1
|
Iyer P, Jasdanwala SS, Wang Y, Bhatia K, Bhatt S. Decoding Acute Myeloid Leukemia: A Clinician's Guide to Functional Profiling. Diagnostics (Basel) 2024; 14:2560. [PMID: 39594226 PMCID: PMC11593197 DOI: 10.3390/diagnostics14222560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a complex clonal disorder characterized by clinical, genetic, metabolomic, and epigenetic heterogeneity resulting in the uncontrolled proliferation of aberrant blood-forming precursor cells. Despite advancements in the understanding of the genetic, metabolic, and epigenetic landscape of AML, it remains a significant therapeutic challenge. Functional profiling techniques, such as BH3 profiling (BP), gene expression profiling (GEP), proteomics, metabolomics, drug sensitivity/resistance testing (DSRT), CRISPR/Cas9, and RNAi screens offer valuable insights into the functional behavior of leukemia cells. BP evaluates the mitochondrial response to pro-apoptotic BH3 peptides, determining a cell's apoptotic threshold and its reliance on specific anti-apoptotic proteins. This knowledge can pinpoint vulnerabilities in the mitochondria-mediated apoptotic pathway in leukemia cells, potentially informing treatment strategies and predicting therapeutic responses. GEP, particularly RNA sequencing, evaluates the transcriptomic landscape and identifies gene expression alterations specific to AML subtypes. Proteomics and metabolomics, utilizing mass spectrometry and nuclear magnetic resonance (NMR), provide a detailed view of the active proteins and metabolic pathways in leukemia cells. DSRT involves exposing leukemia cells to a panel of chemotherapeutic and targeted agents to assess their sensitivity or resistance profiles and potentially guide personalized treatment strategies. CRISPR/Cas9 and RNAi screens enable systematic disruption of genes to ascertain their roles in leukemia cell survival and proliferation. These techniques facilitate precise disease subtyping, uncover novel biomarkers and therapeutic targets, and provide a deeper understanding of drug-resistance mechanisms. Recent studies utilizing functional profiling have identified specific mutations and gene signatures associated with aggressive AML subtypes, aberrant signaling pathways, and potential opportunities for drug repurposing. The integration of multi-omics approaches, advances in single-cell sequencing, and artificial intelligence is expected to refine the precision of functional profiling and ultimately improve patient outcomes in AML. This review highlights the diverse landscape of functional profiling methods and emphasizes their respective advantages and limitations. It highlights select successes in how these methods have further advanced our understanding of AML biology, identifies druggable targets that have improved outcomes, delineates challenges associated with these techniques, and provides a prospective view of the future where these techniques are likely to be increasingly incorporated into the routine care of patients with AML.
Collapse
Affiliation(s)
- Prasad Iyer
- Children’s Blood and Cancer Centre, KK Women’s and Children’s Hospital, Singapore 229899, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Shaista Shabbir Jasdanwala
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| | - Yuhan Wang
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| | - Karanpreet Bhatia
- Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| |
Collapse
|
2
|
Pan Q, Xin X, Mahto S, Dong Y, Kumar V, Hyde RK, Gupta N, Bhatt VR, Mahato RI. Anti-CLL1 liposome loaded with miR-497-5p and venetoclax as a novel therapeutic strategy in acute myeloid leukemia. Mol Ther 2024; 32:4058-4074. [PMID: 39369272 PMCID: PMC11573752 DOI: 10.1016/j.ymthe.2024.09.036] [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: 05/19/2024] [Revised: 09/11/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024] Open
Abstract
Acute myeloid leukemia (AML) is a lethal hematologic malignancy. Chemotherapy resistance results in a dismal survival rate of 1-2 years in older adults with AML. Therefore, novel therapies are urgently required. In this context, microRNA (miRNA)-based treatments remain an untapped strategy in AML. Using patient-derived specimens, we found increased inflammatory cytokines, including interleukin-6 (IL-6) in the serum of older adults with AML, and decreased miR-497-5p in CD34+ leukemic blasts. Target prediction revealed that miR-497-5p could directly target mitogen-activated protein kinase-1 (MAP2K1) mRNA to indirectly target cytokines and the JAK/STAT signaling pathway through the p38-MAPK signaling pathway, potentially inhibiting leukemic growth and overcoming chemoresistance from venetoclax. To improve miRNA delivery and minimize off-target effects, which represent key barriers to clinical translation, we developed liposomes for co-delivery of miR-497-5p and venetoclax. We decorated our liposomes with a peptide targeting CLL1, which is present on 92% of leukemia blasts while being absent in normal hematopoietic cells. This targeted approach demonstrated high efficacy in inhibiting AML growth in mice with minimal toxicity, as well as reduced exposure to chemoresistance. Our findings suggested that anti-CLL1-decorated, miR-497-5p, and venetoclax-loaded liposomes represent a promising novel miRNA-based therapeutic, which should be investigated further as a strategy to reduce venetoclax resistance in AML.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/administration & dosage
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Sulfonamides/pharmacology
- Sulfonamides/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Liposomes
- Animals
- Mice
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/administration & dosage
- Disease Models, Animal
- Female
- Drug Resistance, Neoplasm/genetics
- Signal Transduction/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Male
Collapse
Affiliation(s)
- Qiaoyu Pan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Xiaofei Xin
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sohan Mahto
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yuxiang Dong
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - R Katherine Hyde
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Neha Gupta
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Vijaya R Bhatt
- Division of Hematology and Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
3
|
Sauter C, Morin T, Guidez F, Simonet J, Fournier C, Row C, Masnikov D, Pernon B, Largeot A, Aznague A, Hérault Y, Sauvageau G, Maynadié M, Callanan M, Bastie JN, Aucagne R, Delva L. Protein arginine methyltransferase 2 controls inflammatory signaling in acute myeloid leukemia. Commun Biol 2024; 7:753. [PMID: 38902349 PMCID: PMC11190286 DOI: 10.1038/s42003-024-06453-6] [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: 01/19/2024] [Accepted: 06/14/2024] [Indexed: 06/22/2024] Open
Abstract
Arginine methylation is catalyzed by protein arginine methyltransferases (PRMTs) and is involved in various cellular processes, including cancer development. PRMT2 expression is increased in several cancer types although its role in acute myeloid leukemia (AML) remains unknown. Here, we investigate the role of PRMT2 in a cohort of patients with AML, PRMT2 knockout AML cell lines as well as a Prmt2 knockout mouse model. In patients, low PRMT2 expressors are enriched for inflammatory signatures, including the NF-κB pathway, and show inferior survival. In keeping with a role for PRMT2 in control of inflammatory signaling, bone marrow-derived macrophages from Prmt2 KO mice display increased pro-inflammatory cytokine signaling upon LPS treatment. In PRMT2-depleted AML cell lines, aberrant inflammatory signaling has been linked to overproduction of IL6, resulting from a deregulation of the NF-κB signaling pathway, therefore leading to hyperactivation of STAT3. Together, these findings identify PRMT2 as a key regulator of inflammation in AML.
Collapse
Affiliation(s)
- Camille Sauter
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France.
| | - Thomas Morin
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
| | - Fabien Guidez
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
| | - John Simonet
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
| | - Cyril Fournier
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology, Dijon University Hospital, Dijon, France
| | - Céline Row
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology, Dijon University Hospital, Dijon, France
- Department of Hematology Biology, University Hospital Dijon Bourgogne François-Mitterrand, Dijon, France
| | - Denis Masnikov
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
| | - Baptiste Pernon
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
| | - Anne Largeot
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Aziza Aznague
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- Inserm UMS 58 BioSanD, CRISPR Functional Genomics (CRIGEN) facility, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
| | - Yann Hérault
- Université de Strasbourg, CNRS UMR7104, Inserm U1258, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch-Graffenstaden, France
| | - Guy Sauvageau
- Molecular Genetics of Stem Cells, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada
| | - Marc Maynadié
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- Department of Hematology Biology, University Hospital Dijon Bourgogne François-Mitterrand, Dijon, France
| | - Mary Callanan
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology, Dijon University Hospital, Dijon, France
- Inserm UMS 58 BioSanD, CRISPR Functional Genomics (CRIGEN) facility, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
| | - Jean-Noël Bastie
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- Department of Clinical Hematology, University Hospital Dijon Bourgogne François-Mitterrand, Dijon, France
| | - Romain Aucagne
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology, Dijon University Hospital, Dijon, France
- Inserm UMS 58 BioSanD, CRISPR Functional Genomics (CRIGEN) facility, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
| | - Laurent Delva
- Inserm UMR 1231, Epi2THM team, LabEx LipSTIC Team, UFR des Sciences de Santé, Université de Bourgogne, Dijon, France.
| |
Collapse
|
4
|
Peterlin P, Garnier A, Le Bourgeois A, Guillaume T, Le Bris Y, Theisen O, Béné MC, Eveillard M, Rimbert M, Jullien M, Planche L, Gaschet J, Chevallier P. Tocilizumab in combination with a standard induction chemotherapy in acute myeloid leukaemia patients (TOCILAM study): a single-centre, single-arm, phase 1 trial. EClinicalMedicine 2023; 64:102254. [PMID: 37786451 PMCID: PMC10542006 DOI: 10.1016/j.eclinm.2023.102254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/01/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023] Open
Abstract
Background In acute myeloid leukaemia (AML), interleukin-6 (IL-6) promotes chemo-resistance and its levels correlate with poor prognosis. IL-6 blockade may represent a promising therapeutic strategy. We aimed to test, tocilizumab, an anti-IL-6 receptor (R) monoclonal antibody in combination with standard intensive AML induction chemotherapy. Methods This investigator-initiated single-centre phase 1 trial was conducted at Nantes University Hospital in France. According to a continual reassessment method, three escalating doses were tested of intravenous (IV) tocilizumab (4, 6, and 8 mg/kg) administered at day (d) 8 of a standard AML induction chemotherapy (IV idarubicine 8 mg/m2 d1 to d5 + IV cytarabine 100 mg/m2 d1 to d7). All adults (aged ≥ 18 years) with an Eastern Cooperative Oncology Group performance status of 0-2 and with a newly diagnosed (excluding patients with a favourable risk according to ELN-2017 classification if <60 year-old) or a relapsed/refractory AML were eligible. The primary objective was to determine the maximum tolerated dose of tocilizumab to administrate with a standard intensive AML induction. Safety outcomes were continuously monitored for at each participant contact. This trial is registered with ClinicalTrials.gov, NCT04547062. Findings Between Dec 29, 2020 and Dec 1, 2022, 12 patients were enrolled, of whom 75% had an ELN-2017 high-risk profile, and were treated with tocilizumab- two patients at 4 mg/kg, two at 6 mg/kg and eight at 8 mg/kg of tocilizumab. No dose-limiting toxicity related to tocilizumab was documented. There were nine serious adverse events, none of which were related to tocilizumab, and there was no treatment-related deaths. MTD was thus not reached. Two deaths occurred during induction. In the remaining ten evaluable patients, nine responded to treatment. Interpretation The combination of tocilizumab with standard AML intensive induction appears to be safe and resulting responses are encouraging. A dose of 8 mg/kg of tocilizumab given at day 8 of induction could be used for further phase 2/3 studies. Funding The Leucémie Espoir Atlantique Famille (LEAF)-"Tous avec Fabien" association.
Collapse
Affiliation(s)
- Pierre Peterlin
- Clinical Hematology, Nantes University Hospital, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Alice Garnier
- Clinical Hematology, Nantes University Hospital, Nantes, France
| | | | - Thierry Guillaume
- Clinical Hematology, Nantes University Hospital, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Yannick Le Bris
- Biology Hematology, Nantes University Hospital, Nantes, France
| | - Olivier Theisen
- Biology Hematology, Nantes University Hospital, Nantes, France
| | - Marie C. Béné
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
- Biology Hematology, Nantes University Hospital, Nantes, France
| | | | - Marie Rimbert
- Immunology Biology, Nantes University Hospital, Nantes, France
- Centre d'ImmunoMonitorage Nantes-Atlantique (CIMNA), Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN CHU Nantes, Nantes, France
| | - Maxime Jullien
- Clinical Hematology, Nantes University Hospital, Nantes, France
| | - Lucie Planche
- Clinical Research Centre, Departmental Hospital Centre, La Roche sur Yon, France
| | - Joelle Gaschet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Patrice Chevallier
- Clinical Hematology, Nantes University Hospital, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| |
Collapse
|
5
|
Luciano M, Krenn PW, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia. Front Immunol 2022; 13:1000996. [PMID: 36248849 PMCID: PMC9554002 DOI: 10.3389/fimmu.2022.1000996] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous malignancy of the blood and bone marrow, characterized by clonal expansion of myeloid stem and progenitor cells and rapid disease progression. Chemotherapy has been the first-line treatment for AML for more than 30 years. Application of recent high-throughput next-generation sequencing technologies has revealed significant molecular heterogeneity to AML, which in turn has motivated efforts to develop new, targeted therapies. However, due to the high complexity of this disease, including multiple driver mutations and the coexistence of multiple competing tumorigenic clones, the successful incorporation of these new agents into clinical practice remains challenging. These continuing difficulties call for the identification of innovative therapeutic approaches that are effective for a larger cohort of AML patients. Recent studies suggest that chronic immune stimulation and aberrant cytokine signaling act as triggers for AML initiation and progression, facets of the disease which might be exploited as promising targets in AML treatment. However, despite the greater appreciation of cytokine profiles in AML, the exact functions of cytokines in AML pathogenesis are not fully understood. Therefore, unravelling the molecular basis of the complex cytokine networks in AML is a prerequisite to develop new therapeutic alternatives based on targeting cytokines and their receptors.
Collapse
Affiliation(s)
- Michela Luciano
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Peter W. Krenn
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
6
|
Kierdorf F, Zimmer P, Joisten N, Kiesl D, Winker M, Rigoux L, Elter T. L-kynurenine as a prognostic marker for early mortality in patients with acute myeloid leukemia. Leuk Lymphoma 2022; 63:2001-2004. [DOI: 10.1080/10428194.2022.2053534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Fiona Kierdorf
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sport Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund, Germany
| | - Niklas Joisten
- Division of Performance and Health (Sport Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund, Germany
| | - David Kiesl
- Department of Hematology and Internal Oncology, Kepler University Linz, Linz, Austria
| | - Matteo Winker
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Lionel Rigoux
- Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Thomas Elter
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Köln, Germany
| |
Collapse
|
7
|
Improving prediction accuracy in acute myeloid leukaemia: micro-environment, immune and metabolic models. Leukemia 2021; 35:3073-3077. [PMID: 34365474 PMCID: PMC8550966 DOI: 10.1038/s41375-021-01377-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 02/02/2023]
|
8
|
Gurney M, O’Dwyer M. Realizing Innate Potential: CAR-NK Cell Therapies for Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:1568. [PMID: 33805422 PMCID: PMC8036691 DOI: 10.3390/cancers13071568] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Next-generation cellular immunotherapies seek to improve the safety and efficacy of approved CD19 chimeric antigen receptor (CAR) T-cell products or apply their principles across a growing list of targets and diseases. Supported by promising early clinical experiences, CAR modified natural killer (CAR-NK) cell therapies represent a complementary and potentially off-the-shelf, allogeneic solution. While acute myeloid leukemia (AML) represents an intuitive disease in which to investigate CAR based immunotherapies, key biological differences to B-cell malignancies have complicated progress to date. As CAR-T cell trials treating AML are growing in number, several CAR-NK cell approaches are also in development. In this review we explore why CAR-NK cell therapies may be particularly suited to the treatment of AML. First, we examine the established role NK cells play in AML biology and the existing anti-leukemic activity of NK cell adoptive transfer. Next, we appraise potential AML target antigens and consider common and unique challenges posed relative to treating B-cell malignancies. We summarize the current landscape of CAR-NK development in AML, and potential targets to augment CAR-NK cell therapies pharmacologically and through genetic engineering. Finally, we consider the broader landscape of competing immunotherapeutic approaches to AML treatment. In doing so we evaluate the innate potential, status and remaining barriers for CAR-NK based AML immunotherapy.
Collapse
Affiliation(s)
- Mark Gurney
- Apoptosis Research Center, National University of Ireland Galway, H91 TK33 Galway, Ireland;
| | - Michael O’Dwyer
- Apoptosis Research Center, National University of Ireland Galway, H91 TK33 Galway, Ireland;
- ONK Therapeutics Ltd., H91 V6KV Galway, Ireland
| |
Collapse
|