1
|
Lecornec N, Duchmann M, Itzykson R. Single-cell sequencing applications in acute myeloid leukemia. Leuk Lymphoma 2025; 66:175-189. [PMID: 39496597 DOI: 10.1080/10428194.2024.2422833] [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: 07/23/2024] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of malignancies with poor prognosis. AML result from the proliferation of immature myeloid cells blocked at a variable stage of differentiation. Beyond inter-patient heterogeneity, AMLs are characterized by genetic and phenotypic intra-patient heterogeneity. Despite major advances in deciphering AML biology with bulk sequencing studies, pivotal questions remain unanswered. Analyses at the single-cell level could thus transform our understanding of these neoplasms. We review recent progresses in single-cell sequencing technologies from cell processing to bioinformatic pipelines. We next discuss how single-cell applications have helped understand the genetic and functional intra-leukemic heterogeneity, emphasizing aspects related to leukemic stem cells, clonal evolution and measurable residual disease (MRD) monitoring. We finally delineate how single-cell technologies could be implemented in routine clinical practice to improve patient management.
Collapse
Affiliation(s)
- Nicolas Lecornec
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Département d'Immuno-Hématologie Pédiatrique, Hôpital Robert-Debré, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Matthieu Duchmann
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Raphael Itzykson
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
2
|
Xie SZ. IRE1α-XBP1 moonlighting to restrict leukemia. Nat Immunol 2025:10.1038/s41590-024-02061-y. [PMID: 39825092 DOI: 10.1038/s41590-024-02061-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Affiliation(s)
- Stephanie Z Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Parry TL, Gilmore LA, Khamoui AV. Pan-cancer secreted proteome and skeletal muscle regulation: insight from a proteogenomic data-driven knowledge base. Funct Integr Genomics 2025; 25:14. [PMID: 39812750 DOI: 10.1007/s10142-024-01524-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025]
Abstract
Large-scale, pan-cancer analysis is enabled by data driven knowledge bases that link tumor molecular profiles with phenotypes. A debilitating cancer-related phenotype is skeletal muscle loss, or cachexia, which occurs partly from tumor products secreted into circulation. Using the LinkedOmicsKB knowledge base assembled from the Clinical Proteomics Tumor Analysis Consortium proteogenomic analysis, along with catalogs of human secretome proteins, ligand-receptor pairs and molecular signatures, we sought to identify candidate pan-cancer proteins secreted to blood that could regulate skeletal muscle phenotypes in multiple solid cancers. Tumor proteins having significant pan-cancer associations with muscle were referenced against secretome proteins secreted to blood from the Human Protein Atlas, then verified as increased in paired tumor vs. normal tissues in pan-cancer manner. This workflow revealed seven secreted proteins from cancers afflicting kidneys, head and neck, lungs and pancreas that classified as protein-binding activity modulator, extracellular matrix protein or intercellular signaling molecule. Concordance of these biomarkers with validated molecular signatures of cachexia and senescence supported relevance to muscle and cachexia disease biology, and high tumor expression of the biomarker set associated with lower overall survival. In this article, we discuss avenues by which skeletal muscle and cachexia may be regulated by these candidate pan-cancer proteins secreted to blood, and conceptualize a strategy that considers them collectively as a biomarker signature with potential for refinement by data analytics and radiogenomics for predictive testing of future risk in a non-invasive, blood-based panel amenable to broad uptake and early management.
Collapse
Affiliation(s)
- Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - L Anne Gilmore
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andy V Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA.
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL, USA.
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA.
| |
Collapse
|
4
|
Melo Garcia L, Gangadharan A, Banerjee P, Li Y, Zeng AGX, Rafei H, Lin P, Kumar B, Acharya S, Daher M, Muniz-Feliciano L, Deyter GM, Dominguez G, Park JM, Reyes Silva F, Nunez Cortes AK, Basar R, Uprety N, Shanley M, Kaplan M, Liu E, Shpall EJ, Rezvani K. Overcoming CD226-related immune evasion in acute myeloid leukemia with CD38 CAR-engineered NK cells. Cell Rep 2025; 44:115122. [PMID: 39754720 DOI: 10.1016/j.celrep.2024.115122] [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: 05/29/2024] [Revised: 10/26/2024] [Accepted: 12/06/2024] [Indexed: 01/06/2025] Open
Abstract
CD226 plays a vital role in natural killer (NK) cell cytotoxicity, interacting with its ligands CD112 and CD155 to initiate immune synapse formation, primarily through leukocyte function-associated-1 (LFA-1). Our study examined the role of CD226 in NK cell surveillance of acute myeloid leukemia (AML). NK cells in patients with AML had lower expression of CD226. CRISPR-Cas9 deletion of CD226 led to reduced LFA-1 recruitment, poor synapse formation, and decreased NK cell anti-leukemic activity. Engineering NK cells to express a chimeric antigen receptor targeting the AML antigen CD38 (CAR38) could overcome the need for CD226 to establish strong immune synapses. LFA-1 blockade reduced CAR38 NK cell activity, and this depended on the CD38 expression levels of AML cells. This suggests parallel but potentially cooperative roles for LFA-1 and CAR38 in synapse formation. Our findings suggest that CAR38 NK cells could be an effective therapeutic strategy to overcome CD226-mediated immune evasion in AML.
Collapse
Affiliation(s)
- Luciana Melo Garcia
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada; Hematology-Oncology Service, CHU de Québec - Université Laval, Quebec City, QC G1V 0A6, Canada
| | - Achintyan Gangadharan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Cancer Biology PhD Program, University of South Florida, Tampa, FL 33616, USA
| | - Pinaki Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ye Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andy G X Zeng
- Princess Margaret Cancer Center, University Healthy Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hind Rafei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bijender Kumar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sunil Acharya
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Luis Muniz-Feliciano
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gary M Deyter
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel Dominguez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeong Min Park
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Francia Reyes Silva
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ana Karen Nunez Cortes
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nadima Uprety
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mayra Shanley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mecit Kaplan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Enli Liu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
5
|
Gamma JM, Liu Q, Beauchamp E, Iyer A, Yap MC, Zak Z, Ekstrom C, Pain R, Kostiuk MA, Mackey JR, Brandwein J, Wang JCY, Berthiaume LG. Zelenirstat Inhibits N-Myristoyltransferases to Disrupt Src Family Kinase Signaling and Oxidative Phosphorylation, Killing Acute Myeloid Leukemia Cells. Mol Cancer Ther 2025; 24:69-80. [PMID: 39382188 DOI: 10.1158/1535-7163.mct-24-0307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/14/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024]
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy with limited treatment options and a high likelihood of recurrence after chemotherapy. We studied N-myristoylation, the myristate modification of proteins linked to survival signaling and metabolism, as a potential therapeutic target for AML. N-myristoylation is catalyzed by two N-myristoyltransferases (NMT), NMT1 and NMT2, with varying expressions in AML cell lines and patient samples. We identified NMT2 expression as a marker for survival of patients with AML, and low NMT2 expression was associated with poor outcomes. We used the first-in-class pan-NMT inhibitor, zelenirstat, to investigate the role of N-myristoylation in AML. Zelenirstat effectively inhibits myristoylation in AML cell lines and patient samples, leading to degradation of Src family kinases, induction of endoplasmic reticulum stress, apoptosis, and cell death. Zelenirstat was well tolerated in vivo and reduced the leukemic burden in an ectopic AML cell line and in multiple orthotopic AML patient-derived xenograft models. The leukemia stem cell-enriched fractions of the hierarchical OCI-AML22 model were highly sensitive to myristoylation inhibition. Zelenirstat also impairs mitochondrial complex I and oxidative phosphorylation, which are critical for leukemia stem cell survival. These findings suggest that targeting N-myristoylation with zelenirstat represents a novel therapeutic approach for AML, with promise in patients with currently poor outcomes.
Collapse
Affiliation(s)
- Jay M Gamma
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Qiang Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | | | - Aishwarya Iyer
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Megan C Yap
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Zoulika Zak
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Cassidy Ekstrom
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Rony Pain
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Morris A Kostiuk
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | | | - Joseph Brandwein
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, Canada
| | - Luc G Berthiaume
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
- Pacylex Pharmaceuticals Inc., Edmonton, Canada
| |
Collapse
|
6
|
Gower M, Li X, Aguilar-Navarro AG, Lin B, Fernandez M, Edun G, Nader M, Rondeau V, Arruda A, Tierens A, Eames Seffernick A, Pölönen P, Durocher J, Wagenblast E, Yang L, Lee HS, Mullighan CG, Teachey D, Rashkovan M, Tremblay CS, Herranz D, Itkin T, Loghavi S, Dick JE, Schwartz G, Perusini MA, Sibai H, Hitzler J, Gruber TA, Minden M, Jones CL, Dolgalev I, Jahangiri S, Tikhonova AN. An inflammatory state defines a high-risk T-lineage acute lymphoblastic leukemia subgroup. Sci Transl Med 2025; 17:eadr2012. [PMID: 39742502 DOI: 10.1126/scitranslmed.adr2012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/04/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
T-lineage acute lymphoblastic leukemia (ALL) is an aggressive cancer comprising diverse subtypes that are challenging to stratify using conventional immunophenotyping. To gain insights into subset-specific therapeutic vulnerabilities, we performed an integrative multiomics analysis of bone marrow samples from newly diagnosed T cell ALL, early T cell precursor ALL, and T/myeloid mixed phenotype acute leukemia. Leveraging cellular indexing of transcriptomes and epitopes in conjunction with T cell receptor sequencing, we identified a subset of patient samples characterized by activation of inflammatory and stem gene programs. These inflammatory T-lineage samples exhibited distinct biological features compared with other T-lineage ALL samples, including the production of proinflammatory cytokines, prevalence of mutations affecting cytokine signaling and chromatin remodeling, an altered immune microenvironment, and poor treatment responses. Moreover, we found that, although inflammatory T-lineage ALL samples were less sensitive to dexamethasone, they exhibited unique sensitivity to a BCL-2 inhibitor, venetoclax. To facilitate classification of patients with T-lineage ALL, we developed a computational inflammatory gene signature scoring system, which stratified patients and was associated with disease prognosis in three additional patient cohorts. By identifying a high-risk T-lineage ALL subtype on the basis of an inflammatory score, our study provides a framework for targeted therapeutic approaches for these challenging-to-treat cancers.
Collapse
Affiliation(s)
- Mark Gower
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Ximing Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | | | - Brian Lin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Minerva Fernandez
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Gibran Edun
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Mursal Nader
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Vincent Rondeau
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Laboratory Medicine Program, University Health Network and University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Anne Tierens
- Laboratory Medicine Program, University Health Network and University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Anna Eames Seffernick
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Petri Pölönen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Juliette Durocher
- CHU Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
- Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Elvin Wagenblast
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029-5674, USA
| | - Lin Yang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Ho Seok Lee
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - David Teachey
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marissa Rashkovan
- CHU Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
- Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Cedric S Tremblay
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Daniel Herranz
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Tomer Itkin
- Sagol Center for Regenerative Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Neufeld Cardiovascular Research Institute, Department of Pathology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 5262179, Israel
- Tamman Cardiovascular Research Institute, Lev Leviev Cardiothoracic and Vascular Center, Sheba Medical Center, Ramat Gan 5262100, Israel
| | - Sanam Loghavi
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gregory Schwartz
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
- Canada Vector Institute, Toronto, ON M5G 1M1, Canada
| | - Maria Agustina Perusini
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON M5B 1W8, Canada
| | - Hassan Sibai
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON M5B 1W8, Canada
| | - Johann Hitzler
- Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Pediatrics, University of Toronto, ON M5G 1X8, Canada
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Tanja A Gruber
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305-5101, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON M5B 1W8, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Courtney L Jones
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Igor Dolgalev
- Cellular Analytics Laboratory, NYU Grossman School of Medicine, New York, NY 10016, USA
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Soheil Jahangiri
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Anastasia N Tikhonova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
7
|
Walter W, Iacobucci I, Meggendorfer M. Diagnosis of acute lymphoblastic leukaemia: an overview of the current genomic classification, diagnostic approaches, and future directions. Histopathology 2025; 86:134-145. [PMID: 39403021 DOI: 10.1111/his.15338] [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] [Indexed: 12/17/2024]
Abstract
B-acute lymphoblastic leukaemia (B-ALL) is a haematological disease resulting from haematopoietic system dysfunction, leading to the unchecked growth of immature B lymphoblasts. The disease's complexity is underscored by the spectrum of genetic aberrations that underlie B-ALL entities, necessitating advanced genetic analyses for precise classification and risk determination. Prior to the adoption of next-generation sequencing into standard diagnostic practices, up to 30% of B-ALL cases were not assigned to specific entities due to the limitations of traditional diagnostic methods. The advent of comprehensive genomic analysis, especially whole-genome transcriptome sequencing, has significantly enhanced our understanding of B-ALL's molecular heterogeneity, paving the way for the exploration of novel, tailored treatment strategies. Furthermore, recent technological innovations, such as optical genome mapping, methylation profiling, and single-cell sequencing, have propelled forward the fields of cancer research and B-ALL management. These innovations introduce novel diagnostic approaches and prognostic markers, facilitating a deeper, more nuanced understanding of individual patient disease profiles. This review focuses on the latest diagnostic standards and assays for B-ALL, the importance of new technologies and biomarkers in enhancing diagnostic accuracy, and the expected role of innovative advancements in the future diagnosis and treatment of B-ALL.
Collapse
Affiliation(s)
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
8
|
Fleming TJ, Antoszewski M, Lambo S, Gundry MC, Piussi R, Wahlster L, Shah S, Reed FE, Dong KD, Paulo JA, Gygi SP, Mimoso C, Goldman SR, Adelman K, Perry JA, Pikman Y, Stegmaier K, Barrachina MN, Machlus KR, Hovestadt V, Arruda A, Minden MD, Voit RA, Sankaran VG. CEBPA repression by MECOM blocks differentiation to drive aggressive leukemias. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.30.630680. [PMID: 39803492 PMCID: PMC11722404 DOI: 10.1101/2024.12.30.630680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Acute myeloid leukemias (AMLs) have an overall poor prognosis with many high-risk cases co-opting stem cell gene regulatory programs, yet the mechanisms through which this occurs remain poorly understood. Increased expression of the stem cell transcription factor, MECOM, underlies one key driver mechanism in largely incurable AMLs. How MECOM results in such aggressive AML phenotypes remains unknown. To address existing experimental limitations, we engineered and applied targeted protein degradation with functional genomic readouts to demonstrate that MECOM promotes malignant stem cell-like states by directly repressing pro-differentiation gene regulatory programs. Remarkably and unexpectedly, a single node in this network, a MECOM-bound cis-regulatory element located 42 kb downstream of the myeloid differentiation regulator CEBPA, is both necessary and sufficient for maintaining MECOM-driven leukemias. Importantly, targeted activation of this regulatory element promotes differentiation of these aggressive AMLs and reduces leukemia burden in vivo, suggesting a broadly applicable differentiation-based approach for improving therapy.
Collapse
Affiliation(s)
- Travis J. Fleming
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mateusz Antoszewski
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- These authors contributed equally to this work
| | - Sander Lambo
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- These authors contributed equally to this work
| | - Michael C. Gundry
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Riccardo Piussi
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lara Wahlster
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sanjana Shah
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Fiona E. Reed
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kevin D. Dong
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Claudia Mimoso
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Seth R. Goldman
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Karen Adelman
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jennifer A. Perry
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Yana Pikman
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kimberly Stegmaier
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Maria N. Barrachina
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kellie R. Machlus
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Volker Hovestadt
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Mark D. Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Richard A. Voit
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Present Address: UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vijay G. Sankaran
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA 02142, USA
- Lead contact
| |
Collapse
|
9
|
Sango J, Carcamo S, Sirenko M, Maiti A, Mansour H, Ulukaya G, Tomalin LE, Cruz-Rodriguez N, Wang T, Olszewska M, Olivier E, Jaud M, Nadorp B, Kroger B, Hu F, Silverman L, Chung SS, Wagenblast E, Chaligne R, Eisfeld AK, Demircioglu D, Landau DA, Lito P, Papaemmanuil E, DiNardo CD, Hasson D, Konopleva M, Papapetrou EP. RAS-mutant leukaemia stem cells drive clinical resistance to venetoclax. Nature 2024; 636:241-250. [PMID: 39478230 PMCID: PMC11618090 DOI: 10.1038/s41586-024-08137-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/30/2024] [Indexed: 12/06/2024]
Abstract
Cancer driver mutations often show distinct temporal acquisition patterns, but the biological basis for this, if any, remains unknown. RAS mutations occur invariably late in the course of acute myeloid leukaemia, upon progression or relapsed/refractory disease1-6. Here, by using human leukaemogenesis models, we first show that RAS mutations are obligatory late events that need to succeed earlier cooperating mutations. We provide the mechanistic explanation for this in a requirement for mutant RAS to specifically transform committed progenitors of the myelomonocytic lineage (granulocyte-monocyte progenitors) harbouring previously acquired driver mutations, showing that advanced leukaemic clones can originate from a different cell type in the haematopoietic hierarchy than ancestral clones. Furthermore, we demonstrate that RAS-mutant leukaemia stem cells (LSCs) give rise to monocytic disease, as observed frequently in patients with poor responses to treatment with the BCL2 inhibitor venetoclax. We show that this is because RAS-mutant LSCs, in contrast to RAS-wild-type LSCs, have altered BCL2 family gene expression and are resistant to venetoclax, driving clinical resistance and relapse with monocytic features. Our findings demonstrate that a specific genetic driver shapes the non-genetic cellular hierarchy of acute myeloid leukaemia by imposing a specific LSC target cell restriction and critically affects therapeutic outcomes in patients.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Cell Lineage/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Monocytes/metabolism
- Monocytes/drug effects
- Mutation
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- ras Proteins/metabolism
- ras Proteins/genetics
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Granulocytes
- Clone Cells/metabolism
- Clone Cells/pathology
- Stem Cells/metabolism
- Stem Cells/pathology
- Recurrence
Collapse
Affiliation(s)
- Junya Sango
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saul Carcamo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Sirenko
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hager Mansour
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gulay Ulukaya
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lewis E Tomalin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nataly Cruz-Rodriguez
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tiansu Wang
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Malgorzata Olszewska
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emmanuel Olivier
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manon Jaud
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bettina Nadorp
- Department of Medicine, Division of Precision Medicine, NYU Grossman School of Medicine, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Benjamin Kroger
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Medical Scientist Training Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Feng Hu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lewis Silverman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen S Chung
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elvin Wagenblast
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronan Chaligne
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Ann-Kathrin Eisfeld
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Deniz Demircioglu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan A Landau
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Piro Lito
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan Hasson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Konopleva
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Patel SB, Moskop DR, Jordan CT, Pietras EM. Understanding MDS stem cells: Advances and limitations. Semin Hematol 2024; 61:409-419. [PMID: 39472255 DOI: 10.1053/j.seminhematol.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/10/2024]
Abstract
In work spanning several decades, extensive studies have focused on the properties of malignant stem cells that drive the pathogenesis of acute myeloid leukemia (AML). However, relatively little attention has been devoted to several serious myeloid malignancies that occur prior to the onset of frank leukemia, including myelodysplastic syndrome (MDS). Like leukemia, MDS is hypothesized to arise from a pool of immature malignant stem and progenitor cells (MDS-SCs) that serve as a reservoir for disease evolution and progression1. While multiple studies have sought to identify and characterize the biology and vulnerabilities of MDS-SCs, yet translation of scientific concepts to therapeutically impactful regimens has been limited. Here, we evaluate the currently known properties of MDS-SCs as well as the post-transcriptional mechanisms that drive MDS pathogenesis at a stem and progenitor level. We highlight limits and gaps in our characterization and understanding of MDS-SCs and address the extent to which the properties of MDS-SC are (and can be) inferred from the characterization of LSCs.
Collapse
Affiliation(s)
- Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Daniel R Moskop
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Craig T Jordan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| |
Collapse
|
11
|
Li T, Gui X, Li B, Hu X, Wang Y. LSP1 promotes the progression of acute myelogenous leukemia by regulating KSR/ERK signaling pathway and cell migration. Hematology 2024; 29:2330285. [PMID: 38511641 DOI: 10.1080/16078454.2024.2330285] [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: 06/03/2023] [Accepted: 03/10/2024] [Indexed: 03/22/2024] Open
Abstract
We aimed to investigate the role and mechanism of LSP1 in the progression of acute myelogenous leukemia. In this study, we established shLSP1 cell line to analyze the function of LSP1 in AML. We observed high expression of LSP1 in AML patients, whereas it showed no expression in normal adults. Furthermore, we found that LSP1 expression was associated with disease prognosis. Our results indicate that LSP1 plays a crucial role in mediating proliferation and survival of leukemia cells through the KSR/ERK signaling pathway. Additionally, LSP1 promotes cell chemotaxis and homing by enhancing cell adhesion and migration. We also discovered that LSP1 confers chemotactic ability to leukemia cells in vivo. Finally, our study identified 12 genes related to LSP1 in AML, which indicated poor survival outcome in AML patients and were enriched in Ras and cell adhesion signaling pathways. Our results revealed that the overexpression of LSP1 is related to the activation of the KSR/ERK signaling pathway, as well as cell adhesion and migration in AML patients. Reducing LSP1 expression impair AML progression, suggesting that LSP1 may serve as a potential drug therapy target for more effective treatment of AML.
Collapse
Affiliation(s)
- Tan Li
- Department of Hematology, Hefei City First People's Hospital, Hefei, People's Republic of China
| | - Xiaochen Gui
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, People's Republic of China
| | - Bin Li
- Department of Hematology, Hefei City First People's Hospital, Hefei, People's Republic of China
| | - Xueying Hu
- Department of Hematology, Hefei City First People's Hospital, Hefei, People's Republic of China
| | - Yuanyin Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, People's Republic of China
| |
Collapse
|
12
|
Zhang X, Liu L. Senescent T Cells: The Silent Culprit in Acute Myeloid Leukemia Progression? Int J Mol Sci 2024; 25:12550. [PMID: 39684260 DOI: 10.3390/ijms252312550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Malignant tumors can evade immune surveillance and elimination through multiple mechanisms, with the induction of immune cell dysfunction serving as a crucial strategy. Mounting evidence indicates that T cell senescence constitutes the primary mechanism underlying T cell dysfunction in acute myeloid leukemia (AML) and represents one of the potential causes of immunotherapy failure. AML usually progresses rapidly and is highly susceptible to drug resistance, thereby resulting in recurrence and patient mortality. Hence, disrupting the immune interface within the bone marrow microenvironment of AML has emerged as a critical objective for synergistically enhancing tumor immunotherapy. In this review, we summarize the general characteristics, distinctive phenotypes, and regulatory signaling networks of senescent T cells and highlight their potential clinical significance in the bone marrow microenvironment of AML. Additionally, we discuss potential therapeutic strategies for alleviating and reversing T cell senescence.
Collapse
Affiliation(s)
- Xiaolan Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
13
|
Bonilla G, Morris A, Kundu S, DuCasse A, Kirkpatrick G, Jeffries NE, Chetal K, Yvanovich EE, Milosevic J, Zhao T, Xia J, Barghout R, Scadden D, Mansour MK, Kingston RE, Sykes DB, Mercier FE, Sadreyev RI. Leukemia aggressiveness is driven by chromatin remodeling and expression changes of core regulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582846. [PMID: 38496490 PMCID: PMC10942317 DOI: 10.1101/2024.02.29.582846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Molecular mechanisms driving clonal aggressiveness in leukemia are not fully understood. We tracked and analyzed MLL-rearranged leukemic clones independently evolving towards higher aggressiveness. More aggressive subclones lost their growth differential ex vivo but restored it upon secondary transplantation, suggesting molecular memory of aggressiveness. Development of aggressiveness was associated with clone-specific gradual modulation of chromatin states and expression levels across the genome, with a surprising preferential trend of reversing the earlier changes between normal and leukemic progenitors. To focus on the core aggressiveness program, we identified genes with consistent changes of expression and chromatin marks that were maintained in vivo and ex vivo in both clones. Overexpressing selected core genes (Smad1 as aggressiveness driver, Irx5 and Plag1 as suppressors) affected leukemic progenitor growth in the predicted way and had convergent downstream effects on central transcription factors and repressive epigenetic modifiers, suggesting a broader regulatory network of leukemic aggressiveness.
Collapse
|
14
|
Hwang J, Kim HN, Kwon JA, Yoon SY, Jeon MJ, Yu ES, Kim DS, Choi CW, Yoon J. Impact of TP53 mutation on genetic and cellular hierarchy profile in complex karyotype AML/MDS with increased blasts. Blood Cancer J 2024; 14:201. [PMID: 39548077 PMCID: PMC11568287 DOI: 10.1038/s41408-024-01188-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Affiliation(s)
- Jinha Hwang
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Ha Nui Kim
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Jung Ah Kwon
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Soo-Young Yoon
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Min Ji Jeon
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - Eun Sang Yu
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - Dae Sik Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - Chul Won Choi
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - Jung Yoon
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul, South Korea.
| |
Collapse
|
15
|
Bowman RL, Dunbar AJ, Mishra T, Xiao W, Waarts MR, Maestre IF, Eisman SE, Cai L, Mowla S, Shah N, Youn A, Bennett L, Fontenard S, Gounder S, Gandhi A, Bowman M, O'Connor K, Zaroogian Z, Sánchez-Vela P, Martinez Benitez AR, Werewski M, Park Y, Csete IS, Krishnan A, Lee D, Boorady N, Potts CR, Jenkins MT, Cai SF, Carroll MP, Meyer SE, Miles LA, Ferrell PB, Trowbridge JJ, Levine RL. In vivo models of subclonal oncogenesis and dependency in hematopoietic malignancy. Cancer Cell 2024; 42:1955-1969.e7. [PMID: 39532065 PMCID: PMC11561369 DOI: 10.1016/j.ccell.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/20/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Cancer evolution is a multifaceted process leading to dysregulation of cellular expansion and differentiation through somatic mutations and epigenetic dysfunction. Clonal expansion and evolution is driven by cell-intrinsic and -extrinsic selective pressures, which can be captured with increasing resolution by single-cell and bulk DNA sequencing. Despite the extensive genomic alterations revealed in profiling studies, there remain limited experimental systems to model and perturb evolutionary processes. Here, we integrate multi-recombinase tools for reversible, sequential mutagenesis from premalignancy to leukemia. We demonstrate that inducible Flt3 mutations differentially cooperate with Dnmt3a, Idh2, and Npm1 mutant alleles, and that changing the order of mutations influences cellular and transcriptional landscapes. We next use a generalizable, reversible approach to demonstrate that mutation reversion results in rapid leukemic regression with distinct differentiation patterns depending upon co-occurring mutations. These studies provide a path to experimentally model sequential mutagenesis, investigate mechanisms of transformation and probe oncogenic dependency in disease evolution.
Collapse
Affiliation(s)
- Robert L Bowman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Andrew J Dunbar
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tanmay Mishra
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wenbin Xiao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael R Waarts
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Inés Fernández Maestre
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shira E Eisman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Louise Cai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shoron Mowla
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nisargbhai Shah
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Angela Youn
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura Bennett
- Department of Cell and Developmental Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Suean Fontenard
- Department of Cell and Developmental Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shreeya Gounder
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anushka Gandhi
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Bowman
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kavi O'Connor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zachary Zaroogian
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Pablo Sánchez-Vela
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anthony R Martinez Benitez
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Matthew Werewski
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Young Park
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isabelle S Csete
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Aishwarya Krishnan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Darren Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nayla Boorady
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chad R Potts
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37212 USA
| | - Matthew T Jenkins
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37212 USA
| | - Sheng F Cai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Martin P Carroll
- Department of Medicine, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara E Meyer
- Department of Cancer Biology, Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Linde A Miles
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - P Brent Ferrell
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37212 USA
| | | | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
16
|
Chen T, Zhang Y, Zhang D, Zhou H. Immune-based subgroups uncover diverse tumor immunogenicity and implications for prognosis and precision therapy in acute myeloid leukemia. Front Immunol 2024; 15:1451486. [PMID: 39582863 PMCID: PMC11581856 DOI: 10.3389/fimmu.2024.1451486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/15/2024] [Indexed: 11/26/2024] Open
Abstract
Background Although a considerable proportion of acute myeloid leukemia (AML) patients achieve remission through chemotherapy, relapse remains a recurring and significant event leading to treatment failure. This study aims to investigate the immune landscape in AML and its potential implications for prognosis and chemo-/immune-therapy. Methods Integrated analyses based on multiple sequencing datasets of AML were performed. Various algorithms estimated immune infiltration in AML samples. A subgroup prediction model was developed, and comprehensive bioinformatics and machine learning algorithms were applied to compare immune-based subgroups in relation to clinical features, mutational landscapes, immune characterizations, drug sensitivities, and cellular hierarchies at the single-cell level. Results Two immune-based AML subgroups, G1 and G2, were identified. G1 demonstrated higher immune infiltration, a more monocytic phenotype, increased proportions of monocytes/macrophages, and higher FLT3, DNMT3A, and NPM1 mutation frequencies. It was associated with a poorer prognosis, lower proportions of various immune cell types and a lower T cell infiltration score (TIS). AML T-cell-based immunotherapy target antigens, including CLEC12A, Folate receptor β, IL1RAP and TIM3, showed higher expression levels in G1, while CD117, CD244, CD96, WT and TERT exhibited higher expression levels in G2. G1 samples demonstrated higher sensitivity to elesclomol and panobinostat but increased resistance to venetoclax compared to G2 samples. Moreover, we observed a positive correlation between sample immune infiltration and sample resistance to elesclomol and panobinostat, whereas a negative correlation was found with venetoclax resistance. Conclusion Our study enriches the current AML risk stratification and provides guidance for precision medicine in AML.
Collapse
Affiliation(s)
| | | | | | - Hebing Zhou
- Department of Hematology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Warfvinge R, Geironson Ulfsson L, Dhapola P, Safi F, Sommarin M, Soneji S, Hjorth-Hansen H, Mustjoki S, Richter J, Thakur RK, Karlsson G. Single-cell multiomics analysis of chronic myeloid leukemia links cellular heterogeneity to therapy response. eLife 2024; 12:RP92074. [PMID: 39503729 PMCID: PMC11540304 DOI: 10.7554/elife.92074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
The advent of tyrosine kinase inhibitors (TKIs) as treatment of chronic myeloid leukemia (CML) is a paradigm in molecularly targeted cancer therapy. Nonetheless, TKI-insensitive leukemia stem cells (LSCs) persist in most patients even after years of treatment and are imperative for disease progression as well as recurrence during treatment-free remission (TFR). Here, we have generated high-resolution single-cell multiomics maps from CML patients at diagnosis, retrospectively stratified by BCR::ABL1IS (%) following 12 months of TKI therapy. Simultaneous measurement of global gene expression profiles together with >40 surface markers from the same cells revealed that each patient harbored a unique composition of stem and progenitor cells at diagnosis. The patients with treatment failure after 12 months of therapy had a markedly higher abundance of molecularly defined primitive cells at diagnosis compared to the optimal responders. The multiomic feature landscape enabled visualization of the primitive fraction as a mixture of molecularly distinct BCR::ABL1+ LSCs and BCR::ABL1-hematopoietic stem cells (HSCs) in variable ratio across patients, and guided their prospective isolation by a combination of CD26 and CD35 cell surface markers. We for the first time show that BCR::ABL1+ LSCs and BCR::ABL1- HSCs can be distinctly separated as CD26+CD35- and CD26-CD35+, respectively. In addition, we found the ratio of LSC/HSC to be higher in patients with prospective treatment failure compared to optimal responders, at diagnosis as well as following 3 months of TKI therapy. Collectively, this data builds a framework for understanding therapy response and adapting treatment by devising strategies to extinguish or suppress TKI-insensitive LSCs.
Collapse
Affiliation(s)
- Rebecca Warfvinge
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | | | - Parashar Dhapola
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Fatemeh Safi
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Mikael Sommarin
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Shamit Soneji
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Henrik Hjorth-Hansen
- Department of Hematology, St Olavs HospitalTrondheimNorway
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Satu Mustjoki
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of HelsinkiHelsinkiFinland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer CenterTrondheimNorway
- iCAN Digital Precision Cancer Medicine FlagshipHelsinkiFinland
| | - Johan Richter
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund UniversityLundSweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University HospitalLundSweden
| | - Ram Krishna Thakur
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| | - Göran Karlsson
- Division of Molecular Hematology, Lund Stem Cell Center, Lund UniversityLundSweden
| |
Collapse
|
18
|
Zeng AG, Iacobucci I, Shah S, Mitchell A, Wong G, Bansal S, Chen D, Gao Q, Kim H, Kennedy JA, Arruda A, Minden MD, Haferlach T, Mullighan CG, Dick JE. Single-cell transcriptional mapping reveals genetic and non-genetic determinants of aberrant differentiation in AML. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.26.573390. [PMID: 38234771 PMCID: PMC10793439 DOI: 10.1101/2023.12.26.573390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
In acute myeloid leukemia (AML), genetic mutations distort hematopoietic differentiation, resulting in the accumulation of leukemic blasts. Yet, it remains unclear how these mutations intersect with cellular origins and whether they converge upon similar differentiation patterns. Single-cell RNA sequencing (scRNA-seq) has enabled high-resolution mapping of the relationship between leukemia and normal cell states, yet this application is hampered by imprecise reference maps of normal hematopoiesis and small sample sizes among patient cohorts. As a first step we constructed a reference atlas of human bone marrow hematopoiesis from 263,519 single-cell transcriptomes spanning 55 cellular states, that was benchmarked against independent datasets of immunophenotypically pure hematopoietic stem and progenitor cells. Using this reference atlas, we mapped over 1.2 million single-cell transcriptomes spanning 318 AML, mixed phenotype acute leukemia (MPAL), and acute erythroid leukemia (AEL) samples. This large-scale analysis, together with systematic mapping of genotype-to-phenotype associations between driver mutations and differentiation landscapes, revealed convergence of diverse genetic alterations on twelve recurrent patterns of aberrant differentiation in AML. This included unconventional lymphoid and erythroid priming linked to RUNX1 and TP53 mutations, respectively. We also identified non-genetic determinants of AML differentiation such as two subgroups of KMT2A-rearranged AML that differ in the identity of their leukemic stem cells (LSCs), likely reflecting distinct cellular origins. Furthermore, distinct LSC-driven hierarchies can co-exist within individual patients, providing insights into AML evolution. Together, precise mapping of normal and malignant cell states provides a framework for advancing the study and disease classification of hematologic malignancies thereby informing therapy development.
Collapse
Affiliation(s)
- Andy G.X. Zeng
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto; Toronto, ON, Canada
| | - Ilaria Iacobucci
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sayyam Shah
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
| | - Amanda Mitchell
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
| | - Gordon Wong
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto; Toronto, ON, Canada
| | - Suraj Bansal
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
| | - David Chen
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
| | - Qingsong Gao
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Hyerin Kim
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto; Toronto, ON, Canada
| | - James A. Kennedy
- Division of Medical Oncology and Hematology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
| | - Mark D. Minden
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Medical Oncology and Hematology, University Health Network, Toronto, ON, Canada
| | | | - Charles G. Mullighan
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN, USA
- Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN
| | - John E. Dick
- Princess Margaret Cancer Centre, University Health Network; Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto; Toronto, ON, Canada
| |
Collapse
|
19
|
Penter L, Cieri N, Maurer K, Kwok M, Lyu H, Lu WS, Oliveira G, Gohil SH, Leshchiner I, Lareau CA, Ludwig LS, Neuberg DS, Kim HT, Li S, Bullinger L, Ritz J, Getz G, Garcia JS, Soiffer RJ, Livak KJ, Wu CJ. Tracking Rare Single Donor and Recipient Immune and Leukemia Cells after Allogeneic Hematopoietic Cell Transplantation Using Mitochondrial DNA Mutations. Blood Cancer Discov 2024; 5:442-459. [PMID: 39236287 PMCID: PMC11528187 DOI: 10.1158/2643-3230.bcd-23-0138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 06/30/2024] [Accepted: 09/03/2024] [Indexed: 09/07/2024] Open
Abstract
Combined tracking of clonal evolution and chimeric cell phenotypes could enable detection of the key cellular populations associated with response following therapy, including after allogeneic hematopoietic stem cell transplantation (HSCT). We demonstrate that mitochondrial DNA (mtDNA) mutations coevolve with somatic nuclear DNA mutations at relapse post-HSCT and provide a sensitive means to monitor these cellular populations. Furthermore, detection of mtDNA mutations via single-cell assay for transposase-accessible chromatin with select antigen profiling by sequencing (ASAP-seq) simultaneously determines not only donor and recipient cells but also their phenotype at frequencies of 0.1% to 1%. Finally, integration of mtDNA mutations, surface markers, and chromatin accessibility profiles enables the phenotypic resolution of leukemic populations from normal immune cells, thereby providing fresh insights into residual donor-derived engraftment and short-term clonal evolution following therapy for post-transplant leukemia relapse. As throughput evolves, we envision future development of single-cell sequencing-based post-transplant monitoring as a powerful approach for guiding clinical decision-making. Significance: mtDNA mutations enable single-cell tracking of leukemic clonal evolution and donor-recipient origin following allogeneic HSCT. This provides unprecedented insight into chimeric cellular phenotypes of early immune reconstitution, incipient relapse, and quality of donor engraftment with immediate translational potential for future clinical post-transplant monitoring and decision-making.
Collapse
Affiliation(s)
- Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Berlin, Germany
| | - Nicoletta Cieri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Katie Maurer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Marwan Kwok
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Haoxiang Lyu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wesley S. Lu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Satyen H. Gohil
- Department of Haematology, University College London Hospitals, London, United Kingdom
| | - Ignaty Leshchiner
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | - Caleb A. Lareau
- Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Leif S. Ludwig
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Donna S. Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Haesook T. Kim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Shuqiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Gad Getz
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Jacqueline S. Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Robert J. Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Kenneth J. Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
Driessen A, Unger S, Nguyen AP, Ries RE, Meshinchi S, Kreutmair S, Alberti C, Sumazin P, Aplenc R, Redell MS, Becher B, Rodríguez Martínez M. Identification of single-cell blasts in pediatric acute myeloid leukemia using an autoencoder. Life Sci Alliance 2024; 7:e202402674. [PMID: 39191488 PMCID: PMC11358707 DOI: 10.26508/lsa.202402674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
Pediatric acute myeloid leukemia (AML) is an aggressive blood cancer with a poor prognosis and high relapse rate. Current challenges in the identification of immunotherapy targets arise from patient-specific blast immunophenotypes and their change during disease progression. To overcome this, we present a new computational research tool to rapidly identify malignant cells. We generated single-cell flow cytometry profiles of 21 pediatric AML patients with matched samples at diagnosis, remission, and relapse. We coupled a classifier to an autoencoder for anomaly detection and classified malignant blasts with 90% accuracy. Moreover, our method assigns a developmental stage to blasts at the single-cell level, improving current classification approaches based on differentiation of the dominant phenotype. We observed major immunophenotype and developmental stage alterations between diagnosis and relapse. Patients with KMT2A rearrangement had more profound changes in their blast immunophenotypes at relapse compared to patients with other molecular features. Our method provides new insights into the immunophenotypic composition of AML blasts in an unbiased fashion and can help to define immunotherapy targets that might improve personalized AML treatment.
Collapse
Affiliation(s)
- Alice Driessen
- Data and AI Research, IBM Research Europe, Zürich, Switzerland
- ETH Zürich, Zürich, Switzerland
| | - Susanne Unger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - An-Phi Nguyen
- Data and AI Research, IBM Research Europe, Zürich, Switzerland
| | - Rhonda E Ries
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Stefanie Kreutmair
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - Chiara Alberti
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Pavel Sumazin
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Richard Aplenc
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michele S Redell
- Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, USA
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
21
|
Colonne CK, Kimble EL, Turtle CJ. Evolving strategies to overcome barriers in CAR-T cell therapy for acute myeloid leukemia. Expert Rev Hematol 2024; 17:797-818. [PMID: 39439295 DOI: 10.1080/17474086.2024.2420614] [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: 06/26/2024] [Revised: 09/05/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a complex and heterogeneous disease characterized by an aggressive clinical course and limited efficacious treatment options in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR)-modified T (CAR-T) cell immunotherapy is an investigational treatment strategy for R/R AML that has shown some promise. However, obstacles to successful CAR-T cell immunotherapy for AML remain. AREAS COVERED In analyses of clinical trials of CAR-T cell therapy for R/R AML, complete responses without measurable residual disease have been reported, but the durability of those responses remains unclear. Significant barriers to successful CAR-T cell therapy in AML include the scarcity of suitable tumor-target antigens (TTA), inherent T cell functional deficits, and the immunoinhibitory and hostile tumor microenvironment (TME). This review will focus on these barriers to successful CAR-T cell therapy in AML, and discuss scientific advancements and evolving strategies to overcome them. EXPERT OPINION Achieving durable remissions in R/R AML will likely require a multifaceted approach that integrates advancements in TTA selection, enhancement of the intrinsic quality of CAR-T cells, and development of strategies to overcome inhibitory mechanisms in the AML TME.
Collapse
Affiliation(s)
- Chanukya K Colonne
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erik L Kimble
- Translational Science and Therapeutic Division, Fred Hutchinson Cancer Center, Seattle, USA
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
| | - Cameron J Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
22
|
Wegmann R, Bonilla X, Casanova R, Chevrier S, Coelho R, Esposito C, Ficek-Pascual J, Goetze S, Gut G, Jacob F, Jacobs A, Kuipers J, Lischetti U, Mena J, Milani ES, Prummer M, Del Castillo JS, Singer F, Sivapatham S, Toussaint NC, Vilinovszki O, Wildschut MHE, Thavayogarajah T, Malani D, Aebersold R, Bacac M, Beerenwinkel N, Beisel C, Bodenmiller B, Heinzelmann-Schwarz V, Koelzer VH, Levesque MP, Moch H, Pelkmans L, Rätsch G, Tolnay M, Wicki A, Wollscheid B, Manz MG, Snijder B, Theocharides APA. Single-cell landscape of innate and acquired drug resistance in acute myeloid leukemia. Nat Commun 2024; 15:9402. [PMID: 39477946 PMCID: PMC11525670 DOI: 10.1038/s41467-024-53535-4] [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: 03/08/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Deep single-cell multi-omic profiling offers a promising approach to understand and overcome drug resistance in relapsed or refractory (rr) acute myeloid leukemia (AML). Here, we combine single-cell ex vivo drug profiling (pharmacoscopy) with single-cell and bulk DNA, RNA, and protein analyses, alongside clinical data from 21 rrAML patients. Unsupervised data integration reveals reduced ex vivo response to the Bcl-2 inhibitor venetoclax (VEN) in patients treated with both a hypomethylating agent (HMA) and VEN, compared to those pre-exposed to chemotherapy or HMA alone. Integrative analysis identifies both known and unreported mechanisms of innate and treatment-related VEN resistance and suggests alternative treatments, like targeting increased proliferation with the PLK inhibitor volasertib. Additionally, high CD36 expression in VEN-resistant blasts associates with sensitivity to CD36-targeted antibody treatment ex vivo. This study demonstrates how single-cell multi-omic profiling can uncover drug resistance mechanisms and treatment vulnerabilities, providing a valuable resource for future AML research.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Single-Cell Analysis
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- CD36 Antigens/metabolism
- CD36 Antigens/genetics
- Female
- Male
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Middle Aged
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Aged
Collapse
Affiliation(s)
- Rebekka Wegmann
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Ximena Bonilla
- Department of Computer Science, ETH Zurich, Zurich, Switzerland
| | - Ruben Casanova
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Stéphane Chevrier
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Ricardo Coelho
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Cinzia Esposito
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | | - Sandra Goetze
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Gabriele Gut
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Francis Jacob
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Andrea Jacobs
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Jack Kuipers
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Ulrike Lischetti
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julien Mena
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Emanuela S Milani
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Michael Prummer
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
| | | | - Franziska Singer
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
| | - Sujana Sivapatham
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Nora C Toussaint
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- Swiss Data Science Center, ETH Zürich, Zurich, Switzerland
| | - Oliver Vilinovszki
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Mattheus H E Wildschut
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - Disha Malani
- Harvard Medical School and Dana-Farber Cancer Institute, Boston, USA
| | - Rudolf Aebersold
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | | | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Faculty of Medicine, Zurich, Switzerland
| | | | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Faculty of Medicine, Zurich, Switzerland
| | - Lucas Pelkmans
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Gunnar Rätsch
- Department of Computer Science, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- AI Center at ETH Zurich, Zurich, Switzerland
| | - Markus Tolnay
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Andreas Wicki
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Faculty of Medicine, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland.
| | - Berend Snijder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | | |
Collapse
|
23
|
Santos de Macedo BG, Albuquerque de Melo M, Pereira-Martins DA, Machado-Neto JA, Traina F. Autophagy and inflammasome activation are associated with poor response to FLT3 inhibitors in patients with FLT3-ITD acute myeloid leukemia. Sci Rep 2024; 14:23882. [PMID: 39396074 PMCID: PMC11470923 DOI: 10.1038/s41598-024-74168-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/24/2024] [Indexed: 10/14/2024] Open
Abstract
Beyond its clinical diversity and severity, acute myeloid leukemia (AML) is known for its complex molecular background and for rewiring biological processes to aid disease onset and maintenance. FLT3 mutations are among the most recurring molecular entities that cooperatively drive AML, and their inhibition is a critical molecularly oriented therapeutic strategy. Despite being a promising avenue, it still faces challenges such as intrinsic and acquired drug resistance, which led us to investigate whether and how autophagy and inflammasome interact and whether this interaction could be leveraged to enhance FLT3 inhibition as a therapeutic strategy. We observed a strong and positive correlation between the expression of key genes associated with autophagy and the inflammasome. Gene set enrichment analysis of the FLT3-ITD samples and their ex vivo response to five different FLT3 inhibitors revealed a common molecular signature compatible with autophagy and inflammasome activation across all poor responders. Inflammasome activation was also shown to strongly increase the likelihood of a poor ex vivo response to the FLT3 inhibitors quizartinib and sorafenib. These findings reveal a distinct molecular pattern within FLT3-ITD AML samples that underscores the necessity for further exploration into how approaching these supportive parallel yet altered pathways could improve therapeutic strategies.
Collapse
Affiliation(s)
- Brunno Gilberto Santos de Macedo
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, Ribeirão Preto, São Paulo, 14040-900, Brazil
| | - Manuela Albuquerque de Melo
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, Ribeirão Preto, São Paulo, 14040-900, Brazil
| | | | | | - Fabíola Traina
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, Ribeirão Preto, São Paulo, 14040-900, Brazil.
| |
Collapse
|
24
|
Minciacchi VR, Karantanou C, Bravo J, Pereira RS, Zanetti C, Krack T, Kumar R, Bankov K, Hartmann S, Huntly BJP, Meduri E, Ruf W, Krause DS. Differential inflammatory conditioning of the bone marrow by acute myeloid leukemia and its impact on progression. Blood Adv 2024; 8:4983-4996. [PMID: 38996202 PMCID: PMC11465066 DOI: 10.1182/bloodadvances.2024012867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/11/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
ABSTRACT Inflammation promotes solid tumor progression, but how regulatory mechanisms of inflammation may affect leukemia is less well studied. Using annexin A5 (ANXA5), a calcium-binding protein known for apoptosis, which we discovered to be differentially expressed in the bone marrow microenvironment (BMM) of mice with acute myeloid (AML) vs chronic myeloid leukemia, as a model system, we unravel here a circuit in which AML-derived tumor necrosis factor α (TNF-α) dose-dependently reduces ANXA5 in the BMM. This creates an inflammatory BMM via elevated levels of prostaglandin E2 (PGE2). Via binding to its EP4 receptor, PGE2 increases β-catenin and hypoxia-inducible factor 1α signaling in AML cells, thereby accelerating PGE2-sensitive AML. Human trephine biopsies may show lower ANXA5 expression and higher PGE2 expression in AML than other hematologic malignancies. Furthermore, syngeneic and xenogeneic transplantation models suggest a survival benefit after treatment with the inhibitor of prostaglandin-endoperoxide synthase 2 (cyclooxygenase 2 [COX2]), celecoxib, plus cytarabine in those AML types highly sensitive to PGE2 compared with cytarabine alone. Taken together, TNF-α/ANXA5/NF-κB/COX2/PGE2-mediated inflammation influences AML course in a highly differential and circular manner, and patients with AML with "inflammatory AML" may benefit from antiphlogistic agents as adjunct therapy.
Collapse
Affiliation(s)
- Valentina R. Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Christina Karantanou
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Jimena Bravo
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Raquel S. Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Costanza Zanetti
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Theresa Krack
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Katrin Bankov
- Department of Pathology, Goethe University, Frankfurt am Main, Germany
| | - Sylvia Hartmann
- Department of Pathology, Goethe University, Frankfurt am Main, Germany
| | - Brian J. P. Huntly
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Eshwar Meduri
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| | - Daniela S. Krause
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Institute of Transfusion Medicine - Transfusion Centre, Johannes Gutenberg University Medical Centre, Mainz, Germany
| |
Collapse
|
25
|
Ianevski A, Nader K, Driva K, Senkowski W, Bulanova D, Moyano-Galceran L, Ruokoranta T, Kuusanmäki H, Ikonen N, Sergeev P, Vähä-Koskela M, Giri AK, Vähärautio A, Kontro M, Porkka K, Pitkänen E, Heckman CA, Wennerberg K, Aittokallio T. Single-cell transcriptomes identify patient-tailored therapies for selective co-inhibition of cancer clones. Nat Commun 2024; 15:8579. [PMID: 39362905 PMCID: PMC11450203 DOI: 10.1038/s41467-024-52980-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/27/2024] [Indexed: 10/05/2024] Open
Abstract
Intratumoral cellular heterogeneity necessitates multi-targeting therapies for improved clinical benefits in advanced malignancies. However, systematic identification of patient-specific treatments that selectively co-inhibit cancerous cell populations poses a combinatorial challenge, since the number of possible drug-dose combinations vastly exceeds what could be tested in patient cells. Here, we describe a machine learning approach, scTherapy, which leverages single-cell transcriptomic profiles to prioritize multi-targeting treatment options for individual patients with hematological cancers or solid tumors. Patient-specific treatments reveal a wide spectrum of co-inhibitors of multiple biological pathways predicted for primary cells from heterogenous cohorts of patients with acute myeloid leukemia and high-grade serous ovarian carcinoma, each with unique resistance patterns and synergy mechanisms. Experimental validations confirm that 96% of the multi-targeting treatments exhibit selective efficacy or synergy, and 83% demonstrate low toxicity to normal cells, highlighting their potential for therapeutic efficacy and safety. In a pan-cancer analysis across five cancer types, 25% of the predicted treatments are shared among the patients of the same tumor type, while 19% of the treatments are patient-specific. Our approach provides a widely-applicable strategy to identify personalized treatment regimens that selectively co-inhibit malignant cells and avoid inhibition of non-cancerous cells, thereby increasing their likelihood for clinical success.
Collapse
Affiliation(s)
- Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kristen Nader
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kyriaki Driva
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Wojciech Senkowski
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Daria Bulanova
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Lidia Moyano-Galceran
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Tanja Ruokoranta
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Foundation for the Finnish Cancer Institute (FCI), Helsinki, Finland
| | - Nemo Ikonen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Philipp Sergeev
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Anil K Giri
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute (FCI), Helsinki, Finland
| | - Anna Vähärautio
- Foundation for the Finnish Cancer Institute (FCI), Helsinki, Finland
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Kontro
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Foundation for the Finnish Cancer Institute (FCI), Helsinki, Finland
| | - Kimmo Porkka
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Esa Pitkänen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Krister Wennerberg
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway.
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
26
|
Man CH, Li C, Xu X, Zhao M. Metabolic regulation in normal and leukemic stem cells. Trends Pharmacol Sci 2024; 45:919-930. [PMID: 39306527 DOI: 10.1016/j.tips.2024.08.004] [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: 07/05/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 10/06/2024]
Abstract
Hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs) are crucial for ensuring hematopoietic homeostasis and driving leukemia progression, respectively. Recent research has revealed that metabolic adaptations significantly regulate the function and survival of these stem cells. In this review, we provide an overview of how metabolic pathways regulate oxidative and proteostatic stresses in HSCs during homeostasis and aging. Furthermore, we highlight targetable metabolic pathways and explore their interactions with epigenetics and the microenvironment in addressing the chemoresistance and immune evasion capacities of LSCs. The metabolic differences between HSCs and LSCs have profound implications for therapeutic strategies.
Collapse
Affiliation(s)
- Cheuk-Him Man
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Changzheng Li
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xi Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510030, China
| | - Meng Zhao
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
27
|
Urrutia S, Takahashi K. Precision medicine in AML: overcoming resistance. Int J Hematol 2024; 120:439-454. [PMID: 39085680 DOI: 10.1007/s12185-024-03827-8] [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: 04/19/2024] [Revised: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
The development of molecularly targeted therapy for acute myeloid leukemia is progressing at an accelerated pace. Therapies targeting FLT3, IDH1, IDH2, and BCL2 have been approved in the last 5 years. As we exploit these biological vulnerabilities, various mechanisms of resistance arise. Emergence of competing clones with different genetic drivers and acquisition of constitutional mutations in the target renders therapies ineffective, and enzymatic isoform changes can lead to reappearance of the disease phenotype. Understanding the timing and circumstances of resistance origination will allow clinicians to develop combinatorial and sequential therapeutic approaches to deepen responses and improve survival. The objective of this review is to illustrate the biological underpinnings of each therapy and the landscape of resistance mechanisms and discuss strategies to overcome on- and off-target resistance.
Collapse
Affiliation(s)
- Samuel Urrutia
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1901 East Road, 4SCR6.2085, Houston, TX, 77030-4009, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
28
|
Schiffman JS, D'Avino AR, Prieto T, Pang Y, Fan Y, Rajagopalan S, Potenski C, Hara T, Suvà ML, Gawad C, Landau DA. Defining heritability, plasticity, and transition dynamics of cellular phenotypes in somatic evolution. Nat Genet 2024; 56:2174-2184. [PMID: 39317739 PMCID: PMC11527590 DOI: 10.1038/s41588-024-01920-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 08/21/2024] [Indexed: 09/26/2024]
Abstract
Single-cell sequencing has characterized cell state heterogeneity across diverse healthy and malignant tissues. However, the plasticity or heritability of these cell states remains largely unknown. To address this, we introduce PATH (phylogenetic analysis of trait heritability), a framework to quantify cell state heritability versus plasticity and infer cell state transition and proliferation dynamics from single-cell lineage tracing data. Applying PATH to a mouse model of pancreatic cancer, we observed heritability at the ends of the epithelial-to-mesenchymal transition spectrum, with higher plasticity at more intermediate states. In primary glioblastoma, we identified bidirectional transitions between stem- and mesenchymal-like cells, which use the astrocyte-like state as an intermediary. Finally, we reconstructed a phylogeny from single-cell whole-genome sequencing in B cell acute lymphoblastic leukemia and delineated the heritability of B cell differentiation states linked with genetic drivers. Altogether, PATH replaces qualitative conceptions of plasticity with quantitative measures, offering a framework to study somatic evolution.
Collapse
Affiliation(s)
- Joshua S Schiffman
- New York Genome Center, New York, NY, USA.
- Weill Cornell Medicine, New York, NY, USA.
| | - Andrew R D'Avino
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, Rockefeller University, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tamara Prieto
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | | | - Yilin Fan
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Srinivas Rajagopalan
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Catherine Potenski
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Toshiro Hara
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mario L Suvà
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Charles Gawad
- Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Dan A Landau
- New York Genome Center, New York, NY, USA.
- Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
29
|
Zhang P, Wang T, Cui G, Ye R, Wan W, Liu T, Zheng Y, Zhong Z. Systemic Multifunctional Nanovaccines for Potent Personalized Immunotherapy of Acute Myeloid Leukemia. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407189. [PMID: 39171954 DOI: 10.1002/adma.202407189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/30/2024] [Indexed: 08/23/2024]
Abstract
Hematological malignancies (HM) like acute myeloid leukemia (AML) are often intractable. Cancer vaccines possibly inducing robust and broad anti-tumor immune responses may be a promising treatment option for HM. Few effective vaccines against blood cancers are, however, developed to date partly owing to insufficient stimulation of dendritic cells (DCs) in the body and lacking appropriate tumor antigens (Ags). Here it is found that systemic multifunctional nanovaccines consisting of nucleotide-binding oligomerization domain-containing protein 2 (NOD2) and Toll-like receptor 9 (TLR9) agonists - muramyl dipeptide (MDP) and CpG, and tumor cell lysate (TCL) as Ags (MCA-NV) induce potent and broad immunity against AML. MCA-NV show complementary stimulation of DCs and prime homing to lymphoid organs following systemic administration. Of note, in orthotopic AML mouse models, intravenous infusion of different vaccine formulations elicits substantially higher anti-AML efficacies than subcutaneous administration. Systemic MCA-NV cure 78% of AML mice and elicit long-term immune memory with 100% protection from rechallenging AML cells. Systemic MCA-NV can also serve as prophylactic vaccines against the same AML. These systemic nanovaccines utilizing patient TCL as Ags and dual adjuvants to elicit strong, durable, and broad immune responses can provide a personalized immunotherapeutic strategy against AML and other HM.
Collapse
Affiliation(s)
- Peng Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Tanzhen Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, P. R. China
| | - Guanhong Cui
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Ruonan Ye
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Wenjun Wan
- College of Pharmaceutical Sciences, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, Soochow University, Suzhou, 215123, P. R. China
| | - Tianhui Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, P. R. China
| | - Yiran Zheng
- College of Pharmaceutical Sciences, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, Soochow University, Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
- College of Pharmaceutical Sciences, Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
30
|
Boutzen H, Murison A, Oriecuia A, Bansal S, Arlidge C, Wang JCY, Lupien M, Kaufmann KB, Dick JE. Identification of leukemia stem cell subsets with distinct transcriptional, epigenetic and functional properties. Leukemia 2024; 38:2090-2101. [PMID: 39169113 PMCID: PMC11436360 DOI: 10.1038/s41375-024-02358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024]
Abstract
The leukemia stem cell (LSC) compartment is a complex reservoir fueling disease progression in acute myeloid leukemia (AML). The existence of heterogeneity within this compartment is well documented but prior studies have focused on genetic heterogeneity without being able to address functional heterogeneity. Understanding this heterogeneity is critical for the informed design of therapies targeting LSC, but has been hampered by LSC scarcity and the lack of reliable cell surface markers for viable LSC isolation. To overcome these challenges, we turned to the patient-derived OCI-AML22 cell model. This model includes functionally, transcriptionally and epigenetically characterized LSC broadly representative of LSC found in primary AML samples. Focusing on the pool of LSC, we used an integrated approach combining xenograft assays with single-cell analysis to identify two LSC subtypes with distinct transcriptional, epigenetic and functional properties. These LSC subtypes differed in depth of quiescence, differentiation potential, repopulation capacity, sensitivity to chemotherapy and could be isolated based on CD112 expression. A majority of AML patient samples had transcriptional signatures reflective of either LSC subtype, and some even showed coexistence within an individual sample. This work provides a framework for investigating the LSC compartment and designing combinatorial therapeutic strategies in AML.
Collapse
Affiliation(s)
- Héléna Boutzen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
| | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Alexa Oriecuia
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Suraj Bansal
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Christopher Arlidge
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A4, Canada
| | - Kerstin B Kaufmann
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
31
|
Li Z, Jin P, Xiang R, Li X, Shen J, He M, Liu X, Zhu H, Wu S, Dong F, Zhao H, Liu H, Jin Z, Li J. A CD8 + T cell related immune score predicts survival and refines the risk assessment in acute myeloid leukemia. Front Immunol 2024; 15:1408109. [PMID: 39346926 PMCID: PMC11428106 DOI: 10.3389/fimmu.2024.1408109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
Although advancements in genomic and epigenetic research have deepened our understanding of acute myeloid leukemia (AML), only one-third of patients can achieve durable remission. Growing evidence suggests that the immune microenvironment in bone marrow influences prognosis and survival in AML. There is a specific association between CD8+ T cells and the prognosis of AML patients. To develop a CD8+ T cell-related immune risk score for AML, we first evaluated the accuracy of CIBERSORTx in predicting the abundance of CD8+ T cells in bulk RNA-seq and found it significantly correlated with observed single-cell RNA sequencing data and the proportions of CD8+ T cells derived from flow cytometry. Next, we constructed the CTCG15, a 15-gene prognostic signature, using univariate and LASSO regression on the differentially expressed genes between CD8+ THigh and CD8+ TLow groups. The CTCG15 was further validated across six datasets in different platforms. The CTCG15 has been shown to be independent of established prognostic markers, and can distill transcriptomic consequences of several genetic abnormalities closely related to prognosis in AML patients. Finally, integrating this model into the 2022 European LeukemiaNet contributed to a higher predictive power for prognosis prediction. Collectively, our study demonstrates that CD8+ T cell-related signature could improve the comprehensive risk stratification and prognosis prediction in AML.
Collapse
Affiliation(s)
- Zeyi Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rufang Xiang
- Department of General Practice, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyang Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Shen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengke He
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaxin Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongming Zhu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shishuang Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyi Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huijin Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junmin Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Wuxi Branch of Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Grenier JMP, Testut C, Bal M, Bardin F, De Grandis M, Gelsi-Boyer V, Vernerey J, Delahaye M, Granjeaud S, Zemmour C, Spinella JF, Chavakis T, Mancini SJC, Boher JM, Hébert J, Sauvageau G, Vey N, Schwaller J, Hospital MA, Fauriat C, Aurrand-Lions M. Genetic deletion of JAM-C in preleukemic cells rewires leukemic stem cell gene expression program in AML. Blood Adv 2024; 8:4662-4678. [PMID: 38954834 PMCID: PMC11402138 DOI: 10.1182/bloodadvances.2023011747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 05/23/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
ABSTRACT The leukemic stem cell (LSC) score LSC-17 based on a stemness-related gene expression signature is an indicator of poor disease outcome in acute myeloid leukemia (AML). However, it is not known whether "niche anchoring" of LSC affects disease evolution. To address this issue, we conditionally inactivated the adhesion molecule JAM-C (Junctional Adhesion Molecule-C) expressed by hematopoietic stem cells (HSCs) and LSCs in an inducible mixed-lineage leukemia (iMLL)-AF9-driven AML mouse model. Deletion of Jam3 (encoding JAM-C) before induction of the leukemia-initiating iMLL-AF9 fusion resulted in a shift from long-term to short-term HSC expansion, without affecting disease initiation and progression. In vitro experiments showed that JAM-C controlled leukemic cell nesting irrespective of the bone marrow stromal cells used. RNA sequencing performed on leukemic HSCs isolated from diseased mice revealed that genes upregulated in Jam3-deficient animals belonged to activation protein-1 (AP-1) and tumor necrosis factor α (TNF-α)/NF-κB pathways. Human orthologs of dysregulated genes allowed to identify a score that was distinct from, and complementary to, the LSC-17 score. Substratification of patients with AML using LSC-17 and AP-1/TNF-α genes signature defined 4 groups with median survival ranging from <1 year to a median of "not reached" after 8 years. Finally, coculture experiments showed that AP-1 activation in leukemic cells was dependent on the nature of stromal cells. Altogether, our results identify the AP-1/TNF-α gene signature as a proxy of LSC anchoring in bone marrow niches, which improves the prognostic value of the LSC-17 score. This trial was registered at www.ClinicalTrials.gov as #NCT02320656.
Collapse
Affiliation(s)
- Julien M. P. Grenier
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
- UMR 7268, Aix-Marseille Université, EFS, CNRS, GENGLOBE, Marseille, France
| | - Céline Testut
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Matthieu Bal
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Florence Bardin
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Maria De Grandis
- Aix-Marseille University, CNRS, EFS, ADES, Biologie des Groupes Sanguins, Marseille, France
- UMR 7268, Aix-Marseille Université, EFS, CNRS, GENGLOBE, Marseille, France
| | - Véronique Gelsi-Boyer
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Julien Vernerey
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Marjorie Delahaye
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Samuel Granjeaud
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Christophe Zemmour
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Jean-François Spinella
- Laboratory of Molecular Genetics of Stem Cells, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Stéphane J. C. Mancini
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Jean-Marie Boher
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Josée Hébert
- Division of Hematology-Oncology, Department of Medicine, Maisonneuve-Rosemont Hospital, Université de Montréal, Montreal, QC, Canada
| | - Guy Sauvageau
- Laboratory of Molecular Genetics of Stem Cells, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Norbert Vey
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Jürg Schwaller
- Department of Biomedicine, University Children’s Hospital, University of Basel, Basel, Switzerland
| | | | - Cyril Fauriat
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Michel Aurrand-Lions
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| |
Collapse
|
33
|
Safina K, van Galen P. New frameworks for hematopoiesis derived from single-cell genomics. Blood 2024; 144:1039-1047. [PMID: 38985829 PMCID: PMC11561540 DOI: 10.1182/blood.2024024006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/12/2024] Open
Abstract
ABSTRACT Recent advancements in single-cell genomics have enriched our understanding of hematopoiesis, providing intricate details about hematopoietic stem cell biology, differentiation, and lineage commitment. Technological advancements have highlighted extensive heterogeneity of cell populations and continuity of differentiation routes. Nevertheless, intermediate "attractor" states signify structure in stem and progenitor populations that link state transition dynamics to fate potential. We discuss how innovative model systems quantify lineage bias and how stress accelerates differentiation, thereby reducing fate plasticity compared with native hematopoiesis. We conclude by offering our perspective on the current model of hematopoiesis and discuss how a more precise understanding can translate to strategies that extend healthy hematopoiesis and prevent disease.
Collapse
Affiliation(s)
- Ksenia Safina
- Division of Hematology, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Ludwig Center at Harvard, Boston, MA
| | - Peter van Galen
- Division of Hematology, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Ludwig Center at Harvard, Boston, MA
| |
Collapse
|
34
|
Marot-Lassauzaie V, Beneyto-Calabuig S, Obermayer B, Velten L, Beule D, Haghverdi L. Identifying cancer cells from calling single-nucleotide variants in scRNA-seq data. BIOINFORMATICS (OXFORD, ENGLAND) 2024; 40:btae512. [PMID: 39163479 PMCID: PMC11379463 DOI: 10.1093/bioinformatics/btae512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/14/2024] [Accepted: 08/15/2024] [Indexed: 08/22/2024]
Abstract
MOTIVATION Single-cell RNA sequencing (scRNA-seq) data are widely used to study cancer cell states and their heterogeneity. However, the tumour microenvironment is usually a mixture of healthy and cancerous cells and it can be difficult to fully separate these two populations based on transcriptomics alone. If available, somatic single-nucleotide variants (SNVs) observed in the scRNA-seq data could be used to identify the cancer population and match that information with the single cells' expression profile. However, calling somatic SNVs in scRNA-seq data is a challenging task, as most variants seen in the short-read data are not somatic, but can instead be germline variants, RNA edits or transcription, sequencing, or processing errors. In addition, only variants present in actively transcribed regions for each individual cell will be seen in the data. RESULTS To address these challenges, we develop CCLONE (Cancer Cell Labelling On Noisy Expression), an interpretable tool adapted to handle the uncertainty and sparsity of SNVs called from scRNA-seq data. CCLONE jointly identifies cancer clonal populations, and their associated variants. We apply CCLONE on two acute myeloid leukaemia datasets and one lung adenocarcinoma dataset and show that CCLONE captures both genetic clones and somatic events for multiple patients. These results show how CCLONE can be used to gather insight into the course of the disease and the origin of cancer cells in scRNA-seq data. AVAILABILITY AND IMPLEMENTATION Source code is available at github.com/HaghverdiLab/CCLONE.
Collapse
Affiliation(s)
- Valérie Marot-Lassauzaie
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Hannoversche Str. 28, 10115 Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Sergi Beneyto-Calabuig
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Benedikt Obermayer
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Lars Velten
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Laleh Haghverdi
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Hannoversche Str. 28, 10115 Berlin, Germany
| |
Collapse
|
35
|
Chen H, Lu J, Wang Z, Wu S, Zhang S, Geng J, Hou C, He P, Lu X. Unlocking reproducible transcriptomic signatures for acute myeloid leukaemia: Integration, classification and drug repurposing. J Cell Mol Med 2024; 28:e70085. [PMID: 39267259 PMCID: PMC11392829 DOI: 10.1111/jcmm.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/25/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a highly heterogeneous disease, which lead to various findings in transcriptomic research. This study addresses these challenges by integrating 34 datasets, including 26 control groups, 6 prognostic datasets and 2 single-cell RNA sequencing (scRNA-seq) datasets to identify 10,000 AML-related genes (ARGs). We focused on genes with low variability and high consistency and successfully discovered 191 AML signatures (ASs). Leveraging machine learning techniques, specifically the XGBoost model and our custom framework, we classified AML subtypes with both scRNA-seq and bulk RNA-seq data, complementing the ELN2022 classification approach. Our research also identified promising treatments for AML through drug repurposing, with solasonine showing potential efficacy for high-risk AML patients, supported by molecular docking and transcriptomic analyses. To enhance reproducibility and customizability, we developed CSAMLdb, a user-friendly database platform. It facilitates the reuse and personalized analysis of nearly all results obtained in this research, including single-gene prognostics, multi-gene scoring, enrichment analysis, machine learning risk assessment, drug repositioning analysis and literature abstract named entity recognition. CSAMLdb is available at http://www.csamldb.com.
Collapse
Affiliation(s)
- Haoran Chen
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- School of Management, Shanxi Medical University, Taiyuan, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Jinqi Lu
- Department of Computer Science, Boston University, Boston, Massachusetts, USA
| | - Zining Wang
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Shengnan Wu
- School of Management, Shanxi Medical University, Taiyuan, China
| | - Shengxiao Zhang
- Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - Jie Geng
- Basic Medicine College, Shanxi Medical University, Taiyuan, China
| | - Chuandong Hou
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Peifeng He
- School of Management, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Xuechun Lu
- School of Management, Shanxi Medical University, Taiyuan, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing, China
| |
Collapse
|
36
|
Radpour R, Simillion C, Wang B, Abbas HA, Riether C, Ochsenbein AF. IL-9 secreted by leukemia stem cells induces Th1-skewed CD4+ T cells, which promote their expansion. Blood 2024; 144:888-903. [PMID: 38941612 DOI: 10.1182/blood.2024024000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
ABSTRACT In acute myeloid leukemia (AML), leukemia stem cells (LSCs) and leukemia progenitor cells (LPCs) interact with various cell types in the bone marrow (BM) microenvironment, regulating their expansion and differentiation. To study the interaction of CD4+ and CD8+ T cells in the BM with LSCs and LPCs, we analyzed their transcriptome and predicted cell-cell interactions by unbiased high-throughput correlation network analysis. We found that CD4+ T cells in the BM of patients with AML were activated and skewed toward T-helper (Th)1 polarization, whereas interleukin-9 (IL-9)-producing (Th9) CD4+ T cells were absent. In contrast to normal hematopoietic stem cells, LSCs produced IL-9, and the correlation modeling predicted IL9 in LSCs as a main hub gene that activates CD4+ T cells in AML. Functional validation revealed that IL-9 receptor signaling in CD4+ T cells leads to activation of the JAK-STAT pathway that induces the upregulation of KMT2A and KMT2C genes, resulting in methylation on histone H3 at lysine 4 to promote genome accessibility and transcriptional activation. This induced Th1-skewing, proliferation, and effector cytokine secretion, including interferon gamma (IFN-γ) and tumor necrosis factor α (TNF-α). IFN-γ and, to a lesser extent, TNF-α produced by activated CD4+ T cells induced the expansion of LSCs. In accordance with our findings, high IL9 expression in LSCs and high IL9R, TNF, and IFNG expression in BM-infiltrating CD4+ T cells correlated with worse overall survival in AML. Thus, IL-9 secreted by AML LSCs shapes a Th1-skewed immune environment that promotes their expansion by secreting IFN-γ and TNF-α.
Collapse
MESH Headings
- Interleukin-9/genetics
- Interleukin-9/metabolism
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/immunology
- Th1 Cells/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Tumor Microenvironment/immunology
- Receptors, Interleukin-9/genetics
- Receptors, Interleukin-9/metabolism
- Interferon-gamma/metabolism
- Histone-Lysine N-Methyltransferase/genetics
Collapse
Affiliation(s)
- Ramin Radpour
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Bofei Wang
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Hussein A Abbas
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX
| | - Carsten Riether
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adrian F Ochsenbein
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
37
|
Caliskan G, Pawitan Y, Vu TN. Similarities and differences of bone marrow and peripheral blood samples from acute myeloid leukemia patients in terms of cellular heterogeneity and ex-vivo drug sensitivity. EJHAEM 2024; 5:721-727. [PMID: 39157629 PMCID: PMC11327724 DOI: 10.1002/jha2.961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 08/20/2024]
Abstract
Background Bone marrow (BM) evaluation is the de facto standard for diagnosis, molecular analysis, risk stratification, and therapy response assessment in acute myeloid leukemia (AML), but in patients with a high number of circulating blast cells, the peripheral blood (PB) sample could provide similar information as BM. However, there is no large-scale molecular study comparing the two specimens in terms of their gene expression profiles, cellular heterogeneities, and ex-vivo drug sensitivity. Methodology We used (i) the BEAT-AML cohort each with detailed molecular data; (ii) cell-type deconvolution to estimate leukemic and immune cell proportions between specimen types; (iii) differential expression (DE) and drug-cell type association analysis; and (iv) logistic regression models to assess the association between induction therapy response, cell-type composition and first-line drug treatment. Results Results: We identified 207 patients having BM and 116 patients having PB samples. There was a total of 1271 DE genes (false discovery rate < 0.05) between BM and PB; the top enriched pathways in terms of DE genes belong to the immune system pathways. Aggregated ex-vivo drug response profiles from the two specimens were largely similar, as were the cellular components, except for the GMP-like cell type (17% in BM vs. 5% in PB, p-value = 2 × 10-7). Among the specimen-specific results, the GMP-like subtype was associated with multiple drug resistance in BM and the ProMono-like subtype in PB. Several cell types were associated with the response to induction therapy, but the impact of specimen type on the interaction of cell type and cytarabine-associated induction therapy was not statistically significant for most cell types. Results Conclusions: Even though there are molecular and cellular differences between BM and PB samples, they show many similarities in ex-vivo drug response profiles, indicating the clinical utility of the substantially less-invasive PB samples.
Collapse
Affiliation(s)
- Gulser Caliskan
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Yudi Pawitan
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Trung Nghia Vu
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| |
Collapse
|
38
|
Cai Y, Li D, Lv D, Yu J, Ma Y, Jiang T, Ding N, Liu Z, Li Y, Xu J. MHC-I-presented non-canonical antigens expand the cancer immunotherapy targets in acute myeloid leukemia. Sci Data 2024; 11:831. [PMID: 39090129 PMCID: PMC11294462 DOI: 10.1038/s41597-024-03660-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024] Open
Abstract
Identification of tumor neoantigens is indispensable for the development of cancer immunotherapies. However, we are still lacking knowledge about the potential neoantigens derived from sequences outside protein-coding regions. Here, we comprehensively characterized the immunopeptidome landscape by integrating multi-omics data in acute myeloid leukemia (AML). Both canonical and non-canonical MHC-associated peptides (MAPs) in AML were identified. We found that the quality and characteristics of ncMAPs are comparable or superior to cMAPs, suggesting ncMAPs are indispensable sources for tumor neoantigens. We further proposed a computational framework to prioritize the neoantigens by integrating additional transcriptome and immunopeptidome in normal tissues. Notably, 6 of prioritized 13 neoantigens were derived from ncMAPs. The expressions of corresponding source genes are highly related to infiltrations of immune cells. Finally, a risk model was developed, which exhibited good performance for clinical prognosis in AML. Our findings expand potential cancer immunotherapy targets and provide in-depth insights into AML treatment, laying a new foundation for precision therapies in AML.
Collapse
Affiliation(s)
- Yangyang Cai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Donghao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Dezhong Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Jiaxin Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Yingying Ma
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Tiantongfei Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Na Ding
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Zhigang Liu
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Guangzhou, China.
| | - Yongsheng Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China.
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, 150001, China.
| |
Collapse
|
39
|
McCarthy N, Gui G, Dumezy F, Roumier C, Andrew G, Green S, Jenkins M, Adams A, Khan N, Craddock C, Hourigan CS, Plesa A, Freeman S. Pre-emptive detection and evolution of relapse in acute myeloid leukemia by flow cytometric measurable residual disease surveillance. Leukemia 2024; 38:1667-1673. [PMID: 38890448 PMCID: PMC11286513 DOI: 10.1038/s41375-024-02300-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024]
Abstract
Measurable residual disease (MRD) surveillance in acute myeloid leukemia (AML) may identify patients destined for relapse and thus provide the option of pre-emptive therapy to improve their outcome. Whilst flow cytometric MRD (Flow-MRD) can be applied to high-risk AML/ myelodysplasia patients, its diagnostic performance for detecting impending relapse is unknown. We evaluated this in a cohort comprising 136 true positives (bone marrows preceding relapse by a median of 2.45 months) and 155 true negatives (bone marrows during sustained remission). At an optimal Flow-MRD threshold of 0.040%, clinical sensitivity and specificity for relapse was 74% and 87% respectively (51% and 98% for Flow-MRD ≥ 0.1%) by 'different-from-normal' analysis. Median relapse kinetics were 0.78 log10/month but significantly higher at 0.92 log10/month for FLT3-mutated AML. Computational (unsupervised) Flow-MRD (C-Flow-MRD) generated optimal MRD thresholds of 0.036% and 0.082% with equivalent clinical sensitivity to standard analysis. C-Flow-MRD-identified aberrancies in HLADRlow or CD34+CD38low (LSC-type) subpopulations contributed the greatest clinical accuracy (56% sensitivity, 90% specificity) and notably, by longitudinal profiling expanded rapidly within blasts in > 40% of 86 paired MRD and relapse samples. In conclusion, flow MRD surveillance can detect MRD relapse in high risk AML and its evaluation may be enhanced by computational analysis.
Collapse
MESH Headings
- Humans
- Neoplasm, Residual/diagnosis
- Flow Cytometry/methods
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Middle Aged
- Male
- Female
- Aged
- Adult
- Recurrence
- Aged, 80 and over
- Young Adult
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/diagnosis
- Prognosis
- Immunophenotyping/methods
Collapse
Affiliation(s)
- Nicholas McCarthy
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Gege Gui
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Florent Dumezy
- Laboratory of Hematology, Lille University Hospital, Lille, France
| | | | - Georgia Andrew
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sarah Green
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | | | - Naeem Khan
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Adriana Plesa
- Lyon University Hospital, CHU-HCL, Lyon Sud, Pierre Benite, France
| | - Sylvie Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
40
|
Safri F, Nguyen R, Zerehpooshnesfchi S, George J, Qiao L. Heterogeneity of hepatocellular carcinoma: from mechanisms to clinical implications. Cancer Gene Ther 2024; 31:1105-1112. [PMID: 38499648 PMCID: PMC11327108 DOI: 10.1038/s41417-024-00764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
Hepatocellular Carcinoma (HCC) is one of the most common types of primary liver cancer. Current treatment options have limited efficacy against this malignancy, primarily owing to difficulties in early detection and the inherent resistance to existing drugs. Tumor heterogeneity is a pivotal factor contributing significantly to treatment resistance and recurrent manifestations of HCC. Intratumoral heterogeneity is an important aspect of the spectrum of complex tumor heterogeneity and contributes to late diagnosis and treatment failure. Therefore, it is crucial to thoroughly understand the molecular mechanisms of how tumor heterogeneity develops. This review aims to summarize the possible molecular dimensions of tumor heterogeneity with an emphasis on intratumoral heterogeneity, evaluate its profound impact on the diagnosis and therapeutic strategies for HCC, and explore the suitability of appropriate pre-clinical models that can be used to best study tumor heterogeneity; thus, opening new avenues for cancer treatment.
Collapse
Affiliation(s)
- Fatema Safri
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Romario Nguyen
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Shadi Zerehpooshnesfchi
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
| | - Liang Qiao
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
41
|
Zhang S, Lou S, Bian W, Liu J, Wang R, Wang Y, Zhao Y, Zou X, Jin D, Liang Y, Sun J, Liu L. Selective eradication of venetoclax-resistant monocytic acute myeloid leukemia with iron oxide nanozymes. Biochem Biophys Res Commun 2024; 719:150117. [PMID: 38761635 DOI: 10.1016/j.bbrc.2024.150117] [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: 04/23/2024] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
The clinical treatment of human acute myeloid leukemia (AML) is rapidly progressing from chemotherapy to targeted therapies led by the BCL-2 inhibitor venetoclax (VEN). Despite its unprecedented success, VEN still encounters clinical resistance. Thus, uncovering the biological vulnerability of VEN-resistant AML disease and identifying effective therapies to treat them are urgently needed. We have previously demonstrated that iron oxide nanozymes (IONE) are capable of overcoming chemoresistance in AML. The current study reports a new activity of IONE in overcoming VEN resistance. Specifically, we revealed an aberrant redox balance with excessive intracellular reactive oxygen species (ROS) in VEN-resistant monocytic AML. Treatment with IONE potently induced ROS-dependent cell death in monocytic AML in both cell lines and primary AML models. In primary AML with developmental heterogeneity containing primitive and monocytic subpopulations, IONE selectively eradicated the VEN-resistant ROS-high monocytic subpopulation, successfully resolving the challenge of developmental heterogeneity faced by VEN. Overall, our study revealed an aberrant redox balance as a therapeutic target for monocytic AML and identified a candidate IONE that could selectively and potently eradicate VEN-resistant monocytic disease.
Collapse
MESH Headings
- Humans
- Sulfonamides/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- Reactive Oxygen Species/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Line, Tumor
- Leukemia, Monocytic, Acute/drug therapy
- Leukemia, Monocytic, Acute/metabolism
- Leukemia, Monocytic, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Ferric Compounds/pharmacology
Collapse
Affiliation(s)
- Shaoqi Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Shang Lou
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Wei Bian
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Jun Liu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Rong Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Yanan Wang
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Yin Zhao
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Xiaoqing Zou
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Diange Jin
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Yue Liang
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Jie Sun
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Lina Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China.
| |
Collapse
|
42
|
Larrue C, Mouche S, Angelino P, Sajot M, Birsen R, Kosmider O, Mckee T, Vergez F, Recher C, Mas VMD, Gu Q, Xu J, Tsantoulis P, Sarry JE, Tamburini J. Targeting ferritinophagy impairs quiescent cancer stem cells in acute myeloid leukemia in vitro and in vivo models. Sci Transl Med 2024; 16:eadk1731. [PMID: 39047119 DOI: 10.1126/scitranslmed.adk1731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
Acute myeloid leukemia (AML) remains a challenging hematological malignancy with poor prognosis and limited treatment options. Leukemic stem cells (LSCs) contribute to therapeutic failure, relapse, and adverse outcome. This study investigates the role of quiescence and related molecular mechanisms in AML pathogenesis and LSC functions to identify potential therapeutic targets. Transcriptomic analysis revealed that the LSC-enriched quiescent cell population has a distinct gene signature with prognostic relevance in patients with AML. Mechanistically, quiescent blasts exhibit increased autophagic activity, which contributes to their sustained viability. Proteomic profiling uncovered differential requirements for iron metabolism between quiescent and cycling cells, revealing a unique dependence of quiescent cells on ferritinophagy, a selective form of autophagy mediated by nuclear receptor coactivator 4 (NCOA4), which regulates iron bioavailability. We evaluated the therapeutic potential of inhibiting NCOA4-mediated ferritinophagy using genetic knockdown and chemical inhibition approaches. In vitro assays showed that suppression of NCOA4 was toxic to leukemic blasts, particularly the CD34+CD38- LSC-enriched population, without affecting normal CD34+ hematopoietic progenitors. In vivo studies using murine patient-derived xenograft (PDX) models of AML confirmed that NCOA4 inhibition reduced tumor burden and impaired LSC viability and self-renewal, indicating a specific vulnerability of these cells to ferritinophagy disruption. Our findings underscore the role of NCOA4-mediated ferritinophagy in maintaining LSC quiescence and function and suggest that targeting this pathway may be an effective therapeutic strategy for AML. This study highlights the potential of NCOA4 inhibition to improve AML outcomes and paves the way for future research and clinical development.
Collapse
Affiliation(s)
- Clement Larrue
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Sarah Mouche
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
| | - Paolo Angelino
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Translational Data Science, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Maxime Sajot
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Rudy Birsen
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, 75014 Paris, France
| | - Olivier Kosmider
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, 75014 Paris, France
| | - Thomas Mckee
- Division of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - François Vergez
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Christian Recher
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Véronique Mansat-De Mas
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Petros Tsantoulis
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Jerome Tamburini
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Oncology Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| |
Collapse
|
43
|
Ma C, Hao Y, Shi B, Wu Z, Jin D, Yu X, Jin B. Unveiling mitochondrial and ribosomal gene deregulation and tumor microenvironment dynamics in acute myeloid leukemia. Cancer Gene Ther 2024; 31:1034-1048. [PMID: 38806621 DOI: 10.1038/s41417-024-00788-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Acute myeloid leukemia (AML) is a malignant clonal hematopoietic disease with a poor prognosis. Understanding the interaction between leukemic cells and the tumor microenvironment (TME) can help predict the prognosis of leukemia and guide its treatment. Re-analyzing the scRNA-seq data from the CSC and G20 cohorts, using a Python-based pipeline including machine-learning-based scVI-tools, recapitulated the distinct hierarchical structure within the samples of AML patients. Weighted correlation network analysis (WGCNA) was conducted to construct a weighted gene co-expression network and to identify gene modules primarily focusing on hematopoietic stem cells (HSCs), multipotent progenitors (MPPs), and natural killer (NK) cells. The analysis revealed significant deregulation in gene modules associated with aerobic respiration and ribosomal/cytoplasmic translation. Cell-cell communications were elucidated by the CellChat package, revealing an imbalance of activating and inhibitory immune signaling pathways. Interception of genes upregulated in leukemic HSCs & MPPs as well as in NKG2A-high NK cells was used to construct prognostic models. Normal Cox and artificial neural network models based on 10 genes were developed. The study reveals the deregulation of mitochondrial and ribosomal genes in AML patients and suggests the co-occurrence of stimulatory and inhibitory factors in the AML TME.
Collapse
Affiliation(s)
- Chao Ma
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Yuchao Hao
- Department of Hematology, The Second Hospital of Dalian Medical University, West Section Lvshun South Road, Dalian, 116027, Liaoning, China
| | - Bo Shi
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Zheng Wu
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Di Jin
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Xiao Yu
- NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, South Jiefang Road, Taiyuan, 030001, Shanxi, China.
| | - Bilian Jin
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China.
| |
Collapse
|
44
|
Alhaj Hussen K, Louis V, Canque B. A new model of human lymphopoiesis across development and aging. Trends Immunol 2024; 45:495-510. [PMID: 38908962 DOI: 10.1016/j.it.2024.05.007] [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: 05/06/2024] [Revised: 05/25/2024] [Accepted: 05/26/2024] [Indexed: 06/24/2024]
Abstract
Over the past decade our research has implemented a multimodal approach to human lymphopoiesis, combining clonal-scale mapping of lymphoid developmental architecture with the monitoring of dynamic changes in the pattern of lymphocyte generation across ontogeny. We propose that lymphopoiesis stems from founder populations of CD127/interleukin (IL)7R- or CD127/IL7R+ early lymphoid progenitors (ELPs) polarized respectively toward the T-natural killer (NK)/innate lymphoid cell (ILC) or B lineages, arising from newly characterized CD117lo multi-lymphoid progenitors (MLPs). Recent data on the lifelong lymphocyte dynamics of healthy donors suggest that, after birth, lymphopoiesis may become increasingly oriented toward the production of B lymphocytes. Stemming from this, we posit that there are three major developmental transitions, the first occurring during the neonatal period, the next at puberty, and the last during aging.
Collapse
Affiliation(s)
- Kutaiba Alhaj Hussen
- Service de Biochimie, Université de Paris Saclay, Hôpital Paul Brousse, AP-HP, Paris, France
| | - Valentine Louis
- INSERM 1151, Université de Paris, École Pratique des Hautes Études/PSL Research University, Institut Necker Enfants Malades (INEM), Paris, France
| | - Bruno Canque
- INSERM 1151, Université de Paris, École Pratique des Hautes Études/PSL Research University, Institut Necker Enfants Malades (INEM), Paris, France.
| |
Collapse
|
45
|
Vadakekolathu J, Rutella S. Escape from T-cell-targeting immunotherapies in acute myeloid leukemia. Blood 2024; 143:2689-2700. [PMID: 37467496 PMCID: PMC11251208 DOI: 10.1182/blood.2023019961] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023] Open
Abstract
ABSTRACT Single-cell and spatial multimodal technologies have propelled discoveries of the solid tumor microenvironment (TME) molecular features and their correlation with clinical response and resistance to immunotherapy. Computational tools are incessantly being developed to characterize tumor-infiltrating immune cells and to model tumor immune escape. These advances have led to substantial research into T-cell hypofunctional states in the TME and their reinvigoration with T-cell-targeting approaches, including checkpoint inhibitors (CPIs). Until recently, we lacked a high-dimensional picture of the acute myeloid leukemia (AML) TME, including compositional and functional differences in immune cells between disease onset and postchemotherapy or posttransplantation relapse, and the dynamic interplay between immune cells and AML blasts at various maturation stages. AML subgroups with heightened interferon gamma (IFN-γ) signaling were shown to derive clinical benefit from CD123×CD3-bispecific dual-affinity retargeting molecules and CPIs, while being less likely to respond to standard-of-care cytotoxic chemotherapy. In this review, we first highlight recent progress into deciphering immune effector states in AML (including T-cell exhaustion and senescence), oncogenic signaling mechanisms that could reduce the susceptibility of AML cells to T-cell-mediated killing, and the dichotomous roles of type I and II IFN in antitumor immunity. In the second part, we discuss how this knowledge could be translated into opportunities to manipulate the AML TME with the aim to overcome resistance to CPIs and other T-cell immunotherapies, building on recent success stories in the solid tumor field, and we provide an outlook for the future.
Collapse
Affiliation(s)
- Jayakumar Vadakekolathu
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
46
|
Abroon S, Nouri M, Mahdavi M. Hesperidin/Salinomycin Combination; a Natural Product for Deactivation of the PI3K/Akt Signaling Pathway and Anti-Apoptotic Factors in KG1a Cells. J Fluoresc 2024:10.1007/s10895-024-03808-4. [PMID: 38916633 DOI: 10.1007/s10895-024-03808-4] [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: 03/27/2024] [Accepted: 06/06/2024] [Indexed: 06/26/2024]
Abstract
AML is a highly aggressive malignant clonal disease of hematopoietic origin. Hesperidin as a polyphenol glycoside, Activates the apoptotic pathway and salinomycin as a k + selective ionophore. We examined how hesperidin and salinomycin induce pro-apoptotic effects in KG1a cells. Cells were divided into four groups; 1) control cells (CRTL), 2) cells treated with hesperidin 85 μM, 3) cells treated with 2 μM salinomycin, 4) cells treated with combination of salinomycin and hesperidin. The MTT assay was implemented to determine the IC50 of hesperidin and salinomycin in KG1a cell lines. Propidium iodide staining and flow cytometry were used to analyze the distribution of the cell cycle. The level of ROS was evaluated by fluorescent microscopy and spectrophotometry. Additionally, Akt, XIAP, Bad, and FOXO1 gene expression was analyzed by real-time PCR. Hesperidin/Salinomycin decreased the viability of KG1a leukemic cells more than Hesperidin and Salinomycin separately. Changes in the shape of apoptotic cells and rise in ROS levels were detected after Hesperidin/Salinomycin treatment. Our findings showed that following Hesperidin/Salinomycin treatment, the expression of PI3K/AKT signaling pathway related genes (AKT, PTEN and FOXO1), were in line with the destruction of KG-1a cells. Furthermore, XIAP and BAD mRNA were regulated to trigger apoptosis in cancer cells. The study discovered that hesperidin and salinomycin, could effectively hinder the PI3K/Akt signaling pathway in leukemia cancer cells. Also, the combination of hesperidin and salinomycin has the potential to be a treatment option for acute myeloid leukemia.
Collapse
Affiliation(s)
- Sina Abroon
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Department of biochemistry and clinical laboratories, Faculty of medicine, Tabriz University of medical sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of biochemistry and clinical laboratories, Faculty of medicine, Tabriz University of medical sciences, Tabriz, Iran
| | - Majid Mahdavi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
47
|
Caulier B, Joaquina S, Gelebart P, Dowling TH, Kaveh F, Thomas M, Tandaric L, Wernhoff P, Katyayini NU, Wogsland C, Gjerstad ME, Fløisand Y, Kvalheim G, Marr C, Kobold S, Enserink JM, Gjertsen BT, McCormack E, Inderberg EM, Wälchli S. CD37 is a safe chimeric antigen receptor target to treat acute myeloid leukemia. Cell Rep Med 2024; 5:101572. [PMID: 38754420 PMCID: PMC11228397 DOI: 10.1016/j.xcrm.2024.101572] [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: 10/30/2023] [Revised: 03/05/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Acute myeloid leukemia (AML) is characterized by the accumulation of immature myeloid cells in the bone marrow and the peripheral blood. Nearly half of the AML patients relapse after standard induction therapy, and new forms of therapy are urgently needed. Chimeric antigen receptor (CAR) T therapy has so far not been successful in AML due to lack of efficacy and safety. Indeed, the most attractive antigen targets are stem cell markers such as CD33 or CD123. We demonstrate that CD37, a mature B cell marker, is expressed in AML samples, and its presence correlates with the European LeukemiaNet (ELN) 2017 risk stratification. We repurpose the anti-lymphoma CD37CAR for the treatment of AML and show that CD37CAR T cells specifically kill AML cells, secrete proinflammatory cytokines, and control cancer progression in vivo. Importantly, CD37CAR T cells display no toxicity toward hematopoietic stem cells. Thus, CD37 is a promising and safe CAR T cell AML target.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Animals
- Immunotherapy, Adoptive/methods
- Mice
- Tetraspanins/immunology
- Cell Line, Tumor
- T-Lymphocytes/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/immunology
- Female
- Male
- Antigens, Neoplasm
Collapse
Affiliation(s)
- Benjamin Caulier
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway; Institute for Cancer Research, Department of Molecular Cell Biology, Oslo University Hospital, Oslo, Norway; Center for Cancer Cell Reprogramming (CanCell), Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sandy Joaquina
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Pascal Gelebart
- Department of Clinical Science, Precision Oncology Research Group, University of Bergen, 5021 Bergen, Norway; Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
| | - Tara Helén Dowling
- Department of Clinical Science, Precision Oncology Research Group, University of Bergen, 5021 Bergen, Norway; Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway; Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
| | - Fatemeh Kaveh
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Moritz Thomas
- Institue of AI for Health, Helmholtz Munich, 85764 Neuherberg, Germany; School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Luka Tandaric
- Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway; Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Patrik Wernhoff
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Niveditha Umesh Katyayini
- Institute for Cancer Research, Department of Molecular Cell Biology, Oslo University Hospital, Oslo, Norway; Center for Cancer Cell Reprogramming (CanCell), Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Cara Wogsland
- Department of Clinical Science, Precision Oncology Research Group, University of Bergen, 5021 Bergen, Norway; Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
| | - May Eriksen Gjerstad
- Department of Clinical Science, Precision Oncology Research Group, University of Bergen, 5021 Bergen, Norway; Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
| | - Yngvar Fløisand
- Institute for Cancer Research, Department of Molecular Cell Biology, Oslo University Hospital, Oslo, Norway
| | - Gunnar Kvalheim
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Carsten Marr
- Institue of AI for Health, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Translational Cancer Research (DKTK), Partner Site Munich, Munich, Germany; Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Jorrit M Enserink
- Institute for Cancer Research, Department of Molecular Cell Biology, Oslo University Hospital, Oslo, Norway; Center for Cancer Cell Reprogramming (CanCell), Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway; Department of Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Emmet McCormack
- Department of Clinical Science, Precision Oncology Research Group, University of Bergen, 5021 Bergen, Norway; Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway; Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
| | - Else Marit Inderberg
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
48
|
Ortiz Rojas CA, Pereira-Martins DA, Bellido More CC, Sternadt D, Weinhäuser I, Hilberink JR, Coelho-Silva JL, Thomé CH, Ferreira GA, Ammatuna E, Huls G, Valk PJ, Schuringa JJ, Rego EM. A 4-gene prognostic index for enhancing acute myeloid leukaemia survival prediction. Br J Haematol 2024; 204:2287-2300. [PMID: 38651345 DOI: 10.1111/bjh.19472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
Despite advancements in utilizing genetic markers to enhance acute myeloid leukaemia (AML) outcome prediction, significant disease heterogeneity persists, hindering clinical management. To refine survival predictions, we assessed the transcriptome of non-acute promyelocytic leukaemia chemotherapy-treated AML patients from five cohorts (n = 975). This led to the identification of a 4-gene prognostic index (4-PI) comprising CYP2E1, DHCR7, IL2RA and SQLE. The 4-PI effectively stratified patients into risk categories, with the high 4-PI group exhibiting TP53 mutations and cholesterol biosynthesis signatures. Single-cell RNA sequencing revealed enrichment for leukaemia stem cell signatures in high 4-PI cells. Validation across three cohorts (n = 671), including one with childhood AML, demonstrated the reproducibility and clinical utility of the 4-PI, even using cost-effective techniques like real-time quantitative polymerase chain reaction. Comparative analysis with 56 established prognostic indexes revealed the superior performance of the 4-PI, highlighting its potential to enhance AML risk stratification. Finally, the 4-PI demonstrated to be potential marker to reclassified patients from the intermediate ELN2017 category to the adverse category. In conclusion, the 4-PI emerges as a robust and straightforward prognostic tool to improve survival prediction in AML patients.
Collapse
Affiliation(s)
- Cesar Alexander Ortiz Rojas
- Hematology Division, Department of Internal Medicine, Laboratório de Investigação Médica (LIM) 31, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Center for Cell-Based Therapy, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Diego Antonio Pereira-Martins
- Hematology Division, Department of Internal Medicine, Laboratório de Investigação Médica (LIM) 31, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Center for Cell-Based Therapy, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Hematology, Cancer Research Centre Groningen, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Candy Christie Bellido More
- Department of Pediatrics, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dominique Sternadt
- Department of Hematology, Cancer Research Centre Groningen, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Isabel Weinhäuser
- Hematology Division, Department of Internal Medicine, Laboratório de Investigação Médica (LIM) 31, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Center for Cell-Based Therapy, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Hematology, Cancer Research Centre Groningen, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Jacobien R Hilberink
- Department of Hematology, Cancer Research Centre Groningen, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Juan Luiz Coelho-Silva
- Center for Cell-Based Therapy, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carolina Hassibe Thomé
- Hematology Division, Department of Internal Medicine, Laboratório de Investigação Médica (LIM) 31, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Germano Aguiar Ferreira
- Hematology Division, Department of Internal Medicine, Laboratório de Investigação Médica (LIM) 31, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Emanuele Ammatuna
- Department of Hematology, Cancer Research Centre Groningen, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerwin Huls
- Department of Hematology, Cancer Research Centre Groningen, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter J Valk
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan Jacob Schuringa
- Department of Hematology, Cancer Research Centre Groningen, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Eduardo Magalhães Rego
- Hematology Division, Department of Internal Medicine, Laboratório de Investigação Médica (LIM) 31, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Center for Cell-Based Therapy, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
49
|
Karami A, Skorski T. DNA damage response genes as biomarkers of therapeutic outcomes in acute myeloid leukemia patients. Leukemia 2024; 38:1407-1410. [PMID: 38734788 PMCID: PMC11147752 DOI: 10.1038/s41375-024-02269-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024]
Affiliation(s)
- Adam Karami
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Tang J, Luo L, Bosco B, Li N, Huang B, Wu R, Lin Z, Hong M, Liu W, Wu L, Wu W, Zhu M, Liu Q, Xia P, Yu M, Yao D, Lv S, Zhang R, Liu W, Wang Q, Li K. Identification of cell surface markers for acute myeloid leukemia prognosis based on multi-model analysis. J Biomed Res 2024; 38:397-412. [PMID: 38807380 PMCID: PMC11300515 DOI: 10.7555/jbr.38.20240065] [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: 03/18/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
Given the extremely high inter-patient heterogeneity of acute myeloid leukemia (AML), the identification of biomarkers for prognostic assessment and therapeutic guidance is critical. Cell surface markers (CSMs) have been shown to play an important role in AML leukemogenesis and progression. In the current study, we evaluated the prognostic potential of all human CSMs in 130 AML patients from The Cancer Genome Atlas (TCGA) based on differential gene expression analysis and univariable Cox proportional hazards regression analysis. By using multi-model analysis, including Adaptive LASSO regression, LASSO regression, and Elastic Net, we constructed a 9-CSMs prognostic model for risk stratification of the AML patients. The predictive value of the 9-CSMs risk score was further validated at the transcriptome and proteome levels. Multivariable Cox regression analysis showed that the risk score was an independent prognostic factor for the AML patients. The AML patients with high 9-CSMs risk scores had a shorter overall and event-free survival time than those with low scores. Notably, single-cell RNA-sequencing analysis indicated that patients with high 9-CSMs risk scores exhibited chemotherapy resistance. Furthermore, PI3K inhibitors were identified as potential treatments for these high-risk patients. In conclusion, we constructed a 9-CSMs prognostic model that served as an independent prognostic factor for the survival of AML patients and held the potential for guiding drug therapy.
Collapse
Affiliation(s)
- Jiaqi Tang
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu 223300, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lin Luo
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu 223300, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bakwatanisa Bosco
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ning Li
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bin Huang
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Rongrong Wu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zihan Lin
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ming Hong
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Wenjie Liu
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lingxiang Wu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wei Wu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Mengyan Zhu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Quanzhong Liu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Peng Xia
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Miao Yu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Diru Yao
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Sali Lv
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ruohan Zhang
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wentao Liu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qianghu Wang
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210002, China
| | - Kening Li
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu 223300, China
| |
Collapse
|