1
|
Iyer P, Jasdanwala SS, Wang Y, Bhatia K, Bhatt S. Decoding Acute Myeloid Leukemia: A Clinician's Guide to Functional Profiling. Diagnostics (Basel) 2024; 14:2560. [PMID: 39594226 PMCID: PMC11593197 DOI: 10.3390/diagnostics14222560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a complex clonal disorder characterized by clinical, genetic, metabolomic, and epigenetic heterogeneity resulting in the uncontrolled proliferation of aberrant blood-forming precursor cells. Despite advancements in the understanding of the genetic, metabolic, and epigenetic landscape of AML, it remains a significant therapeutic challenge. Functional profiling techniques, such as BH3 profiling (BP), gene expression profiling (GEP), proteomics, metabolomics, drug sensitivity/resistance testing (DSRT), CRISPR/Cas9, and RNAi screens offer valuable insights into the functional behavior of leukemia cells. BP evaluates the mitochondrial response to pro-apoptotic BH3 peptides, determining a cell's apoptotic threshold and its reliance on specific anti-apoptotic proteins. This knowledge can pinpoint vulnerabilities in the mitochondria-mediated apoptotic pathway in leukemia cells, potentially informing treatment strategies and predicting therapeutic responses. GEP, particularly RNA sequencing, evaluates the transcriptomic landscape and identifies gene expression alterations specific to AML subtypes. Proteomics and metabolomics, utilizing mass spectrometry and nuclear magnetic resonance (NMR), provide a detailed view of the active proteins and metabolic pathways in leukemia cells. DSRT involves exposing leukemia cells to a panel of chemotherapeutic and targeted agents to assess their sensitivity or resistance profiles and potentially guide personalized treatment strategies. CRISPR/Cas9 and RNAi screens enable systematic disruption of genes to ascertain their roles in leukemia cell survival and proliferation. These techniques facilitate precise disease subtyping, uncover novel biomarkers and therapeutic targets, and provide a deeper understanding of drug-resistance mechanisms. Recent studies utilizing functional profiling have identified specific mutations and gene signatures associated with aggressive AML subtypes, aberrant signaling pathways, and potential opportunities for drug repurposing. The integration of multi-omics approaches, advances in single-cell sequencing, and artificial intelligence is expected to refine the precision of functional profiling and ultimately improve patient outcomes in AML. This review highlights the diverse landscape of functional profiling methods and emphasizes their respective advantages and limitations. It highlights select successes in how these methods have further advanced our understanding of AML biology, identifies druggable targets that have improved outcomes, delineates challenges associated with these techniques, and provides a prospective view of the future where these techniques are likely to be increasingly incorporated into the routine care of patients with AML.
Collapse
Affiliation(s)
- Prasad Iyer
- Children’s Blood and Cancer Centre, KK Women’s and Children’s Hospital, Singapore 229899, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Shaista Shabbir Jasdanwala
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| | - Yuhan Wang
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| | - Karanpreet Bhatia
- Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| |
Collapse
|
2
|
Ruglioni M, Crucitta S, Luculli GI, Tancredi G, Del Giudice ML, Mechelli S, Galimberti S, Danesi R, Del Re M. Understanding mechanisms of resistance to FLT3 inhibitors in adult FLT3-mutated acute myeloid leukemia to guide treatment strategy. Crit Rev Oncol Hematol 2024; 201:104424. [PMID: 38917943 DOI: 10.1016/j.critrevonc.2024.104424] [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: 02/29/2024] [Revised: 06/06/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024] Open
Abstract
The presence of FLT3 mutations, including the most common FLT3-ITD (internal tandem duplications) and FLT3-TKD (tyrosine kinase domain), is associated with an unfavorable prognosis in patients affected by acute myeloid leukemia (AML). In this setting, in recent years, new FLT3 inhibitors have demonstrated efficacy in improving survival and treatment response. Nevertheless, the development of primary and secondary mechanisms of resistance poses a significant obstacle to their efficacy. Understanding these mechanisms is crucial for developing novel therapeutic approaches to overcome resistance and improve the outcomes of patients. In this context, the use of novel FLT3 inhibitors and the combination of different targeted therapies have been studied. This review provides an update on the molecular alterations involved in the resistance to FLT3 inhibitors, and describes how the molecular monitoring may be used to guide treatment strategy in FLT3-mutated AML.
Collapse
Affiliation(s)
- Martina Ruglioni
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Giovanna Irene Luculli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Gaspare Tancredi
- Unit of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Maria Livia Del Giudice
- Unit of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Sandra Mechelli
- Unit of Internal Medicine 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Sara Galimberti
- Unit of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Romano Danesi
- Department of Oncology and Hemato-Oncology, University of Milan, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| |
Collapse
|
3
|
Andersen AN, Brodersen AM, Ayuda-Durán P, Piechaczyk L, Tadele DS, Baken L, Fredriksen J, Stoksflod M, Lenartova A, Fløisand Y, Skånland SS, Enserink JM. Clinical forecasting of acute myeloid leukemia using ex vivo drug-sensitivity profiling. CELL REPORTS METHODS 2023; 3:100654. [PMID: 38065095 PMCID: PMC10753296 DOI: 10.1016/j.crmeth.2023.100654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 09/16/2023] [Accepted: 11/09/2023] [Indexed: 12/21/2023]
Abstract
Current treatment selection for acute myeloid leukemia (AML) patients depends on risk stratification based on cytogenetic and genomic markers. However, the forecasting accuracy of treatment response remains modest, with most patients receiving intensive chemotherapy. Recently, ex vivo drug screening has gained traction in personalized treatment selection and as a tool for mapping patient groups based on relevant cancer dependencies. Here, we systematically evaluated the use of drug sensitivity profiling for predicting patient survival and clinical response to chemotherapy in a cohort of AML patients. We compared computational methodologies for scoring drug efficacy and characterized tools to counter noise and batch-related confounders pervasive in high-throughput drug testing. We show that ex vivo drug sensitivity profiling is a robust and versatile approach to patient prognostics that comprehensively maps functional signatures of treatment response and disease progression. In conclusion, ex vivo drug profiling can assess risk for individual AML patients and may guide clinical decision-making.
Collapse
Affiliation(s)
- Aram N Andersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway; Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Andrea M Brodersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway; Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Pilar Ayuda-Durán
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway
| | - Laure Piechaczyk
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway
| | - Dagim Shiferaw Tadele
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway
| | - Lizet Baken
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway
| | - Julia Fredriksen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway; Department of Haematology, Oslo University Hospital, 0372 Oslo, Norway
| | - Mia Stoksflod
- Department of Haematology, Oslo University Hospital, 0372 Oslo, Norway
| | - Andrea Lenartova
- Department of Haematology, Oslo University Hospital, 0372 Oslo, Norway
| | - Yngvar Fløisand
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway
| | - Sigrid S Skånland
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; K.G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Jorrit M Enserink
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Blindern, 0318 Oslo, Norway; Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| |
Collapse
|
4
|
Park S, Lee H. Molecular data representation based on gene embeddings for cancer drug response prediction. Sci Rep 2023; 13:21898. [PMID: 38081928 PMCID: PMC10713675 DOI: 10.1038/s41598-023-49003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Cancer drug response prediction is a crucial task in precision medicine, but existing models have limitations in effectively representing molecular profiles of cancer cells. Specifically, when these models represent molecular omics data such as gene expression, they employ a one-hot encoding-based approach, where a fixed gene set is selected for all samples and omics data values are assigned to specific positions in a vector. However, this approach restricts the utilization of embedding-vector-based methods, such as attention-based models, and limits the flexibility of gene selection. To address these issues, our study proposes gene embedding-based fully connected neural networks (GEN) that utilizes gene embedding vectors as input data for cancer drug response prediction. The GEN allows for the use of embedding-vector-based architectures and different gene sets for each sample, providing enhanced flexibility. To validate the efficacy of GEN, we conducted experiments on three cancer drug response datasets. Our results demonstrate that GEN outperforms other recently developed methods in cancer drug prediction tasks and offers improved gene representation capabilities. All source codes are available at https://github.com/DMCB-GIST/GEN/ .
Collapse
Affiliation(s)
- Sejin Park
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hyunju Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
- Artificial Intelligence Graduate School, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
5
|
Sheng L, Liu Y, Zhu Y, Zhou J, Hua H. Analysis of the clinical characteristics and prognosis of adult de novo acute myeloid leukemia (none APL) with PTPN11 mutations. Open Med (Wars) 2023; 18:20230830. [PMID: 38025540 PMCID: PMC10655689 DOI: 10.1515/med-2023-0830] [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: 05/28/2023] [Revised: 09/13/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023] Open
Abstract
We discuss the clinical characteristics and prognostic significance of adult individuals with PTPN11 mutations who have developed acute myeloid leukemia (AML) (none acute promyelocytic leukemia). Next generation sequencing and Sanger sequencing were used to detect 51 gene mutations, and multiplex-PCR was used to detect 41 fusion genes from 232 de novo adult AML patients retrospectively. About 7.76% patients harbored PTPN11 mutations, 20 PTPN11 alterations were identified, all of which were missense mutations in the N-SH2 (n = 16) and PTP (n = 4) domains located in exon 3. Patients with PTPN11 mut had significantly higher platelet counts and hemoglobin levels (p < 0.001), which were mainly detected in M5 (n = 12, 66.67%, p < 0.001) subtype. Patients with MLL-AF6 positive showed a higher frequency of PTPN11 mut (p = 0.018) in the 118 AML cases. PTPN11 mut were accompanied by other mutations, which were NPM1 (44.44%), DNMT3A (38.89%), FLT3 (38.89%), and NRAS (17.2%). PTPN11 mut had a negative impact on the complete remission rate in M5 subtype patients (p < 0.001). However, no statistically significant effect on overall survival (OS) with PTPN11 mut patients in the whole cohort and age group (p > 0.05) was observed. Further analysis revealed no significant difference in OS among NPM1 mut/PTPN11 mut, NPM1 mut/PTPN11 wt, DNMT3A mut/PTPN11 mut, and DNMT3A mut/PTPN11 wt patients (p > 0.05). Multivariate analysis showed the proportion of bone marrow blasts ≥65.4% was a factor significantly affecting OS in PTPN11 mut patients (p = 0.043).
Collapse
Affiliation(s)
- Li Sheng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yajiao Liu
- Nursing Department, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, China
| | - Yingying Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jingfen Zhou
- Department of Hematology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Haiying Hua
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
- Department of Hematology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
6
|
Steinhäuser S, Silva P, Lenk L, Beder T, Hartmann A, Hänzelmann S, Fransecky L, Neumann M, Bastian L, Lipinski S, Richter K, Bultmann M, Hübner E, Xia S, Röllig C, Vogiatzi F, Schewe DM, Yumiceba V, Schultz K, Spielmann M, Baldus CD. Isocitrate dehydrogenase 1 mutation drives leukemogenesis by PDGFRA activation due to insulator disruption in acute myeloid leukemia (AML). Leukemia 2023; 37:134-142. [PMID: 36411356 PMCID: PMC9883162 DOI: 10.1038/s41375-022-01751-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/22/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by complex molecular alterations and driver mutations. Elderly patients show increased frequencies of IDH mutations with high chemoresistance and relapse rates despite recent therapeutic advances. Besides being associated with global promoter hypermethylation, IDH1 mutation facilitated changes in 3D DNA-conformation by CTCF-anchor methylation and upregulated oncogene expression in glioma, correlating with poor prognosis. Here, we investigated the role of IDH1 p.R132H mutation in altering 3D DNA-architecture and subsequent oncogene activation in AML. Using public RNA-Seq data, we identified upregulation of tyrosine kinase PDGFRA in IDH1-mutant patients, correlating with poor prognosis. DNA methylation analysis identified CpG hypermethylation within a CTCF-anchor upstream of PDGFRA in IDH1-mutant patients. Increased PDGFRA expression, PDGFRA-CTCF methylation and decreased CTCF binding were confirmed in AML CRISPR cells with heterozygous IDH1 p.R132H mutation and upon exogenous 2-HG treatment. IDH1-mutant cells showed higher sensitivity to tyrosine kinase inhibitor dasatinib, which was supported by reduced blast count in a patient with refractory IDH1-mutant AML after dasatinib treatment. Our data illustrate that IDH1 p.R132H mutation leads to CTCF hypermethylation, disrupting DNA-looping and insulation of PDGFRA, resulting in PDGFRA upregulation in IDH1-mutant AML. Treatment with dasatinib may offer a novel treatment strategy for IDH1-mutant AML.
Collapse
Affiliation(s)
- Sophie Steinhäuser
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Patricia Silva
- Department of Hematology and Oncology, Charité University Hospital, Berlin, Germany
| | - Lennart Lenk
- Department of Pediatrics I, ALL-BFM Study Group, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Beder
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Alina Hartmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sonja Hänzelmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lars Fransecky
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Martin Neumann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lorenz Bastian
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Simone Lipinski
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Kathrin Richter
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Miriam Bultmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Emely Hübner
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Shuli Xia
- Kennedy Krieger Institute, Baltimore, MD, USA
- School of Medicine, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Christoph Röllig
- Department of Internal Medicine I, University Hospital Carl-Gustav-Carus, Dresden, Germany
| | - Fotini Vogiatzi
- Department of Pediatrics I, ALL-BFM Study Group, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Veronica Yumiceba
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Kristin Schultz
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Malte Spielmann
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Claudia Dorothea Baldus
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany.
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
7
|
Sun Y, Zhang F, Huo L, Cai W, Wang Q, Wen L, Yan L, Shen H, Xu X, Chen S. Clinical characteristics and prognostic analysis of acute myeloid leukemia patients with PTPN11 mutations. Hematology 2022; 27:1184-1190. [DOI: 10.1080/16078454.2022.2140274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yueyue Sun
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
- Cyrus Tang hematology center, Soochow University, Suzhou, People’s Republic of China
| | - Fenghong Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Li Huo
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
| | - Wenzhi Cai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
| | - Qinrong Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Lijun Wen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Lingzhi Yan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
| | - Hongjie Shen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
| | - Xiaoyu Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, People’s Republic of China
- Cyrus Tang hematology center, Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
8
|
Wang H, Chan KYY, Cheng CK, Ng MH, Lee PY, Cheng FWT, Lam GKS, Chow TW, Ha SY, Chiang AK, Leung WH, Leung AY, Wang CC, Zhang T, Zhang XB, So CC, Yuen YP, Sun Q, Zhang C, Xu Y, Cheung JTK, Ng WH, Tang PMK, Kang W, To KF, Lee WYW, Wong RS, Poon ENY, Zhao Q, Huang J, Chen C, Yuen PMP, Li CK, Leung AWK, Leung KT. Pharmacogenomic Profiling of Pediatric Acute Myeloid Leukemia to Identify Therapeutic Vulnerabilities and Inform Functional Precision Medicine. Blood Cancer Discov 2022; 3:516-535. [PMID: 35960210 PMCID: PMC9894568 DOI: 10.1158/2643-3230.bcd-22-0011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/31/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the expanding portfolio of targeted therapies for adults with acute myeloid leukemia (AML), direct implementation in children is challenging due to inherent differences in underlying genetics. Here we established the pharmacologic profile of pediatric AML by screening myeloblast sensitivity to approved and investigational agents, revealing candidates of immediate clinical relevance. Drug responses ex vivo correlated with patient characteristics, exhibited age-specific alterations, and concorded with activities in xenograft models. Integration with genomic data uncovered new gene-drug associations, suggesting actionable therapeutic vulnerabilities. Transcriptome profiling further identified gene-expression signatures associated with on- and off-target drug responses. We also demonstrated the feasibility of drug screening-guided treatment for children with high-risk AML, with two evaluable cases achieving remission. Collectively, this study offers a high-dimensional gene-drug clinical data set that could be leveraged to research the unique biology of pediatric AML and sets the stage for realizing functional precision medicine for the clinical management of the disease. SIGNIFICANCE We conducted integrated drug and genomic profiling of patient biopsies to build the functional genomic landscape of pediatric AML. Age-specific differences in drug response and new gene-drug interactions were identified. The feasibility of functional precision medicine-guided management of children with high-risk AML was successfully demonstrated in two evaluable clinical cases. This article is highlighted in the In This Issue feature, p. 476.
Collapse
Affiliation(s)
- Han Wang
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kathy Yuen Yee Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi Keung Cheng
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Margaret H.L. Ng
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Po Yi Lee
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Frankie Wai Tsoi Cheng
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Grace Kee See Lam
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Tin Wai Chow
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Shau Yin Ha
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Alan K.S. Chiang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wing Hang Leung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Anskar Y.H. Leung
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tao Zhang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xiao-Bing Zhang
- Department of Medicine, Loma Linda University, Loma Linda, California
| | - Chi Chiu So
- Department of Pathology, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Yuet Ping Yuen
- Department of Pathology, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Qiwei Sun
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi Zhang
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yaqun Xu
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - John Tak Kit Cheung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wing Hei Ng
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wayne Yuk Wai Lee
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Raymond S.M. Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ellen Ngar Yun Poon
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Qi Zhao
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Junbin Huang
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chun Chen
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Patrick Man Pan Yuen
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi-kong Li
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong.,Corresponding Authors: Kam Tong Leung, E-mail: ; Chi-kong Li, Hong Kong Children's Hospital, 1 Shing Cheong Road, Kowloon Bay, Kowloon, Hong Kong. Phone: 852-3513-3176; Fax: 852-2636-0020; E-mail: ; and Alex Wing Kwan Leung, E-mail:
| | - Alex Wing Kwan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong.,Corresponding Authors: Kam Tong Leung, E-mail: ; Chi-kong Li, Hong Kong Children's Hospital, 1 Shing Cheong Road, Kowloon Bay, Kowloon, Hong Kong. Phone: 852-3513-3176; Fax: 852-2636-0020; E-mail: ; and Alex Wing Kwan Leung, E-mail:
| | - Kam Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong.,Corresponding Authors: Kam Tong Leung, E-mail: ; Chi-kong Li, Hong Kong Children's Hospital, 1 Shing Cheong Road, Kowloon Bay, Kowloon, Hong Kong. Phone: 852-3513-3176; Fax: 852-2636-0020; E-mail: ; and Alex Wing Kwan Leung, E-mail:
| |
Collapse
|
9
|
Lee C, Lee S, Park E, Hong J, Shin DY, Byun JM, Yun H, Koh Y, Yoon SS. Transcriptional signatures of the BCL2 family for individualized acute myeloid leukaemia treatment. Genome Med 2022; 14:111. [PMID: 36171613 PMCID: PMC9520894 DOI: 10.1186/s13073-022-01115-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 09/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although anti-apoptotic proteins of the B-cell lymphoma-2 (BCL2) family have been utilized as therapeutic targets in acute myeloid leukaemia (AML), their complicated regulatory networks make individualized therapy difficult. This study aimed to discover the transcriptional signatures of BCL2 family genes that reflect regulatory dynamics, which can guide individualized therapeutic strategies. METHODS From three AML RNA-seq cohorts (BeatAML, LeuceGene, and TCGA; n = 451, 437, and 179, respectively), we constructed the BCL2 family signatures (BFSigs) by applying an innovative gene-set selection method reflecting biological knowledge followed by non-negative matrix factorization (NMF). To demonstrate the significance of the BFSigs, we conducted modelling to predict response to BCL2 family inhibitors, clustering, and functional enrichment analysis. Cross-platform validity of BFSigs was also confirmed using NanoString technology in a separate cohort of 47 patients. RESULTS We established BFSigs labeled as the BCL2, MCL1/BCL2, and BFL1/MCL1 signatures that identify key anti-apoptotic proteins. Unsupervised clustering based on BFSig information consistently classified AML patients into three robust subtypes across different AML cohorts, implying the existence of biological entities revealed by the BFSig approach. Interestingly, each subtype has distinct enrichment patterns of major cancer pathways, including MAPK and mTORC1, which propose subtype-specific combination treatment with apoptosis modulating drugs. The BFSig-based classifier also predicted response to venetoclax with remarkable performance (area under the ROC curve, AUROC = 0.874), which was well-validated in an independent cohort (AUROC = 0.950). Lastly, we successfully confirmed the validity of BFSigs using NanoString technology. CONCLUSIONS This study proposes BFSigs as a biomarker for the effective selection of apoptosis targeting treatments and cancer pathways to co-target in AML.
Collapse
Affiliation(s)
- Chansub Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sungyoung Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Center for Precision Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Eunchae Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junshik Hong
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Yeop Shin
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ja Min Byun
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Center for Precision Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Youngil Koh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Overcoming Resistance: FLT3 Inhibitors Past, Present, Future and the Challenge of Cure. Cancers (Basel) 2022; 14:cancers14174315. [PMID: 36077850 PMCID: PMC9454516 DOI: 10.3390/cancers14174315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
FLT3 ITD and TKD mutations occur in 20% and 10% of Acute Myeloid Leukemia (AML), respectively, and they represent the target of the first approved anti-leukemic therapies in the 2000s. Type I and type II FLT3 inhibitors (FLT3i) are active against FLT3 TKD/ITD and FLT3 ITD mutations alone respectively, but they still fail remissions in 30-40% of patients due to primary and secondary mechanisms of resistance, with variable relapse rate of 30-50%, influenced by NPM status and FLT3 allelic ratio. Mechanisms of resistance to FLT3i have recently been analyzed through NGS and single cell assays that have identified and elucidated the polyclonal nature of relapse in clinical and preclinical studies, summarized here. Knowledge of tumor escape pathways has helped in the identification of new targeted drugs to overcome resistance. Immunotherapy and combination or sequential use of BCL2 inhibitors and experimental drugs including aurora kinases, menin and JAK2 inhibitors will be the goal of present and future clinical trials, especially in patients with FLT3-mutated (FLT3mut) AML who are not eligible for allogeneic transplantation.
Collapse
|
11
|
Dal Bello R, Pasanisi J, Joudinaud R, Duchmann M, Pardieu B, Ayaka P, Di Feo G, Sodaro G, Chauvel C, Kim R, Vasseur L, Chat L, Ling F, Pacchiardi K, Vaganay C, Berrou J, Benaksas C, Boissel N, Braun T, Preudhomme C, Dombret H, Raffoux E, Fenouille N, Clappier E, Adès L, Puissant A, Itzykson R. A multiparametric niche-like drug screening platform in acute myeloid leukemia. Blood Cancer J 2022; 12:95. [PMID: 35750691 PMCID: PMC9232632 DOI: 10.1038/s41408-022-00689-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/13/2022] [Accepted: 06/07/2022] [Indexed: 02/06/2023] Open
Abstract
Functional precision medicine in AML often relies on short-term in vitro drug sensitivity screening (DSS) of primary patient cells in standard culture conditions. We designed a niche-like DSS assay combining physiologic hypoxia (O2 3%) and mesenchymal stromal cell (MSC) co-culture with multiparameter flow cytometry to enumerate lymphocytes and differentiating (CD11/CD14/CD15+) or leukemic stem cell (LSC)-enriched (GPR56+) cells within the leukemic bulk. After functional validation of GPR56 expression as a surrogate for LSC enrichment, the assay identified three patterns of response, including cytotoxicity on blasts sparing LSCs, induction of differentiation, and selective impairment of LSCs. We refined our niche-like culture by including plasma-like amino-acid and cytokine concentrations identified by targeted metabolomics and proteomics of primary AML bone marrow plasma samples. Systematic interrogation revealed distinct contributions of each niche-like component to leukemic outgrowth and drug response. Short-term niche-like culture preserved clonal architecture and transcriptional states of primary leukemic cells. In a cohort of 45 AML samples enriched for NPM1c AML, the niche-like multiparametric assay could predict morphologically (p = 0.02) and molecular (NPM1c MRD, p = 0.04) response to anthracycline-cytarabine induction chemotherapy. In this cohort, a 23-drug screen nominated ruxolitinib as a sensitizer to anthracycline-cytarabine. This finding was validated in an NPM1c PDX model.
Collapse
Affiliation(s)
- Reinaldo Dal Bello
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Justine Pasanisi
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Romane Joudinaud
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Matthieu Duchmann
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Bryann Pardieu
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Paolo Ayaka
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Giuseppe Di Feo
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Gaetano Sodaro
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Clémentine Chauvel
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Rathana Kim
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Loic Vasseur
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Laureen Chat
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Frank Ling
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Kim Pacchiardi
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Camille Vaganay
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Jeannig Berrou
- Université Paris Cité, EA 3518, IRSL, Hôpital Saint-Louis, F-75010, Paris, France
| | - Chaima Benaksas
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Nicolas Boissel
- Service Hématologie Adolescents Jeunes Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Thorsten Braun
- Université Paris Cité, EA 3518, IRSL, Hôpital Saint-Louis, F-75010, Paris, France.,Service d'Hématologie clinique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Claude Preudhomme
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Hervé Dombret
- Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France.,Université Paris Cité, EA 3518, IRSL, Hôpital Saint-Louis, F-75010, Paris, France
| | - Emmanuel Raffoux
- Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Nina Fenouille
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Emmanuelle Clappier
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Lionel Adès
- Service Hématologie Seniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Alexandre Puissant
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Raphael Itzykson
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France. .,Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France.
| |
Collapse
|
12
|
Ohmoto A, Fuji S. Current status of drug repositioning in hematology. Expert Rev Hematol 2021; 14:1005-1011. [PMID: 34657533 DOI: 10.1080/17474086.2021.1995348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Drug repositioning (DR) is defined as determining new therapeutic applications for existing drugs. This approach is advantageous over de novo drug discovery in accelerating clinical development, in terms of lower costs, a shortened development period, a well-known action mechanism, a feasible dosage, and an acceptable safety profile. AREAS COVERED This work was aimed at reviewing agents with successful DR in hematology. EXPERT OPINION Thalidomide and plerixafor have been successfully repositioned for treating multiple myeloma and harvesting peripheral blood stem cells, respectively. The former was originally developed as a sedative and the latter as an anti-HIV drug. Currently, the feasibility of repositioning various agents is being explored (e.g. an anti-influenza virus drug oseltamivir for primary immune thrombocytopenia, an anti-HIV drug abacavir for adult T-cell leukemia, and a macrolide antibiotic clarithromycin for multiple myeloma). Furthermore, bosutinib for chronic myeloid leukemia or the antiplatelet drug cilostazol have been suggested to have clinical benefits for the management of amyotrophic lateral sclerosis and ischemic stroke, respectively. To promote DR, effective application of artificial intelligence or stem cell models, comprehensive database construction shared between academia and pharmaceutical companies, suitable handling of drug patents, and wide cooperation in the area of specialty are warranted.
Collapse
Affiliation(s)
- Akihiro Ohmoto
- Department of Medical Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shigeo Fuji
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
13
|
White BS, Khan SA, Mason MJ, Ammad-Ud-Din M, Potdar S, Malani D, Kuusanmäki H, Druker BJ, Heckman C, Kallioniemi O, Kurtz SE, Porkka K, Tognon CE, Tyner JW, Aittokallio T, Wennerberg K, Guinney J. Bayesian multi-source regression and monocyte-associated gene expression predict BCL-2 inhibitor resistance in acute myeloid leukemia. NPJ Precis Oncol 2021; 5:71. [PMID: 34302041 PMCID: PMC8302655 DOI: 10.1038/s41698-021-00209-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 06/22/2021] [Indexed: 11/09/2022] Open
Abstract
The FDA recently approved eight targeted therapies for acute myeloid leukemia (AML), including the BCL-2 inhibitor venetoclax. Maximizing efficacy of these treatments requires refining patient selection. To this end, we analyzed two recent AML studies profiling the gene expression and ex vivo drug response of primary patient samples. We find that ex vivo samples often exhibit a general sensitivity to (any) drug exposure, independent of drug target. We observe that this "general response across drugs" (GRD) is associated with FLT3-ITD mutations, clinical response to standard induction chemotherapy, and overall survival. Further, incorporating GRD into expression-based regression models trained on one of the studies improved their performance in predicting ex vivo response in the second study, thus signifying its relevance to precision oncology efforts. We find that venetoclax response is independent of GRD but instead show that it is linked to expression of monocyte-associated genes by developing and applying a multi-source Bayesian regression approach. The method shares information across studies to robustly identify biomarkers of drug response and is broadly applicable in integrative analyses.
Collapse
Affiliation(s)
- Brian S White
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA.
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
| | - Suleiman A Khan
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Mike J Mason
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA
| | - Muhammad Ammad-Ud-Din
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Disha Malani
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Biotech Research & Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Brian J Druker
- Howard Hughes Medical Institute, Portland, OR, USA
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Caroline Heckman
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Scilifelab, Karolinska Institute, Solna, Sweden
| | - Stephen E Kurtz
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kimmo Porkka
- HUS Comprehensive Cancer Center, Hematology Research Unit Helsinki and iCAN Digital Precision Cancer Center Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Cristina E Tognon
- Howard Hughes Medical Institute, Portland, OR, USA
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey W Tyner
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, Oslo, Norway
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Biotech Research & Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Justin Guinney
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
The importance of genomic predictors for clinical outcome of hematological malignancies. BLOOD SCIENCE 2021; 3:93-95. [PMID: 35402837 PMCID: PMC8974908 DOI: 10.1097/bs9.0000000000000075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/17/2022] Open
|
15
|
Ramkissoon LA, Buhlinger K, Nichols A, Coombs CC, Foster MC, Galeotti J, Kaiser-Rogers K, Richardson DR, Montgomery ND, Zeidner JF. Clonal evolution of Philadelphia chromosome in acute myeloid leukemia after enasidenib treatment. Leuk Lymphoma 2021; 62:3035-3038. [PMID: 34151687 DOI: 10.1080/10428194.2021.1941928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Lori A Ramkissoon
- Department of Pathology & Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Kaitlyn Buhlinger
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Angela Nichols
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Catherine C Coombs
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Division of Hematology, Department of Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Matthew C Foster
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Division of Hematology, Department of Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jonathan Galeotti
- Department of Pathology & Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Kathleen Kaiser-Rogers
- Department of Pathology & Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Department of Pediatrics, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Daniel R Richardson
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Division of Hematology, Department of Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Nathan D Montgomery
- Department of Pathology & Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Joshua F Zeidner
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Division of Hematology, Department of Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Affiliation(s)
- John S Welch
- Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|