1
|
Zhang X, Liu Y, Hou Q, Guo Y, He Y. DNA methylation inhibitors adverse reaction characteristic analysis: an analysis based on the European spontaneous adverse event reporting system. Front Pharmacol 2025; 15:1527903. [PMID: 39877384 PMCID: PMC11772285 DOI: 10.3389/fphar.2024.1527903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction DNA methylation inhibitors have been approved for the prevention of Acute Myeloid Leukemia (AML), and their safety profile is not fully characterized. This study was aimed at evaluating the adverse drug reactions (ADRs) of DNA methylation inhibitors by analyzing the individual case safety reports (ICSRs) collected in the EudraVigilance (EV) database. Materials and methods The EV database managed by the European Medicines Agency was adopted. The standardized medical terminology set MedDRA was utilized. The ICSRs data of DNA methylation inhibitors for the treatment of acute myeloid leukemia originated from the EV database (2005-2024). A descriptive exploration of the combined data from EV was undertaken to assess the age, gender of patients, severity and outcome of ADR, event year, geographical origin and the qualification of the reporting source. A comprehensive assessment was made for severe ADR cases. By means of the Reporting Odds Ratio (ROR) and 95% Confidence Interval (CI), a non-proportional analysis was made for MedDRA® SOC in DNA methylation inhibitors. Statistical analysis was executed with SPSS version 23.0, and p < 0.05 was regarded as statistically significant. Result The study reveals that reports related to AZACITIDINE increased from 2005 to 2023, with a slight decline in 2024, while those for DECITABINE have been on the rise since 2007. ICSRs were associated with a majority of males and individuals aged 65-85. Healthcare professionals frequently reported ICSRs related to DNA methylation inhibitors. A significant portion of these ICSRs were serious and completely resolved. The most common ADRs were identified, and certain ADRs had a higher reporting probability with AZACITIDINE (e.g., Febrile neutropenia, Anamia, etc.) and others with DECITABINE (e.g., Myelosuppression, Thrombocytopenia, etc.). Conclusion The analysis regarding ADRs of DNA methylation inhibitors was consistent with the literature information disclosed. AZACITIDINE and DECITABINE each have ADRs with a high probability of being reported. Although the study has the advantage of using the database, it is limited by the spontaneous reporting system. Future improvements are needed to accurately evaluate the safety of the drugs.
Collapse
Affiliation(s)
- Xia Zhang
- Department of Oncology, Guihang Guiyang Hospital, Guiyang, Guizhou, China
| | - Yuyu Liu
- School of Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qingwang Hou
- Department of Cardiology, Henan Provincial People’s Hospital, Hennan University People’s Hospital, Zhengzhou, Henan, China
| | - Yongxin Guo
- Department of Cardiology, Henan Provincial People’s Hospital, Hennan University People’s Hospital, Zhengzhou, Henan, China
| | - Youfu He
- Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
2
|
Wisman GBA, Wojdacz TK, Altucci L, Rots MG, DeMeo DL, Snieder H. Clinical promise and applications of epigenetic biomarkers. Clin Epigenetics 2024; 16:192. [PMID: 39732727 DOI: 10.1186/s13148-024-01806-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024] Open
Affiliation(s)
- G Bea A Wisman
- Department of Gynecologic Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Tomasz K Wojdacz
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University, Szczecin, Poland
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli" Napoli, Napoli, Italy
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- IEOS CNR, Naples, Italy
- Medical Epigenetics Program, Azienda Ospedaliera Universitaria "Luigi Vanvitelli", Naples, Italy
| | - Marianne G Rots
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dawn L DeMeo
- Channing Division of Network Medicine and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
3
|
Lamba J, Marchi F, Landwehr M, Schade AK, Shastri V, Ghavami M, Sckaff F, Marrero R, Nguyen N, Mansinghka V, Cao X, Slayton W, Starostik P, Ribeiro R, Rubnitz J, Klco J, Gamis A, Triche T, Ries R, Kolb EA, Aplenc R, Alonzo T, Pounds S, Meshinchi S, Cogle C, Elsayed A. Long-read epigenomic diagnosis and prognosis of Acute Myeloid Leukemia. RESEARCH SQUARE 2024:rs.3.rs-5450972. [PMID: 39711573 PMCID: PMC11661290 DOI: 10.21203/rs.3.rs-5450972/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Acute Myeloid Leukemia (AML) is an aggressive cancer with dismal outcomes, vast subtype heterogeneity, and suboptimal risk stratification. In this study, we harmonized DNA methylation data from 3,314 patients across 11 cohorts to develop the Acute Leukemia Methylome Atlas (ALMA) of diagnostic relevance that predicted 27 WHO 2022 acute leukemia subtypes with an overall accuracy of 96.3% in discovery and 90.1% in validation cohorts. Specifically, for AML, we also developed AML Epigenomic Risk, a prognostic classifier of overall survival (OS) (HR=4.40; 95% CI=3.45-5.61; P<0.0001), and a targeted 38CpG AML signature using a stepwise EWAS-CoxPH-LASSO model predictive of OS (HR=3.84; 95% CI=3.01-4.91; P<0.0001). Finally, we developed a specimen-to-result protocol for simultaneous whole-genome and epigenome sequencing that accurately predicted diagnoses and prognoses from twelve prospectively collected patient samples using long-read sequencing. Our study unveils a new paradigm in acute leukemia management by leveraging DNA methylation for diagnostic and prognostic applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xueyuan Cao
- University of Tennessee Health Science Center
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Tien FM, Hou HA. CEBPA mutations in acute myeloid leukemia: implications in risk stratification and treatment. Int J Hematol 2024; 120:541-547. [PMID: 38671183 DOI: 10.1007/s12185-024-03773-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024]
Abstract
Mutations in CCAAT enhancer binding protein α (CEBPA) occur in approximately 10% of patients with de novo acute myeloid leukemia (AML). Emerging evidence supports that in-frame mutations in the basic leucine zipper domain of CEBPA (CEBPAbZIP-inf) confer a survival benefit, and CEBPAbZIP-inf replaced CEBPA double mutations (CEBPAdm) as a unique entity in the 2022 World Health Organization (WHO-2022) classification and International Consensus Classification (ICC). However, challenges remain in daily clinical practice since more than 30% patients with CEBPAbZIP-inf die of AML despite intensive treatment. This review aims to provide a comprehensive summary of the heterogeneities observed in AML with CEBPAdm and CEBPAbZIP-inf, and will discuss the prognostic implications of concurrent mutations and novel mechanistic targets that may inform future drug development. The ultimate goal is to optimize clinical management and to provide precision medicine for this category of patients.
Collapse
Affiliation(s)
- Feng-Ming Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Division of General Medicine, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
5
|
Šestáková Š, Šálek C, Kundrát D, Cerovská E, Vydra J, Ježíšková I, Folta A, Mayer J, Cetkovský P, Remešová H. MethScore as a new comprehensive DNA methylation-based value refining the prognosis in acute myeloid leukemia. Clin Epigenetics 2024; 16:17. [PMID: 38254139 PMCID: PMC10802002 DOI: 10.1186/s13148-024-01625-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Changes in DNA methylation are common events in the pathogenesis of acute myeloid leukemia (AML) and have been repeatedly reported as associated with prognosis. However, studies integrating these numerous and potentially prognostically relevant DNA methylation changes are lacking. Therefore, we aimed for an overall evaluation of these epigenetic aberrations to provide a comprehensive NGS-based approach of DNA methylation assessment for AML prognostication. RESULTS We designed a sequencing panel targeting 239 regions (approx. 573 kb of total size) described in the literature as having a prognostic impact or being associated with AML pathogenesis. Diagnostic whole-blood DNA samples of adult AML patients divided into a training (n = 128) and a testing cohort (n = 50) were examined. The libraries were prepared using SeqCap Epi Enrichments System (Roche) and sequenced on MiSeq instrument (Illumina). Altogether, 1935 CpGs affecting the survival (p < 0.05) were revealed in the training cohort. A summarizing value MethScore was then calculated from these significant CpGs. Patients with lower MethScore had markedly longer overall survival (OS) and event-free survival (EFS) than those with higher MethScore (p < 0.001). The predictive ability of MethScore was verified on the independent testing cohort for OS (p = 0.01). Moreover, the proof-of-principle validation was performed using the TCGA dataset. CONCLUSIONS We showed that comprehensive NGS-based approach of DNA methylation assessment revealed a robust epigenetic signature relevant to AML outcome. We called this signature MethScore and showed it might serve as a strong prognostic marker able to refine survival probability of AML patients.
Collapse
Affiliation(s)
- Šárka Šestáková
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 00, Prague, Czech Republic
- Institute of Clinical and Experimental Hematology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cyril Šálek
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 00, Prague, Czech Republic
- Institute of Clinical and Experimental Hematology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dávid Kundrát
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 00, Prague, Czech Republic
| | - Ela Cerovská
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 00, Prague, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Jan Vydra
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 00, Prague, Czech Republic
- Institute of Clinical and Experimental Hematology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ivana Ježíšková
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Masaryk University, School of Medicine, Brno, Czech Republic
| | - Adam Folta
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Masaryk University, School of Medicine, Brno, Czech Republic
| | - Jiří Mayer
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Masaryk University, School of Medicine, Brno, Czech Republic
| | - Petr Cetkovský
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 00, Prague, Czech Republic
- Institute of Clinical and Experimental Hematology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hana Remešová
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 00, Prague, Czech Republic.
| |
Collapse
|
6
|
Targeting DNA Methylation in Leukemia, Myelodysplastic Syndrome, and Lymphoma: A Potential Diagnostic, Prognostic, and Therapeutic Tool. Int J Mol Sci 2022; 24:ijms24010633. [PMID: 36614080 PMCID: PMC9820560 DOI: 10.3390/ijms24010633] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
DNA methylation represents a crucial mechanism of epigenetic regulation in hematologic malignancies. The methylation process is controlled by specific DNA methyl transferases and other regulators, which are often affected by genetic alterations. Global hypomethylation and hypermethylation of tumor suppressor genes are associated with hematologic cancer development and progression. Several epi-drugs have been successfully implicated in the treatment of hematologic malignancies, including the hypomethylating agents (HMAs) decitabine and azacytidine. However, combinations with other treatment modalities and the discovery of new molecules are still the subject of research to increase sensitivity to anti-cancer therapies and improve patient outcomes. In this review, we summarized the main functions of DNA methylation regulators and genetic events leading to changes in methylation landscapes. We provide current knowledge about target genes with aberrant methylation levels in leukemias, myelodysplastic syndromes, and malignant lymphomas. Moreover, we provide an overview of the clinical trials, focused mainly on the combined therapy of HMAs with other treatments and its impact on adverse events, treatment efficacy, and survival rates among hematologic cancer patients. In the era of precision medicine, a transition from genes to their regulation opens up the possibility of an epigenetic-based approach as a diagnostic, prognostic, and therapeutic tool.
Collapse
|
7
|
Gu Y, Xu ZJ, Zhou JD, Wen XM, Jin Y, Yuan Q, Xia PH, Feng Y, Yang L, Lin J, Qian J. SLC22A3 methylation-mediated gene silencing predicts adverse prognosis in acute myeloid leukemia. Clin Epigenetics 2022; 14:162. [PMID: 36461046 PMCID: PMC9716704 DOI: 10.1186/s13148-022-01373-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND We screened out several hypermethylated solute carrier (SLC) family genes in acute myeloid leukemia by reduced representation bisulfite sequencing. SLC22A3 encodes an organic cation transport protein, which is critical for drug transportation and cellular detoxification. SLC22A3 is significantly downregulated and associated with tumor progression and worse prognosis in a variety of solid tumors. However, there are no data available regarding the role of SLC22 in AML. This study aimed to explore the regulatory mechanism of DNA methylation on SLC22A3 expression, as well as its clinical significance in AML prognosis. RESULTS SLC22A3 was identified as the sole prognosis-associated gene among SLCs based on TCGA and Beat AML databases. Bone marrow mononuclear cells (BMMNCs) from AML, MDS patients, and healthy donors were enrolled in this study. SLC22A3 methylation was significantly increased in AML compared with controls and MDS patients; meanwhile, the expression level of SLC22A3 was decreased. SLC22A3 hypermethylation presented an obvious association with some specific clinical characteristics and affected the survival time of AML patients as an independent risk indicator. SLC22A3 expression changed regularly as the disease complete remissions and relapses. Demethylation drug 5-aza-2'-deoxycytidine (DAC) activated transcription and increased mRNA expression of SLC22A3 in leukemia cell lines and AML fresh BMMNCs. Knockdown of SLC22A3 in leukemia cells enhanced cell proliferation and suppressed cell apoptosis. Data from public programs were used for auxiliary screening of probable molecular mechanisms of SLC22A3 in the antileukemia effect. CONCLUSIONS Our results showed that increased methylation and decreased expression of SLC22A3 may be indicators of poor prognosis in AML. Methylation-silenced SLC22A3 expression may have potential guiding significance on antileukemia effect of DAC.
Collapse
Affiliation(s)
- Yu Gu
- grid.452247.2Department of Hematology, Affiliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Zi-jun Xu
- Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,grid.452247.2Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Jing-dong Zhou
- grid.452247.2Department of Hematology, Affiliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Xiang-mei Wen
- Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,grid.452247.2Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Ye Jin
- grid.452247.2Department of Hematology, Affiliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Qian Yuan
- Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,grid.452247.2Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Pei-hui Xia
- Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,grid.452247.2Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Yuan Feng
- grid.452247.2Department of Hematology, Affiliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Lei Yang
- grid.452247.2Department of Hematology, Affiliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Jiang Lin
- Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,grid.452247.2Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| | - Jun Qian
- grid.452247.2Department of Hematology, Affiliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,Zhenjiang Clinical Research Center of Hematology, 8 Dianli Rd., Zhenjiang, 212002 Jiangsu People’s Republic of China ,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu People’s Republic of China
| |
Collapse
|
8
|
Small S, Oh TS, Platanias LC. Role of Biomarkers in the Management of Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:14543. [PMID: 36498870 PMCID: PMC9741257 DOI: 10.3390/ijms232314543] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Despite many recent advances in treatment options, acute myeloid leukemia (AML) still has a high mortality rate. One important issue in optimizing outcomes for AML patients lies in the limited ability to predict response to specific therapies, duration of response, and likelihood of relapse. With evolving genetic characterization and improving molecular definitions, the ability to predict outcomes and long-term prognosis is slowly improving. The majority of the currently used prognostic assessments relate to molecular and chromosomal abnormalities, as well as response to initial therapy. These risk categories, however, do not account for a large amount of the variability in AML. Laboratory techniques now utilized in the clinic extend beyond bone marrow morphology and single gene sequencing, to next-generation sequencing of large gene panels and multiparameter flow cytometry, among others. Other technologic advances, such as gene expression analysis, have yet to demonstrate enough predictive and prognostic power to be employed in clinical medicine outside of clinical trials, but may be incorporated into the clinic in the future. In this review, we discuss the utility of current biomarkers, and present novel biomarker techniques and strategies that are in development for AML patients. Measurable residual disease (MRD) is a powerful prognostic tool that is increasingly being incorporated into clinical practice, and there are some exciting emerging biomarker technologies that have the potential to improve prognostic power in AML. As AML continues to be a difficult-to-treat disease with poor outcomes in many subtypes, advances in biomarkers that lead to better treatment decisions are greatly needed.
Collapse
Affiliation(s)
- Sara Small
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Timothy S. Oh
- Division of Hospital Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|