51
|
Vonk CM, Al Hinai ASA, Hanekamp D, Valk PJM. Molecular Minimal Residual Disease Detection in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:5431. [PMID: 34771594 PMCID: PMC8582498 DOI: 10.3390/cancers13215431] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Initial induction chemotherapy to eradicate the bulk of acute myeloid leukemia (AML) cells results in complete remission (CR) in the majority of patients. However, leukemic cells persisting in the bone marrow below the morphologic threshold remain unaffected and have the potential to proliferate and re-emerge as AML relapse. Detection of minimal/measurable residual disease (MRD) is a promising prognostic marker for AML relapse as it can assess an individual patients' risk profile and evaluate their response to treatment. With the emergence of molecular techniques, such as next generation sequencing (NGS), a more sensitive assessment of molecular MRD markers is available. In recent years, the detection of MRD by molecular assays and its association with AML relapse and survival has been explored and verified in multiple studies. Although most studies show that the presence of MRD leads to a worse clinical outcome, molecular-based methods face several challenges including limited sensitivity/specificity, and a difficult distinction between mutations that are representative of AML rather than clonal hematopoiesis. This review describes the studies that have been performed using molecular-based assays for MRD detection in the context of other MRD detection approaches in AML, and discusses limitations, challenges and opportunities.
Collapse
Affiliation(s)
- Christian M Vonk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
| | - Adil S A Al Hinai
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
- National Genetic Center, Ministry of Health, Muscat 111, Oman
| | - Diana Hanekamp
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
- Department of Hematology, Cancer Center VU University Medical Center, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| | - Peter J M Valk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
| |
Collapse
|
52
|
Wang X, Chen Y, Li Z, Huang B, Xu L, Lai J, Lu Y, Zha X, Liu B, Lan Y, Li Y. Single-Cell RNA-Seq of T Cells in B-ALL Patients Reveals an Exhausted Subset with Remarkable Heterogeneity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101447. [PMID: 34365737 PMCID: PMC8498858 DOI: 10.1002/advs.202101447] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/27/2021] [Indexed: 06/02/2023]
Abstract
Characterization of functional T cell clusters is key to developing strategies for immunotherapy and predicting clinical responses in leukemia. Here, single-cell RNA sequencing is performed with T cells sorted from the peripheral blood of healthy individuals and patients with B cell-acute lymphoblastic leukemia (B-ALL). Unbiased bioinformatics analysis enabled the authors to identify 13 T cell clusters in the patients based on their molecular properties. All 11 major T cell subsets in healthy individuals are found in the patients with B-ALL, with the counterparts in the patients universally showing more activated characteristics. Two exhausted T cell populations, characterized by up-regulation of TIGIT, PDCD1, HLADRA, LAG3, and CTLA4 are specifically discovered in B-ALL patients. Of note, these exhausted T cells possess remarkable heterogeneity, and ten sub-clusters are further identified, which are characterized by different cell cycle phases, naïve states, and GNLY (coding granulysin) expression. Coupled with single-cell T cell receptor repertoire profiling, diverse originations of the exhausted T cells in B-ALL are suggested, and clonally expanded exhausted T cells are likely to originate from CD8+ effector memory/terminal effector cells. Together, these data provide for the first-time valuable insights for understanding exhausted T cell populations in leukemia.
Collapse
Affiliation(s)
- Xiaofang Wang
- Department of HematologyFirst Affiliated HospitalJinan UniversityNo. 601 West of Huangpu AvenueGuangzhou510632China
- Key Laboratory for Regenerative Medicine of Ministry of EducationInstitute of HematologySchool of MedicineJinan UniversityGuangzhou510632China
| | - Yanjuan Chen
- Key Laboratory for Regenerative Medicine of Ministry of EducationInstitute of HematologySchool of MedicineJinan UniversityGuangzhou510632China
| | - Zongcheng Li
- State Key Laboratory of Experimental HematologyInstitute of HematologyFifth Medical Center of Chinese PLA General HospitalBeijing100071China
| | - Bingyan Huang
- Key Laboratory for Regenerative Medicine of Ministry of EducationInstitute of HematologySchool of MedicineJinan UniversityGuangzhou510632China
| | - Ling Xu
- Department of HematologyFirst Affiliated HospitalJinan UniversityNo. 601 West of Huangpu AvenueGuangzhou510632China
- Key Laboratory for Regenerative Medicine of Ministry of EducationInstitute of HematologySchool of MedicineJinan UniversityGuangzhou510632China
| | - Jing Lai
- Department of HematologyFirst Affiliated HospitalJinan UniversityNo. 601 West of Huangpu AvenueGuangzhou510632China
| | - Yuhong Lu
- Department of HematologyFirst Affiliated HospitalJinan UniversityNo. 601 West of Huangpu AvenueGuangzhou510632China
| | - Xianfeng Zha
- Department of Clinical LaboratoryFirst Affiliated HospitalSchool of MedicineJinan UniversityNo. 601 West of Huangpu AvenueGuangzhou510632China
| | - Bing Liu
- State Key Laboratory of Experimental HematologyInstitute of HematologyFifth Medical Center of Chinese PLA General HospitalBeijing100071China
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of EducationInstitute of HematologySchool of MedicineJinan UniversityGuangzhou510632China
| | - Yangqiu Li
- Department of HematologyFirst Affiliated HospitalJinan UniversityNo. 601 West of Huangpu AvenueGuangzhou510632China
- Key Laboratory for Regenerative Medicine of Ministry of EducationInstitute of HematologySchool of MedicineJinan UniversityGuangzhou510632China
| |
Collapse
|
53
|
Dinh KN, Jaksik R, Corey SJ, Kimmel M. Predicting Time to Relapse in Acute Myeloid Leukemia through Stochastic Modeling of Minimal Residual Disease Based on Clonality Data. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2021; 1. [PMID: 34541576 DOI: 10.1002/cso2.1026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Event-free and overall survival remain poor for patients with acute myeloid leukemia. Chemoresistant clones contributing to relapse arise from minimal residual disease (MRD) or newly-acquired mutations. However, the dynamics of clones comprising MRD is poorly understood. We developed a predictive stochastic model, based on a multitype age-dependent Markov branching process, to describe how random events in MRD contribute to the heterogeneity in treatment response. We employed training and validation sets of patients who underwent whole genome sequencing and for whom mutant clone frequencies at diagnosis and relapse were available. The disease evolution and treatment outcome are subject to stochastic fluctuations. Estimates of malignant clone growth rates, obtained by model fitting, are consistent with published data. Using the estimates from the training set, we developed a function linking MRD and time of relapse, with MRD inferred from the model fits to clone frequencies and other data. An independent validation set confirmed our model. In a third data set, we fitted the model to data at diagnosis and remission and predicted the time to relapse. As a conclusion, given bone marrow genome at diagnosis and MRD at or past remission, the model can predict time to relapse, and help guide treatment decisions to mitigate relapse.
Collapse
Affiliation(s)
- Khanh N Dinh
- Irving Institute of Cancer Dynamics, Columbia University, New York, NY, USA
| | - Roman Jaksik
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Seth J Corey
- Departments of Pediatric Hematology/Oncology and Stem Cell Transplantation and Cancer Biology, Cleveland Clinic, Cleveland, OH, USA
| | - Marek Kimmel
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland.,Department of Statistics, Rice University, Houston, TX, USA
| |
Collapse
|
54
|
Patel SA, Lloyd MR, Cerny J, Shi Q, Simin K, Ediriwickrema A, Hutchinson L, Miron PM, Higgins AW, Ramanathan M, Gerber JM. Clinico-genomic profiling and clonal dynamic modeling of TP53-aberrant myelodysplastic syndrome and acute myeloid leukemia. Leuk Lymphoma 2021; 62:3348-3360. [PMID: 34496723 DOI: 10.1080/10428194.2021.1957869] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
TP53-aberrant myelodysplastic syndrome and acute myeloid leukemia have dismal outcomes. Here, we define the clinico-genomic landscape of TP53 disruptions in 40 patients and employ clonal dynamic modeling to map the mutational hierarchy against clinical outcomes. Most TP53 mutations (45.2%) localized to the L3 loop or LSH motif of the DNA-binding domain. TP53 disruptions had high co-occurrence with mutations in epigenetic regulators, spliceosome machinery, and cohesin complex and low co-occurrence with mutations in proliferative signaling genes. Ancestral and descendant TP53 mutations constituted measurable residual disease and fueled relapse. High mutant TP53 gene dosage predicted low durability of remission. The median overall survival (OS) was 280 days. Hypomethylating agent-based therapy served as an effective bridge to transplant, leading to improved median OS compared to patients who did not receive a transplant (14.7 vs. 5.1 months). OS was independent of the genomic location of TP53 disruption, which has implications for rational therapeutic design.
Collapse
Affiliation(s)
- Shyam A Patel
- Department of Medicine-Hematology & Oncology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Maxwell R Lloyd
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jan Cerny
- Department of Medicine-Hematology & Oncology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Qiming Shi
- Department of Medicine-Hematology & Oncology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Population & Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA, USA
| | - Karl Simin
- Department of Molecular, Cell & Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Asiri Ediriwickrema
- Division of Hematology, Department of Medicine, Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Lloyd Hutchinson
- Department of Pathology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Patricia M Miron
- Department of Pathology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Anne W Higgins
- Department of Pathology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Muthalagu Ramanathan
- Department of Medicine-Hematology & Oncology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jonathan M Gerber
- Department of Medicine-Hematology & Oncology, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
55
|
Stiehl T, Marciniak-Czochra A. Computational Reconstruction of Clonal Hierarchies From Bulk Sequencing Data of Acute Myeloid Leukemia Samples. Front Physiol 2021; 12:596194. [PMID: 34497529 PMCID: PMC8419336 DOI: 10.3389/fphys.2021.596194] [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: 08/18/2020] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Acute myeloid leukemia is an aggressive cancer of the blood forming system. The malignant cell population is composed of multiple clones that evolve over time. Clonal data reflect the mechanisms governing treatment response and relapse. Single cell sequencing provides most direct insights into the clonal composition of the leukemic cells, however it is still not routinely available in clinical practice. In this work we develop a computational algorithm that allows identifying all clonal hierarchies that are compatible with bulk variant allele frequencies measured in a patient sample. The clonal hierarchies represent descendance relations between the different clones and reveal the order in which mutations have been acquired. The proposed computational approach is tested using single cell sequencing data that allow comparing the outcome of the algorithm with the true structure of the clonal hierarchy. We investigate which problems occur during reconstruction of clonal hierarchies from bulk sequencing data. Our results suggest that in many cases only a small number of possible hierarchies fits the bulk data. This implies that bulk sequencing data can be used to obtain insights in clonal evolution.
Collapse
Affiliation(s)
- Thomas Stiehl
- Institute for Computational Biomedicine – Disease Modeling, RWTH Aachen University, Aachen, Germany
- Institute of Applied Mathematics, Interdisciplinary Center for Scientific Computing and Bioquant Center, Heidelberg University, Heidelberg, Germany
| | - Anna Marciniak-Czochra
- Institute of Applied Mathematics, Interdisciplinary Center for Scientific Computing and Bioquant Center, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
56
|
Kim N, Eum HH, Lee HO. Clinical Perspectives of Single-Cell RNA Sequencing. Biomolecules 2021; 11:biom11081161. [PMID: 34439827 PMCID: PMC8394304 DOI: 10.3390/biom11081161] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022] Open
Abstract
The ability of single-cell genomics to resolve cellular heterogeneity is highly appreciated in cancer and is being exploited for precision medicine. In the recent decade, we have witnessed the incorporation of cancer genomics into the clinical decision-making process for molecular-targeted therapies. Compared with conventional genomics, which primarily focuses on the specific and sensitive detection of the molecular targets, single-cell genomics addresses intratumoral heterogeneity and the microenvironmental components impacting the treatment response and resistance. As an exploratory tool, single-cell genomics provides an unprecedented opportunity to improve the diagnosis, monitoring, and treatment of cancer. The results obtained upon employing bulk cancer genomics indicate that single-cell genomics is at an early stage with respect to exploration of clinical relevance and requires further innovations to become a widely utilized technology in the clinic.
Collapse
Affiliation(s)
- Nayoung Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (N.K.); (H.H.E.)
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Hye Hyeon Eum
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (N.K.); (H.H.E.)
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Hae-Ock Lee
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (N.K.); (H.H.E.)
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-8155
| |
Collapse
|
57
|
Abstract
PURPOSE OF REVIEW Clonal heterogeneity is a significant obstacle to successful treatment of patients with acute myeloid leukemia (AML). Here, we review new advances in the understanding of genetic heterogeneity in AML using single-cell DNA-sequencing technology. RECENT FINDINGS New genomics and immunologic discovery tools have provided single-cell resolution maps of the clonal architecture of AML. The use of these technologies reveals the mutational landscape of AML at diagnosis, during treatment, and at relapse has an enormous degree of clonal complexity and diversity that is poised to adapt and evolve under environmental pressures. SUMMARY AML is a complex ecosystem of competing and cooperating clones undergoing constant evolution and selection.
Collapse
|
58
|
Madaci L, Colle J, Venton G, Farnault L, Loriod B, Costello R. The contribution of single-cell analysis of acute leukemia in the therapeutic strategy. Biomark Res 2021; 9:50. [PMID: 34176517 PMCID: PMC8237443 DOI: 10.1186/s40364-021-00300-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
After decades during which the treatment of acute myeloblastic leukemia was limited to variations around a skeleton of cytarabine/anthracycline, targeted therapies appeared. These therapies, first based on monoclonal antibodies, also rely on specific inhibitors of various molecular abnormalities. A significant but modest prognosis improvement has been observed thanks to these new treatments that are limited by a high rate of relapse, due to the intrinsic chemo and immune-resistance of leukemia stem cell, together with the acquisition of these resistances by clonal evolution. Relapses are also influenced by the equilibrium between the pro or anti-tumor signals from the bone marrow stromal microenvironment and immune effectors. What should be the place of the targeted therapeutic options in light of the tumor heterogeneity inherent to leukemia and the clonal drift of which this type of tumor is capable? Novel approaches by single cell analysis and next generation sequencing precisely define clonal heterogeneity and evolution, leading to a personalized and time variable adapted treatment. Indeed, the evolution of leukemia, either spontaneous or under therapy selection pressure, is a very complex phenomenon. The model of linear evolution is to be forgotten because single cell analysis of samples at diagnosis and at relapse show that tumor escape to therapy occurs from ancestral as well as terminal clones. The determination by the single cell technique of the trajectories of the different tumor sub-populations allows the identification of clones that accumulate factors of resistance to chemo/immunotherapy ("pan-resistant clones"), making possible to choose the combinatorial agents most likely to eradicate these cells. In addition, the single cell technique identifies the nature of each cell and can analyze, on the same sample, both the tumor cells and their environment. It is thus possible to evaluate the populations of immune effectors (T-lymphocytes, natural killer cells) for the leukemia stress-induced alteration of their functions. Finally, the single cells techniques are an invaluable tool for evaluation of the measurable residual disease since not only able to quantify but also to determine the most appropriate treatment according to the sensitivity profile to immuno-chemotherapy of remaining leukemic cells.
Collapse
Affiliation(s)
- Lamia Madaci
- Laboratoire TAGC/INSERM UMR 1090, Parc Scientifique de Luminy case 928, 163, Avenue de Luminy, Cedex 09, 13288, Marseille, France
| | - Julien Colle
- Laboratoire TAGC/INSERM UMR 1090, Parc Scientifique de Luminy case 928, 163, Avenue de Luminy, Cedex 09, 13288, Marseille, France.,Service d'Hématologie et Thérapie Cellulaire, Hôpital La Conception, Assistance Publique des Hôpitaux de Marseille, 147 boulevard Baille, 13005, Marseille, France
| | - Geoffroy Venton
- Laboratoire TAGC/INSERM UMR 1090, Parc Scientifique de Luminy case 928, 163, Avenue de Luminy, Cedex 09, 13288, Marseille, France.,Service d'Hématologie et Thérapie Cellulaire, Hôpital La Conception, Assistance Publique des Hôpitaux de Marseille, 147 boulevard Baille, 13005, Marseille, France
| | - Laure Farnault
- Laboratoire TAGC/INSERM UMR 1090, Parc Scientifique de Luminy case 928, 163, Avenue de Luminy, Cedex 09, 13288, Marseille, France.,Service d'Hématologie et Thérapie Cellulaire, Hôpital La Conception, Assistance Publique des Hôpitaux de Marseille, 147 boulevard Baille, 13005, Marseille, France
| | - Béatrice Loriod
- Laboratoire TAGC/INSERM UMR 1090, Parc Scientifique de Luminy case 928, 163, Avenue de Luminy, Cedex 09, 13288, Marseille, France.,TGML-TAGC/INSERM UMR1090 Parc Scientifique de Luminy case 928, 163, avenue de Luminy, Cedex 09, 13288, Marseille, France
| | - Régis Costello
- Laboratoire TAGC/INSERM UMR 1090, Parc Scientifique de Luminy case 928, 163, Avenue de Luminy, Cedex 09, 13288, Marseille, France. .,Service d'Hématologie et Thérapie Cellulaire, Hôpital La Conception, Assistance Publique des Hôpitaux de Marseille, 147 boulevard Baille, 13005, Marseille, France.
| |
Collapse
|
59
|
Genomic analysis of cellular hierarchy in acute myeloid leukemia using ultrasensitive LC-FACSeq. Leukemia 2021; 35:3406-3420. [PMID: 34021247 PMCID: PMC8606012 DOI: 10.1038/s41375-021-01295-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022]
Abstract
Hematopoiesis is hierarchical, and it has been postulated that acute myeloid leukemia (AML) is organized similarly with leukemia stem cells (LSCs) residing at the apex. Limited cells acquired by fluorescence activated cell sorting in tandem with targeted amplicon-based sequencing (LC-FACSeq) enables identification of mutations in small subpopulations of cells, such as LSCs. Leveraging this, we studied clonal compositions of immunophenotypically-defined compartments in AML through genomic and functional analyses at diagnosis, remission and relapse in 88 AML patients. Mutations involving DNA methylation pathways, transcription factors and spliceosomal machinery did not differ across compartments, while signaling pathway mutations were less frequent in putative LSCs. We also provide insights into TP53-mutated AML by demonstrating stepwise acquisition of mutations beginning from the preleukemic hematopoietic stem cell stage. In 10 analyzed cases, acquisition of additional mutations and del(17p) led to genetic and functional heterogeneity within the LSC pool with subclones harboring varying degrees of clonogenic potential. Finally, we use LC-FACSeq to track clonal evolution in serial samples, which can also be a powerful tool to direct targeted therapy against measurable residual disease. Therefore, studying clinically significant small subpopulations of cells can improve our understanding of AML biology and offers advantages over bulk sequencing to monitor the evolution of disease.
Collapse
|
60
|
Gohil SH, Iorgulescu JB, Braun DA, Keskin DB, Livak KJ. Applying high-dimensional single-cell technologies to the analysis of cancer immunotherapy. Nat Rev Clin Oncol 2021; 18:244-256. [PMID: 33277626 PMCID: PMC8415132 DOI: 10.1038/s41571-020-00449-x] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
Advances in molecular biology, microfluidics and bioinformatics have empowered the study of thousands or even millions of individual cells from malignant tumours at the single-cell level of resolution. This high-dimensional, multi-faceted characterization of the genomic, transcriptomic, epigenomic and proteomic features of the tumour and/or the associated immune and stromal cells enables the dissection of tumour heterogeneity, the complex interactions between tumour cells and their microenvironment, and the details of the evolutionary trajectory of each tumour. Single-cell transcriptomics, the ability to track individual T cell clones through paired sequencing of the T cell receptor genes and high-dimensional single-cell spatial analysis are all areas of particular relevance to immuno-oncology. Multidimensional biomarker signatures will increasingly be crucial to guiding clinical decision-making in each patient with cancer. High-dimensional single-cell technologies are likely to provide the resolution and richness of data required to generate such clinically relevant signatures in immuno-oncology. In this Perspective, we describe advances made using transformative single-cell analysis technologies, especially in relation to clinical response and resistance to immunotherapy, and discuss the growing utility of single-cell approaches for answering important research questions.
Collapse
Affiliation(s)
- Satyen H Gohil
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Academic Haematology, University College London Cancer Institute, London, UK
| | - J Bryan Iorgulescu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David A Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
61
|
Gonzalez Castro LN, Tirosh I, Suvà ML. Decoding Cancer Biology One Cell at a Time. Cancer Discov 2021; 11:960-970. [PMID: 33811126 PMCID: PMC8030694 DOI: 10.1158/2159-8290.cd-20-1376] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022]
Abstract
Human tumors are composed of diverse malignant and nonmalignant cells, generating a complex ecosystem that governs tumor biology and response to treatments. Recent technological advances have enabled the characterization of tumors at single-cell resolution, providing a compelling strategy to dissect their intricate biology. Here we describe recent developments in single-cell expression profiling and the studies applying them in clinical settings. We highlight some of the powerful insights gleaned from these studies for tumor classification, stem cell programs, tumor microenvironment, metastasis, and response to targeted and immune therapies. SIGNIFICANCE: Intratumor heterogeneity (ITH) has been a major barrier to our understanding of cancer. Single-cell genomics is leading a revolution in our ability to systematically dissect ITH. In this review, we focus on single-cell expression profiling and lessons learned in key aspects of human tumor biology.
Collapse
Affiliation(s)
- L Nicolas Gonzalez Castro
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Mario L Suvà
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
62
|
García-Sanz R, Jiménez C. Time to Move to the Single-Cell Level: Applications of Single-Cell Multi-Omics to Hematological Malignancies and Waldenström's Macroglobulinemia-A Particularly Heterogeneous Lymphoma. Cancers (Basel) 2021; 13:1541. [PMID: 33810569 PMCID: PMC8037673 DOI: 10.3390/cancers13071541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Single-cell sequencing techniques have become a powerful tool for characterizing intra-tumor heterogeneity, which has been reflected in the increasing number of studies carried out and reported. We have rigorously reviewed and compiled the information about these techniques inasmuch as they are relative to the area of hematology to provide a practical view of their potential applications. Studies show how single-cell multi-omics can overcome the limitations of bulk sequencing and be applied at all stages of tumor development, giving insights into the origin and pathogenesis of the tumors, the clonal architecture and evolution, or the mechanisms of therapy resistance. Information at the single-cell level may help resolve questions related to intra-tumor heterogeneity that have not been previously explained by other techniques. With that in mind, we review the existing knowledge about a heterogeneous lymphoma called Waldenström's macroglobulinemia and discuss how single-cell studies may help elucidate the underlying causes of this heterogeneity.
Collapse
Affiliation(s)
- Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain;
| | | |
Collapse
|
63
|
Pfisterer U, Bräunig J, Brattås P, Heidenblad M, Karlsson G, Fioretos T. Single-cell sequencing in translational cancer research and challenges to meet clinical diagnostic needs. Genes Chromosomes Cancer 2021; 60:504-524. [PMID: 33611828 DOI: 10.1002/gcc.22944] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022] Open
Abstract
The ability to capture alterations in the genome or transcriptome by next-generation sequencing has provided critical insight into molecular changes and programs underlying cancer biology. With the rapid technological development in single-cell sequencing, it has become possible to study individual cells at the transcriptional, genetic, epigenetic, and protein level. Using single-cell analysis, an increased resolution of fundamental processes underlying cancer development is obtained, providing comprehensive insights otherwise lost by sequencing of entire (bulk) samples, in which molecular signatures of individual cells are averaged across the entire cell population. Here, we provide a concise overview on the application of single-cell analysis of different modalities within cancer research by highlighting key articles of their respective fields. We furthermore examine the potential of existing technologies to meet clinical diagnostic needs and discuss current challenges associated with this translation.
Collapse
Affiliation(s)
- Ulrich Pfisterer
- Center for Translational Genomics, Lund University, Lund, Sweden.,Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Julia Bräunig
- Center for Translational Genomics, Lund University, Lund, Sweden.,Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Per Brattås
- Center for Translational Genomics, Lund University, Lund, Sweden.,Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Markus Heidenblad
- Center for Translational Genomics, Lund University, Lund, Sweden.,Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Göran Karlsson
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Thoas Fioretos
- Center for Translational Genomics, Lund University, Lund, Sweden.,Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden.,Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
64
|
Analysis of Intratumoral Heterogeneity in Myelodysplastic Syndromes with Isolated del(5q) Using a Single Cell Approach. Cancers (Basel) 2021; 13:cancers13040841. [PMID: 33671317 PMCID: PMC7922695 DOI: 10.3390/cancers13040841] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic stem cell malignancies characterized by ineffective differentiation of one or more bone marrow cell lineages. Only 50% of patients with de novo MDS will be found to have cytogenetic abnormalities, of which del(5q) is the most common. In 10% of MDS cases, del(5q) is found as a sole abnormality. In this work, a single cell approach was used to analyze intratumoral heterogeneity in four patients with MDS with isolated del(5q). We were able to observe that an ancestral event in one patient can appear as a secondary hit in another one, thus reflecting the high intratumoral heterogeneity in MDS with isolated del(5q) and the importance of patient-specific molecular characterization. Abstract Myelodysplastic syndromes (MDS) are a heterogeneous group of hematological diseases. Among them, the most well characterized subtype is MDS with isolated chromosome 5q deletion (MDS del(5q)), which is the only one defined by a cytogenetic abnormality that makes these patients candidates to be treated with lenalidomide. During the last decade, single cell (SC) analysis has emerged as a powerful tool to decipher clonal architecture and to further understand cancer and other diseases at higher resolution level compared to bulk sequencing techniques. In this study, a SC approach was used to analyze intratumoral heterogeneity in four patients with MDS del(5q). Single CD34+CD117+CD45+CD19- bone marrow hematopoietic stem progenitor cells were isolated using the C1 system (Fluidigm) from diagnosis or before receiving any treatment and from available follow-up samples. Selected somatic alterations were further analyzed in SC by high-throughput qPCR (Biomark HD, Fluidigm) using specific TaqMan assays. A median of 175 cells per sample were analyzed. Inferred clonal architectures were relatively simple and either linear or branching. Similar to previous studies based on bulk sequencing to infer clonal architecture, we were able to observe that an ancestral event in one patient can appear as a secondary hit in another one, thus reflecting the high intratumoral heterogeneity in MDS del(5q) and the importance of patient-specific molecular characterization.
Collapse
|
65
|
Acha P, Hoyos M, Pratcorona M, Fuster-Tormo F, Palomo L, Ortega E, Zamora L, Vives S, Granada I, Montoro J, Garcia A, Arnan M, Cervera M, Canet M, Gallardo D, Arenillas L, Esteve J, Baragay J, Salamero O, Motlló C, Ortín X, Sierra J, Solé F. Genetic characterization of acute myeloid leukemia patients with mutations in IDH1/2 genes. Leuk Res 2021; 101:106492. [PMID: 33494038 DOI: 10.1016/j.leukres.2020.106492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 11/16/2022]
Affiliation(s)
- Pamela Acha
- MDS Research Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Montserrat Hoyos
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marta Pratcorona
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francisco Fuster-Tormo
- MDS Research Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Laura Palomo
- MDS Research Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Esther Ortega
- MDS Research Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Lurdes Zamora
- Hematology Service, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Institut de Recerca Contra la Leucèmia Josep Carreras, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Susana Vives
- Hematology Service, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Institut de Recerca Contra la Leucèmia Josep Carreras, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Isabel Granada
- Hematology Service, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Institut de Recerca Contra la Leucèmia Josep Carreras, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Julia Montoro
- Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Antoni Garcia
- Hematology Service, Hospital Universitari Arnau Vilanova, Lleida, Spain
| | - Montserrat Arnan
- Hematology Service, Institut Català d'Oncologia-Hospital Duran i Reynals, Barcelona, Spain
| | - Marta Cervera
- Hematology Service, Hospital Universitari Joan XXIII de Tarragona, Tarragona, Spain
| | - Marta Canet
- Hematology Service, Hospital Universitari Mútua Terrassa, Terrassa, Spain
| | - David Gallardo
- Institut Català d'Oncologia-Hospital Dr. Josep trueta, Girona, Spain
| | | | - Jordi Esteve
- Hematology Service, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Joan Baragay
- Hematology Service, Hospital Universitario Son Llàtzer, Palma de Mallorca, Spain
| | - Olga Salamero
- Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | - Xavier Ortín
- Hematology Service, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Jordi Sierra
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francesc Solé
- MDS Research Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain.
| |
Collapse
|
66
|
Banskota SU, Khanal N, Bhatt VR. A precision medicine approach to management of acute myeloid leukemia in older adults. Curr Opin Oncol 2020; 32:650-655. [PMID: 32826488 PMCID: PMC7737662 DOI: 10.1097/cco.0000000000000673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Therapy selection in older adults with acute myeloid leukemia (AML) can be challenging because of a higher incidence of high-risk cytogenetic and molecular features conferring chemoresistance and poor functional status leading to increased treatment-related toxicities. The purpose of this review is to highlight the recent advances in precision medicine in AML that have shown promise to improve outcomes of older adults. RECENT FINDINGS The utilization of next generation sequencing to identify and target actionable mutations can influence therapy selection in one-third of patients and can result in higher response rates as well as survival compared with those who do not receive targeted therapy. Oral targeted agents are available for AML with IDH 1, IDH2, or FLT3 mutations. Low-intensity venetoclax-based regimens have shown high rates of responses in AML, particularly among those with NPM1 and IDH2 mutations; responses are often durable and associated with minimal residual disease (MRD) negativity. Multiple studies have demonstrated the prognostic significance of flow cytometric MRD, with potential implications for subsequent therapy. SUMMARY Novel approaches for AML risk-stratification, MRD assessment, and a precision medicine approach offer significant promise to improve survival and quality of life of older adults.
Collapse
Affiliation(s)
| | - Nabin Khanal
- Franciscan Physician Network Oncology & Hematology Specialists, St Francis hospital, Indianapolis, IN
| | - Vijaya Raj Bhatt
- Department of Internal Medicine, Division of Hematology-Oncology, University of Nebraska Medical Center, Omaha, NE
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
67
|
Carretta C, Mallia S, Genovese E, Parenti S, Rontauroli S, Bianchi E, Fantini S, Sartini S, Tavernari L, Tagliafico E, Manfredini R. Genomic Analysis of Hematopoietic Stem Cell at the Single-Cell Level: Optimization of Cell Fixation and Whole Genome Amplification (WGA) Protocol. Int J Mol Sci 2020; 21:E7366. [PMID: 33036143 PMCID: PMC7582552 DOI: 10.3390/ijms21197366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/15/2022] Open
Abstract
Single-cell genomics has become the method of choice for the study of heterogeneous cell populations and represents an elective application in defining the architecture and clonal evolution in hematological neoplasms. Reconstructing the clonal evolution of a neoplastic population therefore represents the main way to understand more deeply the pathogenesis of the neoplasm, but it is also a potential tool to understand the evolution of the tumor population with respect to its response to therapy. Pre-analytical phase for single-cell genomics analysis is crucial to obtain a cell population suitable for single-cell sorting, and whole genome amplification is required to obtain the necessary amount of DNA from a single cell in order to proceed with sequencing. Here, we evaluated the impact of different methods of cellular immunostaining, fixation and whole genome amplification on the efficiency and yield of single-cell sequencing.
Collapse
Affiliation(s)
- Chiara Carretta
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Selene Mallia
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Elena Genovese
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Sandra Parenti
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Elisa Bianchi
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Sebastian Fantini
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Stefano Sartini
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Lara Tavernari
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.C.); (S.M.); (E.G.); (S.P.); (S.R.); (E.B.); (S.F.); (S.S.); (L.T.)
| |
Collapse
|
68
|
Testa U, Castelli G, Pelosi E. Isocitrate Dehydrogenase Mutations in Myelodysplastic Syndromes and in Acute Myeloid Leukemias. Cancers (Basel) 2020; 12:E2427. [PMID: 32859092 PMCID: PMC7564409 DOI: 10.3390/cancers12092427] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease generated by the acquisition of multiple genetic and epigenetic aberrations which impair the proliferation and differentiation of hematopoietic progenitors and precursors. In the last years, there has been a dramatic improvement in the understanding of the molecular alterations driving cellular signaling and biochemical changes determining the survival advantage, stimulation of proliferation, and impairment of cellular differentiation of leukemic cells. These molecular alterations influence clinical outcomes and provide potential targets for drug development. Among these alterations, an important role is played by two mutant enzymes of the citric acid cycle, isocitrate dehydrogenase (IDH), IDH1 and IDH2, occurring in about 20% of AMLs, which leads to the production of an oncogenic metabolite R-2-hydroxy-glutarate (R-2-HG); this causes a DNA hypermethylation and an inhibition of hematopoietic stem cell differentiation. IDH mutations differentially affect prognosis of AML patients following the location of the mutation and other co-occurring genomic abnormalities. Recently, the development of novel therapies based on the specific targeting of mutant IDH may contribute to new effective treatments of these patients. In this review, we will provide a detailed analysis of the biological, clinical, and therapeutic implications of IDH mutations.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (G.C.); (E.P.)
| | | | | |
Collapse
|
69
|
Yu X, Li H, Hu P, Qing Y, Wang X, Zhu M, Wang H, Wang Z, Xu J, Guo Q, Hui H. Natural HDAC-1/8 inhibitor baicalein exerts therapeutic effect in CBF-AML. Clin Transl Med 2020; 10:e154. [PMID: 32898337 PMCID: PMC7449246 DOI: 10.1002/ctm2.154] [Citation(s) in RCA: 14] [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: 04/23/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Although targeting histone deacetylases (HDACs) may be an effective strategy for core binding factor-acute myeloid leukemia (CBF-AML) harboring t(8;21) or inv(16), HDAC inhibitors are reported to be limited by drug-resistant characteristic. Our purpose is to evaluate the anti-leukemia effects of Baicalein on CBF-AML and clarify its underlying mechanism. METHODS Enzyme activity assay was used to measure the activity inhibition of HDACs. Rhodamine123 and RT-qPCR were employed to evaluate the distribution of drugs and the change of ATP-binding cassette (ABC) transporter genes. CCK8, Annexin V/PI, and FACS staining certified the effects of Baicalein on cell growth, apoptosis, and differentiation. Duolink and IP assay assessed the interaction between HDAC-1 and ubiquitin, HSP90 and AML1-ETO, and Ac-p53 and CBFβ-MYH11. AML cell lines and primary AML cells-bearing NOD/SCID mice models were used to evaluate the anti-leukemic efficiency and potential mechanism of Baicalein in vivo. RESULTS Baicalein showed HDAC-1/8 inhibition to trigger growth suppression and differentiation induction of AML cell lines and primary AML cells. Although the inhibitory action on HDAC-1 was mild, Baicalein could induce the degradation of HDAC-1 via ubiquitin proteasome pathway, thereby upregulating the acetylation of Histone H3 without promoting ABC transporter genes expression. Meanwhile, Baicalein increased the acetylation of HSP90 and lessened its connection to AML1/ETO, consequently leading to degradation of AML1-ETO in t(8;21)q(22;22) AML cells. In inv(16) AML cells, Baicalein possessed the capacity of apoptosis induction accompanied with p53-mediated apoptosis genes expression. Moreover, CBFβ-MYH11-bound p53 acetylation was restored via HDAC-8 inhibition induced by Baicalein contributing the diminishing of survival of CD34+ inv(16) AML cells. CONCLUSIONS These findings improved the understanding of the epigenetic regulation of Baicalein, and warrant therapeutic potential of Baicalein for CBF-AML.
Collapse
Affiliation(s)
- Xiaoxuan Yu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
- Department of PharmacologySchool of medicine & Holostic integrative medicineNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Hui Li
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Po Hu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Yingjie Qing
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Xiangyuan Wang
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Mengyuan Zhu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Hongzheng Wang
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Zhanyu Wang
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Jingyan Xu
- Department of HematologyThe Affiliated DrumTower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Qinglong Guo
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Hui Hui
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionKey Laboratory of Drug Quality Control and PharmacovigilanceMinistry of EducationJiangsu Key Laboratory of Drug Design and OptimizationChina Pharmaceutical UniversityChina Pharmaceutical UniversityNanjingJiangsuChina
| |
Collapse
|
70
|
Aasebø E, Berven FS, Hovland R, Døskeland SO, Bruserud Ø, Selheim F, Hernandez-Valladares M. The Progression of Acute Myeloid Leukemia from First Diagnosis to Chemoresistant Relapse: A Comparison of Proteomic and Phosphoproteomic Profiles. Cancers (Basel) 2020; 12:cancers12061466. [PMID: 32512867 PMCID: PMC7352627 DOI: 10.3390/cancers12061466] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy. Nearly 50% of the patients who receive the most intensive treatment develop chemoresistant leukemia relapse. Although the leukemogenic events leading to relapse seem to differ between patients (i.e., regrowth from a clone detected at first diagnosis, progression from the original leukemic or preleukemic stem cells), a common characteristic of relapsed AML is increased chemoresistance. The aim of the present study was to investigate at the proteomic level whether leukemic cells from relapsed patients present overlapping molecular mechanisms that contribute to this chemoresistance. We used liquid chromatography–tandem mass spectrometry (LC–MS/MS) to compare the proteomic and phosphoproteomic profiles of AML cells derived from seven patients at the time of first diagnosis and at first relapse. At the time of first relapse, AML cells were characterized by increased levels of proteins important for various mitochondrial functions, such as mitochondrial ribosomal subunit proteins (MRPL21, MRPS37) and proteins for RNA processing (DHX37, RNA helicase; RPP40, ribonuclease P component), DNA repair (ERCC3, DNA repair factor IIH helicase; GTF2F1, general transcription factor), and cyclin-dependent kinase (CDK) activity. The levels of several cytoskeletal proteins (MYH14/MYL6/MYL12A, myosin chains; VCL, vinculin) as well as of proteins involved in vesicular trafficking/secretion and cell adhesion (ITGAX, integrin alpha-X; CD36, platelet glycoprotein 4; SLC2A3, solute carrier family 2) were decreased in relapsed cells. Our study introduces new targetable proteins that might direct therapeutic strategies to decrease chemoresistance in relapsed AML.
Collapse
Affiliation(s)
- Elise Aasebø
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (Ø.B.)
- The Department of Biomedicine, The Proteomics Unit at the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
| | - Frode S. Berven
- The Department of Biomedicine, The Proteomics Unit at the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- The Department of Biomedicine, University of Bergen, 5009 Bergen, Norway;
| | - Randi Hovland
- Department for Medical Genetics, Haukeland University Hospital, 5021 Bergen, Norway;
- Department of Biological Sciences, University of Bergen, 5006 Bergen, Norway
| | | | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (Ø.B.)
| | - Frode Selheim
- The Department of Biomedicine, The Proteomics Unit at the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- The Department of Biomedicine, University of Bergen, 5009 Bergen, Norway;
| | - Maria Hernandez-Valladares
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (Ø.B.)
- The Department of Biomedicine, The Proteomics Unit at the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- Correspondence: ; Tel.: +47-5558-6368
| |
Collapse
|