401
|
Boscoe AN, Rolland C, Kelley RK. Frequency and prognostic significance of isocitrate dehydrogenase 1 mutations in cholangiocarcinoma: a systematic literature review. J Gastrointest Oncol 2019; 10:751-765. [PMID: 31392056 PMCID: PMC6657309 DOI: 10.21037/jgo.2019.03.10] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The recognition of distinct molecular subgroups within cholangiocarcinoma (CC), along with the increasing availability of targeted therapies, suggests that further characterization of the prevalence and prognosis of frequently occurring subgroups may assist with the development of more effective treatment approaches for the management of CC. A systematic review was performed to investigate the prevalence of isocitrate dehydrogenase 1 (IDH1) mutations (mIDH1) in patients with CC, the possible clinical and prognostic significance of mIDH1, and the presence of co-mutations in tumors with mIDH1. METHODS This review was conducted using the Cochrane dual-reviewer methodology and the Preferred Reporting Items for Systematic Reviews and Meta-Analyses protocol (PRISMA-P) guidelines. Searches were performed in Embase, MEDLINE, the Cochrane Central Trials Register and Database of Systematic Reviews, and other Cochrane Library assets using terms for CC and mIDH1 with no language or date restrictions for articles published up to December 31, 2017. Searches were also performed of abstracts presented at the following conferences in 2016 and 2017: American Society of Clinical Oncology (ASCO), ASCO-Gastrointestinal Cancers Symposium (ASCO-GI), the European Society for Medical Oncology (ESMO), and ESMO-Asia. Screening was performed separately by two reviewers and cross-checked. Any discrepancies between reviewers were resolved by a senior researcher. Data from all selected references were recorded in a data extraction grid. RESULTS A total of 46 publications met the inclusion criteria and were included in the systematic review. Of these publications, 45 reported the frequency of mIDH1 among a total sample of 5,393 patients with CC. mIDH1 was enriched in intrahepatic CC (ICC), with 552 (13.1%; 95% CI, 12.1-14.2) of the 4,214 patients with ICC having the mutation compared with 9 (0.8%; 95% CI, 0.4-1.5%) of the 1,123 patients with extrahepatic CC (ECC). The percentage of females with mIDH1 CC (66.2%; 95% CI, 57.7-73.7%) was higher than in the overall CC population (44.4%). The frequency of mIDH1 in patients with ICC reported in individual studies ranged from 4.5-55.6%, and a significantly higher frequency was reported in non-Asian centers compared with Asian centers (weighted mean, 16.5% vs. 8.8%; P<0.001). The prevalence of mIDH1 in patients with ICC at USA centers was 18.0% (95% CI, 16.4-19.8%). Eleven publications reported the prevalence of co-mutations in patients with mIDH1 ICC, with the most frequent being AT-rich interactive domain-containing protein 1A (ARID1A) (22.0%), BRCA1-associated protein 1 (BAP1) (15.5%), and PBRM1 (13.3%). Eight publications investigated the possible prognostic significance of mIDH1. None of the studies reported a statistically significant association between mIDH1 and overall survival (OS), progression-free survival (PFS), or time to progression. CONCLUSIONS This systematic review substantiates the prevalence of mIDH1 in CC and further characterizes clinical, pathologic, and genetic covariates within this sub-population. Co-mutation data may inform future studies of mechanisms of response and resistance to mIDH1-targeted therapies.
Collapse
|
402
|
Martin GH, Roy N, Chakraborty S, Desrichard A, Chung SS, Woolthuis CM, Hu W, Berezniuk I, Garrett-Bakelman FE, Hamann J, Devlin SM, Chan TA, Park CY. CD97 is a critical regulator of acute myeloid leukemia stem cell function. J Exp Med 2019; 216:2362-2377. [PMID: 31371381 PMCID: PMC6781010 DOI: 10.1084/jem.20190598] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/20/2019] [Accepted: 06/27/2019] [Indexed: 12/15/2022] Open
Abstract
Despite significant efforts to improve therapies for acute myeloid leukemia (AML), clinical outcomes remain poor. Understanding the mechanisms that regulate the development and maintenance of leukemic stem cells (LSCs) is important to reveal new therapeutic opportunities. We have identified CD97, a member of the adhesion class of G protein-coupled receptors (GPCRs), as a frequently up-regulated antigen on AML blasts that is a critical regulator of blast function. High levels of CD97 correlate with poor prognosis, and silencing of CD97 reduces disease aggressiveness in vivo. These phenotypes are due to CD97's ability to promote proliferation, survival, and the maintenance of the undifferentiated state in leukemic blasts. Collectively, our data credential CD97 as a promising therapeutic target on LSCs in AML.
Collapse
Affiliation(s)
- Gaëlle H Martin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Pathology, New York University School of Medicine, New York, NY
| | - Nainita Roy
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Sohini Chakraborty
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Alexis Desrichard
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Stephen S Chung
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carolien M Woolthuis
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wenhuo Hu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Iryna Berezniuk
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Francine E Garrett-Bakelman
- Department of Medicine, Division of Hematology/Oncology, University of Virginia, Charlottesville, VA.,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA.,Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY
| | - Jörg Hamann
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sean M Devlin
- Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christopher Y Park
- Department of Pathology, New York University School of Medicine, New York, NY
| |
Collapse
|
403
|
Not Only Mutations Matter: Molecular Picture of Acute Myeloid Leukemia Emerging from Transcriptome Studies. JOURNAL OF ONCOLOGY 2019; 2019:7239206. [PMID: 31467542 PMCID: PMC6699387 DOI: 10.1155/2019/7239206] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/12/2019] [Indexed: 01/08/2023]
Abstract
The last two decades of genome-scale research revealed a complex molecular picture of acute myeloid leukemia (AML). On the one hand, a number of mutations were discovered and associated with AML diagnosis and prognosis; some of them were introduced into diagnostic tests. On the other hand, transcriptome studies, which preceded AML exome and genome sequencing, remained poorly translated into clinics. Nevertheless, gene expression studies significantly contributed to the elucidation of AML pathogenesis and indicated potential therapeutic directions. The power of transcriptomic approach lies in its comprehensiveness; we can observe how genome manifests its function in a particular type of cells and follow many genes in one test. Moreover, gene expression measurement can be combined with mutation detection, as high-impact mutations are often present in transcripts. This review sums up 20 years of transcriptome research devoted to AML. Gene expression profiling (GEP) revealed signatures distinctive for selected AML subtypes and uncovered the additional within-subtype heterogeneity. The results were particularly valuable in the case of AML with normal karyotype which concerns up to 50% of AML cases. With the use of GEP, new classes of the disease were identified and prognostic predictors were proposed. A plenty of genes were detected as overexpressed in AML when compared to healthy control, including KIT, BAALC, ERG, MN1, CDX2, WT1, PRAME, and HOX genes. High expression of these genes constitutes usually an unfavorable prognostic factor. Upregulation of FLT3 and NPM1 genes, independent on their mutation status, was also reported in AML and correlated with poor outcome. However, transcriptome is not limited to the protein-coding genes; other types of RNA molecules exist in a cell and regulate genome function. It was shown that microRNA (miRNA) profiles differentiated AML groups and predicted outcome not worse than protein-coding gene profiles. For example, upregulation of miR-10a, miR-10b, and miR-196b and downregulation of miR-192 were found as typical of AML with NPM1 mutation whereas overexpression of miR-155 was associated with FLT3-internal tandem duplication (FLT3-ITD). Development of high-throughput technologies and microarray replacement by next generation sequencing (RNA-seq) enabled uncovering a real variety of leukemic cell transcriptomes, reflected by gene fusions, chimeric RNAs, alternatively spliced transcripts, miRNAs, piRNAs, long noncoding RNAs (lncRNAs), and their special type, circular RNAs. Many of them can be considered as AML biomarkers and potential therapeutic targets. The relations between particular RNA puzzles and other components of leukemic cells and their microenvironment, such as exosomes, are now under investigation. Hopefully, the results of this research will shed the light on these aspects of AML pathogenesis which are still not completely understood.
Collapse
|
404
|
Stuani L, Sabatier M, Sarry JE. Exploiting metabolic vulnerabilities for personalized therapy in acute myeloid leukemia. BMC Biol 2019; 17:57. [PMID: 31319822 PMCID: PMC6637566 DOI: 10.1186/s12915-019-0670-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Changes in cell metabolism and metabolic adaptation are hallmark features of many cancers, including leukemia, that support biological processes involved into tumor initiation, growth, and response to therapeutics. The discovery of mutations in key metabolic enzymes has highlighted the importance of metabolism in cancer biology and how these changes might constitute an Achilles heel for cancer treatment. In this Review, we discuss the role of metabolic and mitochondrial pathways dysregulated in acute myeloid leukemia, and the potential of therapeutic intervention targeting these metabolic dependencies on the proliferation, differentiation, stem cell function and cell survival to improve patient stratification and outcomes.
Collapse
Affiliation(s)
- Lucille Stuani
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, F-31037, Toulouse, France.
| | - Marie Sabatier
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, F-31037, Toulouse, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, F-31037, Toulouse, France.
| |
Collapse
|
405
|
Immunohistochemical Detection and Molecular Characterization of IDH-mutant Sinonasal Undifferentiated Carcinomas. Am J Surg Pathol 2019; 42:1067-1075. [PMID: 29683816 DOI: 10.1097/pas.0000000000001064] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent studies have identified recurrent isocitrate dehydrogenase 2 (IDH2) mutations in a subset of sinonasal undifferentiated carcinomas (SNUCs); however, the true frequency of IDH mutations in SNUC is unknown. We evaluated the utility of mutation-specific IDH1/2 immunohistochemistry (IHC) in a large multi-institutional cohort of SNUC and morphologic mimics. IHC using a multispecific antibody for IDH1/2 (R132/R172) mutant protein was performed on 193 sinonasal tumors including: 53 SNUCs, 8 poorly differentiated carcinomas (PDCARs) and 132 histologic mimics. Mutant IDH1/2 IHC was positive in 26/53 SNUCs (49%; 20 strongly positive and 6 weak) and 3/8 PDCARs (37.5%; 2 strong; 1 weak) but was absent in all other tumor types (0/132). Targeted next-generation sequencing (NGS) on a subset of SNUC/PDCAR (6 strong and 3 weak positive for IDH1/2 IHC; 7 negative) showed frequent IDH2 R172X mutations (10/16) and a single IDH1 R132C mutation. All 6 cases with strong positive mutant IDH1/2 staining and NGS had IDH2 R172S/G mutations. The 3 IHC-weak cases all had IDH2 R172T mutations. Among the 7 tested cases that were negative for mutant IDH1/2 IHC, NGS detected 1 case each with IDH2 R172T and IDH1 R132C mutation. IDH-mutant carcinomas also had frequent mutations in TP53 (55%) and activating mutations in KIT (45%) or the PI3K pathway (36%). Mutation-specific IDH1/2 IHC identifies IDH mutations in SNUC, however, it lacks sensitivity for the full range of IDH mutations. These findings suggest that IDH-mutant sinonasal carcinoma may represent a distinct pathobiological entity with therapeutic implications that can be identified by a combined approach of multispecific IDH1/2 IHC and sequencing.
Collapse
|
406
|
Bowler TG, Pradhan K, Kong Y, Bartenstein M, Morrone KA, Sridharan A, Kessel RM, Shastri A, Giricz O, Bhagat TD, Gordon-Mitchell S, Rohanizadegan M, Hooda L, Datt I, Przychodzen BP, Parmar S, Maqbool S, Maciejewski JP, Steidl U, Greally JM, Verma A. Misidentification of MLL3 and other mutations in cancer due to highly homologous genomic regions. Leuk Lymphoma 2019; 60:3132-3137. [PMID: 31288594 DOI: 10.1080/10428194.2019.1630620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The MLL3 gene has been shown to be recurrently mutated in many malignancies including in families with acute myeloid leukemia. We demonstrate that many MLL3 variant calls made by exome sequencing are false positives due to misalignment to homologous regions, including a region on chr21, and can only be validated by long-range PCR. Numerous other recurrently mutated genes reported in COSMIC and TCGA databases have pseudogenes and cannot also be validated by conventional short read-based sequencing approaches. Genome-wide identification of pseudogene regions demonstrates that frequency of these homologous regions is increased with sequencing read lengths below 200 bps. To enable identification of poor quality sequencing variants in prospective studies, we generated novel genome-wide maps of regions with poor mappability that can be used in variant calling algorithms. Taken together, our findings reveal that pseudogene regions are a source of false-positive mutations in cancers.
Collapse
Affiliation(s)
- Timothy G Bowler
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Kith Pradhan
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Yu Kong
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | | | - Kerry A Morrone
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Ashwin Sridharan
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Rachel M Kessel
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Aditi Shastri
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Orsi Giricz
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Tushar D Bhagat
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | | | | | - Lauren Hooda
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Ishan Datt
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | | | | | - Shahina Maqbool
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | | | - Ulrich Steidl
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - John M Greally
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Amit Verma
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
407
|
Bala Bhaskara Rao K, Katragunta K, Sarma UM, Jain N. Abundance of
d
‐2‐hydroxyglutarate in G2/M is determined by FOXM1 in mutant IDH1‐expressing cells. FEBS Lett 2019; 593:2177-2193. [DOI: 10.1002/1873-3468.13500] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/22/2019] [Accepted: 06/07/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Kancharana Bala Bhaskara Rao
- Department of Applied Biology CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovative Research (AcSIR) New Delhi India
| | - Kumar Katragunta
- Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
| | - Uma Maheswara Sarma
- Organic Synthesis and Process Chemistry CSIR‐Indian Institute of Chemical Technology Hyderabad India
| | - Nishant Jain
- Department of Applied Biology CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovative Research (AcSIR) New Delhi India
| |
Collapse
|
408
|
Dwight T, Kim E, Novos T, Clifton-Bligh RJ. Metabolomics in the Diagnosis of Pheochromocytoma and Paraganglioma. Horm Metab Res 2019; 51:443-450. [PMID: 31307108 DOI: 10.1055/a-0926-3790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Metabolomics refers to the detection and measurement of small molecules (metabolites) within biological systems, and is therefore a powerful tool for identifying dysfunctional cellular physiologies. For pheochromocytomas and paragangliomas (PPGLs), metabolomics has the potential to become a routine addition to histology and genomics for precise diagnostic evaluation. Initial metabolomic studies of ex vivo tumors confirmed, as expected, succinate accumulation in PPGLs associated with pathogenic variants in genes encoding succinate dehydrogenase subunits or their assembly factors (SDHx). Metabolomics has now shown utility in clarifying SDHx variants of uncertain significance, as well as the accurate diagnosis of PPGLs associated with fumarate hydratase (FH), isocitrate dehydrogenase (IDH), malate dehydrogenase (MDH2) and aspartate transaminase (GOT2). The emergence of metabolomics resembles the advent of genetic testing in this field, which began with single-gene discoveries in research laboratories but is now done by standardized massively parallel sequencing (targeted panel/exome/genome testing) in pathology laboratories governed by strict credentialing and governance requirements. In this setting, metabolomics is poised for rapid translation as it can utilize existing infrastructure, namely liquid chromatography-tandem mass spectrometry (LC-MS/MS), for the measurement of catecholamine metabolites. Metabolomics has also proven tractable to in vivo diagnosis of SDH-deficient PPGLs using magnetic resonance spectroscopy (MRS). The future of metabolomics - embedded as a diagnostic tool - will require adoption by pathologists to shepherd development of standardized assays and sample preparation, reference ranges, gold standards, and credentialing.
Collapse
Affiliation(s)
- Trisha Dwight
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, St Leonards, Australia
- University of Sydney, Sydney, Australia
| | - Edward Kim
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, St Leonards, Australia
- University of Sydney, Sydney, Australia
| | - Talia Novos
- Clinical Chemistry, South Eastern Area Laboratory Services Pathology, Prince of Wales Private Hospital, Randwick, Australia
| | - Roderick J Clifton-Bligh
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, St Leonards, Australia
- University of Sydney, Sydney, Australia
- Department of Endocrinology, Royal North Shore Hospital, St Leonards, Australia
| |
Collapse
|
409
|
The Yin and Yang of cancer genes. Gene 2019; 704:121-133. [DOI: 10.1016/j.gene.2019.04.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/21/2019] [Accepted: 04/08/2019] [Indexed: 12/31/2022]
|
410
|
Unruh D, Zewde M, Buss A, Drumm MR, Tran AN, Scholtens DM, Horbinski C. Methylation and transcription patterns are distinct in IDH mutant gliomas compared to other IDH mutant cancers. Sci Rep 2019; 9:8946. [PMID: 31222125 PMCID: PMC6586617 DOI: 10.1038/s41598-019-45346-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/05/2019] [Indexed: 02/08/2023] Open
Abstract
Mutations in isocitrate dehydrogenases 1 and 2 (IDHmut) are present in a variety of cancers, including glioma, acute myeloid leukemia (AML), melanoma, and cholangiocarcinoma. These mutations promote hypermethylation, yet it is only a favorable prognostic marker in glioma, for reasons that are unclear. We hypothesized that the patterns of DNA methylation, and transcriptome profiles, would vary among IDHmut cancers, especially gliomas. Using Illumina 450K and RNA-Seq data from The Cancer Genome Atlas, we show that of 365,092 analyzed CpG sites, 70,591 (19%) were hypermethylated in IDHmut gliomas compared to wild-type (IDHwt) gliomas, and only 3%, 2%, and 4% of CpG sites were hypermethylated in IDHmut AML, melanoma, and cholangiocarcinoma, relative to each of their IDHwt counterparts. Transcriptome differences showed pro-malignant genes that appear to be unique to IDHmut gliomas. However, genes involved in differentiation and immune response were suppressed in all IDHmut cancers. Additionally, IDHmut caused a greater degree of hypermethylation in undifferentiated neural progenitor cells than in mature astrocytes. These data suggest that the extent and targets of IDHmut-induced genomic hypermethylation vary greatly according to the cellular context and may help explain why IDHmut is only a favorable prognostic marker in gliomas.
Collapse
Affiliation(s)
- Dusten Unruh
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Makda Zewde
- Department of Preventive Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Adam Buss
- Department of Preventive Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Michael R Drumm
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Anh N Tran
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Denise M Scholtens
- Department of Preventive Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA.
- Department of Pathology, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
411
|
The cancer driver genes IDH1/2, JARID1C/ KDM5C, and UTX/ KDM6A: crosstalk between histone demethylation and hypoxic reprogramming in cancer metabolism. Exp Mol Med 2019; 51:1-17. [PMID: 31221981 PMCID: PMC6586683 DOI: 10.1038/s12276-019-0230-6] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 12/12/2018] [Indexed: 12/16/2022] Open
Abstract
Recent studies on mutations in cancer genomes have distinguished driver mutations from passenger mutations, which occur as byproducts of cancer development. The cancer genome atlas (TCGA) project identified 299 genes and 24 pathways/biological processes that drive tumor progression (Cell 173: 371-385 e318, 2018). Of the 299 driver genes, 12 genes are involved in histones, histone methylation, and demethylation. Among these 12 genes, those encoding the histone demethylases JARID1C/KDM5C and UTX/KDM6A were identified as cancer driver genes. Furthermore, gain-of-function mutations in genes encoding metabolic enzymes, such as isocitrate dehydrogenases (IDH)1/2, drive tumor progression by producing an oncometabolite, D-2-hydroxyglutarate (D-2HG), which is a competitive inhibitor of α-ketoglutarate, O2-dependent dioxygenases such as Jumonji domain-containing histone demethylases, and DNA demethylases. Studies on oncometabolites suggest that histone demethylases mediate metabolic changes in chromatin structure. We have reviewed the most recent findings regarding cancer-specific metabolic reprogramming and the tumor-suppressive roles of JARID1C/KDM5C and UTX/KDM6A. We have also discussed mutations in other isoforms such as the JARID1A, 1B, 1D of KDM5 subfamilies and the JMJD3/KDM6B of KDM6 subfamilies, which play opposing roles in tumor progression as oncogenes or tumor suppressors depending on the cancer cell type. Genes involved in the removal of methyl groups from histones associated with DNA can promote or suppress tumor growth depending on the metabolic status of the cancer cell. Hyunsung Park and colleagues at the University of Seoul, South Korea, review current knowledge of two genes encoding histone demethylases which have been identified by The Cancer Genome Atlas (TCGA) project as cancer driver genes. Because these demethylase enzymes rely on cellular metabolites to function, their effect is influenced by metabolic conditions in the tumor microenvironment such as low oxygen. The mechanisms through which changes in histone methylation affect the expression of genes involved in tumor progression remain unknown. Further understanding of how cancer metabolism affects the modification of histones will help guide the development of more effective cancer treatments.
Collapse
|
412
|
Patkar N, Kakirde C, Bhanshe P, Joshi S, Chaudhary S, Badrinath Y, Ghoghale S, Deshpande N, Kadechkar S, Chatterjee G, Kannan S, Shetty D, Gokarn A, Punatkar S, Bonda A, Nayak L, Jain H, Bagal B, Menon H, Sengar M, Khizer SH, Khattry N, Tembhare P, Gujral S, Subramanian P. Utility of Immunophenotypic Measurable Residual Disease in Adult Acute Myeloid Leukemia-Real-World Context. Front Oncol 2019; 9:450. [PMID: 31263671 PMCID: PMC6584962 DOI: 10.3389/fonc.2019.00450] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/13/2019] [Indexed: 01/10/2023] Open
Abstract
Introduction: One of the mainstays of chemotherapy in acute myeloid leukemia (AML) is induction with a goal to achieve morphological complete remission (CR). However, not all patients by this remission criterion achieve long-term remission and a subset relapse. This relapse is explained by the presence of measurable residual disease (MRD). Methods: We accrued 451 consecutive patients of adult AML (from March 2012 to December 2017) after informed consent. All patients received standard chemotherapy. MRD testing was done at post-induction and, if feasible, post-consolidation using 8- and later 10-color FCM. Analysis of MRD was done using a combination of difference from normal and leukemia-associated immunophenotype approaches. Conventional karyotyping and FISH were done as per standard recommendations, and patients were classified into favorable, intermediate, and poor cytogenetic risk groups. The presence of FLT3-ITD, NPM1, and CEBPA mutations was detected by a fragment length analysis-based assay. Results: As compared to Western data, our cohort of patients was younger with a median age of 35 years. There were 62 induction deaths in this cohort (13.7%), and 77 patients (17.1%) were not in morphological remission. The median follow-up was 26.0 months. Poor-risk cytogenetics and the presence of FLT3-ITD were significantly associated with inferior outcome. The presence of post-induction MRD assessment was significantly associated with adverse outcome with respect to OS (p = 0.01) as well as RFS (p = 0.004). Among established genetic subgroups, detection of MRD in intermediate cytogenetic and NPM1 mutated groups was also highly predictive of inferior outcome. On multivariate analysis, immunophenotypic MRD at the end of induction and FLT3-ITD emerged as independent prognostic factors predictive for outcome. Conclusion: This is the first data from a resource-constrained real-world setting demonstrating the utility of AML MRD as well as long-term outcome of AML. Our data is in agreement with other studies that determination of MRD is extremely important in predicting outcome. AML MRD is a very useful guide for guiding post-remission strategies in AML and should be incorporated into routine treatment algorithms.
Collapse
Affiliation(s)
- Nikhil Patkar
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Chinmayee Kakirde
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Prasanna Bhanshe
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Swapnali Joshi
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Shruti Chaudhary
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | | | - Sitaram Ghoghale
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Nilesh Deshpande
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Shraddha Kadechkar
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Gaurav Chatterjee
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Sadhana Kannan
- Biostatistics, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Dhanalaxmi Shetty
- Department of Cytogenetics, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Anant Gokarn
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Sachin Punatkar
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Avinash Bonda
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Lingaraj Nayak
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Hasmukh Jain
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Bhausaheb Bagal
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Hari Menon
- Haemato-Oncology, CyteCare Cancer Hospital, Bangalore, India
| | - Manju Sengar
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Syed Hasan Khizer
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Navin Khattry
- Adult Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Prashant Tembhare
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | - Sumeet Gujral
- Haematopathology Laboratory, ACTREC, Tata Memorial Centre, Mumbai, India
| | | |
Collapse
|
413
|
The multifaceted contribution of α-ketoglutarate to tumor progression: An opportunity to exploit? Semin Cell Dev Biol 2019; 98:26-33. [PMID: 31175937 DOI: 10.1016/j.semcdb.2019.05.031] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 01/25/2023]
Abstract
The thriving field that constitutes cancer metabolism has unveiled some groundbreaking facts over the past two decades, at the heart of which is the TCA cycle and its intermediates. As such and besides its metabolic role, α-ketoglutarate was shown to withstand a wide range of physiological reactions from protection against oxidative stress, collagen and bone maintenance to development and immunity. Most importantly, it constitutes the rate-limiting substrate of 2-oxoglutarate-dependent dioxygenases family enzymes, which are involved in hypoxia sensing and in the shaping of cellular epigenetic landscape, two major drivers of oncogenic transformation. Based on literature reports, we hereby review the benefits of this metabolite as a possible novel adjuvant therapeutic opportunity to target tumor progression. This article is part of the special issue "Mitochondrial metabolic alterations in cancer cells and related therapeutic targets".
Collapse
|
414
|
Ibrahim A, Chopra S. Succinate Dehydrogenase–Deficient Gastrointestinal Stromal Tumors. Arch Pathol Lab Med 2019; 144:655-660. [DOI: 10.5858/arpa.2018-0370-rs] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Succinate dehydrogenase (SDH)–deficient gastrointestinal stromal tumor (GIST) is a subset of wild-type GIST that constitutes approximately 10% of gastric GISTs. SDH-mutated GISTs lack mutations in the proto-oncogene receptor tyrosine kinase (also known as KIT, c-KIT, or CD117) or platelet-derived growth factor receptor α (PDGFR-α). These tumors have female predilection, affect children and young adults, and have a spectrum of behavior from indolent to progressive. These tumors have characteristic morphologic features including multinodular architecture, multiple tumors, lymphovascular involvement, and occasional lymph node metastasis. They can be seen in patients with Carney triad or Carney-Stratakis syndrome. Although a mutation in any one of the SDH subunits can be pathogenic, deficiency of a single subunit leads to loss of detectable SDH subunit B by immunohistochemistry, enabling a convenient, tissue-based screening method. The prognosis and the clinical course of these tumors is different from that of KIT- or PDGFR-α–mutated GISTs. Surgical management is considered the main line of treatment. SDH-mutated GISTs do not respond well to the common targeted therapy, with no objective tumor response to imatinib. The role of the pathologist in diagnosing these cases is imperative in management and subsequent follow-up.
Collapse
Affiliation(s)
- Ahmad Ibrahim
- From the Department of Pathology, LAC + USC Medical Center, University of Southern California, Keck School of Medicine, Los Angeles (Dr Ibrahim); and the Department of Pathology, University of Southern California, Keck School of Medicine, Los Angeles (Dr Chopra)
| | - Shefali Chopra
- From the Department of Pathology, LAC + USC Medical Center, University of Southern California, Keck School of Medicine, Los Angeles (Dr Ibrahim); and the Department of Pathology, University of Southern California, Keck School of Medicine, Los Angeles (Dr Chopra)
| |
Collapse
|
415
|
Exner ND, Valenzuela JAC, Abou-El-Ardat K, Miletic H, Huszthy PC, Radehaus PM, Schröck E, Bjerkvig R, Kaderali L, Klink B, Nigro JM. Deep sequencing of a recurrent oligodendroglioma and the derived xenografts reveals new insights into the evolution of human oligodendroglioma and candidate driver genes. Oncotarget 2019; 10:3641-3653. [PMID: 31217899 PMCID: PMC6557204 DOI: 10.18632/oncotarget.26950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/04/2019] [Indexed: 12/23/2022] Open
Abstract
We previously reported the establishment of a rare xenograft derived from a recurrent oligodendroglioma with 1p/19q codeletion. Here, we analyzed in detail the exome sequencing datasets from the recurrent oligodendroglioma (WHO grade III, recurrent O2010) and the first-generation xenograft (xenograft1). Somatic SNVs and small InDels (n = 80) with potential effects at the protein level in recurrent O2010 included variants in IDH1 (NM_005896:c.395G>A; p. Arg132His), FUBP1 (NM_003902:c.1307_1310delTAGA; p.Ile436fs), and CIC (NM_015125:c.4421T>G; p.Val1474Gly). All but 2 of these 80 variants were also present in xenograft1, along with 7 new variants. Deep sequencing of the 87 SNVs and InDels in the original tumor (WHO grade III, primary O2005) and in a second-generation xenograft (xenograft2) revealed that only 11 variants, including IDH1 (NM_005896:c.395G>A; p. Arg132His), PSKH1 (NM_006742.2:c.650G>A; p.Arg217Gln), and SNX12 (NM_001256188:c.470G>A; p.Arg157His), along with a variant in the TERT promoter (C250T, NM_198253.2: c.-146G>A), were already present in primary O2005. Allele frequencies of the 11 variants were calculated to assess their potential as putative driver genes. A missense change in NDST4 (NM_022569:c.2392C>G; p.Leu798Val) on 4q exhibited an increasing allele frequency (~ 20%, primary O2005, 80%, recurrent O2010 and 100%, xenograft1), consistent with a selection event. Sequencing of NDST4 in a cohort of 15 oligodendrogliomas, however, revealed no additional cases with potential protein disrupting variants. Our analysis illuminated a tumor evolutionary series of events, which included 1p/19q codeletion, IDH1 R132H, and TERT C250T as early events, followed by loss of function of NDST4 and mutations in FUBP1 and CIC as late events.
Collapse
Affiliation(s)
- Nadin D Exner
- Institut für Klinische Genetik, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,University of Applied Sciences Mittweida, Department of Applied Informatics & Biosciences, Mittweida, Germany
| | - Jaime Alberto Campos Valenzuela
- Institut für Medizinische Informatik und Biometrie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Khalil Abou-El-Ardat
- Institut für Klinische Genetik, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hrvoje Miletic
- Department of Pathology, Haukeland University Hospital, Bergen, Norway.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Peter C Huszthy
- Oslo University Hospital-Rikshospitalet, Department of Immunology, Oslo, Norway
| | - Petra M Radehaus
- University of Applied Sciences Mittweida, Department of Applied Informatics & Biosciences, Mittweida, Germany
| | - Evelin Schröck
- Institut für Klinische Genetik, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Molecular Tumor Diagnostics, National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Oncology Department, Luxembourg Institute of Health, Val Fleuri, Luxembourg
| | - Lars Kaderali
- University Medicine Greifswald, Institute of Bioinformatics, Greifswald, Germany
| | - Barbara Klink
- Institut für Klinische Genetik, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Molecular Tumor Diagnostics, National Center for Tumor Diseases (NCT), Dresden, Germany.,Centre national de Génétique, Laboratoire National de Santé, Dudelange, Luxembourg.,Co-senior authors
| | - Janice M Nigro
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Co-senior authors
| |
Collapse
|
416
|
Papayannidis C, Sartor C, Marconi G, Fontana MC, Nanni J, Cristiano G, Parisi S, Paolini S, Curti A. Acute Myeloid Leukemia Mutations: Therapeutic Implications. Int J Mol Sci 2019; 20:ijms20112721. [PMID: 31163594 PMCID: PMC6600275 DOI: 10.3390/ijms20112721] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 01/25/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is an extremely heterogeneous group of hematological neoplasms, for which allogeneic stem cell transplantation (HSCT) still represents the only potentially curative option in the majority of cases. However, elderly age and clinically severe comorbidities may often exclude a wide amount of patients from this therapeutic approach, underlying the urgent need for alternative strategies. Thanks to the introduction of advanced high-throughput techniques, light is being shed on the pathogenesis of AML, identifying molecular recurrent mutations as responsible for the onset, as well as progression, of disease. As a consequence, and in parallel, many new compounds, including targeted therapies (FMS-like tyrosine kinase 3 (FLT3) and Isocitrate dehydrogenase 1-2 (IDH1-2) inhibitors), have found a wide room of application in this setting, and are now available in daily practice, or in late phases of clinical development. Moreover, several further innovative molecules are currently under investigation, and promising results for many of them have already been reported. In this review, we will present an update on the most relevant molecular alterations of AML, focusing on the most frequent genomic mutations of the disease, for which compounds have been approved or are still currently under investigation.
Collapse
Affiliation(s)
- Cristina Papayannidis
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Chiara Sartor
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Giovanni Marconi
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Maria Chiara Fontana
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Jacopo Nanni
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Gianluca Cristiano
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Sarah Parisi
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Stefania Paolini
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Antonio Curti
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| |
Collapse
|
417
|
What is the role of a second allogeneic hematopoietic cell transplant in relapsed acute myeloid leukemia? Bone Marrow Transplant 2019; 55:325-331. [PMID: 31160807 DOI: 10.1038/s41409-019-0584-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/29/2019] [Accepted: 05/15/2019] [Indexed: 11/08/2022]
Abstract
Relapsed acute myeloid leukemia (AML) after an allogeneic hematopoietic cell transplant (allo-HCT) entails a poor prognosis. Treating these cases is challenging due to lack of effective therapies and, in some cases, poor performance status and/or presence of graft-versus-host disease (GVHD), among others. No randomized controlled trial (RCT) has ever been conducted comparing a second allo-HCT against other treatments. Existing data are mainly from observational studies or registries. Success of a second allo-HCT is dependent on appropriately selecting patients who might achieve best outcomes with reasonable non-relapse mortality (NRM) risk. Several factors are associated with worse outcomes, namely a shorter time from first allo-HCT to relapse or to the second allo-HCT, and AML not being in complete hematologic remission (CR). Patients relapsing earlier than 6 months or having active/persistent disease should be enrolled in clinical trials. Limitations of the published literature include retrospective small size studies, a heterogeneous population, and absence of information on somatic mutations, among others. Future studies assessing the role of a second allo-HCT should evaluate the impact of IDH1, IDH2, or others on outcomes; and the feasibility and efficacy of targeted therapies in the pre-, peri-, or post-second allo-HCT setting.
Collapse
|
418
|
Horton S, Huntly BJP. Kinase Networks Regulate Metabolism: I'D(H1) Never Have Guessed! Cancer Discov 2019; 9:699-701. [PMID: 31160331 DOI: 10.1158/2159-8290.cd-19-0373] [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] [Indexed: 11/16/2022]
Abstract
Mutations in isoforms of isocitrate dehydrogenase (IDH) enzymes are described in multiple cancers and both mutant and wild-type IDH are important for the generation and maintenance of tumors, but how their activity is regulated is poorly understood. An article in this issue of Cancer Discovery identifies a novel posttranslational mechanism of IDH1 regulation involving phosphorylation of specific tyrosine residues by a network of kinases that alter the specificity of substrate and cofactor binding, dimer formation, and ultimately enzyme activity.See related article by Chen et al., p. 756.
Collapse
Affiliation(s)
- Sarah Horton
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Brian J P Huntly
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom.
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
419
|
Sinha S, Ray A, Abhilash L, Kumar M, Sreenivasamurthy SK, Keshava Prasad TS, Inamdar MS. Proteomics of Asrij Perturbation in Drosophila Lymph Glands for Identification of New Regulators of Hematopoiesis. Mol Cell Proteomics 2019; 18:1171-1182. [PMID: 30923041 PMCID: PMC6553936 DOI: 10.1074/mcp.ra119.001299] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/07/2019] [Indexed: 11/06/2022] Open
Abstract
Hematopoiesis is the process of differentiation of precursor blood cells into mature blood cells that is controlled by a complex set of molecular interactions. Understanding hematopoiesis is important for the study of hematological disorders. However, a comprehensive understanding of how physiological and genetic mechanisms regulate blood cell precursor maintenance and differentiation is lacking. Owing to simplicity and ease of genetic analysis, the Drosophila melanogaster lymph gland (LG) is an excellent model to study hematopoiesis. Here, we quantitatively analyzed the LG proteome under genetic conditions that either maintain precursors or promote their differentiation in vivo, by perturbing expression of Asrij, a conserved endosomal regulator of hematopoiesis. Using iTRAQ-based quantitative proteomics, we determined the relative expression levels of proteins in Asrij-knockout and overexpressing LGs from 1500 larval dissections compared with wild type. Our data showed that at least 6.5% of the Drosophila proteome is expressed in wild type LGs. Of the 2133 proteins identified, 780 and 208 proteins were common to previously reported cardiac tube and hemolymph proteomes, respectively, resulting in the identification of 1238 proteins exclusive to the LG. Perturbation of Asrij levels led to differential expression of 619 proteins, of which 27% have human homologs implicated in various diseases. Proteins regulating metabolism, immune system, signal transduction and vesicle-mediated transport were significantly enriched. Immunostaining of representative candidates from the enriched categories and previous reports confirmed 73% of our results, indicating the validity of our LG proteome. Our study provides, for the first time, an in vivo proteomics resource for identifying novel regulators of hematopoiesis that will also be applicable to understanding vertebrate blood cell development.
Collapse
Affiliation(s)
- Saloni Sinha
- From the ‡Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064, India
| | - Arindam Ray
- From the ‡Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064, India
| | - Lakshman Abhilash
- From the ‡Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064, India
| | - Manish Kumar
- §Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- ¶Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Sreelakshmi K Sreenivasamurthy
- §Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- ‖NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - T S Keshava Prasad
- §Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- **Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to be University), Mangalore-575018, India
| | - Maneesha S Inamdar
- From the ‡Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064, India;
- ‡‡Institute for Stem Cell Biology and Regenerative Medicine, GKVK, Bellary Road, Bangalore 560065, India
| |
Collapse
|
420
|
Kim GH, Choi SY, Oh TI, Kan SY, Kang H, Lee S, Oh T, Ko HM, Lim JH. IDH1 R132H Causes Resistance to HDAC Inhibitors by Increasing NANOG in Glioblastoma Cells. Int J Mol Sci 2019; 20:ijms20112679. [PMID: 31151327 PMCID: PMC6600637 DOI: 10.3390/ijms20112679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/23/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
The R132H mutation in isocitrate dehydrogenase 1 (IDH1R132H) is commonly observed and associated with better survival in glioblastoma multiforme (GBM), a malignant brain tumor. However, the functional role of IDH1R132H as a molecular target for GBM treatment is not completely understood. In this study, we found that the overexpression of IDH1R132H suppresses cell growth, cell cycle progression and motility in U87MG glioblastoma cells. Based on cell viability and apoptosis assays, we found that IDH1R132H-overexpressing U87MG and U373MG cells are resistant to the anti-cancer effect of histone deacetylase inhibitors (HDACi), such as trichostatin A (TSA), vorinostat (SAHA), and valproic acid. Octyl-(R)-2-hydroxyglutarate (Octyl-2HG), which is a membrane-permeable precursor form of the oncometabolite (R)-2-hydroxyglutarate (R-2HG) produced in IDH1-mutant tumor cells, significantly increased HDACi resistance in glioblastoma cells. Mechanistically, IDH1R132H and Octyl-2HG enhanced the promoter activation of NANOG via increased H3K4-3Me, consequently increasing NANOG mRNA and protein expression. Indeed, HDACi resistance was attenuated in IDH1R132H-expressing glioblastoma cells by the suppression of NANOG using small interfering RNAs. Furthermore, we found that AGI-5198, a selective inhibitor of IDH1R132H, significantly attenuates HDACi resistance and NANOG expression IDH1R132H-expressing glioblastoma cells. These results suggested that IDH1R132H is a potential molecular target for HDACi-based therapy for GBM.
Collapse
Affiliation(s)
- Geon-Hee Kim
- Department of Applied Life Science, Graduate School of Konkuk University, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - So Young Choi
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Taek-In Oh
- Department of Applied Life Science, Graduate School of Konkuk University, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Sang-Yeon Kan
- Department of Applied Life Science, Graduate School of Konkuk University, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Hyeji Kang
- Department of Applied Life Science, Graduate School of Konkuk University, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Sujin Lee
- Department of Applied Life Science, Graduate School of Konkuk University, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Taerim Oh
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Hyun Myung Ko
- Department of Life Science, College of Science and Technology, Woosuk University, 66 Daehak-ro, Jincheon-eup, Chungcheongbuk-do 27841, Korea.
| | - Ji-Hong Lim
- Department of Applied Life Science, Graduate School of Konkuk University, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| |
Collapse
|
421
|
Tang J, Lee T, Sun T. Single-nucleotide editing: From principle, optimization to application. Hum Mutat 2019; 40:2171-2183. [PMID: 31131955 DOI: 10.1002/humu.23819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/30/2019] [Accepted: 05/23/2019] [Indexed: 12/26/2022]
Abstract
Cytosine base editors (CBEs) and adenine base editors (ABEs), which are generally composed of an engineered deaminase and a catalytically impaired CRISPR-Cas9 variant, are new favorite tools for single base substitution in cells and organisms. In this review, we summarize the principle of base-editing systems and elaborate on the evolution of different platforms of CBEs and ABEs, including their deaminase, Cas9 variants, and editing outcomes. Moreover, we highlight their applications in mouse and human cells and discuss the challenges and prospects of base editors. The ABE- and CBE systems have been used in gene silencing, pathogenic gene correction, and functional genetic screening. Single base editing is becoming a new promising genetic tool in biomedical research and gene therapy.
Collapse
Affiliation(s)
- Jinling Tang
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Trevor Lee
- Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, New York
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| |
Collapse
|
422
|
Peeters TH, Lenting K, Breukels V, van Lith SAM, van den Heuvel CNAM, Molenaar R, van Rooij A, Wevers R, Span PN, Heerschap A, Leenders WPJ. Isocitrate dehydrogenase 1-mutated cancers are sensitive to the green tea polyphenol epigallocatechin-3-gallate. Cancer Metab 2019; 7:4. [PMID: 31139406 PMCID: PMC6526618 DOI: 10.1186/s40170-019-0198-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/09/2019] [Indexed: 01/09/2023] Open
Abstract
Background Mutations in isocitrate dehydrogenase 1 (IDH1) occur in various types of cancer and induce metabolic alterations resulting from the neomorphic activity that causes production of D-2-hydroxyglutarate (D-2-HG) at the expense of α-ketoglutarate (α-KG) and NADPH. To overcome metabolic stress induced by these alterations, IDH-mutated (IDHmut) cancers utilize rescue mechanisms comprising pathways in which glutaminase and glutamate dehydrogenase (GLUD) are involved. We hypothesized that inhibition of glutamate processing with the pleiotropic GLUD-inhibitor epigallocatechin-3-gallate (EGCG) would not only hamper D-2-HG production, but also decrease NAD(P)H and α-KG synthesis in IDHmut cancers, resulting in increased metabolic stress and increased sensitivity to radiotherapy. Methods We performed 13C-tracing studies to show that HCT116 colorectal cancer cells with an IDH1R132H knock-in allele depend more on glutaminolysis than on glycolysis for the production of D-2-HG. We treated HCT116 cells, HCT116-IDH1R132H cells, and HT1080 cells (carrying an IDH1R132C mutation) with EGCG and evaluated D-2-HG production, cell proliferation rates, and sensitivity to radiotherapy. Results Significant amounts of 13C from glutamate accumulate in D-2-HG in HCT116-IDH1wt/R132H but not in HCT116-IDH1wt/wt. Preventing glutamate processing in HCT116-IDH1wt/R132H cells with EGCG resulted in reduction of D-2-HG production. In addition, EGCG treatment decreased proliferation rates of IDH1mut cells and at high doses sensitized cancer cells to ionizing radiation. Effects of EGCG in IDH-mutated cell lines were diminished by treatment with the IDH1mut inhibitor AGI-5198. Conclusions This work shows that glutamate can be directly processed into D-2-HG and that reduction of glutamatolysis may be an effective and promising new treatment option for IDHmut cancers. Electronic supplementary material The online version of this article (10.1186/s40170-019-0198-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tom H Peeters
- 1Department of Radiology and Nuclear Medicine, Radboud university medical center, PO Box 9101, 6500 Nijmegen, HB The Netherlands
| | - Krissie Lenting
- 2Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 26, 6525 Nijmegen, GA The Netherlands
| | - Vincent Breukels
- 1Department of Radiology and Nuclear Medicine, Radboud university medical center, PO Box 9101, 6500 Nijmegen, HB The Netherlands
| | - Sanne A M van Lith
- 1Department of Radiology and Nuclear Medicine, Radboud university medical center, PO Box 9101, 6500 Nijmegen, HB The Netherlands
| | - Corina N A M van den Heuvel
- 2Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 26, 6525 Nijmegen, GA The Netherlands
| | - Remco Molenaar
- 3Department of Medical Biology, Cancer Center Amsterdam at the Academic Medical Center, Meibergdreef 15, 1105 Amsterdam, AZ The Netherlands
| | - Arno van Rooij
- 4Department of Laboratory Medicine, Radboud university medical center, PO Box 9101, 6500 Nijmegen, HB The Netherlands
| | - Ron Wevers
- 4Department of Laboratory Medicine, Radboud university medical center, PO Box 9101, 6500 Nijmegen, HB The Netherlands
| | - Paul N Span
- 5Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, PO Box 9101, 6500 Nijmegen, HB The Netherlands
| | - Arend Heerschap
- 1Department of Radiology and Nuclear Medicine, Radboud university medical center, PO Box 9101, 6500 Nijmegen, HB The Netherlands
| | - William P J Leenders
- 2Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 26, 6525 Nijmegen, GA The Netherlands
| |
Collapse
|
423
|
Golub D, Iyengar N, Dogra S, Wong T, Bready D, Tang K, Modrek AS, Placantonakis DG. Mutant Isocitrate Dehydrogenase Inhibitors as Targeted Cancer Therapeutics. Front Oncol 2019; 9:417. [PMID: 31165048 PMCID: PMC6534082 DOI: 10.3389/fonc.2019.00417] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 05/02/2019] [Indexed: 12/15/2022] Open
Abstract
The identification of heterozygous neomorphic isocitrate dehydrogenase (IDH) mutations across multiple cancer types including both solid and hematologic malignancies has revolutionized our understanding of oncogenesis in these malignancies and the potential for targeted therapeutics using small molecule inhibitors. The neomorphic mutation in IDH generates an oncometabolite product, 2-hydroxyglutarate (2HG), which has been linked to the disruption of metabolic and epigenetic mechanisms responsible for cellular differentiation and is likely an early and critical contributor to oncogenesis. In the past 2 years, two mutant IDH (mutIDH) inhibitors, Enasidenib (AG-221), and Ivosidenib (AG-120), have been FDA-approved for IDH-mutant relapsed or refractory acute myeloid leukemia (AML) based on phase 1 safety and efficacy data and continue to be studied in trials in hematologic malignancies, as well as in glioma, cholangiocarcinoma, and chondrosarcoma. In this review, we will summarize the molecular pathways and oncogenic consequences associated with mutIDH with a particular emphasis on glioma and AML, and systematically review the development and preclinical testing of mutIDH inhibitors. Existing clinical data in both hematologic and solid tumors will likewise be reviewed followed by a discussion on the potential limitations of mutIDH inhibitor monotherapy and potential routes for treatment optimization using combination therapy.
Collapse
Affiliation(s)
- Danielle Golub
- Department of Neurosurgery, New York University School of Medicine, NYU Langone Health, New York, NY, United States.,Clinical and Translational Science Institute, New York University School of Medicine, NYU Langone Health, New York, NY, United States
| | - Nishanth Iyengar
- New York University School of Medicine, NYU Langone Health, New York, NY, United States
| | - Siddhant Dogra
- New York University School of Medicine, NYU Langone Health, New York, NY, United States
| | - Taylor Wong
- Department of Neurosurgery, New York University School of Medicine, NYU Langone Health, New York, NY, United States
| | - Devin Bready
- Department of Neurosurgery, New York University School of Medicine, NYU Langone Health, New York, NY, United States
| | - Karen Tang
- Clinical and Translational Science Institute, New York University School of Medicine, NYU Langone Health, New York, NY, United States.,Division of Hematology/Oncology, Department of Pediatrics, New York University School of Medicine, NYU Langone Health, New York, NY, United States
| | - Aram S Modrek
- Department of Radiation Oncology, New York University School of Medicine, NYU Langone Health, New York, NY, United States
| | - Dimitris G Placantonakis
- Department of Neurosurgery, New York University School of Medicine, NYU Langone Health, New York, NY, United States.,Kimmel Center for Stem Cell Biology, New York University School of Medicine, NYU Langone Health, New York, NY, United States.,Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, NY, United States.,Brain Tumor Center, New York University School of Medicine, NYU Langone Health, New York, NY, United States.,Neuroscience Institute, New York University School of Medicine, NYU Langone Health, New York, NY, United States
| |
Collapse
|
424
|
DeZern AE, Malcovati L, Ebert BL. CHIP, CCUS, and Other Acronyms: Definition, Implications, and Impact on Practice. Am Soc Clin Oncol Educ Book 2019; 39:400-410. [PMID: 31099654 DOI: 10.1200/edbk_239083] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Unexplained blood cytopenias can be a clinical challenge for patients and clinicians alike. The relationship between these cytopenias and myeloid neoplasms like myelodysplastic syndromes (MDS) is currently an area of active research. There have been marked developments in our understanding of clonal hematopoiesis based on findings of somatic mutations in genes known to be associated with MDS. This has led to newer terms to describe precursor states to MDS, such as clonal hematopoiesis of indeterminate potential (CHIP) and clonal cytopenia of undetermined significance (CCUS). These conditions may allow earlier diagnosis, modify surveillance for MDS, and guide additional therapies. This review summarizes recent updates in the field for affected patients.
Collapse
Affiliation(s)
- Amy E DeZern
- 1 Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Luca Malcovati
- 2 Department of Molecular Medicine, University of Pavia, and Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Benjamin L Ebert
- 3 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
425
|
Genetic abnormalities and pathophysiology of MDS. Int J Clin Oncol 2019; 24:885-892. [DOI: 10.1007/s10147-019-01462-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 04/28/2019] [Indexed: 12/14/2022]
|
426
|
Degradation of D-2-hydroxyglutarate in the presence of isocitrate dehydrogenase mutations. Sci Rep 2019; 9:7436. [PMID: 31092874 PMCID: PMC6520482 DOI: 10.1038/s41598-019-43891-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 03/20/2019] [Indexed: 11/08/2022] Open
Abstract
D-2-Hydroxyglutarate (D-2-HG) is regarded as an oncometabolite. It is found at elevated levels in certain malignancies such as acute myeloid leukaemia and glioma. It is produced by a mutated isocitrate dehydrogenase IDH1/2, a low-affinity/high-capacity enzyme. Its degradation, in contrast, is catalysed by the high-affinity/low-capacity enzyme D-2-hydroxyglutarate dehydrogenase (D2HDH). So far, it has not been proven experimentally that the accumulation of D-2-HG in IDH mutant cells is the result of its insufficient degradation by D2HDH. Therefore, we developed an LC-MS/MS-based enzyme activity assay that measures the temporal drop in substrate and compared this to the expression of D2HDH protein as measured by Western blot. Our data clearly indicate, that the maximum D-2-HG degradation rate by D2HDH is reached in vivo, as vmax is low in comparison to production of D-2-HG by mutant IDH1/2. The latter seems to be limited only by substrate availability. Further, incubation of IDH wild type cells for up to 48 hours with 5 mM D-2-HG did not result in a significant increase in either D2HDH protein abundance or enzyme activity.
Collapse
|
427
|
Culos K, Byrne M. Salvage Therapy after Allogeneic Hematopoietic Cell Transplantation: Targeted and Low-Intensity Treatment Options in Myelodysplastic Syndrome and Acute Myeloid Leukemia. Clin Hematol Int 2019; 1:94-100. [PMID: 34595416 PMCID: PMC8432395 DOI: 10.2991/chi.d.190503.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/17/2019] [Indexed: 11/01/2022] Open
Abstract
Patients with high-risk myeloid neoplasms, including myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML), are offered allogeneic hematopoietic cell transplantation (alloHCT) to improve the likelihood of long-term disease control. More than 50% of patients with high-risk disease will relapse after HCT and face a poor prognosis with shortened survival. The recent development of targeted therapies and effective, low-intensity treatment strategies will likely improve the outcomes of these patients. In MDS, hypomethylating agents (HMAs) are the mainstay of salvage therapy but new treatments with APR-246 and luspatercept demonstrate excellent results in phase 1 and phase 3 clinical studies, respectively. In AML, new directed agents in the relapsed/refractory setting include gilteritinib (FLT3-ITD/-TKD), ivosidenib (IDH1), and enasidenib (IDH2). In patients without targetable mutations, HMAs may be used, and early data with venetoclax-based regimens are encouraging.
Collapse
Affiliation(s)
- Katie Culos
- Department of Pharmacy, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Byrne
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
428
|
Williams D, Fingleton B. Non-canonical roles for metabolic enzymes and intermediates in malignant progression and metastasis. Clin Exp Metastasis 2019; 36:211-224. [PMID: 31073762 DOI: 10.1007/s10585-019-09967-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/29/2019] [Indexed: 12/16/2022]
Abstract
Metabolic alterations are established as a hallmark of cancer. Such hallmark changes in cancer metabolism are characterized by reprogramming of energy-producing pathways and increases in the generation of biosynthetic intermediates to meet the needs of rapidly proliferating tumor cells. Various metabolic phenotypes such as aerobic glycolysis, increased glutamine consumption, and lipolysis have also been associated with the process of metastasis. However, in addition to the energy and biosynthetic alterations, a number of secondary functions of enzymes and metabolites are emerging that specifically contribute to metastasis. Here, we describe atypical intracellular roles of metabolic enzymes, extracellular functions of metabolic enzymes, roles of metabolites as signaling molecules, and epigenetic regulation mediated by altered metabolism, all of which can affect metastatic progression. We highlight how some of these mechanisms are already being exploited for therapeutic purposes, and discuss how others show similar potential.
Collapse
Affiliation(s)
- Demond Williams
- Program in Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Barbara Fingleton
- Program in Cancer Biology and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
429
|
Largeaud L, Bérard E, Bertoli S, Dufrechou S, Prade N, Gadaud N, Tavitian S, Bories P, Luquet I, Sarry A, De Mas V, Huguet F, Delabesse E, Récher C. Outcome of AML patients with IDH2 mutations in real world before the era of IDH2 inhibitors. Leuk Res 2019; 81:82-87. [PMID: 31055247 DOI: 10.1016/j.leukres.2019.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 11/28/2022]
Abstract
Describing the prognosis of sub-groups of acute myeloid leukemia (AML) patients treated in real world with current therapies is becoming increasingly relevant to estimate the benefit that new targeted drugs will bring in the field. This is particularly the case when novel drugs are registered on the basis of non-randomized studies. IDH2 inhibitors have recently emerged as promising drugs in patients with IDH2R140 or IDH2R172 mutations. Enasidenib, a first-in-class IDH2 inhibitor, has been approved following promising results of a phase 1-2 clinical trial in relapsed or refractory AML patients with IDH2 mutations. In this study, we described the characteristics, treatments and outcome of 75 IDH2 mutated patients both at diagnosis and relapse or refractory disease. Among the 33 relapsed/refractory AML patients with either IDH2R140 or IDH2R172, 28 (84.8%) patients received salvage therapy and 14 achieved a complete response (50%). Median duration of response was 15.2 months. Median, 1-y, 3-y and 5-y OS were 15.1 months (IQR, 4.6-37.7), 53.1% (95% CI, 33.2-69.5), 29.2% (95% CI, 12.6-48.1) and 24.4% (95% CI, 9.3-43.1), respectively. In responding patients, median OS was 37.7 months and 1-y, 3-y and 5-y OS was 85.7%, 57.1% and 47.6%, respectively. In non-responding patients, median OS was 5.0 months (IQR, 4.5-8.6) and 1-y and 3-y OS was 17.9% and 0%, respectively. Thus, a substantial number of R/R AML patients with IDH2 mutations can be salvaged by current treatments and benefit from prolonged survival. It is expected that novel targeted agents such as enasidenib will further improve efficacy and safety in the next future.
Collapse
Affiliation(s)
- Laetitia Largeaud
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France
| | - Emilie Bérard
- Service d'Epidémiologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France; UMR 1027, INSERM-Université de Toulouse III, Toulouse, France
| | - Sarah Bertoli
- Université Toulouse III Paul Sabatier, Toulouse, France; Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France; Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Stéphanie Dufrechou
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Naïs Prade
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Noémie Gadaud
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Suzanne Tavitian
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Pierre Bories
- Réseau Onco-occitanie, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Isabelle Luquet
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Audrey Sarry
- Service d'Epidémiologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Véronique De Mas
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France
| | - Françoise Huguet
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Eric Delabesse
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France
| | - Christian Récher
- Université Toulouse III Paul Sabatier, Toulouse, France; Cancer Research Center of Toulouse, UMR1037-INSERM, ERL5294 CNRS, Toulouse, France; Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.
| |
Collapse
|
430
|
Antoniou SX, Gaude E, Ruparel M, van der Schee MP, Janes SM, Rintoul RC. The potential of breath analysis to improve outcome for patients with lung cancer. J Breath Res 2019; 13:034002. [PMID: 30822771 DOI: 10.1088/1752-7163/ab0bee] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lung cancer remains the most common cause of cancer related death in both the UK and USA. Development of diagnostic approaches that have the ability to detect lung cancer early are a research priority with potential to improve survival. Analysis of exhaled breath metabolites, or volatile organic compounds (VOCs) is an area of considerable interest as it could fulfil such requirements. Numerous studies have shown that VOC profiles are different in the breath of patients with lung cancer compared to healthy individuals or those with non-malignant lung diseases. This review provides a scientific and clinical assessment of the potential value of a breath test in lung cancer. It discusses the current understanding of metabolic pathways that contribute to exhaled VOC production in lung cancer and reviews the research conducted to date. Finally, we highlight important areas for future research and discuss how a breath test could be incorporated into various clinical pathways.
Collapse
Affiliation(s)
- S X Antoniou
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom.,Equal contribution
| | - E Gaude
- Owlstone Medical, Cambridge, United Kingdom,Equal contribution
| | - M Ruparel
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | | | - S M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - R C Rintoul
- Papworth Trials Unit Collaboration, Royal Papworth Hospital, Cambridge, United Kingdom,Department of Oncology, University of Cambridge, United Kingdom
| | | |
Collapse
|
431
|
Dai D, Yang H, Nabhan S, Liu H, Hickman D, Liu G, Zacher J, Vutikullird A, Prakash C, Agresta S, Bowden C, Fan B. Effect of itraconazole, food, and ethnic origin on the pharmacokinetics of ivosidenib in healthy subjects. Eur J Clin Pharmacol 2019; 75:1099-1108. [PMID: 31011758 DOI: 10.1007/s00228-019-02673-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/21/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE To assess the effect of ethnicity, food, and itraconazole (strong CYP3A4 inhibitor) on the pharmacokinetics of ivosidenib after single oral doses in healthy subjects. METHODS Three phase 1 open-label studies were performed. Study 1: Japanese and Caucasian subjects received single doses of 250, 500, or 1000 mg ivosidenib (NCT03071770). Part 1 of study 2 (a two-period crossover study): subjects received 500 mg ivosidenib after either an overnight fast or a high-fat meal. Subjects received 1000 mg ivosidenib after an overnight fast in the single period of part 2 (NCT02579707). Study 3: in period 1, subjects received 250 mg ivosidenib; then, in period 2, subjects received oral itraconazole (200 mg once daily) on days 1-18, plus 250 mg ivosidenib on day 5 (NCT02831972). RESULTS Ivosidenib was well tolerated in all three studies. Study 1: pharmacokinetic profiles were generally comparable, although AUC and Cmax were slightly lower in Japanese subjects than in Caucasian subjects, by ~ 30 and 17%, respectively. Study 2: AUC increased by ~ 25% and Cmax by ~ 98%, when ivosidenib was administered with a high-fat meal compared with a fasted state. Study 3: co-administration of itraconazole increased ivosidenib AUC by 169% (90% CI 145-195) but had no effect on ivosidenib Cmax. CONCLUSIONS No ivosidenib dose adjustment is deemed necessary for Japanese subjects. High-fat meals should be avoided when ivosidenib is taken with food. When co-administered with strong CYP3A4 inhibitors, monitoring for QT interval prolongation (a previously defined adverse event of interest) is recommended and an ivosidenib dose interruption or reduction may be considered. CLINICALTRIALS.GOV : NCT03071770, NCT02579707, and NCT02831972.
Collapse
Affiliation(s)
- David Dai
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA
| | - Hua Yang
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA
| | - Salah Nabhan
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA
| | - Hua Liu
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA
| | - Denice Hickman
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA
| | - Guowen Liu
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA
| | | | | | - Chandra Prakash
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA
| | - Samuel Agresta
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA.,Infinity Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Chris Bowden
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA
| | - Bin Fan
- Agios Pharmaceuticals, Inc., 88 Sidney Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
432
|
Toth LN, Green D, Peterson J, Deharvengt SJ, de Abreu FB, Loo EY. Variant allele frequencies do not correlate well with myeloblast counts in a clinically validated gene sequencing panel for routine acute myeloid leukemia workup. Leuk Lymphoma 2019; 60:2415-2422. [DOI: 10.1080/10428194.2019.1587757] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Laura N. Toth
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center One Medical Center Drive, Lebanon, NH, USA
| | - Donald Green
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center One Medical Center Drive, Lebanon, NH, USA
| | - Jason Peterson
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center One Medical Center Drive, Lebanon, NH, USA
| | - Sophie J. Deharvengt
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center One Medical Center Drive, Lebanon, NH, USA
| | - Francine B. de Abreu
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center One Medical Center Drive, Lebanon, NH, USA
| | - Eric Y. Loo
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center One Medical Center Drive, Lebanon, NH, USA
| |
Collapse
|
433
|
Affiliation(s)
- Kiran Naqvi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
434
|
Treatment optimization and genomic outcomes in refractory severe aplastic anemia treated with eltrombopag. Blood 2019; 133:2575-2585. [PMID: 30992268 DOI: 10.1182/blood.2019000478] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
Eltrombopag (EPAG) received approval from the US Food and Drug Administration for the treatment of refractory severe aplastic anemia (rSAA) based on treatment of 43 patients with doses escalating from 50 to 150 mg daily for 12 weeks. Response kinetics suggested that more prolonged administration of EPAG at a dose of 150 mg could speed and improve response rates. We enrolled 40 patients with rSAA in a study of EPAG 150 mg daily, with a primary end point of response at 24 weeks. Twenty (50%) of 40 patients responded at 24 weeks; 5 (25%) of 20 would have been deemed nonresponders at 12 weeks, the end point of the previous study. Fifteen of the 19 responding patients continuing on EPAG had drug discontinued for robust response; 5 of the 15 required EPAG re-initiation for relapse, with all recovering response. To analyze risk of clonal progression, we combined long-term data from the 83 patients with rSAA enrolled in both studies. Evolution to an abnormal karyotype occurred in 16 (19%), most within 6 months of EPAG initiation. Targeted deep sequencing/whole-exome sequencing was performed pre-EPAG and at primary response end point and/or time of clonal evolution or longest follow-up. Cytogenetic evolution did not correlate with mutational status, and overall mutated allele fractions of myeloid cancer genes did not increase on EPAG. In summary, extended administration of EPAG at a dose of 150 mg for 24 weeks rescued responses in some patients with rSAA not responding at 12 weeks. The temporal relationship between clonal evolution and drug exposure suggests that EPAG may promote expansion of dormant preexisting clones with an aberrant karyotype. The studies were registered at www.clinicaltrials.gov as #NCT00922883 and #NCT01891994.
Collapse
|
435
|
Hu CW, Qiu Y, Ligeralde A, Raybon AY, Yoo SY, Coombes KR, Qutub AA, Kornblau SM. A quantitative analysis of heterogeneities and hallmarks in acute myelogenous leukaemia. Nat Biomed Eng 2019; 3:889-901. [PMID: 30988472 PMCID: PMC7051028 DOI: 10.1038/s41551-019-0387-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 03/08/2019] [Indexed: 01/18/2023]
Abstract
Acute myelogenous leukaemia (AML) is associated with risk factors that are largely unknown and with a heterogeneous response to treatment. Here, we provide a comprehensive quantitative understanding of AML proteomic heterogeneities and hallmarks by using the AML proteome atlas, a proteomics database that we have newly derived from MetaGalaxy analyses, for the proteomic profiling of 205 AML patients and 111 leukaemia cell lines. The analysis of the dataset revealed 154 functional patterns based on common molecular pathways, 11 constellations of correlated functional patterns, and 13 signatures that stratify the patients’ outcomes. We find limited overlap between proteomics data and both cytogenetics and genetic mutations, and also that leukaemia cell lines show limited proteomic similarities with cells from AML patients, suggesting that a deeper focus on patient-derived samples is needed to gain disease-relevant insights. The AML proteome atlas provides a knowledge base for proteomic patterns in AML, a guide to leukaemia cell-line selection, and a broadly applicable computational approach for quantifying the heterogeneities of protein expression and proteomic hallmarks in AML.
Collapse
Affiliation(s)
- C W Hu
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Y Qiu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Ligeralde
- Biophysics Graduate Program, University of California, Berkeley, CA, USA
| | - A Y Raybon
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, TX, USA
| | - S Y Yoo
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K R Coombes
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - A A Qutub
- Department of Bioengineering, Rice University, Houston, TX, USA. .,Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, TX, USA.
| | - S M Kornblau
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
436
|
Tiong IS, Wei AH. New drugs creating new challenges in acute myeloid leukemia. Genes Chromosomes Cancer 2019; 58:903-914. [PMID: 30861214 DOI: 10.1002/gcc.22750] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/20/2019] [Accepted: 03/01/2019] [Indexed: 12/31/2022] Open
Abstract
The therapeutic landscape is rapidly changing, with eight new drugs approved by the Food and Drug Administration within the last 2 years, including midostaurin and gilteritinib for FLT3 mutant newly diagnosed and relapsed/refractory (R/R) acute myeloid leukemia (AML), respectively; CPX-351 (liposomal cytarabine and daunorubicin) for therapy-related AML and AML with myelodysplasia-related changes; gemtuzumab ozogamicin (anti-CD33 monoclonal antibody conjugated with calicheamicin) for newly diagnosed and R/R CD33-positive AML; enasidenib and ivosidenib for IDH2 and IDH1 mutant R/R AML, respectively. Novel therapies have also emerged for newly diagnosed AML in adults who are age 75 years or older, or who have comorbidities that preclude the use of intensive induction chemotherapy. These include venetoclax (BCL-2 inhibitor) in combination with hypomethylating agents (azacitidine or decitabine) or low-dose cytarabine (LDAC), and glasdegib (sonic hedgehog pathway inhibitor) in combination with LDAC. This flurry of new drug approvals has markedly altered the treatment landscape in AML and provided new opportunities, as well as new challenges for treating clinicians. This review will focus on how these drugs might shape clinical practice and the hurdles likely to be faced by new therapies seeking entry into this dynamic and rapidly changing therapeutic landscape.
Collapse
Affiliation(s)
- Ing S Tiong
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Andrew H Wei
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
437
|
Lee S, Urman A, Desai P. Emerging drug profile: Krebs cycle and cancer: IDH mutations and therapeutic implications. Leuk Lymphoma 2019; 60:2635-2645. [PMID: 30958073 DOI: 10.1080/10428194.2019.1602260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Mutations in IDH1 and IDH2 genes occur frequently in myeloid malignancies and certain solid tumors. IDH1 and IDH2 are enzymes that are involved in the tricarboxylic acid (TCA) cycle. Activating mutations in IDH1 and IDH2 leads to increased production of 2-hydroxygluterate and epigenetic modification, affecting cell differentiation. Small molecule inhibitors of mutated IDH1 and IDH2 have shown promising anti-cancer activity in both preclinical models and early clinical trials. Recently, enasidenib and ivosidenib, oral inhibitors of mutated IDH2 and IDH1 genes, respectively, were approved for use in relapsed or refractory acute myeloid leukemia. This review will focus on the underlying biological mechanism and clinical relevance of IDH mutations in cancer.
Collapse
Affiliation(s)
- Sangmin Lee
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Arielle Urman
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Pinkal Desai
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
438
|
Metabolic Imaging Reveals a Unique Preference of Symmetric Cell Division and Homing of Leukemia-Initiating Cells in an Endosteal Niche. Cell Metab 2019; 29:950-965.e6. [PMID: 30581117 DOI: 10.1016/j.cmet.2018.11.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 07/23/2018] [Accepted: 11/21/2018] [Indexed: 01/08/2023]
Abstract
The metabolic properties of leukemia-initiating cells (LICs) in distinct bone marrow niches and their relationships to cell-fate determinations remain largely unknown. Using a metabolic imaging system with a highly responsive genetically encoded metabolic sensor, SoNar, we reveal that SoNar-high cells are more glycolytic, enriched for higher LIC frequency, and develop leukemia much faster than SoNar-low counterparts in an MLL-AF9-induced murine acute myeloid leukemia model. SoNar-high cells mainly home to and locate in the hypoxic endosteal niche and maintain their activities through efficient symmetric division. SoNar can indicate the dynamics of metabolic changes of LICs in the endosteal niche. SoNar-high human leukemia cells or primary samples have enhanced clonogenic capacities in vitro or leukemogenesis in vivo. PDK2 fine-tunes glycolysis, homing, and symmetric division of LICs. These findings provide a unique angle for the study of metabolisms in stem cells, and may lead to development of novel strategies for cancer treatment.
Collapse
|
439
|
Aoki K, Natsume A. Overview of DNA methylation in adult diffuse gliomas. Brain Tumor Pathol 2019; 36:84-91. [PMID: 30937703 DOI: 10.1007/s10014-019-00339-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 03/19/2019] [Indexed: 12/30/2022]
Abstract
Adult diffuse gliomas form a heterogeneous group of tumors of the central nervous system that vary greatly in histology and prognosis. A significant advance during the last decade has been the identification of a set of genetic lesions that correlate well with histology and clinical outcome in diffuse gliomas. Most characteristic driver mutations consist of isocitrate dehydrogenase 1 (IDH1) and IDH2, and H3 histone family member 3A, which are strongly associated with DNA and histone methylation patterns. A well-characterized DNA methylation aberration is on the O6-methylguanine-DNA methyltransferase promoter. This aberration is associated with an improved response to the DNA alkylating agent, temozolomide. Methylation alterations are used for classification or treatment decisions of diffuse gliomas. This supports the importance of considering epigenomic aberrations in the pathogenesis of gliomas. Recent DNA methylation analyses revealed a small group of IDH mutant diffuse gliomas exhibiting decreased DNA hypermethylation resulting in substantial unfavorable prognosis comparable to glioblastoma. Thus, DNA methylation patterns may become a new standard that replaces the conventional grading system based on histological diagnosis. In this review, we summarize recent developments regarding the contributions of methylation patterns to the pathogenesis of adult diffuse glioma, the interactions between methylation patterns and driver mutations, and potential epigenomic targeted therapies.
Collapse
Affiliation(s)
- Kosuke Aoki
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
440
|
Hayashi Y, Yokota A, Harada H, Huang G. Hypoxia/pseudohypoxia-mediated activation of hypoxia-inducible factor-1α in cancer. Cancer Sci 2019; 110:1510-1517. [PMID: 30844107 PMCID: PMC6501028 DOI: 10.1111/cas.13990] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 12/12/2022] Open
Abstract
Since the first identification of hypoxic cells in sections of carcinomas in the 1950s, hypoxia has been known as a central hallmark of cancer cells and their microenvironment. Indeed, hypoxia benefits cancer cells in their growth, survival, and metastasis. The historical discovery of hypoxia‐inducible factor‐1α (HIF1A) in the early 1990s had a great influence on the field as many phenomena in hypoxia could be explained by HIF1A. However, not all regions or types of tumors are necessarily hypoxic. Thus, it is difficult to explain whole cancer pathobiology by hypoxia, especially in the early stage of cancer. Upregulation of glucose metabolism in cancer cells has been well known. Oxygen‐independent glycolysis is activated in cancer cells even in the normoxia condition, which is known as the Warburg effect. Accumulating evidence and recent advances in cancer metabolism research suggest that hypoxia‐independent mechanisms for HIF signaling activation is a hallmark for cancer. There are various mechanisms that generate pseudohypoxic conditions, even in normoxia. Given the importance of HIF1A for cancer pathobiology, the pseudohypoxia concept could shed light on the longstanding mystery of the Warburg effect and accelerate better understanding of the diverse phenomena seen in a variety of cancers.
Collapse
Affiliation(s)
- Yoshihiro Hayashi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Asumi Yokota
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hironori Harada
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
441
|
Abstract
An emergent theme in cancer biology is that dysregulated energy metabolism may directly influence oncogenic gene expression. This is due to the fact that many enzymes involved in gene regulation use cofactors derived from primary metabolism, including acetyl-CoA, S-adenosylmethionine, and 2-ketoglutarate. While this phenomenon was first studied through the prism of histone and DNA modifications (the epigenome), recent work indicates metabolism can also impact gene regulation by disrupting the balance of RNA post-transcriptional modifications (the epitranscriptome). Here we review recent studies that explore how metabolic regulation of writers and erasers of the epitranscriptome (FTO, TET2, NAT10, MTO1, and METTL16) helps shape gene expression through three distinct mechanisms: cofactor inhibition, cofactor depletion, and writer localization. Our brief survey underscores similarities and differences between the metabolic regulation of the epigenome and epitranscriptome, and highlights fertile ground for future investigation.
Collapse
Affiliation(s)
- Justin M. Thomas
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Pedro J. Batista
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Jordan L. Meier
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
442
|
Chen D, Xia S, Wang M, Lin R, Li Y, Mao H, Aguiar M, Famulare CA, Shih AH, Brennan CW, Gao X, Pan Y, Liu S, Fan J, Jin L, Song L, Zhou A, Mukherjee J, Pieper RO, Mishra A, Peng J, Arellano M, Blum WG, Lonial S, Boggon TJ, Levine RL, Chen J. Mutant and Wild-Type Isocitrate Dehydrogenase 1 Share Enhancing Mechanisms Involving Distinct Tyrosine Kinase Cascades in Cancer. Cancer Discov 2019; 9:756-777. [PMID: 30862724 DOI: 10.1158/2159-8290.cd-18-1040] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/10/2019] [Accepted: 03/07/2019] [Indexed: 01/03/2023]
Abstract
Isocitrate dehydrogenase 1 (IDH1) is important for reductive carboxylation in cancer cells, and the IDH1 R132H mutation plays a pathogenic role in cancers including acute myeloid leukemia (AML). However, the regulatory mechanisms modulating mutant and/or wild-type (WT) IDH1 function remain unknown. Here, we show that two groups of tyrosine kinases (TK) enhance the activation of mutant and WT IDH1 through preferential Y42 or Y391 phosphorylation. Mechanistically, Y42 phosphorylation occurs in IDH1 monomers, which promotes dimer formation with enhanced substrate (isocitrate or α-ketoglutarate) binding, whereas Y42-phosphorylated dimers show attenuated disruption to monomers. Y391 phosphorylation occurs in both monomeric and dimeric IDH1, which enhances cofactor (NADP+ or NADPH) binding. Diverse oncogenic TKs phosphorylate IDH1 WT at Y42 and activate Src to phosphorylate IDH1 at Y391, which contributes to reductive carboxylation and tumor growth, whereas FLT3 or the FLT3-ITD mutation activates JAK2 to enhance mutant IDH1 activity through phosphorylation of Y391 and Y42, respectively, in AML cells. SIGNIFICANCE: We demonstrated an intrinsic connection between oncogenic TKs and activation of WT and mutant IDH1, which involves distinct TK cascades in related cancers. In particular, these results provide an additional rationale supporting the combination of FLT3 and mutant IDH1 inhibitors as a promising clinical treatment of mutant IDH1-positive AML.See related commentary by Horton and Huntly, p. 699.This article is highlighted in the In This Issue feature, p. 681.
Collapse
Affiliation(s)
- Dong Chen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Siyuan Xia
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Mei Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, China
| | - Ruiting Lin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Yuancheng Li
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Department of Radiology and Imaging Sciences, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Hui Mao
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Department of Radiology and Imaging Sciences, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Mike Aguiar
- Cell Signaling Technology, Inc., Danvers, Massachusetts
| | | | - Alan H Shih
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Xue Gao
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Yaozhu Pan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,General Hospital of Lanzhou Military Region, Lanzhou, China
| | - Shuangping Liu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Department of Pathology, Medical College, Dalian University, Dalian, China
| | - Jun Fan
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Lingtao Jin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Lina Song
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia
| | - An Zhou
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Joydeep Mukherjee
- Department of Neurological Surgery, University of California, San Francisco, California
| | - Russell O Pieper
- Department of Neurological Surgery, University of California, San Francisco, California
| | - Ashutosh Mishra
- Department of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Junmin Peng
- Department of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Martha Arellano
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - William G Blum
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Titus J Boggon
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Ross L Levine
- Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Jing Chen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia. .,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
443
|
Wang P, Chen X, Yang J, Pei Y, Bian M, Zhu G. Characterization of the nicotinamide adenine dinucleotides (NAD + and NADP +) binding sites of the monomeric isocitrate dehydrogenases from Campylobacter species. Biochimie 2019; 160:148-155. [PMID: 30876971 DOI: 10.1016/j.biochi.2019.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/10/2019] [Indexed: 12/15/2022]
Abstract
Monomeric isocitrate dehydrogenases (IDHs) have once been proposed to be exclusively NADP+-specific. Intriguingly, we recently have reported an NAD+-specific monomeric IDH from Campylobacter sp. FOBRC14 (CaIDH). Moreover, bioinformatic analysis revealed at least three different coenzyme-binding motifs among Campylobacter IDHs. Besides the NAD+-binding motif in CaIDH (Leu584/Asp595/Ser644), a typical NADP+-binding motif was also identified in Campylobacter corcagiensis IDH (CcoIDH, His582/Arg593/Arg638). Meanwhile, a third putative NAD+-binding motif was found in Campylobacter concisus IDH (CcIDH, Leu580/Leu591/Ala640). In this study, CcIDH was overexpressed in Escherichia coli and purified to homogeneity. Gel filtration chromatography demonstrated that the recombinant CcIDH exists as a monomer in solution. Kinetic analysis showed that the Km value of CcIDH for NADP+ was over 49-fold higher than that for NAD+ and the catalytic efficiency (kcat/Km) of CcIDH is 115-fold greater for NAD+ than NADP+. Thus, CcIDH is indeed an NAD+-specific enzyme. However, the catalytic efficiency (kcat/Km = 0.886 μM-1 s-1) of CcIDH for NAD+ is much lower (<5%) when compared to that of the typical monomeric NADP-IDHs for NADP+. Then, the three core NAD+-binding sites were evaluated by site-directed mutagenesis. The mutant CcIDH (H580R591R640) showed a 51-fold higher Km value for NAD+ and 21-fold lower Km value for NADP+ as compared to that of the wild type enzyme, respectively. The overall specificity of the mutant CcIDH was 12-fold greater for NADP+ than that for NAD+. Thus, the coenzyme specificity of CcIDH was converted from NAD+ to NADP+. Isocitrate dependence of enzyme kinetics showed that although the mutant H580R591R640 preferred NADP+ as its coenzyme, its catalytic efficiency for isocitrate reduced to half of that for the wild-type CcIDH as using NAD+. The finding of both NAD+ and NADP+-binding sites in monomeric IDHs from Campylobacter species will provide us a chance to explore the evolution of the coenzyme specificity in monomeric IDH subfamily.
Collapse
Affiliation(s)
- Peng Wang
- Institute of Molecular Biology and Biotechnology and the Research Center of Life Omics and Health, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Xuefei Chen
- Institute of Molecular Biology and Biotechnology and the Research Center of Life Omics and Health, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Jing Yang
- Institute of Molecular Biology and Biotechnology and the Research Center of Life Omics and Health, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Yunyun Pei
- Institute of Molecular Biology and Biotechnology and the Research Center of Life Omics and Health, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Mingjie Bian
- Institute of Molecular Biology and Biotechnology and the Research Center of Life Omics and Health, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Guoping Zhu
- Institute of Molecular Biology and Biotechnology and the Research Center of Life Omics and Health, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.
| |
Collapse
|
444
|
Consequences of IDH1/2 Mutations in Gliomas and an Assessment of Inhibitors Targeting Mutated IDH Proteins. Molecules 2019; 24:molecules24050968. [PMID: 30857299 PMCID: PMC6429355 DOI: 10.3390/molecules24050968] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 11/16/2022] Open
Abstract
Isocitrate dehydrogenases (IDH) 1 and 2 are key metabolic enzymes that generate reduced nicotinamide adenine dinucleotide phosphate (NADPH) to maintain a pool of reduced glutathione and peroxiredoxin, and produce α-ketoglutarate, a co-factor of numerous enzymes. IDH1/2 is mutated in ~70–80% of lower-grade gliomas and the majority of secondary glioblastomas. The mutant IDH1 (R132H), in addition to losing its normal catalytic activity, gains the function of producing the d-(R)-2-hydroxyglutarate (2-HG). Overproduction of 2-HG in cancer cells interferes with cellular metabolism and inhibits histone and DNA demethylases, which results in histone and DNA hypermethylation and the blockade of cellular differentiation. We summarize recent findings characterizing molecular mechanisms underlying oncogenic alterations associated with mutated IDH1/2, and their impact on tumor microenvironment and antitumor immunity. Isoform-selective IDH inhibitors which suppress 2-HG production and induce antitumor responses in cells with IDH1 and IDH2 mutations were developed and validated in preclinical settings. Inhibitors of mutated IDH1/2 enzymes entered clinical trials and represent a novel drug class for targeted therapy of gliomas. We describe the development of small-molecule compounds and peptide vaccines targeting IDH-mutant gliomas and the results of their testing in preclinical and clinical studies. All those results support the translational potential of strategies targeting gliomas carrying IDH1 mutations.
Collapse
|
445
|
SPINT2 is hypermethylated in both IDH1 mutated and wild-type glioblastomas, and exerts tumor suppression via reduction of c-Met activation. J Neurooncol 2019; 142:423-434. [PMID: 30838489 DOI: 10.1007/s11060-019-03126-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/09/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Both IDH1-mutated and wild-type gliomas abundantly display aberrant CpG island hypermethylation. However, the potential role of hypermethylation in promoting gliomas, especially the most aggressive form, glioblastoma (GBM), remains poorly understood. METHODS We analyzed RRBS-generated methylation profiles for 11 IDH1WT gliomas (including 7 GBMs), 24 IDH1MUT gliomas (including 6 GBMs), and 5 normal brain samples and employed TCGA GBM methylation profiles as a validation set. Upon classification of differentially methylated CpG islands by IDH1 status, we used integrated analysis of methylation and gene expression to identify SPINT2 as a top cancer related gene. To explore functional consequences of SPINT2 methylation in GBM, we validated SPINT2 methylation status using targeted bisulfite sequencing in a large cohort of GBM samples. We assessed DNA methylation-mediated SPINT2 gene regulation using 5-aza-2'-deoxycytidine treatment, DNMT1 knockdown and luciferase reporter assays. We conducted functional analyses of SPINT2 in GBM cell lines in vitro and in vivo. RESULTS We identified SPINT2 as a candidate tumor-suppressor gene within a group of CpG islands (designated GT-CMG) that are hypermethylated in both IDH1MUT and IDH1WT gliomas but not in normal brain. We established that SPINT2 downregulation results from promoter hypermethylation, and that restoration of SPINT2 expression reduces c-Met activation and tumorigenic properties of GBM cells. CONCLUSIONS We defined a previously under-recognized group of coordinately methylated CpG islands common to both IDH1WT and IDH1MUT gliomas (GT-CMG). Within GT-CMG, we identified SPINT2 as a top cancer-related candidate and demonstrated that SPINT2 suppressed GBM via down-regulation of c-Met activation.
Collapse
|
446
|
Iyer G, Rosenberg JE. Lessons learned from exceptional responders. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1578171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Gopa Iyer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jonathan E. Rosenberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
447
|
|
448
|
Alonso CM, Llop M, Sargas C, Pedrola L, Panadero J, Hervás D, Cervera J, Such E, Ibáñez M, Ayala R, Martínez-López J, Onecha E, de Juan I, Palanca S, Martínez-Cuadrón D, Rodríguez-Veiga R, Boluda B, Montesinos P, Sanz G, Sanz MA, Barragán E. Clinical Utility of a Next-Generation Sequencing Panel for Acute Myeloid Leukemia Diagnostics. J Mol Diagn 2019; 21:228-240. [DOI: 10.1016/j.jmoldx.2018.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 09/06/2018] [Accepted: 09/20/2018] [Indexed: 10/27/2022] Open
|
449
|
Sim HW, Nejad R, Zhang W, Nassiri F, Mason W, Aldape KD, Zadeh G, Chen EX. Tissue 2-Hydroxyglutarate as a Biomarker for Isocitrate Dehydrogenase Mutations in Gliomas. Clin Cancer Res 2019; 25:3366-3373. [PMID: 30777876 DOI: 10.1158/1078-0432.ccr-18-3205] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/12/2018] [Accepted: 02/14/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Isocitrate dehydrogenase (IDH) mutations are common in low-grade gliomas and the IDH mutation status is now integrated into the WHO classification of gliomas. IDH mutations lead to preferential accumulation of the R- relative to the S-enantiomer of 2-hydroxyglutarate (2-HG). We investigated the utility of tissue total 2-HG, R-2-HG, and the R-2-HG/S-2-HG ratio (rRS) as diagnostic and prognostic biomarkers for IDH mutations in gliomas.Experimental Design: Glioma tissue and blood samples from 87 patients were analyzed with HPLC-MS/MS coupled with a CHIROBIOTIC column to quantify both enantiomers of 2-HG. ROC analysis was conducted to evaluate the sensitivity and specificity of 2-HG, R-2-HG, and rRS. The feasibility of real-time determination of IDH status was evaluated in 11 patients intraoperatively. The prognostic value of rRS was evaluated using the Kaplan-Meier method. RESULTS The rRS in glioma tissues clearly distinguished patients with IDH-mutant versus wild-type tumors (P < 0.001). Sensitivity and specificity using an rRS cut-off value of 32.26 were 97% and 100%, respectively. None of total 2-HG, R-2-HG, or rRS was elevated in serum samples. Among patients with IDH-mutant tumors, tissue rRS stratifies overall survival. The duration of tissue analysis is approximately 60 minutes. CONCLUSIONS Our study demonstrates that rRS is a reliable biomarker of IDH mutation status. This technique can be used to determine IDH mutation status intraoperatively, and to guide treatment decisions based on IDH mutation status in real time. Finally, rRS values may provide additional prognostic information and further validation is required.
Collapse
Affiliation(s)
- Hao-Wen Sim
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Romina Nejad
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Wenjiang Zhang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Farshad Nassiri
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Warren Mason
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Kenneth D Aldape
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
| | - Eric X Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
450
|
Brunner AM, Neuberg DS, Wander SA, Sadrzadeh H, Ballen KK, Amrein PC, Attar E, Hobbs GS, Chen YB, Perry A, Connolly C, Joseph C, Burke M, Ramos A, Galinsky I, Yen K, Yang H, Straley K, Agresta S, Adamia S, Borger DR, Iafrate A, Graubert TA, Stone RM, Fathi AT. Isocitrate dehydrogenase 1 and 2 mutations, 2-hydroxyglutarate levels, and response to standard chemotherapy for patients with newly diagnosed acute myeloid leukemia. Cancer 2019; 125:541-549. [PMID: 30422308 DOI: 10.1002/cncr.31729] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/16/2018] [Accepted: 07/24/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) cells harboring mutations in isocitrate dehydrogenase 1 (IDH1) and isocitrate dehydrogenase 2 (IDH2) produce the oncometabolite 2-hydroxyglutarate (2HG). This study prospectively evaluated the 2HG levels, IDH1/2 mutational status, and outcomes of patients receiving standard chemotherapy for newly diagnosed AML. METHODS Serial samples of serum, urine, and bone marrow aspirates were collected from patients newly diagnosed with AML, and 2HG levels were measured with mass spectrometry. Patients with baseline serum 2HG levels greater than 1000 ng/mL or marrow pellet 2HG levels greater than 1000 ng/2 × 106 cells, which suggested the presence of an IDH1/2 mutation, underwent serial testing. IDH1/2 mutations and estimated variant allele frequencies were identified. AML characteristics were compared with the Wilcoxon test and Fisher's exact test. Disease-free survival and overall survival (OS) were evaluated with log-rank tests and Cox regression. RESULTS Two hundred and two patients were treated for AML; 51 harbored IDH1/2 mutations. IDH1/2-mutated patients had significantly higher 2HG levels in serum, urine, bone marrow aspirates, and aspirate cell pellets than wild-type patients. A serum 2HG level greater than 534.5 ng/mL was 98.8% specific for the presence of an IDH1/2 mutation. Patients with IDH1/2-mutated AML treated with 7+3-based induction had a 2-year event-free survival (EFS) rate of 44% and a 2-year OS rate of 57%. There was no difference in complete remission rates, EFS, or OS between IDH1/2-mutated and wild-type patients. Decreased serum 2HG levels on day 14 as a proportion of the baseline were significantly associated with improvements in EFS (P = .047) and OS (P = .019) in a multivariate analysis. CONCLUSIONS Among patients with IDH1/2-mutated AML, 2HG levels are highly specific for the mutational status at diagnosis, and they have prognostic relevance in patients receiving standard chemotherapy.
Collapse
Affiliation(s)
| | | | - Seth A Wander
- Massachusetts General Hospital, Boston, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | | | - Eyal Attar
- Massachusetts General Hospital, Boston, Massachusetts.,Agios Pharmaceuticals, Cambridge, Massachusetts
| | | | - Yi-Bin Chen
- Massachusetts General Hospital, Boston, Massachusetts
| | - Ashley Perry
- Massachusetts General Hospital, Boston, Massachusetts
| | | | | | - Meghan Burke
- Massachusetts General Hospital, Boston, Massachusetts
| | - Aura Ramos
- Massachusetts General Hospital, Boston, Massachusetts
| | | | | | - Hua Yang
- Agios Pharmaceuticals, Cambridge, Massachusetts
| | | | - Sam Agresta
- Agios Pharmaceuticals, Cambridge, Massachusetts
| | | | | | | | | | | | - Amir T Fathi
- Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|