have been identified in renal cell carcinoma (RCC). The Xp11 translocation RCCs were first officially recognized in the 2004 WHO renal tumor classification, and harbor gene fusions involving TFE3. The t(6;11) RCCs harbor a specific Alpha-TFEB gene fusion and were first officially recognized in the 2013 International Society of Urologic Pathology (ISUP) Vancouver classification of renal neoplasia. These two subtypes of translocation RCC have many similarities. Both were initially described in and disproportionately involve young patients, though adult translocation RCC may overall outnumber pediatric cases. Both often have unusual and distinctive morphologies; the Xp11 translocation RCCs frequently have clear cells with papillary architecture and abundant psammomatous bodies, while the t(6;11) RCCs frequently have a biphasic appearance with both large and small epithelioid cells and nodules of basement membrane material. However, the morphology of these two neoplasms can overlap, with one mimicking the other. Both of these RCCs underexpress epithelial immunohistochemical markers like cytokeratin and epithelial membrane antigen (EMA) relative to most other RCCs. Unlike other RCCs, both frequently express the cysteine protease cathepsin k and often express melanocytic markers like HMB45 and Melan A. Finally, TFE3 and TFEB have overlapping functional activity as these two transcription factors frequently heterodimerize and bind to the same targets. Therefore, on the basis of clinical, morphologic, immunohistochemical, and genetic similarities, the 2013 ISUP Vancouver classification of renal neoplasia grouped these two neoplasms together under the heading of "MiT family translocation RCC." This review summarizes our current knowledge of these recently described RCCs.
Collapse
Affiliation(s)
- Pedram Argani
- Department of Pathology, The Johns Hopkins University School of Medicine, The Johns Hopkins Hospital, 401 North Broadway, Weinberg 2242, Baltimore, Maryland 21231.
| |
Collapse
|
49
|
Goodwin ML, Jin H, Straessler K, Smith-Fry K, Zhu JF, Monument MJ, Grossmann A, Randall RL, Capecchi MR, Jones KB. Modeling alveolar soft part sarcomagenesis in the mouse: a role for lactate in the tumor microenvironment. Cancer Cell 2014; 26:851-862. [PMID: 25453902 PMCID: PMC4327935 DOI: 10.1016/j.ccell.2014.10.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 05/08/2014] [Accepted: 10/03/2014] [Indexed: 12/16/2022]
Abstract
Alveolar soft part sarcoma (ASPS), a deadly soft tissue malignancy with a predilection for adolescents and young adults, associates consistently with t(X;17) translocations that generate the fusion gene ASPSCR1-TFE3. We proved the oncogenic capacity of this fusion gene by driving sarcomagenesis in mice from conditional ASPSCR1-TFE3 expression. The completely penetrant tumors were indistinguishable from human ASPS by histology and gene expression. They formed preferentially in the anatomic environment highest in lactate, the cranial vault, expressed high levels of lactate importers, harbored abundant mitochondria, metabolized lactate as a metabolic substrate, and responded to the administration of exogenous lactate with tumor cell proliferation and angiogenesis. These data demonstrate lactate's role as a driver of alveolar soft part sarcomagenesis.
Collapse
Affiliation(s)
- Matthew L Goodwin
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA
| | - Huifeng Jin
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA; Center for Children's Cancer Research at the Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Krystal Straessler
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Kyllie Smith-Fry
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA; Center for Children's Cancer Research at the Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Ju-Fen Zhu
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA; Center for Children's Cancer Research at the Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Michael J Monument
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA; Center for Children's Cancer Research at the Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Allie Grossmann
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - R Lor Randall
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA; Center for Children's Cancer Research at the Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Mario R Capecchi
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Kevin B Jones
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA; Center for Children's Cancer Research at the Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
50
|
Clinical heterogeneity of Xp11 translocation renal cell carcinoma: impact of fusion subtype, age, and stage. Mod Pathol 2014; 27:875-86. [PMID: 24309327 DOI: 10.1038/modpathol.2013.208] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/13/2013] [Accepted: 09/24/2013] [Indexed: 12/16/2022]
Abstract
Xp11 translocation renal cell carcinomas harbor chromosome translocations involving the Xp11 breakpoint, resulting in gene fusions involving the TFE3 gene. The most common subtypes are the ASPSCR1-TFE3 renal cell carcinomas resulting from t(X;17)(p11;q25) translocation, and the PRCC-TFE3 renal cell carcinomas, resulting from t(X;1)(p11;q21) translocation. A formal clinical comparison of these two subtypes of Xp11 translocation renal cell carcinomas has not been performed. We report one new genetically confirmed Xp11 translocation renal cell carcinoma of each type. We also reviewed the literature for all published cases of ASPSCR1-TFE3 and PRCC-TFE3 renal cell carcinomas and contacted all corresponding authors to obtain or update the published follow-up information. Study of two new, unpublished cases, and review of the literature revealed that 8/8 patients who presented with distant metastasis had ASPSCR1-TFE3 renal cell carcinomas, and all but one of these patients either died of disease or had progressive disease. Regional lymph nodes were involved by metastasis in 24 of the 32 ASPSCR1-TFE3 cases in which nodes were resected, compared with 5 of 14 PRCC-TFE3 cases (P=0.02).; however, 11 of 13 evaluable patients with ASPSCR1-TFE3 renal cell carcinomas who presented with N1M0 disease remained disease free. Two PRCC-TFE3 renal cell carcinomas recurred late (at 20 and 30 years, respectively). In multivariate analysis, only older age or advanced stage at presentation (not fusion subtype) predicted death. In conclusion, ASPSCR1-TFE3 renal cell carcinomas are more likely to present at advanced stage (particularly node-positive disease) than are PRCC-TFE3 renal cell carcinomas. Although systemic metastases portend a grim prognosis, regional lymph node involvement does not, at least in short-term follow-up. The tendency for PRCC-TFE3 renal cell carcinomas to recur late warrants long-term follow-up.
Collapse
|