1
|
Yao J, Yao P, Li Y, He K, Ma X, Yang Q, Jia J, Chen Z, Yu S, Gu S, Chen K, Zhao Y, Li W, Wang G, Guo M. Integration of multi-omics data revealed the orphan CpG islands and enhancer-dominated c is-regulatory network in glioma. iScience 2024; 27:110946. [PMID: 39391717 PMCID: PMC11465130 DOI: 10.1016/j.isci.2024.110946] [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: 03/18/2024] [Revised: 07/12/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
The complex transcriptional regulatory network leads to the poor prognosis of glioma. The role of orphan CpG islands (oCGIs) in the transcriptional regulatory network has been overlooked. We conducted a comprehensive exploration of the cis-regulatory roles of oCGIs and enhancers by integrating multi-omics data. Direct regulation of target genes by oCGIs or enhancers is of great importance in the cis-regulatory network. Furthermore, based on single-cell multi-omics data, we found that the highly activated cis-regulatory network in cluster 2 (C2) sustains the high proliferative potential of glioma cells. The upregulation of oCGIs and enhancers related genes in C2 results in glioma patients exhibiting resistance to radiotherapy and chemotherapy. These findings were further validated through glioma cell line related experiments. Our study offers insight into the pathogenesis of glioma and provides a strategy to treat this challenging disease.
Collapse
Affiliation(s)
- Jiawei Yao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Penglei Yao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yang Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ke He
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xinqi Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Qingsong Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Junming Jia
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Zeren Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Shan Yu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Shuqing Gu
- Department of Neurosurgery, The First Hospital of Qiqihar, Qiqihar 161005, China
| | - Kunliang Chen
- Department of Neurosurgery, People’s Hospital of the Daxing’an Mountain Range, Daxing’an Mountain Range 165300, China
| | - Yan Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Weihua Li
- Medical Imaging Department, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Guangzhi Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Mian Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
2
|
Sprinzen L, Garcia F, Mela A, Lei L, Upadhyayula P, Mahajan A, Humala N, Manier L, Caprioli R, Quiñones-Hinojosa A, Casaccia P, Canoll P. EZH2 Inhibition Sensitizes IDH1R132H-Mutant Gliomas to Histone Deacetylase Inhibitor. Cells 2024; 13:219. [PMID: 38334611 PMCID: PMC10854521 DOI: 10.3390/cells13030219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Abstract
Isocitrate Dehydrogenase-1 (IDH1) is commonly mutated in lower-grade diffuse gliomas. The IDH1R132H mutation is an important diagnostic tool for tumor diagnosis and prognosis; however, its role in glioma development, and its impact on response to therapy, is not fully understood. We developed a murine model of proneural IDH1R132H-mutated glioma that shows elevated production of 2-hydroxyglutarate (2-HG) and increased trimethylation of lysine residue K27 on histone H3 (H3K27me3) compared to IDH1 wild-type tumors. We found that using Tazemetostat to inhibit the methyltransferase for H3K27, Enhancer of Zeste 2 (EZH2), reduced H3K27me3 levels and increased acetylation on H3K27. We also found that, although the histone deacetylase inhibitor (HDACi) Panobinostat was less cytotoxic in IDH1R132H-mutated cells (either isolated from murine glioma or oligodendrocyte progenitor cells infected in vitro with a retrovirus expressing IDH1R132H) compared to IDH1-wild-type cells, combination treatment with Tazemetostat is synergistic in both mutant and wild-type models. These findings indicate a novel therapeutic strategy for IDH1-mutated gliomas that targets the specific epigenetic alteration in these tumors.
Collapse
Affiliation(s)
- Lisa Sprinzen
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; (L.S.); (F.G.); (A.M.)
| | - Franklin Garcia
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; (L.S.); (F.G.); (A.M.)
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; (L.S.); (F.G.); (A.M.)
| | - Liang Lei
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10032, USA; (L.L.); (P.U.); (N.H.)
| | - Pavan Upadhyayula
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10032, USA; (L.L.); (P.U.); (N.H.)
| | - Aayushi Mahajan
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10032, USA; (L.L.); (P.U.); (N.H.)
| | - Nelson Humala
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10032, USA; (L.L.); (P.U.); (N.H.)
| | - Lisa Manier
- Department of Chemistry, Vanderbilt School of Medicine, Nashville, TN 37240, USA; (L.M.); (R.C.)
| | - Richard Caprioli
- Department of Chemistry, Vanderbilt School of Medicine, Nashville, TN 37240, USA; (L.M.); (R.C.)
| | | | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center, City University of New York, New York, NY 10031, USA;
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; (L.S.); (F.G.); (A.M.)
| |
Collapse
|
3
|
Zage PE, Huo Y, Subramonian D, Le Clorennec C, Ghosh P, Sahoo D. Identification of a novel gene signature for neuroblastoma differentiation using a Boolean implication network. Genes Chromosomes Cancer 2023; 62:313-331. [PMID: 36680522 PMCID: PMC10257350 DOI: 10.1002/gcc.23124] [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: 04/01/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
Although induction of differentiation represents an effective strategy for neuroblastoma treatment, the mechanisms underlying neuroblastoma differentiation are poorly understood. We generated a computational model of neuroblastoma differentiation consisting of interconnected gene clusters identified based on symmetric and asymmetric gene expression relationships. We identified a differentiation signature consisting of series of gene clusters comprised of 1251 independent genes that predicted neuroblastoma differentiation in independent datasets and in neuroblastoma cell lines treated with agents known to induce differentiation. This differentiation signature was associated with patient outcomes in multiple independent patient cohorts and validated the role of MYCN expression as a marker of neuroblastoma differentiation. Our results further identified novel genes associated with MYCN via asymmetric Boolean implication relationships that would not have been identified using symmetric computational approaches and that were associated with both neuroblastoma differentiation and patient outcomes. Our differentiation signature included a cluster of genes involved in intracellular signaling and growth factor receptor trafficking pathways that is strongly associated with neuroblastoma differentiation, and we validated the associations of UBE4B, a gene within this cluster, with neuroblastoma cell and tumor differentiation. Our findings demonstrate that Boolean network analyses of symmetric and asymmetric gene expression relationships can identify novel genes and pathways relevant for neuroblastoma tumor differentiation that could represent potential therapeutic targets.
Collapse
Affiliation(s)
- Peter E. Zage
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
| | - Yuchen Huo
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
| | - Divya Subramonian
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
| | - Christophe Le Clorennec
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
| | - Pradipta Ghosh
- Department of Medicine, UCSD, La Jolla, CA
- Department of Cellular and Molecular Medicine, UCSD, La Jolla, CA
- Veterans Affairs Medical Center, La Jolla, CA
| | - Debashis Sahoo
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
- Department of Computer Science and Engineering, Jacobs School of Engineering, UCSD, La Jolla, CA
| |
Collapse
|
4
|
Zhang P, Rashidi A, Zhao J, Silvers C, Wang H, Castro B, Ellingwood A, Han Y, Lopez-Rosas A, Zannikou M, Dmello C, Levine R, Xiao T, Cordero A, Sonabend AM, Balyasnikova IV, Lee-Chang C, Miska J, Lesniak MS. STING agonist-loaded, CD47/PD-L1-targeting nanoparticles potentiate antitumor immunity and radiotherapy for glioblastoma. Nat Commun 2023; 14:1610. [PMID: 36959214 PMCID: PMC10036562 DOI: 10.1038/s41467-023-37328-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/08/2023] [Indexed: 03/25/2023] Open
Abstract
As a key component of the standard of care for glioblastoma, radiotherapy induces several immune resistance mechanisms, such as upregulation of CD47 and PD-L1. Here, leveraging these radiotherapy-elicited processes, we generate a bridging-lipid nanoparticle (B-LNP) that engages tumor-associated myeloid cells (TAMCs) to glioblastoma cells via anti-CD47/PD-L1 dual ligation. We show that the engager B-LNPs block CD47 and PD-L1 and promote TAMC phagocytic activity. To enhance subsequent T cell recruitment and antitumor responses after tumor engulfment, the B-LNP was encapsulated with diABZI, a non-nucleotidyl agonist for stimulator of interferon genes. In vivo treatment with diABZI-loaded B-LNPs induced a transcriptomic and metabolic switch in TAMCs, turning these immunosuppressive cells into antitumor effectors, which induced T cell infiltration and activation in brain tumors. In preclinical murine models, B-LNP/diABZI administration synergized with radiotherapy to promote brain tumor regression and induce immunological memory against glioma. In summary, our study describes a nanotechnology-based approach that hijacks irradiation-triggered immune checkpoint molecules to boost potent and long-lasting antitumor immunity against glioblastoma.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Aida Rashidi
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Junfei Zhao
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Caylee Silvers
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hanxiang Wang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brandyn Castro
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Abby Ellingwood
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yu Han
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Aurora Lopez-Rosas
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Markella Zannikou
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rebecca Levine
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ting Xiao
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Alex Cordero
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam M Sonabend
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason Miska
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Maciej S Lesniak
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
5
|
Larionova TD, Bastola S, Aksinina TE, Anufrieva KS, Wang J, Shender VO, Andreev DE, Kovalenko TF, Arapidi GP, Shnaider PV, Kazakova AN, Latyshev YA, Tatarskiy VV, Shtil AA, Moreau P, Giraud F, Li C, Wang Y, Rubtsova MP, Dontsova OA, Condro M, Ellingson BM, Shakhparonov MI, Kornblum HI, Nakano I, Pavlyukov MS. Alternative RNA splicing modulates ribosomal composition and determines the spatial phenotype of glioblastoma cells. Nat Cell Biol 2022; 24:1541-1557. [PMID: 36192632 PMCID: PMC10026424 DOI: 10.1038/s41556-022-00994-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/15/2022] [Indexed: 02/08/2023]
Abstract
Glioblastoma (GBM) is characterized by exceptionally high intratumoral heterogeneity. However, the molecular mechanisms underlying the origin of different GBM cell populations remain unclear. Here, we found that the compositions of ribosomes of GBM cells in the tumour core and edge differ due to alternative RNA splicing. The acidic pH in the core switches before messenger RNA splicing of the ribosomal gene RPL22L1 towards the RPL22L1b isoform. This allows cells to survive acidosis, increases stemness and correlates with worse patient outcome. Mechanistically, RPL22L1b promotes RNA splicing by interacting with lncMALAT1 in the nucleus and inducing its degradation. Contrarily, in the tumour edge region, RPL22L1a interacts with ribosomes in the cytoplasm and upregulates the translation of multiple messenger RNAs including TP53. We found that the RPL22L1 isoform switch is regulated by SRSF4 and identified a compound that inhibits this process and decreases tumour growth. These findings demonstrate how distinct GBM cell populations arise during tumour growth. Targeting this mechanism may decrease GBM heterogeneity and facilitate therapy.
Collapse
Affiliation(s)
- Tatyana D Larionova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation
| | - Soniya Bastola
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Tatiana E Aksinina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation
| | - Ksenia S Anufrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, Russian Federation
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical and Biological Agency, Moscow, Russian Federation
| | - Jia Wang
- Department of Neurosurgery, Centre of Brain Science, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Victoria O Shender
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, Russian Federation
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical and Biological Agency, Moscow, Russian Federation
| | - Dmitriy E Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Tatiana F Kovalenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation
| | - Georgij P Arapidi
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, Russian Federation
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical and Biological Agency, Moscow, Russian Federation
| | - Polina V Shnaider
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, Russian Federation
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical and Biological Agency, Moscow, Russian Federation
| | - Anastasia N Kazakova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical and Biological Agency, Moscow, Russian Federation
| | - Yaroslav A Latyshev
- N.N. Burdenko National Medical Research Center of Neurosurgery, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | - Alexander A Shtil
- Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation
| | - Pascale Moreau
- Institute of Chemistry of Clermont-Ferrand, CNRS, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Francis Giraud
- Institute of Chemistry of Clermont-Ferrand, CNRS, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Chaoxi Li
- Department of Neurosurgery, School of Medicine and O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yichan Wang
- Department of Neurosurgery, Centre of Brain Science, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Maria P Rubtsova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Olga A Dontsova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russian Federation
| | - Michael Condro
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Benjamin M Ellingson
- Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Harley I Kornblum
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ichiro Nakano
- Department of Neurosurgery, Medical Institute of Hokuto, Hokkaido, Japan.
| | - Marat S Pavlyukov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation.
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.
| |
Collapse
|
6
|
Ke X, Wu H, Chen YX, Guo Y, Yao S, Guo MR, Duan YY, Wang NN, Shi W, Wang C, Dong SS, Kang H, Dai Z, Yang TL. Individualized pathway activity algorithm identifies oncogenic pathways in pan-cancer analysis. EBioMedicine 2022; 79:104014. [PMID: 35487057 PMCID: PMC9117264 DOI: 10.1016/j.ebiom.2022.104014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 02/07/2023] Open
Abstract
Background Accumulative evidences have shown that dysregulation of biological pathways contributed to the initiation and progression of malignant tumours. Several methods for pathway activity measurement have been proposed, but they are restricted to making comparisons between groups or sensitive to experimental batch effects. Methods We introduced a novel method for individualized pathway activity measurement (IPAM) that is based on the ranking of gene expression levels in individual sample. Taking advantage of IPAM, we calculated the pathway activity of 318 pathways from KEGG database in the 10528 tumour/normal samples of 33 cancer types from TCGA to identify characteristic dysregulated pathways among different cancer types. Findings IPAM precisely quantified the level of activity of each pathway in pan-cancer analysis and exhibited better performance in cancer classification and prognosis prediction over five widely used tools. The average ROC-AUC of cancer diagnostic model using tumour-educated platelets (TEPs) reached 92.84%, suggesting the potential of our algorithm in early diagnosis of cancer. We identified several pathways significantly deregulated and associated with patient survival in a large fraction of cancer types, such as tyrosine metabolism, fatty acid degradation, cell cycle, p53 signalling pathway and DNA replication. We also confirmed the dominant role of metabolic pathways in cancer pathway dysregulation and identified the driving factors of specific pathway dysregulation, such as PPARA for branched-chain amino acid metabolism and NR1I2, NR1I3 for fatty acid metabolism. Interpretation Our study will provide novel clues for understanding the pathological mechanisms of cancer, ultimately paving the way for personalized medicine of cancer. Funding A full list of funding can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Xin Ke
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Hao Wu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Yi-Xiao Chen
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Shi Yao
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Ming-Rui Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Yuan-Yuan Duan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Nai-Ning Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Wei Shi
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Chen Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China; National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China.
| |
Collapse
|
7
|
La Ferlita A, Alaimo S, Ferro A, Pulvirenti A. Pathway Analysis for Cancer Research and Precision Oncology Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:143-161. [DOI: 10.1007/978-3-030-91836-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
8
|
Yu CY, Mitrofanova A. Mechanism-Centric Approaches for Biomarker Detection and Precision Therapeutics in Cancer. Front Genet 2021; 12:687813. [PMID: 34408770 PMCID: PMC8365516 DOI: 10.3389/fgene.2021.687813] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Biomarker discovery is at the heart of personalized treatment planning and cancer precision therapeutics, encompassing disease classification and prognosis, prediction of treatment response, and therapeutic targeting. However, many biomarkers represent passenger rather than driver alterations, limiting their utilization as functional units for therapeutic targeting. We suggest that identification of driver biomarkers through mechanism-centric approaches, which take into account upstream and downstream regulatory mechanisms, is fundamental to the discovery of functionally meaningful markers. Here, we examine computational approaches that identify mechanism-centric biomarkers elucidated from gene co-expression networks, regulatory networks (e.g., transcriptional regulation), protein-protein interaction (PPI) networks, and molecular pathways. We discuss their objectives, advantages over gene-centric approaches, and known limitations. Future directions highlight the importance of input and model interpretability, method and data integration, and the role of recently introduced technological advantages, such as single-cell sequencing, which are central for effective biomarker discovery and time-cautious precision therapeutics.
Collapse
Affiliation(s)
- Christina Y. Yu
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Antonina Mitrofanova
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
9
|
Zhao N, Wang F, Ahmed S, Liu K, Zhang C, Cathcart SJ, DiMaio DJ, Punsoni M, Guan B, Zhou P, Wang S, Batra SK, Bronich T, Hei TK, Lin C, Zhang C. Androgen Receptor, Although Not a Specific Marker For, Is a Novel Target to Suppress Glioma Stem Cells as a Therapeutic Strategy for Glioblastoma. Front Oncol 2021; 11:616625. [PMID: 34094902 PMCID: PMC8175980 DOI: 10.3389/fonc.2021.616625] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/23/2021] [Indexed: 11/13/2022] Open
Abstract
Targeting androgen receptor (AR) has been shown to be promising in treating glioblastoma (GBM) in cell culture and flank implant models but the mechanisms remain unclear. AR antagonists including enzalutamide are available for treating prostate cancer patients in clinic and can pass the blood-brain barrier, thus are potentially good candidates for GBM treatment but have not been tested in GBM orthotopically. Our current studies confirmed that in patients, a majority of GBM tumors overexpress AR in both genders. Enzalutamide inhibited the proliferation of GBM cells both in vitro and in vivo. Although confocal microscopy demonstrated that AR is expressed but not specifically in glioma cancer stem cells (CSCs) (CD133+), enzalutamide treatment significantly decreased CSC population in cultured monolayer cells and spheroids, suppressed tumor sphere-forming capacity of GBM cells, and downregulated CSC gene expression at mRNA and protein levels in a dose- and time-dependent manner. We have, for the first time, demonstrated that enzalutamide treatment decreased the density of CSCs in vivo and improved survival in an orthotopic GBM mouse model. We conclude that AR antagonists potently target glioma CSCs in addition to suppressing the overall proliferation of GBM cells as a mechanism supporting their repurposing for clinical applications treating GBM.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Radiation Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fei Wang
- Department of Radiation Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shaheen Ahmed
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kan Liu
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Sahara J Cathcart
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Dominick J DiMaio
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michael Punsoni
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Bingjie Guan
- Department of Radiation Oncology, Union Hospital of Fujian Medical University, Fuzhou, China
| | - Ping Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shuo Wang
- Department of Radiation Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, United States
| | - Tatiana Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Tom K Hei
- Center for Radiological Research, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Chi Lin
- Department of Radiation Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Chi Zhang
- Department of Radiation Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
10
|
Convection Enhanced Delivery of Topotecan for Gliomas: A Single-Center Experience. Pharmaceutics 2020; 13:pharmaceutics13010039. [PMID: 33396668 PMCID: PMC7823846 DOI: 10.3390/pharmaceutics13010039] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022] Open
Abstract
A key limitation to glioma treatment involves the blood brain barrier (BBB). Convection enhanced delivery (CED) is a technique that uses a catheter placed directly into the brain parenchyma to infuse treatments using a pressure gradient. In this manuscript, we describe the physical principles behind CED along with the common pitfalls and methods for optimizing convection. Finally, we highlight our institutional experience using topotecan CED for the treatment of malignant glioma.
Collapse
|
11
|
Fang J, Pian C, Xu M, Kong L, Li Z, Ji J, Zhang L, Chen Y. Revealing Prognosis-Related Pathways at the Individual Level by a Comprehensive Analysis of Different Cancer Transcription Data. Genes (Basel) 2020; 11:genes11111281. [PMID: 33138076 PMCID: PMC7692404 DOI: 10.3390/genes11111281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Identifying perturbed pathways at an individual level is important to discover the causes of cancer and develop individualized custom therapeutic strategies. Though prognostic gene lists have had success in prognosis prediction, using single genes that are related to the relevant system or specific network cannot fully reveal the process of tumorigenesis. We hypothesize that in individual samples, the disruption of transcription homeostasis can influence the occurrence, development, and metastasis of tumors and has implications for patient survival outcomes. Here, we introduced the individual-level pathway score, which can measure the correlation perturbation of the pathways in a single sample well. We applied this method to the expression data of 16 different cancer types from The Cancer Genome Atlas (TCGA) database. Our results indicate that different cancer types as well as their tumor-adjacent tissues can be clearly distinguished by the individual-level pathway score. Additionally, we found that there was strong heterogeneity among different cancer types and the percentage of perturbed pathways as well as the perturbation proportions of tumor samples in each pathway were significantly different. Finally, the prognosis-related pathways of different cancer types were obtained by survival analysis. We demonstrated that the individual-level pathway score (iPS) is capable of classifying cancer types and identifying some key prognosis-related pathways.
Collapse
Affiliation(s)
- Jingya Fang
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Cong Pian
- Department of Mathematics, College of Science, Nanjing Agricultural University, Nanjing 210095, China;
| | - Mingmin Xu
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Lingpeng Kong
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Zutan Li
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Jinwen Ji
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
| | - Liangyun Zhang
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; (J.F.); (M.X.); (L.K.); (Z.L.); (J.J.)
- Correspondence: (L.Z.); (Y.C.)
| | - Yuanyuan Chen
- Department of Mathematics, College of Science, Nanjing Agricultural University, Nanjing 210095, China;
- Correspondence: (L.Z.); (Y.C.)
| |
Collapse
|
12
|
Kane JR, Zhao J, Tsujiuchi T, Laffleur B, Arrieta VA, Mahajan A, Rao G, Mela A, Dmello C, Chen L, Zhang DY, González-Buendia E, Lee-Chang C, Xiao T, Rothschild G, Basu U, Horbinski C, Lesniak MS, Heimberger AB, Rabadan R, Canoll P, Sonabend AM. CD8 + T-cell-Mediated Immunoediting Influences Genomic Evolution and Immune Evasion in Murine Gliomas. Clin Cancer Res 2020; 26:4390-4401. [PMID: 32430477 DOI: 10.1158/1078-0432.ccr-19-3104] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 03/27/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023]
Abstract
PURPOSE Cancer immunoediting shapes tumor progression by the selection of tumor cell variants that can evade immune recognition. Given the immune evasion and intratumor heterogeneity characteristic of gliomas, we hypothesized that CD8+ T cells mediate immunoediting in these tumors. EXPERIMENTAL DESIGN We developed retrovirus-induced PDGF+ Pten -/- murine gliomas and evaluated glioma progression and tumor immunogenicity in the absence of CD8+ T cells by depleting this immune cell population. Furthermore, we characterized the genomic alterations present in gliomas that developed in the presence and absence of CD8+ T cells. RESULTS Upon transplantation, gliomas that developed in the absence of CD8+ T cells engrafted poorly in recipients with intact immunity but engrafted well in those with CD8+ T-cell depletion. In contrast, gliomas that developed under pressure from CD8+ T cells were able to fully engraft in both CD8+ T-cell-depleted mice and immunocompetent mice. Remarkably, gliomas developed in the absence of CD8+ T cells exhibited increased aneuploidy, MAPK pathway signaling, gene fusions, and macrophage/microglial infiltration, and showed a proinflammatory phenotype. MAPK activation correlated with macrophage/microglia recruitment in this model and in the human disease. CONCLUSIONS Our studies indicate that, in these tumor models, CD8+ T cells influence glioma oncogenic pathways, tumor genotype, and immunogenicity. This suggests immunoediting of immunogenic tumor clones through their negative selection by CD8+ T cells during glioma formation.
Collapse
Affiliation(s)
- Joshua R Kane
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Junfei Zhao
- Department of Systems Biology, Columbia University, New York City, New York.,Department of Biomedical Informatics, Columbia University, New York City, New York
| | - Takashi Tsujiuchi
- Department of Neurosurgery, Columbia University, New York City, New York
| | - Brice Laffleur
- Department of Microbiology and Immunology, Columbia University, New York City, New York
| | - Víctor A Arrieta
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aayushi Mahajan
- Department of Neurosurgery, Columbia University, New York City, New York
| | - Ganesh Rao
- Department of Neurological Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University, New York City, New York
| | - Crismita Dmello
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Li Chen
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Daniel Y Zhang
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Edgar González-Buendia
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Catalina Lee-Chang
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ting Xiao
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Gerson Rothschild
- Department of Microbiology and Immunology, Columbia University, New York City, New York
| | - Uttiya Basu
- Department of Microbiology and Immunology, Columbia University, New York City, New York
| | - Craig Horbinski
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Maciej S Lesniak
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Amy B Heimberger
- Department of Neurological Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raul Rabadan
- Department of Systems Biology, Columbia University, New York City, New York.,Department of Biomedical Informatics, Columbia University, New York City, New York.,Department of Mathematical Genomics, Columbia University, New York City, New York
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York City, New York
| | - Adam M Sonabend
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
13
|
Gampa G, Kenchappa RS, Mohammad AS, Parrish KE, Kim M, Crish JF, Luu A, West R, Hinojosa AQ, Sarkaria JN, Rosenfeld SS, Elmquist WF. Enhancing Brain Retention of a KIF11 Inhibitor Significantly Improves its Efficacy in a Mouse Model of Glioblastoma. Sci Rep 2020; 10:6524. [PMID: 32300151 PMCID: PMC7162859 DOI: 10.1038/s41598-020-63494-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/21/2020] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma, the most lethal primary brain cancer, is extremely proliferative and invasive. Tumor cells at tumor/brain-interface often exist behind a functionally intact blood-brain barrier (BBB), and so are shielded from exposure to therapeutic drug concentrations. An ideal glioblastoma treatment needs to engage targets that drive proliferation as well as invasion, with brain penetrant therapies. One such target is the mitotic kinesin KIF11, which can be inhibited with ispinesib, a potent molecularly-targeted drug. Although, achieving durable brain exposures of ispinesib is critical for adequate tumor cell engagement during mitosis, when tumor cells are vulnerable, for efficacy. Our results demonstrate that the delivery of ispinesib is restricted by P-gp and Bcrp efflux at BBB. Thereby, ispinesib distribution is heterogeneous with concentrations substantially lower in invasive tumor rim (intact BBB) compared to glioblastoma core (disrupted BBB). We further find that elacridar—a P-gp and Bcrp inhibitor—improves brain accumulation of ispinesib, resulting in remarkably reduced tumor growth and extended survival in a rodent model of glioblastoma. Such observations show the benefits and feasibility of pairing a potentially ideal treatment with a compound that improves its brain accumulation, and supports use of this strategy in clinical exploration of cell cycle-targeting therapies in brain cancers.
Collapse
Affiliation(s)
- Gautham Gampa
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | | | - Afroz S Mohammad
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Karen E Parrish
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Minjee Kim
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - James F Crish
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA
| | - Amanda Luu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Rita West
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
14
|
Lentiviral Vector Induced Modeling of High-Grade Spinal Cord Glioma in Minipigs. Sci Rep 2020; 10:5291. [PMID: 32210315 PMCID: PMC7093438 DOI: 10.1038/s41598-020-62167-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/09/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Prior studies have applied driver mutations targeting the RTK/RAS/PI3K and p53 pathways to induce the formation of high-grade gliomas in rodent models. In the present study, we report the production of a high-grade spinal cord glioma model in pigs using lentiviral gene transfer. METHODS Six Gottingen Minipigs received thoracolumbar (T14-L1) lateral white matter injections of a combination of lentiviral vectors, expressing platelet-derived growth factor beta (PDGF-B), constitutive HRAS, and shRNA-p53 respectively. All animals received injection of control vectors into the contralateral cord. Animals underwent baseline and endpoint magnetic resonance imaging (MRI) and were evaluated daily for clinical deficits. Hematoxylin and eosin (H&E) and immunohistochemical analysis was conducted. Data are presented using descriptive statistics including relative frequencies, mean, standard deviation, and range. RESULTS 100% of animals (n = 6/6) developed clinical motor deficits ipsilateral to the oncogenic lentiviral injections by a three-week endpoint. MRI scans at endpoint demonstrated contrast enhancing mass lesions at the site of oncogenic lentiviral injection and not at the site of control injections. Immunohistochemistry demonstrated positive staining for GFAP, Olig2, and a high Ki-67 proliferative index. Histopathologic features demonstrate consistent and reproducible growth of a high-grade glioma in all animals. CONCLUSIONS Lentiviral gene transfer represents a feasible pathway to glioma modeling in higher order species. The present model is the first lentiviral vector induced pig model of high-grade spinal cord glioma and may potentially be used in preclinical therapeutic development programs.
Collapse
|
15
|
Das P, Peterson CB, Do KA, Akbani R, Baladandayuthapani V. NExUS: Bayesian simultaneous network estimation across unequal sample sizes. Bioinformatics 2020; 36:798-804. [PMID: 31504175 PMCID: PMC8215919 DOI: 10.1093/bioinformatics/btz636] [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: 11/08/2018] [Revised: 06/25/2019] [Accepted: 08/26/2019] [Indexed: 01/31/2023] Open
Abstract
MOTIVATION Network-based analyses of high-throughput genomics data provide a holistic, systems-level understanding of various biological mechanisms for a common population. However, when estimating multiple networks across heterogeneous sub-populations, varying sample sizes pose a challenge in the estimation and inference, as network differences may be driven by differences in power. We are particularly interested in addressing this challenge in the context of proteomic networks for related cancers, as the number of subjects available for rare cancer (sub-)types is often limited. RESULTS We develop NExUS (Network Estimation across Unequal Sample sizes), a Bayesian method that enables joint learning of multiple networks while avoiding artefactual relationship between sample size and network sparsity. We demonstrate through simulations that NExUS outperforms existing network estimation methods in this context, and apply it to learn network similarity and shared pathway activity for groups of cancers with related origins represented in The Cancer Genome Atlas (TCGA) proteomic data. AVAILABILITY AND IMPLEMENTATION The NExUS source code is freely available for download at https://github.com/priyamdas2/NExUS. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Priyam Das
- Department of Biostatistics, TX 77030, USA
| | | | - Kim-Anh Do
- Department of Biostatistics, TX 77030, USA
| | - Rehan Akbani
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
16
|
Gill BJA, Wu X, Khan FA, Sosunov AA, Liou JY, Dovas A, Eissa TL, Banu MA, Bateman LM, McKhann GM, Canoll P, Schevon C. Ex vivo multi-electrode analysis reveals spatiotemporal dynamics of ictal behavior at the infiltrated margin of glioma. Neurobiol Dis 2019; 134:104676. [PMID: 31731042 PMCID: PMC8147009 DOI: 10.1016/j.nbd.2019.104676] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/22/2019] [Accepted: 11/11/2019] [Indexed: 01/02/2023] Open
Abstract
The purpose of this study is to develop a platform in which the cellular and molecular underpinnings of chronic focal neocortical lesional epilepsy can be explored and use it to characterize seizure-like events (SLEs) in an ex vivo model of infiltrating high-grade glioma. Microelectrode arrays were used to study electrophysiologic changes in ex vivo acute brain slices from a PTEN/p53 deleted, PDGF-B driven mouse model of high-grade glioma. Electrode locations were co-registered to the underlying histology to ascertain the influence of the varying histologic landscape on the observed electrophysiologic changes. Peritumoral, infiltrated, and tumor sites were sampled in tumor-bearing slices. Following the addition of zero Mg2+ solution, all three histologic regions in tumor-bearing slices showed significantly greater increases in firing rates when compared to the control sites. Tumor-bearing slices demonstrated increased proclivity for SLEs, with 40 events in tumor-bearing slices and 5 events in control slices (p-value = .0105). Observed SLEs were characterized by either low voltage fast (LVF) onset patterns or short bursts of repetitive widespread, high amplitude low frequency discharges. Seizure foci comprised areas from all three histologic regions. The onset electrode was found to be at the infiltrated margin in 50% of cases and in the peritumoral region in 36.9% of cases. These findings reveal a landscape of histopathologic and electrophysiologic alterations associated with ictogenesis and spread of tumor-associated seizures.
Collapse
Affiliation(s)
- Brian J A Gill
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA.
| | - Xiaoping Wu
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Farhan A Khan
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Alexander A Sosunov
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Jyun-You Liou
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Tahra L Eissa
- Department of Applied Mathematics, University of Colorado at Boulder, Boulder, CO, USA
| | - Matei A Banu
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Lisa M Bateman
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Guy M McKhann
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Catherine Schevon
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
17
|
Myosin IIA suppresses glioblastoma development in a mechanically sensitive manner. Proc Natl Acad Sci U S A 2019; 116:15550-15559. [PMID: 31235578 DOI: 10.1073/pnas.1902847116] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The ability of glioblastoma to disperse through the brain contributes to its lethality, and blocking this behavior has been an appealing therapeutic approach. Although a number of proinvasive signaling pathways are active in glioblastoma, many are redundant, so targeting one can be overcome by activating another. However, these pathways converge on nonredundant components of the cytoskeleton, and we have shown that inhibiting one of these-the myosin II family of cytoskeletal motors-blocks glioblastoma invasion even with simultaneous activation of multiple upstream promigratory pathways. Myosin IIA and IIB are the most prevalent isoforms of myosin II in glioblastoma, and we now show that codeleting these myosins markedly impairs tumorigenesis and significantly prolongs survival in a rodent model of this disease. However, while targeting just myosin IIA also impairs tumor invasion, it surprisingly increases tumor proliferation in a manner that depends on environmental mechanics. On soft surfaces myosin IIA deletion enhances ERK1/2 activity, while on stiff surfaces it enhances the activity of NFκB, not only in glioblastoma but in triple-negative breast carcinoma and normal keratinocytes as well. We conclude myosin IIA suppresses tumorigenesis in at least two ways that are modulated by the mechanics of the tumor and its stroma. Our results also suggest that inhibiting tumor invasion can enhance tumor proliferation and that effective therapy requires targeting cellular components that drive both proliferation and invasion simultaneously.
Collapse
|
18
|
Aouiche C, Chen B, Shang X. Predicting stage-specific cancer related genes and their dynamic modules by integrating multiple datasets. BMC Bioinformatics 2019; 20:194. [PMID: 31074385 PMCID: PMC6509867 DOI: 10.1186/s12859-019-2740-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The mechanism of many complex diseases has not been detected accurately in terms of their stage evolution. Previous studies mainly focus on the identification of associations between genes and individual diseases, but less is known about their associations with specific disease stages. Exploring biological modules through different disease stages could provide valuable knowledge to genomic and clinical research. RESULTS In this study, we proposed a powerful and versatile framework to identify stage-specific cancer related genes and their dynamic modules by integrating multiple datasets. The discovered modules and their specific-signature genes were significantly enriched in many relevant known pathways. To further illustrate the dynamic evolution of these clinical-stages, a pathway network was built by taking individual pathways as vertices and the overlapping relationship between their annotated genes as edges. CONCLUSIONS The identified pathway network not only help us to understand the functional evolution of complex diseases, but also useful for clinical management to select the optimum treatment regimens and the appropriate drugs for patients.
Collapse
Affiliation(s)
- Chaima Aouiche
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University Ministry of Industry and Information Technology, Xi'an, China
| | - Bolin Chen
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China. .,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University Ministry of Industry and Information Technology, Xi'an, China.
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University Ministry of Industry and Information Technology, Xi'an, China
| |
Collapse
|
19
|
Lukas RV, Wainwright DA, Ladomersky E, Sachdev S, Sonabend AM, Stupp R. Newly Diagnosed Glioblastoma: A Review on Clinical Management. ONCOLOGY (WILLISTON PARK, N.Y.) 2019; 33:91-100. [PMID: 30866031 PMCID: PMC7278092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Glioblastoma is an aggressive primary tumor of the central nervous system. This review will focus on clinical developments and management of newly diagnosed disease, including a discussion about the incorporation of molecular features into the classification of glioblastoma. Such advances will continue to shape our thinking about the disease and how to best manage it. With regards to treatment, the role of surgical resection, radiotherapy, chemotherapy, and tumor-treating fields will be presented. Pivotal studies defining our current standard of care will be highlighted, as will key ongoing trials that may influence our management of glioblastoma in the near future.
Collapse
|
20
|
Bansal M, He J, Peyton M, Kustagi M, Iyer A, Comb M, White M, Minna JD, Califano A. Elucidating synergistic dependencies in lung adenocarcinoma by proteome-wide signaling-network analysis. PLoS One 2019; 14:e0208646. [PMID: 30615629 PMCID: PMC6322741 DOI: 10.1371/journal.pone.0208646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/20/2018] [Indexed: 12/26/2022] Open
Abstract
To understand drug combination effect, it is necessary to decipher the interactions between drug targets-many of which are signaling molecules. Previously, such signaling pathway models are largely based on the compilation of literature data from heterogeneous cellular contexts. Indeed, de novo reconstruction of signaling interactions from large-scale molecular profiling is still lagging, compared to similar efforts in transcriptional and protein-protein interaction networks. To address this challenge, we introduce a novel algorithm for the systematic inference of protein kinase pathways, and applied it to published mass spectrometry-based phosphotyrosine profile data from 250 lung adenocarcinoma (LUAD) samples. The resulting network includes 43 TKs and 415 inferred, LUAD-specific substrates, which were validated at >60% accuracy by SILAC assays, including "novel' substrates of the EGFR and c-MET TKs, which play a critical oncogenic role in lung cancer. This systematic, data-driven model supported drug response prediction on an individual sample basis, including accurate prediction and validation of synergistic EGFR and c-MET inhibitor activity in cells lacking mutations in either gene, thus contributing to current precision oncology efforts.
Collapse
Affiliation(s)
- Mukesh Bansal
- Psychogenics Inc., Paramus, New Jersey, United States of America
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Jing He
- Department of Systems Biology, Columbia University, New York, NY, United States of America
- Center for Computational Biology and Bioinformatics (C2B2), Columbia University, New York, NY, United States of America
- Department of Biomedical Informatics (DBMI), Columbia University, New York, NY, United States of America
| | - Michael Peyton
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Manjunath Kustagi
- Department of Systems Biology, Columbia University, New York, NY, United States of America
| | - Archana Iyer
- Department of Systems Biology, Columbia University, New York, NY, United States of America
| | - Michael Comb
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA, United States of America
| | - Michael White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Departments of Pharmacology, and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, United States of America
- Center for Computational Biology and Bioinformatics (C2B2), Columbia University, New York, NY, United States of America
- Department of Biomedical Informatics (DBMI), Columbia University, New York, NY, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, United States of America
- Institute for Cancer Genetics, Columbia University, New York, NY, United States of America
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, United States of America
| |
Collapse
|
21
|
Pan-cancer analysis of transcriptional metabolic dysregulation using The Cancer Genome Atlas. Nat Commun 2018; 9:5330. [PMID: 30552315 PMCID: PMC6294258 DOI: 10.1038/s41467-018-07232-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/18/2018] [Indexed: 12/21/2022] Open
Abstract
Understanding metabolic dysregulation in different disease settings is vital for the safe and effective incorporation of metabolism-targeted therapeutics in the clinic. Here, using transcriptomic data for 10,704 tumor and normal samples from The Cancer Genome Atlas, across 26 disease sites, we present a novel bioinformatics pipeline that distinguishes tumor from normal tissues, based on differential gene expression for 114 metabolic pathways. We confirm pathway dysregulation in separate patient populations, demonstrating the robustness of our approach. Bootstrapping simulations were then applied to assess the biological significance of these alterations. We provide distinct examples of the types of analysis that can be accomplished with this tool to understand cancer specific metabolic dysregulation, highlighting novel pathways of interest, and patterns of metabolic flux, in both common and rare disease sites. Further, we show that Master Metabolic Transcriptional Regulators explain why metabolic differences exist, can segregate patient populations, and predict responders to different metabolism-targeted therapeutics.
Collapse
|
22
|
Abstract
Glioma cells diffusely infiltrate the surrounding brain tissue where they intermingle with nonneoplastic brain cells, including astrocytes, microglia, oligodendrocytes and neurons. The infiltrative margins of glioma represent the structural and functional interface between neoplastic and nonneoplastic brain tissue that underlies neurologic alterations associated with glioma, including epilepsy and neurologic deficits. Technological advancements in molecular analysis, including single cell sequencing, now allow us to assess alterations in specific cell types in the brain tumor microenvironment, which can enhance the development of novel therapies that target glioma growth and glioma-induced neurologic symptoms.
Collapse
|
23
|
Testa U, Castelli G, Pelosi E. Genetic Abnormalities, Clonal Evolution, and Cancer Stem Cells of Brain Tumors. Med Sci (Basel) 2018; 6:E85. [PMID: 30279357 PMCID: PMC6313628 DOI: 10.3390/medsci6040085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
Brain tumors are highly heterogeneous and have been classified by the World Health Organization in various histological and molecular subtypes. Gliomas have been classified as ranging from low-grade astrocytomas and oligodendrogliomas to high-grade astrocytomas or glioblastomas. These tumors are characterized by a peculiar pattern of genetic alterations. Pediatric high-grade gliomas are histologically indistinguishable from adult glioblastomas, but they are considered distinct from adult glioblastomas because they possess a different spectrum of driver mutations (genes encoding histones H3.3 and H3.1). Medulloblastomas, the most frequent pediatric brain tumors, are considered to be of embryonic derivation and are currently subdivided into distinct subgroups depending on histological features and genetic profiling. There is emerging evidence that brain tumors are maintained by a special neural or glial stem cell-like population that self-renews and gives rise to differentiated progeny. In many instances, the prognosis of the majority of brain tumors remains negative and there is hope that the new acquisition of information on the molecular and cellular bases of these tumors will be translated in the development of new, more active treatments.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
24
|
Scaglione A, Patzig J, Liang J, Frawley R, Bok J, Mela A, Yattah C, Zhang J, Teo SX, Zhou T, Chen S, Bernstein E, Canoll P, Guccione E, Casaccia P. PRMT5-mediated regulation of developmental myelination. Nat Commun 2018; 9:2840. [PMID: 30026560 PMCID: PMC6053423 DOI: 10.1038/s41467-018-04863-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Oligodendrocytes (OLs) are the myelin-forming cells of the central nervous system. They are derived from differentiation of oligodendrocyte progenitors through a process requiring cell cycle exit and histone modifications. Here we identify the histone arginine methyl-transferase PRMT5, a molecule catalyzing symmetric methylation of histone H4R3, as critical for developmental myelination. PRMT5 pharmacological inhibition, CRISPR/cas9 targeting, or genetic ablation decrease p53-dependent survival and impair differentiation without affecting proliferation. Conditional ablation of Prmt5 in progenitors results in hypomyelination, reduced survival and differentiation. Decreased histone H4R3 symmetric methylation is followed by increased nuclear acetylation of H4K5, and is rescued by pharmacological inhibition of histone acetyltransferases. Data obtained using purified histones further validate the results obtained in mice and in cultured oligodendrocyte progenitors. Together, these results identify PRMT5 as critical for oligodendrocyte differentiation and developmental myelination by modulating the cross-talk between histone arginine methylation and lysine acetylation. Myelin-forming cells derive from oligodendrocyte progenitors. Here the authors identify histone arginine methyl-transferase PRMT5 as critical for developmental myelination by modulating the cross-talk between histone arginine methylation and lysine acetylation, to favor differentiation.
Collapse
Affiliation(s)
- Antonella Scaglione
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Julia Patzig
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Jialiang Liang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Rebecca Frawley
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Jabez Bok
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Camila Yattah
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Graduate Program in Biochemistry, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA
| | - Jingxian Zhang
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Shun Xie Teo
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Ting Zhou
- Room A-829, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Shuibing Chen
- Room A-829, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Emily Bernstein
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Ernesto Guccione
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Patrizia Casaccia
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA. .,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA. .,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA. .,Graduate Program in Biochemistry, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA.
| |
Collapse
|
25
|
Vitucci M, Irvin DM, McNeill RS, Schmid RS, Simon JM, Dhruv HD, Siegel MB, Werneke AM, Bash RE, Kim S, Berens ME, Miller CR. Genomic profiles of low-grade murine gliomas evolve during progression to glioblastoma. Neuro Oncol 2018; 19:1237-1247. [PMID: 28398584 DOI: 10.1093/neuonc/nox050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Gliomas are diverse neoplasms with multiple molecular subtypes. How tumor-initiating mutations relate to molecular subtypes as these tumors evolve during malignant progression remains unclear. Methods We used genetically engineered mouse models, histopathology, genetic lineage tracing, expression profiling, and copy number analyses to examine how genomic tumor diversity evolves during the course of malignant progression from low- to high-grade disease. Results Knockout of all 3 retinoblastoma (Rb) family proteins was required to initiate low-grade tumors in adult mouse astrocytes. Mutations activating mitogen-activated protein kinase signaling, specifically KrasG12D, potentiated Rb-mediated tumorigenesis. Low-grade tumors showed mutant Kras-specific transcriptome profiles but lacked copy number mutations. These tumors stochastically progressed to high-grade, in part through acquisition of copy number mutations. High-grade tumor transcriptomes were heterogeneous and consisted of 3 subtypes that mimicked human mesenchymal, proneural, and neural glioblastomas. Subtypes were confirmed in validation sets of high-grade mouse tumors initiated by different driver mutations as well as human patient-derived xenograft models and glioblastoma tumors. Conclusion These results suggest that oncogenic driver mutations influence the genomic profiles of low-grade tumors and that these, as well as progression-acquired mutations, contribute strongly to the genomic heterogeneity across high-grade tumors.
Collapse
Affiliation(s)
- Mark Vitucci
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - David M Irvin
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Robert S McNeill
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Ralf S Schmid
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Jeremy M Simon
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Harshil D Dhruv
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Marni B Siegel
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Andrea M Werneke
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Ryan E Bash
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Seungchan Kim
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - Michael E Berens
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| | - C Ryan Miller
- Curriculum in Genetics and Molecular Biology, Pathobiology and Translational Science Graduate Program, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Carolina Institute for Developmental Disabilities and Department of Genetics, Lineberger Comprehensive Cancer Center, Neurosciences Center, and Department of Neurology, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina;Cancer & Cell Biology Division, Translational Genomics Institute (TGen), Phoenix, Arizona
| |
Collapse
|
26
|
Arrieta VA, Cacho-Díaz B, Zhao J, Rabadan R, Chen L, Sonabend AM. The possibility of cancer immune editing in gliomas. A critical review. Oncoimmunology 2018; 7:e1445458. [PMID: 29900059 PMCID: PMC5993488 DOI: 10.1080/2162402x.2018.1445458] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/20/2018] [Accepted: 02/20/2018] [Indexed: 01/21/2023] Open
Abstract
The relationship between anti-tumoral immunity and cancer progression is complex. Recently, immune editing has emerged as a model to explain the interplay between the immune system and the selection of genetic alterations in cancer. In this model, the immune system selects cancer cells that grow as these are fit to escape immune surveillance during tumor development. Gliomas and glioblastoma, the most aggressive and most common of all primary malignant brain tumors are genetically heterogeneous, are relatively less antigenic, and are less responsive to immunotherapy than other cancers. In this review, we provide an overview of the relationship between glioma´s immune suppressive features, anti-tumoral immunity and cancer genomics. In this context, we provide a critical discussion of evidence suggestive of immune editing in this disease and discuss possible alternative explanations for these findings.
Collapse
Affiliation(s)
- Víctor A Arrieta
- PECEM, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Junfei Zhao
- Department of Systems Biology, Herbert Irving Comprehensive Center, Columbia University, New York City, New York, USA
| | - Raul Rabadan
- Department of Systems Biology, Herbert Irving Comprehensive Center, Columbia University, New York City, New York, USA
| | - Li Chen
- Department of Neurosurgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adam M Sonabend
- Department of Neurosurgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
27
|
King CJ, Woodward J, Schwartzman J, Coleman DJ, Lisac R, Wang NJ, Van Hook K, Gao L, Urrutia J, Dane MA, Heiser LM, Alumkal JJ. Integrative molecular network analysis identifies emergent enzalutamide resistance mechanisms in prostate cancer. Oncotarget 2017; 8:111084-111095. [PMID: 29340039 PMCID: PMC5762307 DOI: 10.18632/oncotarget.22560] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/29/2017] [Indexed: 12/26/2022] Open
Abstract
Recent work demonstrates that castration-resistant prostate cancer (CRPC) tumors harbor countless genomic aberrations that control many hallmarks of cancer. While some specific mutations in CRPC may be actionable, many others are not. We hypothesized that genomic aberrations in cancer may operate in concert to promote drug resistance and tumor progression, and that organization of these genomic aberrations into therapeutically targetable pathways may improve our ability to treat CRPC. To identify the molecular underpinnings of enzalutamide-resistant CRPC, we performed transcriptional and copy number profiling studies using paired enzalutamide-sensitive and resistant LNCaP prostate cancer cell lines. Gene networks associated with enzalutamide resistance were revealed by performing an integrative genomic analysis with the PAthway Representation and Analysis by Direct Reference on Graphical Models (PARADIGM) tool. Amongst the pathways enriched in the enzalutamide-resistant cells were those associated with MEK, EGFR, RAS, and NFKB. Functional validation studies of 64 genes identified 10 candidate genes whose suppression led to greater effects on cell viability in enzalutamide-resistant cells as compared to sensitive parental cells. Examination of a patient cohort demonstrated that several of our functionally-validated gene hits are deregulated in metastatic CRPC tumor samples, suggesting that they may be clinically relevant therapeutic targets for patients with enzalutamide-resistant CRPC. Altogether, our approach demonstrates the potential of integrative genomic analyses to clarify determinants of drug resistance and rational co-targeting strategies to overcome resistance.
Collapse
Affiliation(s)
- Carly J. King
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Josha Woodward
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jacob Schwartzman
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Daniel J. Coleman
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Robert Lisac
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Nicholas J. Wang
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kathryn Van Hook
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Lina Gao
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Joshua Urrutia
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Mark A. Dane
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Laura M. Heiser
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Joshi J. Alumkal
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
28
|
Pisapia DJ. The Updated World Health Organization Glioma Classification: Cellular and Molecular Origins of Adult Infiltrating Gliomas. Arch Pathol Lab Med 2017; 141:1633-1645. [PMID: 29189064 DOI: 10.5858/arpa.2016-0493-ra] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - In the recently updated World Health Organization (WHO) classification of central nervous system tumors, our concept of infiltrating gliomas as a molecular dichotomy between oligodendroglial and astrocytic tumors has been codified. Advances in animal models of glioma and a wealth of sophisticated molecular analyses of human glioma tissue have led to a greater understanding of some of the biologic underpinnings of gliomagenesis. OBJECTIVE - To review our understanding of gliomagenesis in the setting of the recently updated WHO classification of central nervous system tumors. Topics addressed include a summary of an updated diagnostic schema for infiltrating gliomas, the crucial importance of isocitrate dehydrogenase mutations, candidate cells of origin for gliomas, environmental and other posited contributing factors to gliomagenesis, and the possible role of chromatin topology in setting the stage for gliomagenesis. DATA SOURCES - We conducted a primary literature search using PubMed. CONCLUSIONS - With multidimensional molecular data sets spanning increasingly larger numbers of patients with infiltrating gliomas, our understanding of the disease at the point of surgical resection has improved dramatically and this understanding is reflected in the updated WHO classification. Animal models have demonstrated a diversity of candidates for glioma cells of origin, but crucial questions remain, including the role of neural stem cells, more differentiated progenitor cells, and glioma stem cells. At this stage the increase in data generated from human samples will hopefully inform the creation of newer animal models that will recapitulate more accurately the diversity of gliomas and provide novel insights into the biologic mechanisms underlying tumor initiation and progression.
Collapse
|
29
|
Kosty J, Lu F, Kupp R, Mehta S, Lu QR. Harnessing OLIG2 function in tumorigenicity and plasticity to target malignant gliomas. Cell Cycle 2017; 16:1654-1660. [PMID: 28806136 DOI: 10.1080/15384101.2017.1361062] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Glioblastoma (GBM) is the most prevalent and malignant brain tumor, displaying notorious resistance to conventional therapy, partially due to molecular and genetic heterogeneity. Understanding the mechanisms for gliomagenesis, tumor stem/progenitor cell propagation and phenotypic diversity is critical for devising effective and targeted therapy for this lethal disease. The basic helix-loop-helix transcription factor OLIG2, which is universally expressed in gliomas, has emerged as an important player in GBM cell reprogramming, genotoxic resistance, and tumor phenotype plasticity. In an animal model of proneural GBM, elimination of mitotic OLIG2+ progenitors blocks tumor growth, suggesting that these progenitors are a seeding source for glioma propagation. OLIG2 deletion reduces tumor growth and causes an oligodendrocytic to astrocytic phenotype shift, with PDGFRα downregulation and reciprocal EGFR signaling upregulation, underlying alternative pathways in tumor recurrence. In patient-derived glioma stem cells (GSC), knockdown of OLIG2 leads to downregulation of PDGFRα, while OLIG2 silencing results in a shift from proneural-to-classical gene expression pattern or a proneural-to-mesenchymal transition in distinct GSC cell lines, where OLIG2 appears to regulate EGFR expression in a context-dependent manner. In addition, post-translational modifications such as phosphorylation by a series of protein kinases regulates OLIG2 activity in glioma cell growth and invasive behaviors. In this perspective, we will review the role of OLIG2 in tumor initiation, proliferation and phenotypic plasticity in animal models of gliomas and human GSC cell lines, and discuss the underlying mechanisms in the control of tumor growth and potential therapeutic strategies to target OLIG2 in malignant gliomas.
Collapse
Affiliation(s)
- Jennifer Kosty
- a Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,b Department of Neurosurgery , University of Cincinnati , Cincinnati , OH , USA
| | - Fanghui Lu
- a Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,c National Centre for International Research in Cell and Gene Therapy, Centre for Cell and Gene Therapy of Academy of Medical Sciences , Zhengzhou University , Zhengzhou , Henan , China
| | - Robert Kupp
- d Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix , AZ , USA.,e Cancer Research UK Cambridge Institute , University of Cambridge, Li Ka Shing Centre , Cambridge , UK
| | - Shwetal Mehta
- d Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix , AZ , USA
| | - Q Richard Lu
- a Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA
| |
Collapse
|
30
|
Lescarbeau RS, Lei L, Bakken KK, Sims PA, Sarkaria JN, Canoll P, White FM. Quantitative Phosphoproteomics Reveals Wee1 Kinase as a Therapeutic Target in a Model of Proneural Glioblastoma. Mol Cancer Ther 2016; 15:1332-43. [PMID: 27196784 PMCID: PMC4893926 DOI: 10.1158/1535-7163.mct-15-0692] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 02/24/2016] [Indexed: 01/09/2023]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain cancer. With a median survival of about a year, new approaches to treating this disease are necessary. To identify signaling molecules regulating GBM progression in a genetically engineered murine model of proneural GBM, we quantified phosphotyrosine-mediated signaling using mass spectrometry. Oncogenic signals, including phosphorylated ERK MAPK, PI3K, and PDGFR, were found to be increased in the murine tumors relative to brain. Phosphorylation of CDK1 pY15, associated with the G2 arrest checkpoint, was identified as the most differentially phosphorylated site, with a 14-fold increase in phosphorylation in the tumors. To assess the role of this checkpoint as a potential therapeutic target, syngeneic primary cell lines derived from these tumors were treated with MK-1775, an inhibitor of Wee1, the kinase responsible for CDK1 Y15 phosphorylation. MK-1775 treatment led to mitotic catastrophe, as defined by increased DNA damage and cell death by apoptosis. To assess the extensibility of targeting Wee1/CDK1 in GBM, patient-derived xenograft (PDX) cell lines were also treated with MK-1775. Although the response was more heterogeneous, on-target Wee1 inhibition led to decreased CDK1 Y15 phosphorylation and increased DNA damage and apoptosis in each line. These results were also validated in vivo, where single-agent MK-1775 demonstrated an antitumor effect on a flank PDX tumor model, increasing mouse survival by 1.74-fold. This study highlights the ability of unbiased quantitative phosphoproteomics to reveal therapeutic targets in tumor models, and the potential for Wee1 inhibition as a treatment approach in preclinical models of GBM. Mol Cancer Ther; 15(6); 1332-43. ©2016 AACR.
Collapse
Affiliation(s)
- Rebecca S Lescarbeau
- Department of Biological Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Liang Lei
- Department of Pathology and Cell Biology and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Katrina K Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Peter A Sims
- Department of Systems Biology and Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Peter Canoll
- Department of Pathology and Cell Biology and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Forest M White
- Department of Biological Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
31
|
Lescarbeau RS, Lei L, Bakken KK, Sims PA, Sarkaria JN, Canoll P, White FM. Quantitative Phosphoproteomics Reveals Wee1 Kinase as a Therapeutic Target in a Model of Proneural Glioblastoma. Mol Cancer Ther 2016. [PMID: 27196784 DOI: 10.1158/1535-7163.mct-15-0692-t] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain cancer. With a median survival of about a year, new approaches to treating this disease are necessary. To identify signaling molecules regulating GBM progression in a genetically engineered murine model of proneural GBM, we quantified phosphotyrosine-mediated signaling using mass spectrometry. Oncogenic signals, including phosphorylated ERK MAPK, PI3K, and PDGFR, were found to be increased in the murine tumors relative to brain. Phosphorylation of CDK1 pY15, associated with the G2 arrest checkpoint, was identified as the most differentially phosphorylated site, with a 14-fold increase in phosphorylation in the tumors. To assess the role of this checkpoint as a potential therapeutic target, syngeneic primary cell lines derived from these tumors were treated with MK-1775, an inhibitor of Wee1, the kinase responsible for CDK1 Y15 phosphorylation. MK-1775 treatment led to mitotic catastrophe, as defined by increased DNA damage and cell death by apoptosis. To assess the extensibility of targeting Wee1/CDK1 in GBM, patient-derived xenograft (PDX) cell lines were also treated with MK-1775. Although the response was more heterogeneous, on-target Wee1 inhibition led to decreased CDK1 Y15 phosphorylation and increased DNA damage and apoptosis in each line. These results were also validated in vivo, where single-agent MK-1775 demonstrated an antitumor effect on a flank PDX tumor model, increasing mouse survival by 1.74-fold. This study highlights the ability of unbiased quantitative phosphoproteomics to reveal therapeutic targets in tumor models, and the potential for Wee1 inhibition as a treatment approach in preclinical models of GBM. Mol Cancer Ther; 15(6); 1332-43. ©2016 AACR.
Collapse
Affiliation(s)
- Rebecca S Lescarbeau
- Department of Biological Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Liang Lei
- Department of Pathology and Cell Biology and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Katrina K Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Peter A Sims
- Department of Systems Biology and Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Peter Canoll
- Department of Pathology and Cell Biology and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Forest M White
- Department of Biological Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
32
|
Lu F, Chen Y, Zhao C, Wang H, He D, Xu L, Wang J, He X, Deng Y, Lu EE, Liu X, Verma R, Bu H, Drissi R, Fouladi M, Stemmer-Rachamimov AO, Burns D, Xin M, Rubin JB, Bahassi EM, Canoll P, Holland EC, Lu QR. Olig2-Dependent Reciprocal Shift in PDGF and EGF Receptor Signaling Regulates Tumor Phenotype and Mitotic Growth in Malignant Glioma. Cancer Cell 2016; 29:669-683. [PMID: 27165742 PMCID: PMC4946168 DOI: 10.1016/j.ccell.2016.03.027] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 01/05/2016] [Accepted: 03/31/2016] [Indexed: 02/05/2023]
Abstract
Malignant gliomas exhibit extensive heterogeneity and poor prognosis. Here we identify mitotic Olig2-expressing cells as tumor-propagating cells in proneural gliomas, elimination of which blocks tumor initiation and progression. Intriguingly, deletion of Olig2 resulted in tumors that grow, albeit at a decelerated rate. Genome occupancy and expression profiling analyses reveal that Olig2 directly activates cell-proliferation machinery to promote tumorigenesis. Olig2 deletion causes a tumor phenotypic shift from an oligodendrocyte precursor-correlated proneural toward an astroglia-associated gene expression pattern, manifest in downregulation of platelet-derived growth factor receptor-α and reciprocal upregulation of epidermal growth factor receptor (EGFR). Olig2 deletion further sensitizes glioma cells to EGFR inhibitors and extends the lifespan of animals. Thus, Olig2-orchestrated receptor signaling drives mitotic growth and regulates glioma phenotypic plasticity. Targeting Olig2 may circumvent resistance to EGFR-targeted drugs.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cell Transformation, Neoplastic/genetics
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic
- Glioma/genetics
- Glioma/metabolism
- Glioma/pathology
- Humans
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Oligodendroglia/metabolism
- Phenotype
- Receptors, Platelet-Derived Growth Factor/genetics
- Receptors, Platelet-Derived Growth Factor/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/genetics
- Spheroids, Cellular/metabolism
- Survival Analysis
Collapse
Affiliation(s)
- Fanghui Lu
- Laboratory of Pathology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu 610041, China; Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Ying Chen
- School of Life Sciences, Xiamen University, Fujian 361102, China
| | - Chuntao Zhao
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Haibo Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Danyang He
- Department of Pathology & Integrative Biology Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lingli Xu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Jincheng Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Xuelian He
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Yaqi Deng
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Ellen E Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Xue Liu
- School of Life Sciences, Xiamen University, Fujian 361102, China
| | - Ravinder Verma
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Hong Bu
- Laboratory of Pathology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu 610041, China
| | - Rachid Drissi
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Maryam Fouladi
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Anat O Stemmer-Rachamimov
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dennis Burns
- Department of Pathology & Integrative Biology Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mei Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA
| | - Joshua B Rubin
- Departments of Pediatrics and Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - El Mustapha Bahassi
- Department of Internal Medicine, UC Brain Tumor Center, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Peter Canoll
- Department of Pathology & Cellular Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Eric C Holland
- Division of Human Biology and Solid Tumor Translational Research, Fred Hutchinson Cancer Research Center, Alvord Brain Tumor Center, University of Washington, Seattle, WA 98109, USA
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 25229, USA; Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
33
|
Zhang Q, Li J, Xue H, Kong L, Wang Y. Network-based methods for identifying critical pathways of complex diseases: a survey. MOLECULAR BIOSYSTEMS 2016; 12:1082-1089. [DOI: 10.1039/c5mb00815h] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
We review seven major network-based pathway analysis methods and enumerate their benefits and limitations from an algorithmic perspective to provide a reference for the next generation of pathway analysis methods.
Collapse
Affiliation(s)
- Qiaosheng Zhang
- School of Computer Science and Technology
- Harbin Institute of Technology
- China
- Heilongjiang Bayi Agricultural University
- China
| | - Jie Li
- School of Computer Science and Technology
- Harbin Institute of Technology
- China
| | - Hanqing Xue
- School of Computer Science and Technology
- Harbin Institute of Technology
- China
| | - Leilei Kong
- School of Computer Science and Technology
- Heilongjiang Institute of Technology
- China
| | - Yadong Wang
- School of Computer Science and Technology
- Harbin Institute of Technology
- China
| |
Collapse
|
34
|
Construction and analysis of dynamic transcription factor regulatory networks in the progression of glioma. Sci Rep 2015; 5:15953. [PMID: 26526635 PMCID: PMC4630656 DOI: 10.1038/srep15953] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/02/2015] [Indexed: 12/21/2022] Open
Abstract
The combinatorial cross-regulation of transcription factors (TFs) plays an important role in cellular identity and function; however, the dynamic regulation of TFs during glioma progression remains largely unknown. Here, we used the genome-wide expression of TFs to construct an extensive human TF network comprising interactions among 513 TFs and to analyse the dynamics of the TF-TF network during glioma progression. We found that the TF regulatory networks share a common architecture and that the topological structures are conserved. Strikingly, despite the conservation of the network architecture, TF regulatory networks are highly grade specific, and TF circuitry motifs are dynamically rewired during glioma progression. In addition, the most frequently observed structure in the grade-specific TF networks was the feedforward loop (FFL). We described applications that show how investigating the behaviour of FFLs in glioblastoma can reveal FFLs (such as RARG-NR1I2-CDX2) that are associated with prognosis. We constructed comprehensive TF-TF networks and systematically analysed the circuitry, dynamics, and topological principles of the networks during glioma progression, which will further enhance our understanding of the functions of TFs in glioma.
Collapse
|
35
|
Creixell P, Reimand J, Haider S, Wu G, Shibata T, Vazquez M, Mustonen V, Gonzalez-Perez A, Pearson J, Sander C, Raphael BJ, Marks DS, Ouellette BFF, Valencia A, Bader GD, Boutros PC, Stuart JM, Linding R, Lopez-Bigas N, Stein LD. Pathway and network analysis of cancer genomes. Nat Methods 2015; 12:615-621. [PMID: 26125594 DOI: 10.1038/nmeth.3440] [Citation(s) in RCA: 218] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/27/2015] [Indexed: 12/26/2022]
Abstract
Genomic information on tumors from 50 cancer types cataloged by the International Cancer Genome Consortium (ICGC) shows that only a few well-studied driver genes are frequently mutated, in contrast to many infrequently mutated genes that may also contribute to tumor biology. Hence there has been large interest in developing pathway and network analysis methods that group genes and illuminate the processes involved. We provide an overview of these analysis techniques and show where they guide mechanistic and translational investigations.
Collapse
Affiliation(s)
- Pau Creixell
- Cellular Signal Integration Group (C-SIG), Technical University of Denmark, Lyngby, Denmark
| | - Jüri Reimand
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Syed Haider
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Guanming Wu
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center, Chuo-ku, Tokyo, Japan
| | - Miguel Vazquez
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Ville Mustonen
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Abel Gonzalez-Perez
- Research Unit on Biomedical Informatics, University Pompeu Fabra, Barcelona, Spain
| | - John Pearson
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Chris Sander
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Benjamin J Raphael
- Department of Computer Science and Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Debora S Marks
- Department of Systems Biology, Harvard Medical School, Boston, MA USA
| | - B F Francis Ouellette
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Alfonso Valencia
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Paul C Boutros
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Joshua M Stuart
- Department of Biomolecular Engineering, University of California, Santa Cruz, California, USA.,Center for Biomolecular Science and Engineering, University of California, Santa Cruz, California, USA
| | - Rune Linding
- Cellular Signal Integration Group (C-SIG), Technical University of Denmark, Lyngby, Denmark.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen (UCPH), DK-2200 Copenhagen, Denmark
| | - Nuria Lopez-Bigas
- Research Unit on Biomedical Informatics, University Pompeu Fabra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Lincoln D Stein
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
36
|
Molecular subtypes, stem cells and heterogeneity: Implications for personalised therapy in glioma. J Clin Neurosci 2015; 22:1219-26. [DOI: 10.1016/j.jocn.2015.02.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/14/2015] [Indexed: 01/08/2023]
|
37
|
Biomarkers for glioma immunotherapy: the next generation. J Neurooncol 2015; 123:359-72. [PMID: 25724916 DOI: 10.1007/s11060-015-1746-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/16/2015] [Indexed: 12/11/2022]
Abstract
The term "biomarker" historically refers to a single parameter, such as the expression level of a gene or a radiographic pattern, used to indicate a broader biological state. Molecular indicators have been applied to several aspects of cancer therapy: to describe the genotypic and phenotypic state of neoplastic tissue for prognosis, to predict susceptibility to anti-proliferative agents, to validate the presence of specific drug targets, and to evaluate responsiveness to therapy. For glioblastoma (GBM), immunohistochemical and radiographic biomarkers accessible to the clinical lab have informed traditional regimens, but while immunotherapies have emerged as potentially disruptive weapons against this diffusely infiltrating, heterogeneous tumor, biomarkers with strong predictive power have not been fully established. The cancer immunotherapy field, through the recently accelerated expansion of trials, is currently leveraging this wealth of clinical and biological data to define and revise the use of biomarkers for improving prognostic accuracy, personalization of therapy, and evaluation of responses across the wide variety of tumors. Technological advancements in DNA sequencing, cytometry, and microscopy have facilitated the exploration of more integrated, high-dimensional profiling of the disease system-incorporating both immune and tumor parameters-rather than single metrics, as biomarkers for therapeutic sensitivity. Here we discuss the utility of traditional GBM biomarkers in immunotherapy and how the impending transformation of the biomarker paradigm-from single markers to integrated profiles-may offer the key to bringing predictive, personalized immunotherapy to GBM patients.
Collapse
|
38
|
Abstract
Glioma growth is driven by signaling that ultimately regulates protein synthesis. Gliomas are also complex at the cellular level and involve multiple cell types, including transformed and reactive cells in the brain tumor microenvironment. The distinct functions of the various cell types likely lead to different requirements and regulatory paradigms for protein synthesis. Proneural gliomas can arise from transformation of glial progenitors that are driven to proliferate via mitogenic signaling that affects translation. To investigate translational regulation in this system, we developed a RiboTag glioma mouse model that enables cell-type-specific, genome-wide ribosome profiling of tumor tissue. Infecting glial progenitors with Cre-recombinant retrovirus simultaneously activates expression of tagged ribosomes and delivers a tumor-initiating mutation. Remarkably, we find that although genes specific to transformed cells are highly translated, their translation efficiencies are low compared with normal brain. Ribosome positioning reveals sequence-dependent regulation of ribosomal activity in 5'-leaders upstream of annotated start codons, leading to differential translation in glioma compared with normal brain. Additionally, although transformed cells express a proneural signature, untransformed tumor-associated cells, including reactive astrocytes and microglia, express a mesenchymal signature. Finally, we observe the same phenomena in human disease by combining ribosome profiling of human proneural tumor and non-neoplastic brain tissue with computational deconvolution to assess cell-type-specific translational regulation.
Collapse
|
39
|
Starke RM, Komotar RJ, Connolly ES. The regulatory network of proneural glioma in tumor progression. Neurosurgery 2014; 75:N15-6. [PMID: 25406622 DOI: 10.1227/01.neu.0000457195.86910.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Robert M Starke
- University of Virginia, School of Medicine, Charlottesville, Virginia University of Miami, School of Medicine, Miami, Florida Columbia University College of Physicians and Surgeons, New York, New York
| | | | | |
Collapse
|
40
|
MRI-localized biopsies reveal subtype-specific differences in molecular and cellular composition at the margins of glioblastoma. Proc Natl Acad Sci U S A 2014; 111:12550-5. [PMID: 25114226 DOI: 10.1073/pnas.1405839111] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Glioblastomas (GBMs) diffusely infiltrate the brain, making complete removal by surgical resection impossible. The mixture of neoplastic and nonneoplastic cells that remain after surgery form the biological context for adjuvant therapeutic intervention and recurrence. We performed RNA-sequencing (RNA-seq) and histological analysis on radiographically guided biopsies taken from different regions of GBM and showed that the tissue contained within the contrast-enhancing (CE) core of tumors have different cellular and molecular compositions compared with tissue from the nonenhancing (NE) margins of tumors. Comparisons with the The Cancer Genome Atlas dataset showed that the samples from CE regions resembled the proneural, classical, or mesenchymal subtypes of GBM, whereas the samples from the NE regions predominantly resembled the neural subtype. Computational deconvolution of the RNA-seq data revealed that contributions from nonneoplastic brain cells significantly influence the expression pattern in the NE samples. Gene ontology analysis showed that the cell type-specific expression patterns were functionally distinct and highly enriched in genes associated with the corresponding cell phenotypes. Comparing the RNA-seq data from the GBM samples to that of nonneoplastic brain revealed that the differentially expressed genes are distributed across multiple cell types. Notably, the patterns of cell type-specific alterations varied between the different GBM subtypes: the NE regions of proneural tumors were enriched in oligodendrocyte progenitor genes, whereas the NE regions of mesenchymal GBM were enriched in astrocytic and microglial genes. These subtype-specific patterns provide new insights into molecular and cellular composition of the infiltrative margins of GBM.
Collapse
|
41
|
Sonabend AM, Carminucci AS, Amendolara B, Bansal M, Leung R, Lei L, Realubit R, Li H, Karan C, Yun J, Showers C, Rothcock R, O J, Califano A, Canoll P, Bruce JN. Convection-enhanced delivery of etoposide is effective against murine proneural glioblastoma. Neuro Oncol 2014; 16:1210-9. [PMID: 24637229 DOI: 10.1093/neuonc/nou026] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Glioblastoma subtypes have been defined based on transcriptional profiling, yet personalized care based on molecular classification remains unexploited. Topoisomerase II (TOP2) contributes to the transcriptional signature of the proneural glioma subtype. Thus, we targeted TOP2 pharmacologically with etoposide in proneural glioma models. METHODS TOP2 gene expression was evaluated in mouse platelet derived growth factor (PDGF)(+)phosphatase and tensin homolog (PTEN)(-/-)p53(-/-) and PDGF(+)PTEN(-/-) proneural gliomas and cell lines, as well as human glioblastoma from The Cancer Genome Atlas. Correlation between TOP2 transcript levels and etoposide susceptibility was investigated in 139 human cancer cell lines from the Cancer Cell Line Encyclopedia public dataset and in mouse proneural glioma cell lines. Convection-enhanced delivery (CED) of etoposide was tested on cell-based PDGF(+)PTEN(-/-)p53(-/-) and retroviral-based PDGF(+)PTEN(-/-) mouse proneural glioma models. RESULTS TOP2 expression was significantly higher in human proneural glioblastoma and in mouse proneural tumors at early as well as late stages of development compared with normal brain. TOP2B transcript correlated with susceptibility to etoposide in mouse proneural cell lines and in 139 human cancer cell lines from the Cancer Cell Line Encyclopedia. Intracranial etoposide CED treatment (680 μM) was well tolerated by mice and led to a significant survival benefit in the PDGF(+)PTEN(-/-)p53(-/-) glioma model. Moreover, etoposide CED treatment at 80 μM but not 4 μM led to a significant survival advantage in the PDGF(+)PTEN(-/-) glioma model. CONCLUSIONS TOP2 is highly expressed in proneural gliomas, rendering its pharmacological targeting by intratumoral administration of etoposide by CED effective on murine proneural gliomas. We provide evidence supporting clinical testing of CED of etoposide with a molecular-based patient selection approach.
Collapse
Affiliation(s)
- Adam M Sonabend
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Arthur S Carminucci
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Benjamin Amendolara
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Mukesh Bansal
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Richard Leung
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Liang Lei
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Ronald Realubit
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Hai Li
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Charles Karan
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Jonathan Yun
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Christopher Showers
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Robert Rothcock
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Jane O
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Andrea Califano
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Peter Canoll
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| | - Jeffrey N Bruce
- Gabriele Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (A.M.S., A.S.C., B.A., R.L., J.Y., C.S., R.R., J.O, P.C., J.N.B.); Department of Systems Biology, Columbia University, New York, New York (M.B., A.C.); Center for Computational Biology and Bioinformatics, Columbia University, New York, New York (M.B., A.C.); Department of Pathology and Cell Biology, Irving Research Cancer Center, Columbia University Medical Center, New York, New York (L.L.); High Throughput Screening Center, Columbia University Medical Center Judith P. Sulzberger Genome Center, New York, New York (R.R., H.L., C.K.); Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, New York (A.C.); Department of Biomedical Informatics, Columbia University, New York, New York (A.C.); Institute for Cancer Genetics, Columbia University, New York, New York (A.C.); Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York (A.C.)
| |
Collapse
|