1
|
Lee J, Kang H, Chen B, Na Y, Khatri I, Soares F, He HH, Law AD, Pan T, Gerbitz A, Zhu X, Minden MD, Zhang L. Allogeneic DNT cell therapy synergizes with T cells to promote anti-leukemic activities while suppressing GvHD. J Exp Clin Cancer Res 2025; 44:28. [PMID: 39876025 PMCID: PMC11773727 DOI: 10.1186/s13046-024-03247-w] [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: 08/28/2024] [Accepted: 12/02/2024] [Indexed: 01/30/2025] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a second-line treatment with curative potential for leukemia patients. However, the prognosis of allo-HSCT patients with disease relapse or graft-versus-host disease (GvHD) is poor. CD4+ or CD8+ conventional T (Tconv) cells are critically involved in mediating anti-leukemic immune responses to prevent relapse and detrimental GvHD. Hence, treatment for one increases the risk of the other. Thus, therapeutic strategies that can address relapse and GvHD are considered the Holy Grail of allo-HSCT. CD3+CD4-CD8- double-negative T cells (DNTs) are unconventional mature T cells with potent anti-leukemia effects with "off-the-shelf" potential. A phase I clinical trial demonstrated the feasibility, safety, and potential efficacy of allogeneic DNT therapy for patients with relapsing acute myeloid leukemia (AML) post-allo-HSCT. Here, we studied the impact of DNTs on the anti-leukemic and GvHD-inducing activities of Tconv cells. DNTs synergized with Tconv cells to mediate superior anti-leukemic activity. Mechanistically, DNTs released soluble factors which activated and evoked potent anti-leukemic activities of Tconv cells. In contrast, DNTs suppressed GvHD-inducing activities of Tconv cells in a CD18-dependent manner by mediating cytotoxicity against proliferative Tconv cells. The seemingly opposite immunological activities of DNTs were dictated by the presence or absence of AML cells. Collectively, these results support the potential of DNTs as an adjuvant to allo-HSCT to address both disease relapse and GvHD.
Collapse
Affiliation(s)
- Jongbok Lee
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Hyeonjeong Kang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Branson Chen
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Yoosu Na
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Ismat Khatri
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Fraser Soares
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Housheng Hansen He
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Arjun D Law
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Tianzhong Pan
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Armin Gerbitz
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xiaoyu Zhu
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Li Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Jin P, Shen J, Zhao M, Yu J, Jin W, Jiang G, Li Z, He M, Liu X, Wu S, Dong F, Cao Y, Zhu H, Li X, Wang X, Zhang Y, Jin Z, Li J, Wang K. Driver mutation landscape of acute myeloid leukemia provides insights for neoantigen-based immunotherapy. Cancer Lett 2024; 611:217427. [PMID: 39725148 DOI: 10.1016/j.canlet.2024.217427] [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: 09/16/2024] [Revised: 11/27/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Acute myeloid leukemia (AML) has lagged in benefiting from immunotherapies, primarily due to the scarcity of actionable AML-specific antigens. Driver mutations represent promising immunogenic targets, but a comprehensive characterization of the AML neoantigen landscape and their impact on patient outcomes and the AML immune microenvironment remain unclear. Herein, we conducted matched DNA and RNA sequencing on 304 AML patients and extensively integrated data from additional ∼2500 AML cases, identifying 49 driver genes, notably characterized by a significant proportion of insertions and deletions (indels). Neoantigen analysis showed that indels triggered a higher abundance of neoantigens both in quantity and quality compared to single nucleotide variants (SNVs) and gene fusions. By integrating peptide features pertinent to neoantigen presentation and T cell recognition, we developed two robust models of epitope immunogenicity that significantly enriched immunogenic neoepitopes. We validated 30 neoantigens through in vitro direct binding assays of predicted peptides to MHC proteins and confirmed the immunogenicity of 20 neoantigens using interferon-γ ELISpot and tetramer assays. Moreover, we demonstrated that patients with higher neoantigen loads, derived from driver mutations, exhibited poor clinical outcomes and an IFN-driven adaptive immune response, which was associated with immune suppression and tumor evasion. Through deconvolution of large-scale bulk transcriptomes, integration of single-cell RNA sequencing and multiparametric flow cytometry, we confirmed a strong association between neoantigen load and CD8+ T cell exhaustion. This study provides a comprehensive landscape of AML neoantigens derived from driver mutations, offering putative immunogenic targets and emphasizing the need for strategies to revitalize the immunosuppressive milieu.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Ming 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; School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jinyi Yu
- 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
| | - Wen 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; CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ge Jiang
- 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
| | - 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
| | - 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
| | - 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
| | - Yuncan Cao
- 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
| | - 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
| | - Xiaoling Wang
- Department of Reproductive Medical Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yunxiang Zhang
- 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, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Kankan Wang
- 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; CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Radich J. Mutations and MRD: clinical implications of clonal ontogeny. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:150-157. [PMID: 39644022 DOI: 10.1182/hematology.2024000541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Measurable residual disease (MRD) is a strong but imprecise predictor of relapse in acute myeloid leukemia. Many patients fall into the outlier categories of MRD positivity without relapse or MRD negativity with relapse. Why? We will discuss these states in the context of "clonal ontogeny" examining how mutations, clonal structure, and Darwinian rules impact response, resistance, and relapse.
Collapse
Affiliation(s)
- Jerald Radich
- Translational Science & Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
4
|
Ebelt ND, Loganathan S, Avsharian LC, Manuel ER. Effects of Hypomethylating Agents on Gene Modulation in the Leukemic Microenvironment and Disease Trajectory in a Mouse Model of AML. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626276. [PMID: 39677768 PMCID: PMC11642732 DOI: 10.1101/2024.12.01.626276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Hypomethylating agents (HMAs), such as decitabine and 5-azacytidine (AZA), are valuable treatment options for patients with acute myeloid leukemia that are ineligible for intensive chemotherapy. Despite providing significant extensions in survival when used alone or in combination, eventual relapse and resistance to HMAs are observed. The mechanisms leading to these outcomes are still not well defined and may, in part, be due to a focus on leukemic populations with limited information on the effects of HMAs on non-leukemic cells in the blood and other tissue compartments. In this study, we elucidated effects on immune-related gene expression in non-leukemic blood cells and the spleen during AZA treatment in leukemia-challenged mice. We observed significant changes in pathways regulating adhesion, thrombosis, and angiogenesis as well as a dichotomy in extramedullary disease sites that manifests during relapse. We also identify several genes that may contribute to the anti-leukemic activity of AZA in blood and spleen. Overall, this work has identified novel gene targets and pathways that could be further modulated to augment efficacy of HMA treatment.
Collapse
Affiliation(s)
- Nancy D. Ebelt
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | - Lara C. Avsharian
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010, USA
| | - Edwin R. Manuel
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
5
|
Della Porta MG, Martinelli G, Rambaldi A, Santoro A, Voso MT. A practical algorithm for acute myeloid leukaemia diagnosis following the updated 2022 classifications. Crit Rev Oncol Hematol 2024; 198:104358. [PMID: 38615870 DOI: 10.1016/j.critrevonc.2024.104358] [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: 11/02/2023] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024] Open
Abstract
Disease classification of complex and heterogenous diseases, such as acute myeloid leukaemia (AML), is continuously updated to define diagnoses, appropriate treatments, and assist research and education. Recent availability of molecular profiling techniques further benefits the classification of AML. The World Health Organization (WHO) classification of haematolymphoid tumours and the International Consensus Classification of myeloid neoplasms and acute leukaemia from 2022 are two updated versions of the WHO 2016 classification. As a consequence, the European LeukemiaNet 2022 recommendations on the diagnosis and management of AML in adults have been also updated. The current review provides a practical interpretation of these guidelines to facilitate the diagnosis of AML and discusses genetic testing, disease genetic heterogeneity, and FLT3 mutations. We propose a practical algorithm for the speedy diagnosis of AML. Future classifications may need to incorporate gene mutation combinations to enable personalised treatment regimens in the management of patients with AML.
Collapse
Affiliation(s)
- Matteo Giovanni Della Porta
- Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy.
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Armadori", Meldola, Italy; University of Bologna, Bologna, Italy
| | - Alessandro Rambaldi
- Department of Oncology and Hematology, University of Milan, Milan, Italy and Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Alessandra Santoro
- UOSD Laboratory of Oncohematology, Cellular Manipulation and Cytogenetics, Department of Genetic, Oncohematology a Rare Disease, AOR "Villa Sofia-Cervello", Palermo, Italy
| | - Maria Teresa Voso
- UOSD Diagnostica Avanzata Oncoematologia, Policlinico Tor Vergata, and Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
6
|
Pino JC, Posso C, Joshi SK, Nestor M, Moon J, Hansen JR, Hutchinson-Bunch C, Gritsenko MA, Weitz KK, Watanabe-Smith K, Long N, McDermott JE, Druker BJ, Liu T, Tyner JW, Agarwal A, Traer E, Piehowski PD, Tognon CE, Rodland KD, Gosline SJC. Mapping the proteogenomic landscape enables prediction of drug response in acute myeloid leukemia. Cell Rep Med 2024; 5:101359. [PMID: 38232702 PMCID: PMC10829797 DOI: 10.1016/j.xcrm.2023.101359] [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: 05/25/2023] [Revised: 10/20/2023] [Accepted: 12/10/2023] [Indexed: 01/19/2024]
Abstract
Acute myeloid leukemia is a poor-prognosis cancer commonly stratified by genetic aberrations, but these mutations are often heterogeneous and fail to consistently predict therapeutic response. Here, we combine transcriptomic, proteomic, and phosphoproteomic datasets with ex vivo drug sensitivity data to help understand the underlying pathophysiology of AML beyond mutations. We measure the proteome and phosphoproteome of 210 patients and combine them with genomic and transcriptomic measurements to identify four proteogenomic subtypes that complement existing genetic subtypes. We build a predictor to classify samples into subtypes and map them to a "landscape" that identifies specific drug response patterns. We then build a drug response prediction model to identify drugs that target distinct subtypes and validate our findings on cell lines representing various stages of quizartinib resistance. Our results show how multiomics data together with drug sensitivity data can inform therapy stratification and drug combinations in AML.
Collapse
Affiliation(s)
- James C Pino
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Camilo Posso
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Sunil K Joshi
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Michael Nestor
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jamie Moon
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Joshua R Hansen
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Chelsea Hutchinson-Bunch
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Marina A Gritsenko
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Karl K Weitz
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Kevin Watanabe-Smith
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Nicola Long
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Jason E McDermott
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA; Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Brian J Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA; Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Tao Liu
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jeffrey W Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA; Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Elie Traer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Paul D Piehowski
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Cristina E Tognon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Karin D Rodland
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
| | - Sara J C Gosline
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
7
|
García-Álvarez M, Yeguas A, Jiménez C, Medina-Herrera A, González-Calle V, Hernández-Ruano M, Maldonado R, Aires I, Casquero C, Sánchez-Villares I, Balanzategui A, Sarasquete ME, Alcoceba M, Vidriales MB, González-Díaz M, García-Sanz R, Chillón MC. Single-Cell DNA Sequencing and Immunophenotypic Profiling to Track Clonal Evolution in an Acute Myeloid Leukemia Patient. Biomedicines 2023; 12:66. [PMID: 38255173 PMCID: PMC10813288 DOI: 10.3390/biomedicines12010066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Single-cell DNA sequencing can address the sequence of somatic genetic events during myeloid transformation in relapsed acute myeloid leukemia (AML). We present an NPM1-mutated AML patient with an initial low ratio of FLT3-ITD (low-risk ELN-2017), treated with midostaurin combined with standard chemotherapy as front-line treatment, and with salvage therapy plus gilteritinib following allogenic stem cell transplantation after relapse. Simultaneous single-cell DNA sequencing and cell-surface immunophenotyping was used in diagnostic and relapse samples to understand the clinical scenario of this patient and to reconstruct the clonal composition of both tumors. Four independent clones were present before treatment: DNMT3A/DNMT3A/NPM1 (63.9%), DNMT3A/DNMT3A (13.9%), DNMT3A/DNMT3A/NPM1/FLT3 (13.8%), as well as a wild-type clone (8.3%), but only the minor clone with FLT3-ITD survived and expanded after therapy, being the most represented one (58.6%) at relapse. FLT3-ITD was subclonal and was found only in the myeloid blast population (CD38/CD117/CD123). Our study shows the usefulness of this approach to reveal the clonal architecture of the leukemia and the identification of small subclones at diagnosis and relapse that may explain how the neoplastic cells can escape from the activity of different treatments in a stepwise process that impedes the disease cure despite different stages of complete remission.
Collapse
Affiliation(s)
- María García-Álvarez
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Ana Yeguas
- Hematology Department, Complejo Asistencial Universitario de Palencia, 34005 Palencia, Spain;
| | - Cristina Jiménez
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Alejandro Medina-Herrera
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Verónica González-Calle
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Montserrat Hernández-Ruano
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Rebeca Maldonado
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Irene Aires
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Cristina Casquero
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Inmaculada Sánchez-Villares
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Ana Balanzategui
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - María Eugenia Sarasquete
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Miguel Alcoceba
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - María Belén Vidriales
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Marcos González-Díaz
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| | - María Carmen Chillón
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.G.-Á.); (C.J.); (A.M.-H.); (V.G.-C.); (M.H.-R.); (R.M.); (I.A.); (C.C.); (I.S.-V.); (A.B.); (M.E.S.); (M.A.); (M.B.V.); (M.G.-D.); (M.C.C.)
| |
Collapse
|
8
|
Arévalo C, Rojas L, Santamaria M, Molina L, Arbeláez L, Sánchez P, Ballesteros-Ramírez R, Arevalo-Zambrano M, Quijano S, Cala MP, Fiorentino S. Untargeted metabolomic and lipidomic analyses reveal lipid dysregulation in the plasma of acute leukemia patients. Front Mol Biosci 2023; 10:1235160. [PMID: 38028534 PMCID: PMC10667492 DOI: 10.3389/fmolb.2023.1235160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Acute leukemias (AL) are aggressive neoplasms with high mortality rates. Metabolomics and oxidative status have emerged as important tools to identify new biomarkers with clinical utility. To identify the metabolic differences between healthy individuals (HI) and patients with AL, a multiplatform untargeted metabolomic and lipidomic approach was conducted using liquid and gas chromatography coupled with quadrupole-time-of-flight mass spectrometry (LC-QTOF-MS or GC-QTOF-MS). Additionally, the total antioxidant capacity (TAC) was measured. A total of 20 peripheral blood plasma samples were obtained from patients with AL and 18 samples from HI. Our analysis revealed 135 differentially altered metabolites in the patients belonging to 12 chemical classes; likewise, the metabolic pathways of glycerolipids and sphingolipids were the most affected in the patients. A decrease in the TAC of the patients with respect to the HI was evident. This study conducted with a cohort of Colombian patients is consistent with observations from other research studies that suggest dysregulation of lipid compounds. Furthermore, metabolic differences between patients and HI appear to be independent of lifestyle, race, or geographic location, providing valuable information for future advancements in understanding the disease and developing more global therapies.
Collapse
Affiliation(s)
- Cindy Arévalo
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Laura Rojas
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Mary Santamaria
- MetCore—Metabolomics Core Facility, Vice-Presidency for Research, Universidad de Los Andes, Bogotá, Colombia
| | | | - Lina Arbeláez
- Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Paula Sánchez
- Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Ricardo Ballesteros-Ramírez
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
- Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Mónica P. Cala
- MetCore—Metabolomics Core Facility, Vice-Presidency for Research, Universidad de Los Andes, Bogotá, Colombia
| | - Susana Fiorentino
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
9
|
Chianese U, Papulino C, Megchelenbrink W, Tambaro FP, Ciardiello F, Benedetti R, Altucci L. Epigenomic machinery regulating pediatric AML: clonal expansion mechanisms, therapies, and future perspectives. Semin Cancer Biol 2023; 92:84-101. [PMID: 37003397 DOI: 10.1016/j.semcancer.2023.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/07/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a genetic, epigenetic, and transcriptional etiology mainly presenting somatic and germline abnormalities. AML incidence rises with age but can also occur during childhood. Pediatric AML (pAML) accounts for 15-20% of all pediatric leukemias and differs considerably from adult AML. Next-generation sequencing technologies have enabled the research community to "paint" the genomic and epigenomic landscape in order to identify pathology-associated mutations and other prognostic biomarkers in pAML. Although current treatments have improved the prognosis for pAML, chemoresistance, recurrence, and refractory disease remain major challenges. In particular, pAML relapse is commonly caused by leukemia stem cells that resist therapy. Marked patient-to-patient heterogeneity is likely the primary reason why the same treatment is successful for some patients but, at best, only partially effective for others. Accumulating evidence indicates that patient-specific clonal composition impinges significantly on cellular processes, such as gene regulation and metabolism. Although our understanding of metabolism in pAML is still in its infancy, greater insights into these processes and their (epigenetic) modulation may pave the way toward novel treatment options. In this review, we summarize current knowledge on the function of genetic and epigenetic (mis)regulation in pAML, including metabolic features observed in the disease. Specifically, we describe how (epi)genetic machinery can affect chromatin status during hematopoiesis, leading to an altered metabolic profile, and focus on the potential value of targeting epigenetic abnormalities in precision and combination therapy for pAML. We also discuss the possibility of using alternative epidrug-based therapeutic approaches that are already in clinical practice, either alone as adjuvant treatments and/or in combination with other drugs.
Collapse
Affiliation(s)
- Ugo Chianese
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Chiara Papulino
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Wout Megchelenbrink
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy; Princess Máxima Center, Heidelberglaan 25, 3584 CS, Utrecht, the Netherlands.
| | - Francesco Paolo Tambaro
- Bone Marrow Transplant Unit, Pediatric Oncology Department AORN Santobono Pausilipon, 80129, Naples Italy.
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy; Biogem Institute of Molecular and Genetic Biology, 83031 Ariano Irpino, Italy; IEOS, Institute for Endocrinology and Oncology "Gaetano Salvatore" (IEOS), 80131 Naples, Italy.
| |
Collapse
|
10
|
Serroukh Y, Hébert J, Busque L, Mercier F, Rudd CE, Assouline S, Lachance S, Delisle JS. Blasts in context: the impact of the immune environment on acute myeloid leukemia prognosis and treatment. Blood Rev 2023; 57:100991. [PMID: 35941029 DOI: 10.1016/j.blre.2022.100991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
Acute myeloid leukemia (AML) is a cancer that originates from the bone marrow (BM). Under physiological conditions, the bone marrow supports the homeostasis of immune cells and hosts memory lymphoid cells. In this review, we summarize our present understanding of the role of the immune microenvironment on healthy bone marrow and on the development of AML, with a focus on T cells and other lymphoid cells. The types and function of different immune cells involved in the AML microenvironment as well as their putative role in the onset of disease and response to treatment are presented. We also describe how the immune context predicts the response to immunotherapy in AML and how these therapies modulate the immune status of the bone marrow. Finally, we focus on allogeneic stem cell transplantation and summarize the current understanding of the immune environment in the post-transplant bone marrow, the factors associated with immune escape and relevant strategies to prevent and treat relapse.
Collapse
Affiliation(s)
- Yasmina Serroukh
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Erasmus Medical center Cancer Institute, University Medical Center Rotterdam, Department of Hematology, Rotterdam, the Netherlands; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada.
| | - Josée Hébert
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada; The Quebec Leukemia Cell Bank, Canada
| | - Lambert Busque
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - François Mercier
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, 3755 Côte-Sainte-Catherine Road, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte-Sainte-Catherine Road, Montreal, Canada
| | - Christopher E Rudd
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Sarit Assouline
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, 3755 Côte-Sainte-Catherine Road, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte-Sainte-Catherine Road, Montreal, Canada
| | - Silvy Lachance
- Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Jean-Sébastien Delisle
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| |
Collapse
|
11
|
Redavid I, Conserva MR, Anelli L, Zagaria A, Specchia G, Musto P, Albano F. Single-Cell Sequencing: Ariadne’s Thread in the Maze of Acute Myeloid Leukemia. Diagnostics (Basel) 2022; 12:diagnostics12040996. [PMID: 35454044 PMCID: PMC9024495 DOI: 10.3390/diagnostics12040996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 02/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is a haematological neoplasm resulting from the accumulation of genetic and epigenetic alterations. Patients’ prognoses vary with AML genetic heterogeneity, which hampers successful treatments. Single-cell approaches have provided new insights of the clonal architecture of AML, revealing the mutational history from diagnosis, during treatment and to relapse. In this review, we imagine single-cell technologies as the Ariadne’s thread that will guide us out of the AML maze, provide a precise identikit of the leukemic cell at single-cell resolution and explore genomic, transcriptomic, epigenetic and proteomic levels.
Collapse
Affiliation(s)
- Immacolata Redavid
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (I.R.); (M.R.C.); (L.A.); (A.Z.); (P.M.)
| | - Maria Rosa Conserva
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (I.R.); (M.R.C.); (L.A.); (A.Z.); (P.M.)
| | - Luisa Anelli
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (I.R.); (M.R.C.); (L.A.); (A.Z.); (P.M.)
| | - Antonella Zagaria
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (I.R.); (M.R.C.); (L.A.); (A.Z.); (P.M.)
| | - Giorgina Specchia
- School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| | - Pellegrino Musto
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (I.R.); (M.R.C.); (L.A.); (A.Z.); (P.M.)
| | - Francesco Albano
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (I.R.); (M.R.C.); (L.A.); (A.Z.); (P.M.)
- Correspondence:
| |
Collapse
|
12
|
Vicente-Garcés C, Esperanza-Cebollada E, Montesdeoca S, Torrebadell M, Rives S, Dapena JL, Català A, Conde N, Camós M, Vega-García N. Technical Validation and Clinical Utility of an NGS Targeted Panel to Improve Molecular Characterization of Pediatric Acute Leukemia. Front Mol Biosci 2022; 9:854098. [PMID: 35463953 PMCID: PMC9021638 DOI: 10.3389/fmolb.2022.854098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 12/03/2022] Open
Abstract
Development of next-generation sequencing (NGS) has provided useful genetic information to redefine diagnostic, prognostic, and therapeutic strategies for the management of acute leukemia (AL). However, the application in the clinical setting is still challenging. Our aim was to validate the AmpliSeq™ for Illumina® Childhood Cancer Panel, a pediatric pan-cancer targeted NGS panel that includes the most common genes associated with childhood cancer, and assess its utility in the daily routine of AL diagnostics. In terms of sequencing metrics, the assay reached all the expected values. We obtained a mean read depth greater than 1000×. The panel demonstrated a high sensitivity for DNA (98.5% for variants with 5% variant allele frequency (VAF)) and RNA (94.4%), 100% of specificity and reproducibility for DNA and 89% of reproducibility for RNA. Regarding clinical utility, 49% of mutations and 97% of the fusions identified were demonstrated to have clinical impact. Forty-one percent of mutations refined diagnosis, while 49% of them were considered targetable. Regarding RNA, fusion genes were more clinically impactful in terms of refining diagnostic (97%). Overall, the panel found clinically relevant results in the 43% of patients tested in this cohort. To sum up, we validated a reliable and reproducible method to refine pediatric AL diagnosis, prognosis, and treatment, and demonstrated the feasibility of incorporating a targeted NGS panel into pediatric hematology practice.
Collapse
Affiliation(s)
- Clara Vicente-Garcés
- Hematology Laboratory, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Barcelona, Spain
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Elena Esperanza-Cebollada
- Hematology Laboratory, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Barcelona, Spain
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Sara Montesdeoca
- Hematology Laboratory, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Barcelona, Spain
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Montserrat Torrebadell
- Hematology Laboratory, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Barcelona, Spain
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Susana Rives
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Hematology and Oncology Department, Hospital Sant Joan de Déu Barcelona, University of Barcelona, Barcelona, Spain
| | - José Luis Dapena
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Pediatric Hematology and Oncology Department, Hospital Sant Joan de Déu Barcelona, University of Barcelona, Barcelona, Spain
| | - Albert Català
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Hematology and Oncology Department, Hospital Sant Joan de Déu Barcelona, University of Barcelona, Barcelona, Spain
| | - Nuria Conde
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Pediatric Hematology and Oncology Department, Hospital Sant Joan de Déu Barcelona, University of Barcelona, Barcelona, Spain
| | - Mireia Camós
- Hematology Laboratory, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Barcelona, Spain
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Nerea Vega-García
- Hematology Laboratory, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Barcelona, Spain
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- *Correspondence: Nerea Vega-García,
| |
Collapse
|
13
|
Savanur MA, Weinstein-Marom H, Gross G. Implementing Logic Gates for Safer Immunotherapy of Cancer. Front Immunol 2021; 12:780399. [PMID: 34804073 PMCID: PMC8600566 DOI: 10.3389/fimmu.2021.780399] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022] Open
Abstract
Targeting solid tumors with absolute precision is a long-standing challenge in cancer immunotherapy. The identification of antigens, which are expressed by a large fraction of tumors of a given type and, preferably, across various types, but not by normal cells, holds the key to developing safe, off-the-shelf immunotherapies. Although the quest for widely shared, strictly tumor-specific antigens has been the focus of tremendous effort, only few such candidates have been implicated. Almost all antigens that are currently explored as targets for chimeric antigen receptor (CAR) or T cell receptor (TCR)-T cell therapy are also expressed by healthy cells and the risk of on-target off-tumor toxicity has remained a major concern. Recent studies suggest that this risk could be obviated by targeting instead combinations of two or more antigens, which are co-expressed by tumor but not normal cells and, as such, are tumor-specific. Moreover, the expression of a shared tumor antigen along with the lack of a second antigen that is expressed by normal tissues can also be exploited for precise recognition. Additional cues, antigenic or non-antigenic ones, which characterize the tumor microenvironment, could be harnessed to further increase precision. This review focuses on attempts to define the targetable signatures of tumors and assesses different strategies employing advanced synthetic biology for translating such information into safer modes of immunotherapy, implementing the principles of Boolean logic gates.
Collapse
Affiliation(s)
- Mohammed Azharuddin Savanur
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Hadas Weinstein-Marom
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Gideon Gross
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| |
Collapse
|
14
|
Grandits AM, Wieser R. Gene expression changes contribute to stemness and therapy resistance of relapsed acute myeloid leukemia: roles of SOCS2, CALCRL, MTSS1, and KDM6A. Exp Hematol 2021; 99:1-11. [PMID: 34029637 PMCID: PMC7612147 DOI: 10.1016/j.exphem.2021.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022]
Abstract
Relapse is associated with therapy resistance and is a major cause of death in acute myeloid leukemia (AML). It is thought to result from the accretion of therapy-refractory leukemic stem cells. Genetic and transcriptional changes that are recurrently gained at relapse are likely to contribute to the increased stemness and decreased therapy responsiveness at this disease stage. Despite the recent approval of several targeted drugs, chemotherapy with cytosine arabinoside and anthracyclines is still the mainstay of AML therapy. Accordingly, a number of studies have investigated genetic and gene expression changes between diagnosis and relapse of patients subjected to such treatment. Genetic alterations recurrently acquired at relapse were identified, but were restricted to small proportions of patients, and their functional characterization is still largely pending. In contrast, the expression of a substantial number of genes was altered consistently between diagnosis and recurrence of AML. Recent studies corroborated the roles of the upregulation of SOCS2 and CALCRL and of the downregulation of MTSS1 and KDM6A in therapy resistance and/or stemness of AML. These findings spur the assumption that functional investigations of genes consistently altered at recurrence of AML have the potential to promote the development of novel targeted drugs that may help to improve the outcome of this currently often fatal disease.
Collapse
Affiliation(s)
- Alexander M Grandits
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Vienna, Austria
| | - Rotraud Wieser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Vienna, Austria.
| |
Collapse
|
15
|
Epigenetic Effects of Benzene in Hematologic Neoplasms: The Altered Gene Expression. Cancers (Basel) 2021; 13:cancers13102392. [PMID: 34069279 PMCID: PMC8156840 DOI: 10.3390/cancers13102392] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Benzene is produced by diverse petroleum transformation processes and it is widely employed in industry despite its oncogenic effects. In fact, occupational exposure to benzene may cause hematopoietic malignancy. The leukemogenic action of benzene is particularly complex. Possible processes of onset of hematological malignancies have been recognized as a genotoxic action and the provocation of immunosuppression. However, benzene can induce modifications that do not involve alterations in the DNA sequence, the so-called epigenetics changes. Acquired epigenetic modification may also induce leukemogenesis, as benzene may alter nuclear receptors, and cause changes at the protein level, thereby modifying the function of regulatory proteins, including oncoproteins and tumor suppressor proteins. Abstract Benzene carcinogenic ability has been reported, and chronic exposure to benzene can be one of the risk elements for solid cancers and hematological neoplasms. Benzene is acknowledged as a myelotoxin, and it is able to augment the risk for the onset of acute myeloid leukemia, myelodysplastic syndromes, aplastic anemia, and lymphomas. Possible mechanisms of benzene initiation of hematological tumors have been identified, as a genotoxic effect, an action on oxidative stress and inflammation and the provocation of immunosuppression. However, it is becoming evident that genetic alterations and the other causes are insufficient to fully justify several phenomena that influence the onset of hematologic malignancies. Acquired epigenetic alterations may participate with benzene leukemogenesis, as benzene may affect nuclear receptors, and provoke post-translational alterations at the protein level, thereby touching the function of regulatory proteins, comprising oncoproteins and tumor suppressor proteins. DNA hypomethylation correlates with stimulation of oncogenes, while the hypermethylation of CpG islands in promoter regions of specific tumor suppressor genes inhibits their transcription and stimulates the onset of tumors. The discovery of the systems of epigenetic induction of benzene-caused hematological tumors has allowed the possibility to operate with pharmacological interventions able of stopping or overturning the negative effects of benzene.
Collapse
|
16
|
Sendker S, Waack K, Reinhardt D. Far from Health: The Bone Marrow Microenvironment in AML, A Leukemia Supportive Shelter. CHILDREN (BASEL, SWITZERLAND) 2021; 8:371. [PMID: 34066861 PMCID: PMC8150304 DOI: 10.3390/children8050371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/28/2022]
Abstract
Acute myeloid leukemia (AML) is the second most common leukemia among children. Although significant progress in AML therapy has been achieved, treatment failure is still associated with poor prognosis, emphasizing the need for novel, innovative therapeutic approaches. To address this major obstacle, extensive knowledge about leukemogenesis and the complex interplay between leukemic cells and their microenvironment is required. The tremendous role of this bone marrow microenvironment in providing a supportive and protective shelter for leukemic cells, leading to disease development, progression, and relapse, has been emphasized by recent research. It has been revealed that the interplay between leukemic cells and surrounding cellular as well as non-cellular components is critical in the process of leukemogenesis. In this review, we provide a comprehensive overview of recently gained knowledge about the importance of the microenvironment in AML whilst focusing on promising future therapeutic targets. In this context, we describe ongoing clinical trials and future challenges for the development of targeted therapies for AML.
Collapse
Affiliation(s)
| | | | - Dirk Reinhardt
- Department of Pediatric Hematology and Oncology, Clinic of Pediatrics III, Essen University Hospital, 45147 Essen, Germany; (S.S.); (K.W.)
| |
Collapse
|