1
|
Nguyen NHK, Rafiee R, Parcha PK, Tagmount A, Rubnitz J, Ribeiro R, Cao X, Pounds SB, Vulpe CD, Lamba JK. Genome-wide CRISPR/Cas9 screen identifies AraC-daunorubicin-etoposide response modulators associated with outcomes in pediatric AML. Blood Adv 2025; 9:1078-1091. [PMID: 39715471 PMCID: PMC11914169 DOI: 10.1182/bloodadvances.2024014157] [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: 07/29/2024] [Revised: 11/18/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
ABSTRACT Cytarabine, daunorubicin, and etoposide (ADE) have been the standard backbone of induction chemotherapy regimen for patients with pediatric acute myeloid leukemia (pAML) for >5 decades. However, chemoresistance is still a major concern, and a significant proportion of pAML becomes resistant to ADE treatment and relapse, leading to poor survival. Therefore, there is a considerable need to identify mechanisms mediating drug resistance for overcoming chemoresistance. Herein, we performed synthetic lethal CRISPR/Cas9 screens using the ADE components to identify response markers. We further integrated significant markers in 3 independent pAML clinical cohorts treated with only an ADE regimen to identify drug response biomarkers with prognostic significance. We were able to identify several mediators that represent clinically and biologically significant marker genes for ADE treatment, such as BCL2, CLIP2, and VAV3, which are resistant markers to ADE, with high expression associated with poor outcomes in pAML treated with ADE, and GRPEL1, HCFC1, and TAF10, which are sensitive markers to ADE, with high expression showing beneficial outcomes. Notably, BCL2, CLIP2, and VAV3 knockdowns in their expression in AML cell lines sensitized the cells more to the ADE components, suggesting that these modulators should be further studied as potential therapeutic targets to overcome chemoresistance.
Collapse
Affiliation(s)
- Nam H. K. Nguyen
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| | - Roya Rafiee
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| | - Phani K. Parcha
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| | - Abderrahmane Tagmount
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL
| | - Jeffrey Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Raul Ribeiro
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Xueyuan Cao
- Departments of Health Promotion and Disease Prevention, and Preventive Medicine, The University of Tennessee Health Science Center, Memphis, TN
| | - Stanley B. Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Christopher D. Vulpe
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL
| | - Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| |
Collapse
|
2
|
Iyer P. Pediatric AML: state of the Art and Future Directions. Pediatr Hematol Oncol 2025; 42:126-145. [PMID: 39889807 DOI: 10.1080/08880018.2025.2453861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
Pediatric acute myeloid leukemia (AML) is a heterogeneous and aggressive hematological malignancy. Despite advances in treatment, the survival rates remain unsatisfactory, emphasizing the need for innovative therapeutic approaches. This narrative review presents a comprehensive overview of the current approach and likely future directions for pediatric AML. The distinct genetic, epigenetic, and molecular features of pediatric AML contribute to its complex pathophysiology and impact on prognosis. Current treatment practices involve a multifaceted approach combining chemotherapy, molecularly targeted therapies, and hematopoietic stem cell transplantation. However, intensive treatment often leads to significant acute and long-term toxicity. Emerging strategies, including precision medicine, immunotherapy, and novel agents, hold promise for improving outcomes and minimizing adverse effects. Ongoing clinical trials are investigating the potential of these innovative approaches to transform pediatric AML care. By highlighting the evolving treatment paradigms and future perspectives, this review underscores the importance of continued research and development in pediatric AML to enhance the survival rates and quality of life of these young patients.
Collapse
Affiliation(s)
- Prasad Iyer
- Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore
- Duke NUS Medical School, Singapore
| |
Collapse
|
3
|
Rao M, Luo W, Luo C, Wu B, Xu T, Wei Z, Deng H, Li K, Zhou D. Prognostic factors and outcomes in pediatric acute myeloid leukemia: a comprehensive bibliometric analysis of global research trends. Front Oncol 2025; 15:1466818. [PMID: 40034590 PMCID: PMC11873564 DOI: 10.3389/fonc.2025.1466818] [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: 07/26/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pediatric AML prognosis research has advanced significantly, yet gaps in understanding genetic and molecular interactions persist. Despite improved outcomes, relapse/refractory cases and personalized treatment integration remain critical clinical challenges. Objective To analyze the global research landscape on pediatric AML prognosis, highlight influential components and collaborations, and identify major potential research trends. Methods Publications on pediatric AML prognosis research from 1999 to 2023 were retrieved from the Clarivate Analytics Web of Science Core Collection (WoSCC) database. Bibliometric analysis was conducted using CiteSpace and VOSviewer to identify leading countries, prominent institutions, high-impact journals, key research categories, influential authors, and emerging research topics. Results The bibliometric analysis encompassed 924 publications, with St. Jude Children's Research Hospital emerging as the most prolific institution. The United States leads globally in terms of countries, institutions, journals, and authors. Todd A. Alonzo ranks highest in publication volume, while U. Creutzig leads in citations. The top research categories were Oncology, Hematology, and Pediatrics. Key research topics included genomics, transcriptomics, epigenomics, targeted therapies, immune therapy, and integrative diagnostic approaches. Conclusion This bibliometric analysis highlights significant advancements in pediatric AML prognosis over the past 25 years, driven by the integration of genetic markers, immunological insights, transcriptomics, and epigenomics, which have collectively transformed risk stratification and treatment strategies. Overcoming challenges, such as discovering new therapeutic targets and enhancing treatment combinations, will depend on global collaboration and advanced technologies to propel the field forward.
Collapse
Affiliation(s)
- Mingliang Rao
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenna Luo
- Department of Laboratory Medicine, Heyuan People’s Hospital, Heyuan, China
| | - Caiju Luo
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Baojing Wu
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tiantian Xu
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ziqian Wei
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haolan Deng
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kejing Li
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dunhua Zhou
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Yotsomnuk P, Rajendran AP, Sundaram DNM, Morales LC, Kucharski C, Nasrullah M, Skolpap W, Jiang X, Gibson SB, Brandwein J, Uludağ H. Lipopolymers as the Basis of Non-Viral Delivery of Therapeutic siRNA Nanoparticles in a Leukemia (MOLM-13) Model. Biomolecules 2025; 15:115. [PMID: 39858509 PMCID: PMC11763671 DOI: 10.3390/biom15010115] [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: 10/22/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Small interfering RNA (siRNA) therapy in acute myeloid leukemia (AML) is a promising strategy as the siRNA molecule can specifically target proteins involved in abnormal cell proliferation. The development of a clinically applicable method for delivering siRNA molecules is imperative due to the challenges involved in effectively delivering the siRNA into cells. We investigated the delivery of siRNA to AML MOLM-13 cells with the use of two lipid-substituted polyethyleneimines (PEIs), a commercially available reagent (Prime-Fect) and a recently reported reagent with improved lipid substitution (PEI1.2k-PHPA-Lin9). The siRNAs utilized in this study were targeting the oncogenes FLT3 and KMT2A::MLLT3. Both lipopolymers gave similar-size siRNA complexes (210-220 nm) with positive ζ-potentials (+17 to +25 mV). While the binding efficiency of both lipopolymers to siRNA were similar, PEI1.2k-PHPA-Lin9 complexes were more resistant to heparin-induced dissociation. The quantitative analysis of gene silencing performed by qPCR as well as immunostaining/flow cytometry indicated significant reduction in both FLT3 expression and FLT3 protein after specific siRNA delivery. The desired inhibition of cell growth was attained with both FLT3 and KMT2A::MLLT3 siRNAs, and the combination provided more potent effects in both cell growth and colony formation assays. Induction of apoptosis was confirmed after specific siRNA treatments using the Annexin V assay. Using Luc(+) MOLM-13 cells, the growth of the xenografted cells was shown to be retarded with Prime-Fect-delivered FLT3 siRNA, unlike the siRNA delivered with PEI1.2k-PHPA-Lin9. These results demonstrate the potential of designed lipopolymers in implementing RNAi (via delivery of siRNA) for inhibition of leukemia growth and provide evidence for the feasibility of targeting different oncogenes using siRNA-mediated therapy.
Collapse
MESH Headings
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/pharmacology
- Humans
- Animals
- Nanoparticles/chemistry
- Mice
- Cell Line, Tumor
- Polyethyleneimine/chemistry
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors
- Lipids/chemistry
- Polymers/chemistry
- Gene Silencing
- Histone-Lysine N-Methyltransferase
Collapse
Affiliation(s)
- Panadda Yotsomnuk
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
- Department of Chemical Engineering, Faculty of Engineering, Thammasat University, Pathumthani 12120, Thailand
| | - Amarnath Praphakar Rajendran
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
| | - Daniel Nisakar Meenakshi Sundaram
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
| | - Luis Carlos Morales
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
| | - Cezary Kucharski
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
| | - Mohammad Nasrullah
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H1, Canada
| | - Wanwisa Skolpap
- Department of Chemical Engineering, Faculty of Engineering, Thammasat University, Pathumthani 12120, Thailand
| | - Xiaoyan Jiang
- Department of Medical Genetics, Terry Fox Laboratory, British Columbia Cancer Research Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Spencer B. Gibson
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Joseph Brandwein
- Division of Hematology, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Hasan Uludağ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H1, Canada
| |
Collapse
|
5
|
Kato S, Tsujimoto SI, Matsubayashi J, Iwamoto S, Hiramatsu H, Okuno Y, Kamitori T, Ohki K, Deguchi T, Kiyokawa N, Kato M, Takita J, Tanaka S, Adachi S, Tomizawa D, Shiba N. Adverse prognostic impact of KIT exon 17 mutations despite negative flow cytometric measurable residual disease in pediatric acute myeloid leukemia with RUNX1::RUNX1T1. Haematologica 2025; 110:251-256. [PMID: 39234860 PMCID: PMC11694115 DOI: 10.3324/haematol.2024.286243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Shota Kato
- Department of Pediatrics, Graduate School of Medicine, the University of Tokyo, Tokyo
| | - Shin-Ichi Tsujimoto
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama
| | - Jun Matsubayashi
- Center for Clinical Research and Advanced Medicine, Shiga University of Medical Science, Otsu
| | - Shotaro Iwamoto
- Department of Pediatrics, Graduate School of Medicine Mie University, Tsu
| | - Hidefumi Hiramatsu
- Department of Pediatrics, Graduate School of Medicine Kyoto University, Kyoto
| | - Yusuke Okuno
- Department of Virology, Nagoya City University Graduate School of Medical Sciences, Nagoya
| | - Tatsuya Kamitori
- Department of Pediatrics, Graduate School of Medicine Kyoto University, Kyoto
| | - Kentaro Ohki
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo
| | - Takao Deguchi
- Division of Cancer Immunodiagnostics, Children's Cancer Center, National Center for Child Health and Development, Tokyo
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo
| | - Motohiro Kato
- Department of Pediatrics, Graduate School of Medicine, the University of Tokyo, Tokyo
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine Kyoto University, Kyoto
| | - Shiro Tanaka
- Department of Clinical Biostatistics, Graduate School of Medicine, Kyoto University, Kyoto
| | - Souichi Adachi
- Human Health Science, Graduate School of Medicine Kyoto University, Kyoto, Japan; and
| | - Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo
| | - Norio Shiba
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama.
| |
Collapse
|
6
|
Lamba J, Marchi F, Landwehr M, Schade AK, Shastri V, Ghavami M, Sckaff F, Marrero R, Nguyen N, Mansinghka V, Cao X, Slayton W, Starostik P, Ribeiro R, Rubnitz J, Klco J, Gamis A, Triche T, Ries R, Kolb EA, Aplenc R, Alonzo T, Pounds S, Meshinchi S, Cogle C, Elsayed A. Long-read epigenomic diagnosis and prognosis of Acute Myeloid Leukemia. RESEARCH SQUARE 2024:rs.3.rs-5450972. [PMID: 39711573 PMCID: PMC11661290 DOI: 10.21203/rs.3.rs-5450972/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Acute Myeloid Leukemia (AML) is an aggressive cancer with dismal outcomes, vast subtype heterogeneity, and suboptimal risk stratification. In this study, we harmonized DNA methylation data from 3,314 patients across 11 cohorts to develop the Acute Leukemia Methylome Atlas (ALMA) of diagnostic relevance that predicted 27 WHO 2022 acute leukemia subtypes with an overall accuracy of 96.3% in discovery and 90.1% in validation cohorts. Specifically, for AML, we also developed AML Epigenomic Risk, a prognostic classifier of overall survival (OS) (HR=4.40; 95% CI=3.45-5.61; P<0.0001), and a targeted 38CpG AML signature using a stepwise EWAS-CoxPH-LASSO model predictive of OS (HR=3.84; 95% CI=3.01-4.91; P<0.0001). Finally, we developed a specimen-to-result protocol for simultaneous whole-genome and epigenome sequencing that accurately predicted diagnoses and prognoses from twelve prospectively collected patient samples using long-read sequencing. Our study unveils a new paradigm in acute leukemia management by leveraging DNA methylation for diagnostic and prognostic applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xueyuan Cao
- University of Tennessee Health Science Center
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
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
|
8
|
Hashii Y, Kawaguchi K, Kurakami H, Umeda K, Hasegawa D, Taki T, Hyakuna N, Ishida H, Takahashi Y, Nagasawa M, Yabe H, Yano M, Nakazawa Y, Fujisaki H, Matsumoto K, Yanagimachi M, Yoshida N, Kakuda H, Satou A, Tabuchi K, Tomizawa D, Taga T, Adachi S, Koh K, Kato K. A Retrospective Study of Pediatric Patients With Low- or Intermediate-Risk Acute Myeloid Leukemia Who Underwent Allogeneic Hematopoietic Cell Transplantation for the AML-05 Study Conducted by the Japanese Pediatric Leukemia/Lymphoma Study Group. Transplant Cell Ther 2024; 30:1102.e1-1102.e12. [PMID: 39181536 DOI: 10.1016/j.jtct.2024.08.011] [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: 05/30/2024] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024]
Abstract
The AML-05 study aimed to examine the efficacy and safety of a therapeutic strategy based on risk stratification for low-, intermediate-, or high-risk acute myeloid leukemia (AML) pediatric patients. Allogeneic hematopoietic cell transplantation (allo-HCT) was not indicated for low- or intermediate-risk AML patients in first complete remission. The present retrospective study for the AML-05 study aimed to identify prognostic factors for survival and to determine optimal allo-HCT according to multivariate analysis on overall survival (OS), event-free survival (EFS), cumulative incidence of relapse (CIR), and cumulative incidence of nonrelapse mortality for and between low- and intermediate-risk AML group patients in the AML-05 study who had undergone allo-HCT after its completion and relapse. The unique patient numbers (UPNs) of the AML-05 study were matched with the Transplant Registry Unified Management Program (TRUMP)-registered numbers, and the tied data on the AML-05 study's UPNs and the TRUMP-registered numbers were analyzed. The primary endpoint was 3-yr OS. Among 443 AML patients in the AML-05 study, 79 (32 low-risk AML and 47 intermediate-risk AML) were analyzed. The following statistically favorable prognostic factors were identified by multivariate analysis on the low- and intermediate-risk AML groups, respectively: UCB (OS-hazard ratio [HR], 0.105; 95% CI, 0.011 to 0.941; P = .004 and EFS-HR, 0.065, 95% CI, 0.007 to 0.577, P = .014) and late relapse (OS-HR, 0.212; 95% CI, 0.072 to 0.626; P = .005 and EFS-HR, 0.236; 95% CI, 0.088 to 0.630; P = .004). Three-year OS, 3-yr EFS, and 3-yr CIR were significantly different between the low- and intermediate-risk AML groups. UCB may be a safe and beneficial donor source for low-risk AML patients, while late relapse was a favorable prognostic factor for intermediate-risk AML patients. Intermediate-risk AML patients with late relapse and low-risk AML patients may benefit from allo-HCT after relapse.
Collapse
Affiliation(s)
- Yoshiko Hashii
- Department of Pediatrics, Osaka International Cancer Institute, Osaka, Japan; Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Koji Kawaguchi
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Hiroyuki Kurakami
- Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Tomohiko Taki
- Department of Medical Technology, Kyorin University Faculty of Health Sciences, Mitaka, Japan
| | - Nobuyuki Hyakuna
- Department of Pediatrics, University of the Ryukyus Hospital, Nishihara, Japan
| | | | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masayuki Nagasawa
- Department of Pediatrics, Musashino Red Cross Hospital, Musashino, Japan
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Michihiro Yano
- Department of Pediatrics, Akita University Hospital, Akita, Japan
| | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hiroyuki Fujisaki
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Kimikazu Matsumoto
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Masakatsu Yanagimachi
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Nao Yoshida
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Aichi Medical Center Nagoya First Hospital, Nagoya, Japan
| | - Harumi Kakuda
- Department of Hematology/Oncology, Chiba Children's Hospital, Chiba, Japan
| | - Atsushi Satou
- Department of Hematology and Oncology, Miyagi Children's Hospital, Sendai, Japan
| | - Ken Tabuchi
- Japanese Data Center for Hematopoietic Cell Transplantation
| | - Daisuke Tomizawa
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsuyoshi Koh
- Department of Hematology and Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Koji Kato
- Central Japan Cord Blood Bank, Seto, Japan
| |
Collapse
|
9
|
Pawińska-Wąsikowska K, Czogała M, Bukowska-Strakova K, Surman M, Rygielska M, Książek T, Sadowska B, Pac A, Skalska-Sadowska J, Samborska M, Wachowiak J, Ciebiera M, Chaber R, Tomaszewska R, Szczepański T, Zielezińska K, Urasiński T, Rodziewicz-Konarska A, Kałwak K, Kozłowska M, Irga-Jaworska N, Sikorska-Fic B, Chyżyński B, Łaguna P, Muszyńska-Rosłan K, Krawczuk-Rybak M, Deleszkiewicz P, Drabko K, Bobeff K, Młynarski W, Chodała-Grzywacz A, Karolczyk G, Mycko K, Badowska W, Bartoszewicz N, Styczyński J, Machnik K, Stolpa W, Mizia-Malarz A, Balwierz W, Skoczeń S. Analysis of early and treatment related deaths among children and adolescents with acute myeloid leukemia in Poland: 2005-2023. Front Pediatr 2024; 12:1482720. [PMID: 39483533 PMCID: PMC11524810 DOI: 10.3389/fped.2024.1482720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Background A personalised approach to the treatment of acute myeloid leukemia (AML) in children and adolescents, as well as the development of supportive therapies, has significantly improved survival. Despite this, some patients still die before starting treatment or in an early phase of therapy before achieving remission. The study analysed the frequency, clinical features and risk factors for early deaths (ED) and treatment related deaths (TRD) of children and adolescents with AML. Methods From January 2005 to November 2023, 646 children with AML treated in the centers of the Polish Pediatric Leukemia and Lymphoma Study Group according to three subsequent therapeutic protocols were evaluated: AML-BFM 2004 Interim (385 children), AML-BFM 2012 Registry (131 children) and AML-BFM 2019 (130 children). Results Out of 646 children, early death occurred in 30 children, including 15 girls. The median age was 10.7 years (1 day to 18 years). More than half of the patients (53%) were diagnosed with acute myelomonocytic leukemia (M5) and 13% with acute promyelocytic leukemia (M3). The ED rate for the three consecutive AML-BFM protocols was 4.9% vs. 5.3% vs. 3.1%, respectively. In 19 patients, death occurred before the 15th day of treatment, in 11 between the 15th and 42nd day. The most common cause of death before the 15th day (ED15) was leukostasis and bleeding, whereas between the 15th and 42nd day (ED15-42), infections, mainly bacterial sepsis. A significant association was found between ED15 and high leukocyte count (>10 × 109/L), M3 leukemia (p < 0.001), and ED15-42 and age <1 year (p = 0.029). In the univariate analysis only initial high leukocyte count >100 × 109/L, was a significant predictor of early death. The overall TRD for the entire study period was 3.4%. The main cause of death were infections, mainly bacterial sepsis (10 children out of 22, 45.4%). Conclusions Hyperleukocytosis remains significant factor of early mortality in patients with AML, despite the introduction of various cytoreductive methods. Infections are still the main cause of treatment related deaths. A more individualized approach by using new targeted drugs may be the therapeutic option of choice in the future.
Collapse
Affiliation(s)
- Katarzyna Pawińska-Wąsikowska
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children Hospital of Krakow, Krakow, Poland
| | - Małgorzata Czogała
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children Hospital of Krakow, Krakow, Poland
| | - Karolina Bukowska-Strakova
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Marta Surman
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Monika Rygielska
- Department of Pediatric Oncology and Hematology, Hematology Laboratory, University Children’s Hospital, Krakow, Poland
| | - Teofila Książek
- Department of Molecular Genetics, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Beata Sadowska
- Department of Pediatric Oncology and Hematology, Cytogenetics and Molecular Genetics Laboratory, University Children’s Hospital, Krakow, Poland
| | - Agnieszka Pac
- Department of Epidemiology and Preventive Medicine, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Jolanta Skalska-Sadowska
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| | - Magdalena Samborska
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jacek Wachowiak
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| | - Małgorzata Ciebiera
- Department of Pediatric Oncohematology, Clinical Province Hospital of Rzeszow, Rzeszow, Poland
- Department of Pediatrics, Institute of Medical Sciences, Medical College, University of Rzeszow, Rzeszow, Poland
| | - Radosław Chaber
- Department of Pediatric Oncohematology, Clinical Province Hospital of Rzeszow, Rzeszow, Poland
- Department of Pediatrics, Institute of Medical Sciences, Medical College, University of Rzeszow, Rzeszow, Poland
| | - Renata Tomaszewska
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia, Katowice, Poland
| | - Tomasz Szczepański
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia, Katowice, Poland
| | - Karolina Zielezińska
- Department of Pediatrics, Hemato-Oncology and Gastroenterology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Tomasz Urasiński
- Department of Pediatrics, Hemato-Oncology and Gastroenterology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Anna Rodziewicz-Konarska
- Clinical Department of Pediatric Bone Marrow Transplantation, Oncology and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Kałwak
- Clinical Department of Pediatric Bone Marrow Transplantation, Oncology and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Marta Kozłowska
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdansk, Gdansk, Poland
| | - Ninela Irga-Jaworska
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdansk, Gdansk, Poland
| | - Barbara Sikorska-Fic
- Department of Oncology, Pediatric Hematology, Transplantology and Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Bartosz Chyżyński
- Department of Oncology, Pediatric Hematology, Transplantology and Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Paweł Łaguna
- Department of Oncology, Pediatric Hematology, Transplantology and Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | | | - Maryna Krawczuk-Rybak
- Departament of Pediatrics, Oncology and Hematology Medical University of Bialystok, Bialystok, Poland
| | - Paulina Deleszkiewicz
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Katarzyna Drabko
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Katarzyna Bobeff
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | - Wojciech Młynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | | | - Grażyna Karolczyk
- Department of Pediatric Hematology and Oncology, Regional Polyclinic Hospital in Kielce, Kielce, Poland
| | - Katarzyna Mycko
- Department of Pediatrics and Hematology and Oncology, Province Children’s Hospital, Olsztyn, Poland
| | - Wanda Badowska
- Department of Pediatrics and Hematology and Oncology, Province Children’s Hospital, Olsztyn, Poland
| | - Natalia Bartoszewicz
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University Torun, Bydgoszcz, Poland
| | - Jan Styczyński
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University Torun, Bydgoszcz, Poland
| | - Katarzyna Machnik
- Department of Pediatrics, Hematology and Oncology, City Hospital, Chorzow, Poland
| | - Weronika Stolpa
- Department of Pediatrics, Upper Silesia Children’s Care Health Centre, Medical University of Silesia, Katowice, Poland
| | - Agnieszka Mizia-Malarz
- Department of Pediatrics, Upper Silesia Children’s Care Health Centre, Medical University of Silesia, Katowice, Poland
| | - Walentyna Balwierz
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children Hospital of Krakow, Krakow, Poland
| | - Szymon Skoczeń
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children Hospital of Krakow, Krakow, Poland
| |
Collapse
|
10
|
Marrero RJ, Wu H, Cao X, Parcha PK, Elsayed AH, Inaba H, Kuo DJ, Degar BA, Heym K, Taub JW, Lacayo N, Pui CH, Ribeiro RC, Rubnitz JE, Pounds SB, Lamba JK. Pharmacogenomic Score Effectively Personalizes Treatment of Acute Myeloid Leukemia. Clin Cancer Res 2024; 30:4388-4396. [PMID: 39078289 PMCID: PMC11444877 DOI: 10.1158/1078-0432.ccr-24-0863] [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: 03/15/2024] [Revised: 04/23/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024]
Abstract
PURPOSE Cytarabine (also known as ara-C) has been the backbone of acute myeloid leukemia (AML) chemotherapy for more than five decades. Recent pharmacogenomics-based 10-SNP ara-C (ACS10) scores showed low ACS10 (≤0) to be associated with poor outcomes in patients with AML treated with standard chemotherapy. Here, we evaluated the ACS10 score in the context of three different induction I regimens in patients with pediatric AML. EXPERIMENTAL DESIGN ACS10 score groups (low, ≤0, or high, >0) were evaluated for association with event-free survival (EFS) and overall survival (OS) by three randomized treatment arms in patients treated on the AML02 (NCT00136084) and AML08 (NCT00703820) clinical trials: AML02 low-dose ara-C (LDAC arm, n = 91), AML02 + AML08 high-dose ara-C (HDAC arm, n = 194), and AML08 clofarabine + ara-C (Clo/ara-C arm, n = 105) induction I regimens. RESULTS Within the low-ACS10 score (≤0) group, significantly improved EFS and OS were observed among patients treated with Clo/ara-C as compared with LDAC (EFS, HR = 0.45; 95% CI, 0.23-0.88; P = 0.020; OS, HR = 0.44; 95% CI, 0.19-0.99; P = 0.048). In contrast, within the high-ACS10 score group (score >0), augmentation with Clo/ara-C was not favorable as compared with LDAC (Clo/ara-C vs. LDAC, EFS, HR = 1.95; 95% CI, 1.05-3.63; P = 0.035; OS, HR = 2.10; 95% CI, 0.96-4.59; P = 0.063). Personalization models predicted 9% improvement in the outcome in ACS10 score-based tailored induction (Clo/ara-C for low and LDAC for high-ACS10 score groups) as compared with nonpersonalized approaches (P < 0.002). CONCLUSIONS Our findings suggest that tailoring induction regimens using ACS10 scores can significantly improve outcomes in patients with AML. Given the SNPs are germline, preemptive genotyping can accelerate matching the most effective remission induction regimen.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Cytarabine/therapeutic use
- Cytarabine/administration & dosage
- Female
- Male
- Child
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Adolescent
- Precision Medicine/methods
- Child, Preschool
- Polymorphism, Single Nucleotide
- Pharmacogenetics/methods
- Infant
- Prognosis
Collapse
Affiliation(s)
- Richard J. Marrero
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Huiyun Wu
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Xueyuan Cao
- Department of Health Promotion and Disease Prevention, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Phani Krishna Parcha
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Abdelrahman H. Elsayed
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Dennis John Kuo
- Division of Pediatric Hematology-Oncology, Rady Children’s Hospital San Diego/University of California, San Diego, CA, USA
| | - Barbara A. Degar
- Dana-Farber Cancer Institute, Boston Children’s Hospital, Boston, MA, USA
| | - Kenneth Heym
- Cook Children’s Medical Center, Fort Worth, TX, USA
| | | | - Norman Lacayo
- Lucile Packard Children’s Hospital and Stanford Cancer Institute, Palo Alto, CA, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Raul C. Ribeiro
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jeffrey E. Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Stanley B. Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL, USA
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
11
|
H Elsayed A, Cao X, Marrero RJ, Nguyen NHK, Wu H, Ni Y, Ribeiro RC, Tobias H, Valk PJ, Béliveau F, Richard-Carpentier G, Hébert J, Zwaan CM, Gamis A, Kolb EA, Aplenc R, Alonzo TA, Meshinchi S, Rubnitz J, Pounds S, Lamba JK. Integrated drug resistance and leukemic stemness gene-expression scores predict outcomes in large cohort of over 3500 AML patients from 10 trials. NPJ Precis Oncol 2024; 8:168. [PMID: 39090192 PMCID: PMC11294346 DOI: 10.1038/s41698-024-00643-5] [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: 01/05/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024] Open
Abstract
In this study, we leveraged machine-learning tools by evaluating expression of genes of pharmacological relevance to standard-AML chemotherapy (ara-C/daunorubicin/etoposide) in a discovery-cohort of pediatric AML patients (N = 163; NCT00136084 ) and defined a 5-gene-drug resistance score (ADE-RS5) that was predictive of outcome (high MRD1 positivity p = 0.013; lower EFS p < 0.0001 and OS p < 0.0001). ADE-RS5 was integrated with a previously defined leukemic-stemness signature (pLSC6) to classify patients into four groups. ADE-RS5, pLSC6 and integrated-score was evaluated for association with outcome in one of the largest assembly of ~3600 AML patients from 10 independent cohorts (1861 pediatric and 1773 adult AML). Patients with high ADE-RS5 had poor outcome in validation cohorts and the previously reported pLSC6 maintained strong significant association in all validation cohorts. For pLSC6/ADE-RS5-integrated-score analysis, using Group-1 (low-scores for ADE-RS5 and pLSC6) as reference, Group-4 (high-scores for ADE-RS5 and pLSC6) showed worst outcome (EFS: p < 0.0001 and OS: p < 0.0001). Groups-2/3 (one high and one low-score) showed intermediate outcome (p < 0.001). Integrated score groups remained an independent predictor of outcome in multivariable-analysis after adjusting for established prognostic factors (EFS: Group 2 vs. 1, HR = 4.68, p < 0.001, Group 3 vs. 1, HR = 3.22, p = 0.01, and Group 4 vs. 1, HR = 7.26, p < 0.001). These results highlight the significant prognostic value of transcriptomics-based scores capturing disease aggressiveness through pLSC6 and drug resistance via ADE-RS5. The pLSC6 stemness score is a significant predictor of outcome and associates with high-risk group features, the ADE-RS5 drug resistance score adds further value, reflecting the clinical utility of simultaneous testing of both for optimizing treatment strategies.
Collapse
Affiliation(s)
- Abdelrahman H Elsayed
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Xueyuan Cao
- Department of Health Promotion and Disease Prevention, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Richard J Marrero
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Nam H K Nguyen
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Huiyun Wu
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yonhui Ni
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Raul C Ribeiro
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Herold Tobias
- Department of Medicine III, Ludwig Maximillans University Hospital, LMU Munich, Germany
| | - Peter J Valk
- Department of Hematology, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - François Béliveau
- Quebec leukemia cell bank, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Guillaume Richard-Carpentier
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of Medical Oncology and Hematology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Josée Hébert
- Quebec leukemia cell bank, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Division of Hematology and Oncology, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Alan Gamis
- Division of Hematology/Oncology, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Edward Anders Kolb
- Nemours Center for Cancer and Blood Disorders, Alfred I. DuPont Hospital for Children, Wilmington, DE, USA
| | - Richard Aplenc
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Todd A Alonzo
- COG Statistics and Data Center, Monrovia, CA, USA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jeffrey Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jatinder K Lamba
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA.
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
van Weelderen RE, Harrison CJ, Klein K, Jiang Y, Abrahamsson J, Alonzo T, Aplenc R, Arad-Cohen N, Bart-Delabesse E, Buldini B, De Moerloose B, Dworzak MN, Elitzur S, Fernández Navarro JM, Gamis A, Gerbing RB, Goemans BF, de Groot-Kruseman HA, Guest E, Ha SY, Hasle H, Kelaidi C, Lapillonne H, Leverger G, Locatelli F, Miyamura T, Norén-Nyström U, Polychronopoulou S, Rasche M, Rubnitz JE, Stary J, Tierens A, Tomizawa D, Zwaan CM, Kaspers GJL. Optimized cytogenetic risk-group stratification of KMT2A-rearranged pediatric acute myeloid leukemia. Blood Adv 2024; 8:3200-3213. [PMID: 38621200 PMCID: PMC11225675 DOI: 10.1182/bloodadvances.2023011771] [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: 09/24/2023] [Revised: 03/04/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
ABSTRACT A comprehensive international consensus on the cytogenetic risk-group stratification of KMT2A-rearranged (KMT2A-r) pediatric acute myeloid leukemia (AML) is lacking. This retrospective (2005-2016) International Berlin-Frankfurt-Münster Study Group study on 1256 children with KMT2A-r AML aims to validate the prognostic value of established recurring KMT2A fusions and additional cytogenetic aberrations (ACAs) and to define additional, recurring KMT2A fusions and ACAs, evaluating their prognostic relevance. Compared with our previous study, 3 additional, recurring KMT2A-r groups were defined: Xq24/KMT2A::SEPT6, 1p32/KMT2A::EPS15, and 17q12/t(11;17)(q23;q12). Across 13 KMT2A-r groups, 5-year event-free survival probabilities varied significantly (21.8%-76.2%; P < .01). ACAs occurred in 46.8% of 1200 patients with complete karyotypes, correlating with inferior overall survival (56.8% vs 67.9%; P < .01). Multivariable analyses confirmed independent associations of 4q21/KMT2A::AFF1, 6q27/KMT2A::AFDN, 10p12/KMT2A::MLLT10, 10p11.2/KMT2A::ABI1, and 19p13.3/KMT2A::MLLT1 with adverse outcomes, but not those of 1q21/KMT2A::MLLT11 and trisomy 19 with favorable and adverse outcomes, respectively. Newly identified ACAs with independent adverse prognoses were monosomy 10, trisomies 1, 6, 16, and X, add(12p), and del(9q). Among patients with 9p22/KMT2A::MLLT3, the independent association of French-American-British-type M5 with favorable outcomes was confirmed, and those of trisomy 6 and measurable residual disease at end of induction with adverse outcomes were identified. We provide evidence to incorporate 5 adverse-risk KMT2A fusions into the cytogenetic risk-group stratification of KMT2A-r pediatric AML, to revise the favorable-risk classification of 1q21/KMT2A::MLLT11 to intermediate risk, and to refine the risk-stratification of 9p22/KMT2A::MLLT3 AML. Future studies should validate the associations between the newly identified ACAs and outcomes and unravel the underlying biological pathogenesis of KMT2A fusions and ACAs.
Collapse
Affiliation(s)
- Romy E. van Weelderen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Emma Children’s Hospital, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Christine J. Harrison
- Leukemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle-upon-Tyne, United Kingdom
| | - Kim Klein
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Emma Children’s Hospital, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Pediatrics, Wilhelmina Children’s Hospital/University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yilin Jiang
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jonas Abrahamsson
- Department of Pediatrics, Institute of Clinical Sciences, Salgrenska University Hospital, Gothenburg, Sweden
| | - Todd Alonzo
- Division of Biostatistics, University of Southern California, Los Angeles, CA
| | - Richard Aplenc
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Nira Arad-Cohen
- Department of Pediatric Hematology-Oncology, Ruth Rappaport Children’s Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Emmanuelle Bart-Delabesse
- Institut Universitaire du Cancer Toulouse-Oncopole, Laboratoire d’Hématologie secteur Génétique des Hémopathies, Toulouse, France
| | - Barbara Buldini
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplant, Department of Maternal and Child Health, Padua University, Padua, Italy
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Michael N. Dworzak
- Department of Pediatrics, St. Anna Children’s Hospital, Medical University of Vienna & St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Sarah Elitzur
- Department of Pediatric Hematology and Oncology, Schneider Children’s Medical Center & Tel Aviv University, Tel Aviv, Israel
| | | | - Alan Gamis
- Department of Hematology and Oncology, Children’s Mercy Hospital, Kansas City, MO
| | | | - Bianca F. Goemans
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Hester A. de Groot-Kruseman
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- DCOG, Dutch Childhood Oncology Group, Utrecht, The Netherlands
| | - Erin Guest
- Department of Hematology and Oncology, Children’s Mercy Hospital, Kansas City, MO
| | - Shau-Yin Ha
- Department of Pediatrics & Adolescent Medicine, Hong Kong Children’s Hospital, Kowloon Bay, Hong Kong
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Charikleia Kelaidi
- Department of Pediatric Hematology and Oncology, Aghia Sophia Children’s Hospital, Athens, Greece
| | - Hélène Lapillonne
- Department of Pediatric Hematology and Oncology, Hôpital Armand Trousseau, Paris, France
| | - Guy Leverger
- Department of Pediatric Hematology and Oncology, Hôpital Armand Trousseau, Paris, France
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome, Italy
| | - Takako Miyamura
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Sophia Polychronopoulou
- Department of Pediatric Hematology and Oncology, Aghia Sophia Children’s Hospital, Athens, Greece
| | - Mareike Rasche
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Jeffrey E. Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jan Stary
- Department of Pediatric Hematology and Oncology, University Hospital Motol and Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Anne Tierens
- Department of Pathobiology and Laboratory Medicine, University Health Network, Toronto General Hospital, Toronto, ON, Canada
| | - Daisuke Tomizawa
- Children’s Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - C. Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus Medical Center Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Gertjan J. L. Kaspers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Emma Children’s Hospital, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Tierens A, Arad-Cohen N, Cheuk D, De Moerloose B, Fernandez Navarro JM, Hasle H, Jahnukainen K, Juul-Dam KL, Kaspers G, Kovalova Z, Lausen B, Norén-Nyström U, Palle J, Pasauliene R, Jan Pronk C, Saks K, Zeller B, Abrahamsson J. Mitoxantrone Versus Liposomal Daunorubicin in Induction of Pediatric AML With Risk Stratification Based on Flow Cytometry Measurement of Residual Disease. J Clin Oncol 2024; 42:2174-2185. [PMID: 38603646 DOI: 10.1200/jco.23.01841] [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: 08/25/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 04/13/2024] Open
Abstract
PURPOSE Measurable residual disease (MRD) by using flow cytometry after induction therapy is strongly prognostic in pediatric AML, and hematopoietic stem-cell transplant (hSCT) may counteract a poor response. We designed a phase III study with intensified response-guided induction and MRD-based risk stratification and treated poor induction response with hSCT. The efficacy of liposomal daunorubicin (DNX) in induction was compared with mitoxantrone. METHODS The study planned to randomly assign 300 patients, but the production of DNX ceased in 2017. One hundred ninety-four patients were randomly assigned to mitoxantrone or experimental DNX in induction 1. Ninety-three non-randomly assigned patients served as an observation cohort. Primary end point was fraction of patients with MRD <0.1% on day 22 after induction 1. Patients with MRD ≥15% after induction 1 or ≥0.1% after induction 2 or FLT3-ITD with NPM1 wildtype were stratified to high-risk therapy, including hSCT. RESULTS Outcome for all 287 children was good with 5-year event-free survival (EFS5y) 66.7% (CI, 61.4 to 72.4) and 5-year overall survival (OS5y) 79.6% (CI, 75.0 to 84.4). Overall, 75% were stratified to standard-risk and 19% to high-risk. There was no difference in the proportion of patients with MRD <0.1% on day 22 after induction 1 (34% mitoxantrone, etoposide, araC [MEC], 30% DNX, P = .65), but the proportion increased to 61% for MEC versus 47% for DNX (P = .061) at the last evaluation before induction 2. EFS5y was significantly lower, 56.6% (CI, 46.7 to 66.5) versus 71.9% (CI, 63.0 to 80.9), and cumulative incidence of relapse (CIR) was higher, 35.1% (CI, 25.7 to 44.7) versus 18.8% (CI, 11.6 to 27.2) for DNX. The inferior outcome for DNX was only in standard-risk patients with EFS5y 55.3% (CI, 45.1 to 67.7) versus 79.9% (CI, 71.1 to 89.9), CIR 39.5% (CI, 28.4 to 50.3) versus 18.7% (CI, 10.5 to 28.7), and OS5y 76.2% (CI, 67.2 to 86.4) versus 88.6% (CI, 81.4 to 96.3). As-treated analyses, including the observation cohort, supported these results. For all high-risk patients, 85% received hSCT, and EFS5y was 77.7 (CI, 67.3 to 89.7) and OS5y was 83.0 (CI, 73.5 to 93.8). CONCLUSION The intensification of induction therapy with risk stratification on the basis of response to induction and hSCT for high-risk patients led to improved outcomes. Mitoxantrone had a superior anti-leukemic effect than liposomal daunorubicin.
Collapse
Affiliation(s)
- Anne Tierens
- Laboratory Medicine Program, University Health Network, Toronto General Hospital, Toronto, ON, Canada
| | - Nira Arad-Cohen
- Department of Pediatric Hemato-Oncology, Rambam Health Care Campus, Haifa, Israel
| | - Daniel Cheuk
- Department of Pediatrics and Adolescent Medicine, Hong Kong Children's Hospital and Hong Kong Pediatric Hematology and Oncology Study Group (HKPHOSG), Hong Kong, China
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology, Ghent University Hospital, Gent, Belgium
| | | | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Kirsi Jahnukainen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Gertjan Kaspers
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, the Netherlands
| | - Zanna Kovalova
- Department of Paediatric Oncology/Haematology, Children's Clinical University Hospital, Riga, Latvia
| | - Birgitte Lausen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Josefine Palle
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Ramune Pasauliene
- Center of Oncology and Hematology, BMT Unit, Vilnius University Children's Hospital, Vilnius, Lithuania
| | | | - Kadri Saks
- Department of Paediatrics, SA Tallinna Lastehaigla, Tallinn, Estonia
| | - Bernward Zeller
- Department of Pediatrics, Oslo University Hospital, Oslo, Norway
| | - Jonas Abrahamsson
- Institution for Clinical Sciences, Department of Pediatrics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Li JN, Chen YJ, Fan Z, Li QR, Liao LH, Ke ZY, Li Y, Wang LN, Yang CY, Luo XQ, Tang YL, Zhang XL, Huang LB. Intensive chemotherapy with dual induction and ALL-like consolidation for childhood acute myeloid leukemia: a respective report from multiple centers in China. Ther Adv Hematol 2024; 15:20406207241256894. [PMID: 38828002 PMCID: PMC11143855 DOI: 10.1177/20406207241256894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 05/07/2024] [Indexed: 06/05/2024] Open
Abstract
Background Pediatric acute myeloid leukemia (AML) has poor prognosis and high rate of relapse and mortality, and exploration of new treatment options is still critically needed. Objectives To summarize the outcome of our new treatment strategies for pediatric AML, which is characterized by dual induction and acute lymphoblastic leukemia (ALL) elements consolidation. Design Retrospective, single-arm study. Methods From July 2012 to December 2019, an intensive chemotherapy protocol was used for newly diagnosed children with AML, which contains dual induction, three courses of consolidations based on high-dose cytarabine, and two courses of consolidations composed of high-dose methotrexate, vincristine, asparaginase, and mercaptopurine (ALL-like elements). Blasts were monitored by bone marrow smears at intervals, and two lumbar punctures were performed during chemotherapy. We retrospectively analyzed the efficacy and safety of this study. The last follow-up was on 26 May 2023. Results A total of 70 pediatric AMLs were included. The median age at diagnosis was 6.7 (0.5-16.0) years. The median initial WBC count was 23.74 × 109/L, 11 of whom ⩾100 × 109/L. After dual induction, there were 62 cases of complete remission (CR), 5 cases of partial remission, and 3 cases of nonremission. The CR rate was 88.57%. The median follow-up time was 5.8 (0.2-9.4) years, the 5-year overall survival was 78.2% ± 5%, the event-free survival (EFS) was 71.2% ± 5.6%, and the cumulative recurrence rate was 27.75%. The 5-year EFS of patients with initial WBC < 100 × 109/L (n = 59) and ⩾100 × 109/L (n = 11) were 76.4% ± 5.7% and 45.5% ± 15% (p = 0.013), respectively. A total of 650 hospital infections occurred. The main causes of infection were respiratory tract infection (26.92%), septicemia (18.46%), stomatitis (11.85%), and skin and soft-tissue infection (10.46%). Conclusion This intensive treatment protocol with dual induction and ALL-like elements is effective and safe for childhood AML. Initial WBC ⩾ 100 × 109/L was the only independent risk factor in this cohort. Trial registration It is a retrospective study, and no registration on ClinicalTrials.gov.
Collapse
Affiliation(s)
- Jia-Nan Li
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi-Jun Chen
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Fan
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiao-Ru Li
- Department of Pediatrics, Zhongshan People’s Hospital, Zhongshan, China
| | - Liu-Hua Liao
- Department of Pediatrics, Huizhou Central People’s Hospital, Huizhou, China
| | - Zhi-Yong Ke
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Li
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Na Wang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cui-Yun Yang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xue-Qun Luo
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Lai Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou 510080, China
| | - Xiao-Li Zhang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou 510080, China
| | - Li-Bin Huang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou 510080, China
| |
Collapse
|
15
|
Lamba JK, Marrero R, Wu H, Cao X, Parcha PK, Karol SE, Inaba H, Kuo DJ, Degar BA, Heym K, Taub JW, Lacayo NJ, Pui CH, Ribeiro RC, Pounds SB, Rubnitz JE. Pharmacogenomics, Race, and Treatment Outcome in Pediatric Acute Myeloid Leukemia. JAMA Netw Open 2024; 7:e2411726. [PMID: 38753328 PMCID: PMC11099689 DOI: 10.1001/jamanetworkopen.2024.11726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/15/2024] [Indexed: 05/19/2024] Open
Abstract
Importance Disparities in outcomes exist between Black and White patients with acute myeloid leukemia (AML), with Black patients experiencing poorer prognosis compared with their White counterparts. Objective To assess whether varying intensity of induction therapy to treat pediatric AML is associated with reduced disparities in treatment outcome by race. Design, Setting, and Participants A comparative effectiveness analysis was conducted of 86 Black and 359 White patients with newly diagnosed AML who were enrolled in the AML02 trial from 2002 to 2008 or the AML08 trial from 2008 to 2017. Statistical analysis was conducted from July 2023 through January 2024. Interventions Patients in AML02 were randomly assigned to receive standard low-dose cytarabine-based induction therapy or augmented high-dose cytarabine-based induction therapy, whereas patients in AML08 received high-dose cytarabine-based therapy. Main Outcomes and Measures Cytarabine pharmacogenomic 10-single-nucleotide variant (ACS10) scores were evaluated for association with outcome according to race and treatment arm. Results This analysis included 86 Black patients (mean [SD] age, 8.8 [6.5] years; 54 boys [62.8%]; mean [SD] leukocyte count, 52 600 [74 000] cells/µL) and 359 White patients (mean [SD] age, 9.1 [6.2] years; 189 boys [52.6%]; mean [SD] leukocyte count, 54 500 [91 800] cells/µL); 70 individuals with other or unknown racial and ethnic backgrounds were not included. Among all patients without core binding factor AML who received standard induction therapy, Black patients had significantly worse outcomes compared with White patients (5-year event-free survival rate, 25% [95% CI, 9%-67%] compared with 56% [95% CI, 46%-70%]; P = .03). By contrast, among all patients who received augmented induction therapy, there were no differences in outcome according to race (5-year event-free survival rate, Black patients, 50% [95% CI, 38%-67%]; White patients, 48% [95% CI, 42%-55%]; P = .78). Among patients who received standard induction therapy, those with low ACS10 scores had a significantly worse 5-year event-free survival rate compared with those with high scores (42.4% [95% CI, 25.6%-59.3%] and 70.0% [95% CI, 56.6%-83.1%]; P = .004); however, among patients who received augmented induction therapy, there were no differences in 5-year event-free survival rates according to ACS10 score (low score, 60.6% [95% CI, 50.9%-70.2%] and high score, 54.8% [95% CI, 47.1%-62.5%]; P = .43). Conclusions and Relevance In this comparative effectiveness study of pediatric patients with AML treated in 2 consecutive clinical trials, Black patients had worse outcomes compared with White patients after treatment with standard induction therapy, but this disparity was eliminated by treatment with augmented induction therapy. When accounting for ACS10 scores, no outcome disparities were seen between Black and White patients. Our results suggest that using pharmacogenomics parameters to tailor induction regimens for both Black and White patients may narrow the racial disparity gap in patients with AML.
Collapse
Affiliation(s)
- Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville
- University of Florida Health Cancer Center, University of Florida, Gainesville
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville
| | - Richard Marrero
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville
| | - Huiyun Wu
- Department of Biostatistics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Xueyuan Cao
- Department of Health Promotion and Disease Prevention, University of Tennessee Health Science Center, Memphis
| | - Phani Krishna Parcha
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville
| | - Seth E. Karol
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Hiroto Inaba
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Dennis John Kuo
- Division of Pediatric Hematology-Oncology, Rady Children’s Hospital San Diego/University of California, San Diego
| | - Barbara A. Degar
- Hematology/Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Hematology/Oncology, Boston Children’s Hospital, Boston, Massachusetts
| | - Kenneth Heym
- Hematology/Oncology, Cook Children’s Medical Center, Fort Worth, Texas
| | - Jeffrey W. Taub
- Hematology/Oncology, Children’s Hospital of Michigan, Detroit
| | - Norman J. Lacayo
- Hematology/Oncology, Lucile Packard Children’s Hospital, Palo Alto, California
- Hematology/Oncology, Stanford Cancer Institute, Palo Alto, California
| | - Ching-Hon Pui
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Raul C. Ribeiro
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Stanley B. Pounds
- Department of Biostatistics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Jeffrey E. Rubnitz
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee
| |
Collapse
|
16
|
Abla O, Ries RE, Triche T, Gerbing RB, Hirsch B, Raimondi S, Cooper T, Farrar JE, Buteyn N, Harmon LM, Wen H, Deshpande AJ, Kolb EA, Gamis AS, Aplenc R, Alonzo T, Meshinchi S. Structural variants involving MLLT10 fusion are associated with adverse outcomes in pediatric acute myeloid leukemia. Blood Adv 2024; 8:2005-2017. [PMID: 38306602 PMCID: PMC11024924 DOI: 10.1182/bloodadvances.2023010805] [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: 05/23/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024] Open
Abstract
ABSTRACT MLLT10 gene rearrangements with KMT2A occur in pediatric acute myeloid leukemia (AML) and confer poor prognosis, but the prognostic impact of MLLT10 in partnership with other genes is unknown. We conducted a retrospective study with 2080 children and young adults with AML registered on the Children's Oncology Group AAML0531 (NCT00372593) and AAML1031 trials (NCT01371981). Transcriptome profiling and/or karyotyping were performed to identify leukemia-associated fusions associated with prognosis. Collectively, 127 patients (6.1%) were identified with MLLT10 fusions: 104 (81.9%) with KMT2A::MLLT10, 13 (10.2%) with PICALM::MLLT10, and 10 (7.9%) X::MLLT10: (2 each of DDX3X and TEC), with 6 partners (DDX3Y, CEP164, SCN2B, TREH, NAP1L1, and XPO1) observed in single patients. Patients with MLLT10 (n = 127) demonstrated adverse outcomes, with 5-year event-free survival (EFS) of 18.6% vs 49% in patients without MLLT10 (n = 1953, P < .001), inferior 5-year overall survival (OS) of 38.2% vs 65.7% (P ≤ .001), and a higher relapse risk of 76% vs 38.6% (P < .001). Patients with KMT2A::MLLT10 had an EFS from study entry of 19.5% vs 12.7% (P = .628), and an OS from study entry of 40.4% vs 27.6% (P = .361) in those with other MLLT10 fusion partners. Patients with PICALM::MLLT10 had an EFS of 9.2% vs 20% in other MLLT10- without PICALM (X::MLLT10; P = .788). Patients with PICALM::MLLT10 and X::MLLT10 fusions exhibit a DNA hypermethylation signature resembling NUP98::NSD1 fusions, whereas patients with KMT2A::MLLT10 bear aberrations primarily affecting distal regulatory elements. Regardless of the fusion partner, patients with AML harboring MLLT10 fusions exhibit very high-risk features and should be prioritized for alternative therapeutic interventions.
Collapse
Affiliation(s)
- Oussama Abla
- Division of Hematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Rhonda E. Ries
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Tim Triche
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI
| | | | - Betsy Hirsch
- Division of Laboratory Medicine, University of Minnesota Medical Center, Minneapolis, MN
| | - Susana Raimondi
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Todd Cooper
- Division of Hematology-Oncology, Seattle Children's Hospital, University of Washington, Seattle, WA
| | - Jason E. Farrar
- Department of Pediatrics, Hematology-Oncology Section, Arkansas Children's Research Institute, Little Rock, AR
| | | | | | - Hong Wen
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI
| | | | - E. Anders Kolb
- Nemours Center for Cancer and Blood Disorders and Alfred I. DuPont Hospital for Children, Wilmington, DE
| | - Alan S. Gamis
- Division of Hematology, Oncology and Bone Marrow Transplantation, Children's Mercy Hospitals and Clinics, Kansas City, MO
| | | | - Todd Alonzo
- Department of Translational Genomics, University of Southern California, Los Angeles, CA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology-Oncology, Seattle Children's Hospital, University of Washington, Seattle, WA
| |
Collapse
|
17
|
Lavasidis G, Papaioannou K, Anagnostou N, Ketteler P, Bechrakis NE, Ntzani E. Evidence in Focus: The Sparse Landscape of Randomized Trials on Retinoblastoma Treatment. Ocul Oncol Pathol 2024; 10:53-62. [PMID: 38751498 PMCID: PMC11095627 DOI: 10.1159/000536410] [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: 09/04/2023] [Accepted: 01/15/2024] [Indexed: 05/18/2024] Open
Abstract
Background Retinoblastoma, although rare, is one of the most common intraocular malignancies worldwide. Its prognosis has improved significantly in the past few decades, thanks to modern treatments, like systemic, intra-arterial, and intravitreal chemotherapy. However, regarding survival, there are significant differences between high- and low-income countries, eye salvage is still a challenge worldwide, and treatment-related toxicity needs to be carefully and sufficiently managed. Summary To appraise the strength of supporting evidence, we performed a systematic review of randomized controlled trials investigating any therapeutic protocol for retinoblastoma. Four trials with 174 participants (188 eyes) were eligible, all pertaining to different intravenous chemotherapy regimens. Vincristine, etoposide, and carboplatin (VEC) appear superior to a 5-drug combination for stage III retinoblastoma. Moreover, etoposide and carboplatin as neoadjuvant chemotherapy followed by thermochemotherapy seem to offer better local control than vincristine and carboplatin. However, increasing carboplatin dose in the VEC protocol failed to improve treatment efficacy. Key Messages Retinoblastoma is a success story of modern medicine. However, only intravenous chemotherapy has been studied through randomized trials, while evidence for the most novel retinoblastoma treatments has mainly stemmed from observational studies. International collaborations for multicenter randomized trials could overcome difficulties and increase certainty and precision in the field.
Collapse
Affiliation(s)
- Georgios Lavasidis
- Evidence-based Medicine Unit, Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
- Department of Ophthalmology, Elpis General Hospital of Athens, Athens, Greece
| | - Kyriaki Papaioannou
- Department of Pediatric Hematology and Oncology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Nikolaos Anagnostou
- Evidence-based Medicine Unit, Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Petra Ketteler
- Department of Pediatric Hematology and Oncology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Nikolaos E. Bechrakis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Evangelia Ntzani
- Evidence-based Medicine Unit, Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
- Center for Evidence Synthesis in Health, Brown University School of Public Health, Providence, RI, USA
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| |
Collapse
|
18
|
Hasle H, Juul-Dam KL. Maximal benefit of minimal residual disease monitoring in pediatric acute myeloid leukemia. Haematologica 2024; 109:701-703. [PMID: 37675508 PMCID: PMC10905066 DOI: 10.3324/haematol.2023.283765] [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: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Henrik Hasle
- Department of pediatrics and adolescent medicine, Aarhus University Hospital, Aarhus.
| | | |
Collapse
|
19
|
Zheng Y, Pan L, Li J, Feng X, Li C, Zheng M, Mai H, Yang L, He Y, He X, Xu H, Wen H, Le S. Prognostic significance of multiparametric flow cytometry minimal residual disease at two time points after induction in pediatric acute myeloid leukemia. BMC Cancer 2024; 24:46. [PMID: 38195455 PMCID: PMC10775489 DOI: 10.1186/s12885-023-11784-4] [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: 05/09/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Prompt response to induction chemotherapy is a prognostic factor in pediatric acute myeloid leukemia. In this study, we aimed to evaluate the prognostic significance of multiparametric flow cytometry-minimal residual disease (MFC-MRD), assessed at the end of the first and second induction courses. METHODS MFC-MRD was performed at the end of the first induction (TP1) in 524 patients and second induction (TP2) in 467 patients who were treated according to the modified Medical Research Council (UK) acute myeloid leukemia 15 protocol. RESULTS Using a 0.1% cutoff level, patients with MFC-MRD at the two time points had lower event-free survival and overall survival. Only the TP2 MFC-MRD level could predict the outcome in a separate analysis of high and intermediate risks based on European LeukemiaNet risk stratification and KMT2A rearrangement. The TP2 MFC-MRD level could further differentiate the prognosis of patients into complete remission or non-complete remission based on morphological evaluation. Multivariate analysis indicated the TP2 MFC-MRD level as an independent adverse prognostic factor for event-free survival and overall survival. When comparing patients with MFC-MRD ≥ 0.1%, those who underwent hematopoietic stem cell transplant during the first complete remission had significantly higher 5-year event-free survival and overall survival and lower cumulative incidence of relapse than those who only received consolidation chemotherapy. CONCLUSIONS The TP2 MFC-MRD level can predict the outcomes in pediatric patients with acute myeloid leukemia and help stratify post-remission treatment.
Collapse
Affiliation(s)
- Yongzhi Zheng
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lili Pan
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian Li
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoqin Feng
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunfu Li
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Mincui Zheng
- Department of Pediatric Hematology/Oncology, Hematology and Oncology, Hunan Children's Hospital, Changsha, China
| | - Huirong Mai
- Department of Pediatric Hematology/Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Lihua Yang
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yingyi He
- Department of Pediatric Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Xiangling He
- People's Hospital of Hunan Province, Changsha, China
| | - Honggui Xu
- Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Hong Wen
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Shaohua Le
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
20
|
Tao Y, Wei L, Shiba N, Tomizawa D, Hayashi Y, Ogawa S, Chen L, You H. Development and validation of a promising 5-gene prognostic model for pediatric acute myeloid leukemia. MOLECULAR BIOMEDICINE 2024; 5:1. [PMID: 38163849 PMCID: PMC10758381 DOI: 10.1186/s43556-023-00162-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: 08/26/2023] [Accepted: 12/03/2023] [Indexed: 01/03/2024] Open
Abstract
Risk classification in pediatric acute myeloid leukemia (P-AML) is crucial for personalizing treatments. Thus, we aimed to establish a risk-stratification tool for P-AML patients and eventually guide individual treatment. A total of 256 P-AML patients with accredited mRNA-seq data from the TARGET database were divided into training and internal validation datasets. A gene-expression-based prognostic score was constructed for overall survival (OS), by using univariate Cox analysis, LASSO regression analysis, Kaplan-Meier (K-M) survival, and multivariate Cox analysis. A P-AML-5G prognostic score bioinformatically derived from expression levels of 5 genes (ZNF775, RNFT1, CRNDE, COL23A1, and TTC38), clustered P-AML patients in training dataset into high-risk group (above optimal cut-off) with shorter OS, and low-risk group (below optimal cut-off) with longer OS (p < 0.0001). Meanwhile, similar results were obtained in internal validation dataset (p = 0.005), combination dataset (p < 0.001), two treatment sub-groups (p < 0.05), intermediate-risk group defined with the Children's Oncology Group (COG) (p < 0.05) and an external Japanese P-AML dataset (p = 0.005). The model was further validated in the COG study AAML1031(p = 0.001), and based on transcriptomic analysis of 943 pediatric patients and 70 normal bone marrow samples from this dataset, two genes in the model demonstrated significant differential expression between the groups [all log2(foldchange) > 3, p < 0.001]. Independent of other prognostic factors, the P-AML-5G groups presented the highest concordance-index values in training dataset, chemo-therapy only treatment subgroups of the training and internal validation datasets, and whole genome-sequencing subgroup of the combined dataset, outperforming two Children's Oncology Group (COG) risk stratification systems, 2022 European LeukemiaNet (ELN) risk classification tool and two leukemic stem cell expression-based models. The 5-gene prognostic model generated by a single assay can further refine the current COG risk stratification system that relies on numerous tests and may have the potential for the risk judgment and identification of the high-risk pediatric AML patients receiving chemo-therapy only treatment.
Collapse
Affiliation(s)
- Yu Tao
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wei
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, China
- Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Norio Shiba
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Yasuhide Hayashi
- Department of Hematology/Oncology, Gunma and Institute of Physiology and Medicine, Gunma Children's Medical Center, Jobu University, Gunma, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institute, 17177, Stockholm, Sweden
| | - Li Chen
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Hua You
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
21
|
Zaliova M, Zuna J, Winkowska L, Janotova I, Skorepova J, Lukes J, Meyer C, Marschalek R, Novak Z, Domansky J, Stary J, Sramkova L, Trka J. Genomic DNA-based measurable residual disease monitoring in pediatric acute myeloid leukemia: unselected consecutive cohort study. Leukemia 2024; 38:21-30. [PMID: 38001170 PMCID: PMC10776399 DOI: 10.1038/s41375-023-02083-9] [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: 09/15/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023]
Abstract
Measurable residual disease (MRD) monitoring in childhood acute myeloid leukemia (AML) is used to assess response to treatment and for early detection of imminent relapse. In childhood AML, MRD is typically evaluated using flow cytometry, or by quantitative detection of leukemia-specific aberrations at the mRNA level. Both methods, however, have significant limitations. Recently, we demonstrated the feasibility of MRD monitoring in selected subgroups of AML at the genomic DNA (gDNA) level. To evaluate the potential of gDNA-based MRD monitoring across all AML subtypes, we conducted a comprehensive analysis involving 133 consecutively diagnosed children. Integrating next-generation sequencing into the diagnostic process, we identified (presumed) primary genetic aberrations suitable as MRD targets in 97% of patients. We developed patient-specific quantification assays and monitored MRD in 122 children. The gDNA-based MRD monitoring via quantification of primary aberrations with a sensitivity of at least 10-4 was possible in 86% of patients; via quantification with sensitivity of 5 × 10-4, of secondary aberrations, or at the mRNA level in an additional 8%. Importantly, gDNA-based MRD exhibited independent prognostic value at early time-points in patients stratified to intermediate-/high-risk treatment arms. Our study demonstrates the broad applicability, feasibility, and clinical significance of gDNA-based MRD monitoring in childhood AML.
Collapse
Affiliation(s)
- Marketa Zaliova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic.
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
- University Hospital Motol, Prague, Czech Republic.
| | - Jan Zuna
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Lucie Winkowska
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | | | - Justina Skorepova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Julius Lukes
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Claus Meyer
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt am Main, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt am Main, Germany
| | - Zbynek Novak
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jiri Domansky
- Pediatric Oncology Department, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Lucie Sramkova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Jan Trka
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic.
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
- University Hospital Motol, Prague, Czech Republic.
| |
Collapse
|
22
|
Tomizawa D, Matsubayashi J, Iwamoto S, Hiramatsu H, Hasegawa D, Moritake H, Hasegawa D, Terui K, Hama A, Tsujimoto SI, Kiyokawa N, Miyachi H, Deguchi T, Hashii Y, Iijima-Yamashita Y, Taki T, Noguchi Y, Koike K, Koh K, Yuza Y, Moriya Saito A, Horibe K, Taga T, Tanaka S, Adachi S. High-dose cytarabine induction therapy and flow cytometric measurable residual disease monitoring for children with acute myeloid leukemia. Leukemia 2024; 38:202-206. [PMID: 37926712 DOI: 10.1038/s41375-023-02075-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/07/2023]
Affiliation(s)
- Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan.
| | - Jun Matsubayashi
- Center for Clinical Research and Advanced Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Shotaro Iwamoto
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidefumi Hiramatsu
- Department of Pediatrics, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Hiroshi Moritake
- Division of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Kiminori Terui
- Department of Pediatrics, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Asahito Hama
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Aichi Medical Center Nagoya First Hospital, Nagoya, Japan
| | - Shin-Ichi Tsujimoto
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hayato Miyachi
- Department of Laboratory Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Takao Deguchi
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
- Division of Cancer Immunodiagnostics, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Pediatrics, Osaka International Cancer Institute, Osaka, Japan
| | | | - Tomohiko Taki
- Department of Medical Technology, Kyorin University Faculty of Health Sciences, Mitaka, Japan
| | - Yasushi Noguchi
- Department of Pediatrics, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Kazutoshi Koike
- Department of Pediatric Hematology/Oncology, Ibaraki Children's Hospital, Mito, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Yuki Yuza
- Department of Hematology and Oncology, Tokyo Metropolitan Children's Medical Center, Fuchu, Japan
| | | | - Keizo Horibe
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Shiro Tanaka
- Department of Clinical Biostatistics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
23
|
Kim H, Ikuse T, Matsui T, Sakaguchi H, Ishiguro A, Shoji K. Factors distinguishing leukemoid reaction from hematological malignancy in children. Pediatr Int 2024; 66:e15837. [PMID: 39692201 DOI: 10.1111/ped.15837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 07/02/2024] [Accepted: 07/31/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Information on the etiology and prognosis for leukemoid reaction (LR) in children is still limited and little is known about the factors that distinguish LR from hematological malignancy (HM). METHODS This was a single-center, case-control study. Pediatric patients (<18 years) with a white blood cell (WBC) count of 50,000/μL or more were included in the study. Clinical information and laboratory test results were extracted from the electronic medical records. Patients were divided into the LR and HM groups. Logistic regression analysis was performed to investigate the factors that discriminated LR from HM. RESULTS We found 214 cases (115 cases in the LR group and 99 cases in the HM group) eligible for analysis. Approximately half of the LR cases were due to infectious diseases (n = 58, 50%); bacteremia and respiratory infections were the most common infections. Age younger than 2 years (odds ratio [95% confidence interval]) (2.154 [0.690-6.727]), presence of known underlying diseases (10.006 [3.119-32.102]), WBC count <60,555/μL (20.676 [6.357-67.251]), platelet count 118,000/μL or higher (15.059 [3.876-58.504]), lactate dehydrogenase (LDH) below 781 U/L (4.219 [1.378-12.915]), and C-reactive protein (CRP) ≥0.91 mg/dL (10.568 [2.736-40.825]) were identified as the predictive factors for LR by logistic regression analysis. Thirty-day mortality was higher in the LR group than in the HM group but the difference was not statistically significant (13% vs. 6%, p = 0.087). CONCLUSIONS Approximately half of the etiology of LR was infectious diseases and prognosis was poorer for LR than for HM. Age, presence of known underlying conditions, and laboratory tests, including WBC count, platelet count, LDH, and CRP, may be useful in distinguishing LR from HM.
Collapse
Affiliation(s)
- Heeyung Kim
- Center for Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| | - Tatsuki Ikuse
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| | - Toshihiro Matsui
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| | - Hirotoshi Sakaguchi
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Akira Ishiguro
- Center for Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| | - Kensuke Shoji
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
24
|
Lee JH, Ju HY, Hyun JK, Kim SJ, Cho HW, Lee JK, Lee JW, Sung KW, Yoo KH. Treatment outcome and prognostic factors in relapsed pediatric acute myeloid leukemia. Blood Res 2023; 58:181-186. [PMID: 37926557 PMCID: PMC10758629 DOI: 10.5045/br.2023.2023152] [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: 08/09/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Background Despite improved outcomes for pediatric patients with acute myeloid leukemia (AML), the prognosis for relapse remains poor. This study aimed to examine the clinical factors associated with prognosis in relapsed pediatric AML. Methods We conducted a chart review of pediatric patients with AML who experienced their first relapse and received treatment at our institution between 2008 and 2019. Risk stratification at diagnosis was performed according to the definition suggested by the ongoing AML 2012 study in Korea, and the clinical factors associated with prognosis were analyzed. Results A total of 27 pediatric patients with relapsed AML were identified. The 5-year overall survival (OS) and event-free survival (EFS) rates were 32.9% and 32.9%, respectively. A duration ≥12 months from diagnosis to relapse had a favorable impact on survival outcomes (5-yr OS, 64.0% vs. 15.7%; P=0.007). Patients who achieved complete remission (CR) after 1 course of chemotherapy following relapse (N=15) had a 5-year OS rate of 59.3%, while none of the other patients survived (P<0.0001). Additionally, the 5-year OS differed significantly based on the risk group at initial diagnosis (62.3% [favorable and intermediate prognosis groups, N=11] vs. 13.3% [poor prognosis group, N=15]; P=0.014). Conclusion Patients with a longer duration of CR before relapse, who achieved CR following 1 course of reinduction chemotherapy, and were in the favorable or intermediate prognosis group at diagnosis demonstrated better outcomes. These findings emphasize the importance of tailoring treatment strategies based on the expected prognosis at relapse in pediatric patients with AML.
Collapse
Affiliation(s)
- Jung Hwan Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ju Kyung Hyun
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Jin Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Kyung Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Science and Technology, SAIHST, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| |
Collapse
|
25
|
D’Alessandro G, Morales-Juarez DA, Richards SL, Nitiss KC, Serrano-Benitez A, Wang J, Thomas JC, Gupta V, Voigt A, Belotserkovskaya R, Goh CG, Bowden AR, Galanty Y, Beli P, Nitiss JL, Zagnoli-Vieira G, Jackson SP. RAD54L2 counters TOP2-DNA adducts to promote genome stability. SCIENCE ADVANCES 2023; 9:eadl2108. [PMID: 38055822 PMCID: PMC10699776 DOI: 10.1126/sciadv.adl2108] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/07/2023] [Indexed: 12/08/2023]
Abstract
The catalytic cycle of topoisomerase 2 (TOP2) enzymes proceeds via a transient DNA double-strand break (DSB) intermediate termed the TOP2 cleavage complex (TOP2cc), in which the TOP2 protein is covalently bound to DNA. Anticancer agents such as etoposide operate by stabilizing TOP2ccs, ultimately generating genotoxic TOP2-DNA protein cross-links that require processing and repair. Here, we identify RAD54 like 2 (RAD54L2) as a factor promoting TOP2cc resolution. We demonstrate that RAD54L2 acts through a novel mechanism together with zinc finger protein associated with tyrosyl-DNA phosphodiesterase 2 (TDP2) and TOP2 (ZATT/ZNF451) and independent of TDP2. Our work suggests a model wherein RAD54L2 recognizes sumoylated TOP2 and, using its ATPase activity, promotes TOP2cc resolution and prevents DSB exposure. These findings suggest RAD54L2-mediated TOP2cc resolution as a potential mechanism for cancer therapy resistance and highlight RAD54L2 as an attractive candidate for drug discovery.
Collapse
Affiliation(s)
- Giuseppina D’Alessandro
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Sean L. Richards
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Almudena Serrano-Benitez
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Juanjuan Wang
- Institute of Molecular Biology (IMB), Chromatin Biology & Proteomics, Mainz, Germany
| | - John C. Thomas
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Vipul Gupta
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Andrea Voigt
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Rimma Belotserkovskaya
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Chen Gang Goh
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Anne Ramsay Bowden
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Yaron Galanty
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Petra Beli
- Institute of Molecular Biology (IMB), Chromatin Biology & Proteomics, Mainz, Germany
- Institute of Developmental Biology and Neurobiology (IDN), Johannes Gutenberg-Universität, Mainz, Germany
| | | | - Guido Zagnoli-Vieira
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Stephen P. Jackson
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- The Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
26
|
Juul-Dam KL, Shukla NN, Cooper TM, Cuglievan B, Heidenreich O, Kolb EA, Rasouli M, Hasle H, Zwaan CM. Therapeutic targeting in pediatric acute myeloid leukemia with aberrant HOX/MEIS1 expression. Eur J Med Genet 2023; 66:104869. [PMID: 38174649 PMCID: PMC11195042 DOI: 10.1016/j.ejmg.2023.104869] [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: 08/31/2022] [Revised: 05/21/2023] [Accepted: 10/22/2023] [Indexed: 01/05/2024]
Abstract
Despite advances in the clinical management of childhood acute myeloid leukemia (AML) during the last decades, outcome remains fatal in approximately one third of patients. Primary chemoresistance, relapse and acute and long-term toxicities to conventional myelosuppressive therapies still constitute significant challenges and emphasize the unmet need for effective targeted therapies. Years of scientific efforts have translated into extensive insights on the heterogeneous spectrum of genetics and oncogenic signaling pathways of AML and identified a subset of patients characterized by upregulation of HOXA and HOXB homeobox genes and myeloid ecotropic virus insertion site 1 (MEIS1). Aberrant HOXA/MEIS1 expression is associated with genotypes such as rearrangements in Histone-lysine N-methyltransferase 2A (KMT2A-r), nucleoporin 98 (NUP98-r) and mutated nucleophosmin (NPM1c) that are found in approximately one third of children with AML. AML with upregulated HOXA/MEIS1 shares a number of molecular vulnerabilities amenable to recently developed molecules targeting the assembly of protein complexes or transcriptional regulators. The interaction between the nuclear scaffold protein menin and KMT2A has gained particular interest and constitutes a molecular dependency for maintenance of the HOXA/MEIS1 transcription program. Menin inhibitors disrupt the menin-KMT2A complex in preclinical models of KMT2A-r, NUP98-r and NPM1c acute leukemias and its occupancy at target genes leading to leukemic cell differentiation and apoptosis. Early-phase clinical trials are either ongoing or in development and preliminary data suggests tolerable toxicities and encouraging efficacy of menin inhibitors in adults with relapsed or refractory KMT2A-r and NPM1c AML. The Pediatric Acute Leukemia/European Pediatric Acute Leukemia (PedAL/EUPAL) project is focused to advance and coordinate informative clinical trials with new agents and constitute an ideal framework for testing of menin inhibitors in pediatric study populations. Menin inhibitors in combination with standard chemotherapy or other targeting agents may enhance anti-leukemic effects and constitute rational treatment strategies for select genotypes of childhood AML, and provide enhanced safety to avoid differentiation syndrome. In this review, we discuss the pathophysiological mechanisms in KMT2A-r, NUP98-r and NPM1c AML, emerging molecules targeting the HOXA/MEIS1 transcription program with menin inhibitors as the most prominent examples and future therapeutic implications of these agents in childhood AML.
Collapse
Affiliation(s)
- Kristian L Juul-Dam
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | - Neerav N Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Todd M Cooper
- Division of Hematology/Oncology, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Branko Cuglievan
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Olaf Heidenreich
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - E Anders Kolb
- Division of Oncology, Nemours/Alfred I. Dupont Hospital for Children, Wilmington, DE, USA
| | - Milad Rasouli
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| |
Collapse
|
27
|
Li J, Zong S, Wan Y, Ruan M, Zhang L, Yang W, Chen X, Zou Y, Chen Y, Guo Y, Wu P, Zhang Y, Zhu X. Integration of Transcriptomic Features to Improve Prognosis Prediction of Pediatric Acute Myeloid Leukemia With KMT2A Rearrangement. Hemasphere 2023; 7:e979. [PMID: 38026790 PMCID: PMC10666994 DOI: 10.1097/hs9.0000000000000979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Lysine methyltransferase 2A-rearranged acute myeloid leukemia (KMT2A-r AML) is a special entity in the 2022 World Health Organization classification of myeloid neoplasms, characterized by high relapse rate and adverse outcomes. Current risk stratification was established on the treatment response and translocation partner of KMT2A. To study the transcriptomic feature and refine the current stratification of pediatric KMT2A-r AML, we analyzed clinical and RNA sequencing data of 351 patients. By implementing least absolute shrinkage and selection operator algorithm, we identified 7 genes (KIAA1522, SKAP2, EGFL7, GAB2, HEBP1, FAM174B, and STARD8) of which the expression levels were strongly associated with outcomes. We then developed a transcriptome-based score, dividing patients into 2 groups with distinct gene expression patterns and prognosis, which was further validated in an independent cohort and outperformed the LSC17 score. We also found cell cycle, oxidative phosphorylation, and metabolism pathways were upregulated in patients with inferior outcomes. By integrating clinical characteristics, we proposed a simple-to-use prognostic scoring system with excellent discriminability, which allowed us to distinguish allogeneic hematopoietic stem cell transplantation candidates more precisely. In conclusion, pediatric KMT2A-r AML is heterogenous on transcriptomic level and the newly proposed scoring system combining clinical characteristics and transcriptomic features can be instructive in clinical routines.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Suyu Zong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yang Wan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Min Ruan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Li Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenyu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaojuan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yao Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yumei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ye Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Peng Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
28
|
Raghuram N, Hasegawa D, Nakashima K, Rahman S, Antoniou E, Skajaa T, Merli P, Verma A, Rabin KR, Aftandilian C, Kotecha RS, Cheuk D, Jahnukainen K, Kolenova A, Balwierz W, Norton A, O’Brien M, Cellot S, Chopek A, Arad-Cohen N, Goemans B, Rojas-Vasquez M, Ariffin H, Bartram J, Kolb EA, Locatelli F, Klusmann JH, Hasle H, McGuire B, Hasnain A, Sung L, Hitzler J. Survival outcomes of children with relapsed or refractory myeloid leukemia associated with Down syndrome. Blood Adv 2023; 7:6532-6539. [PMID: 36735769 PMCID: PMC10632607 DOI: 10.1182/bloodadvances.2022009381] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Children with Down syndrome (DS) are at a significantly higher risk of developing acute myeloid leukemia, also termed myeloid leukemia associated with DS (ML-DS). In contrast to the highly favorable prognosis of primary ML-DS, the limited data that are available for children who relapse or who have refractory ML-DS (r/r ML-DS) suggest a dismal prognosis. There are few clinical trials and no standardized treatment approach for this population. We conducted a retrospective analysis of international study groups and pediatric oncology centers and identified 62 patients who received treatment with curative intent for r/r ML-DS between year 2000 to 2021. Median time from diagnosis to relapse was 6.8 (range, 1.1-45.5) months. Three-year event-free survival (EFS) and overall survival (OS) were 20.9 ± 5.3% and 22.1 ± 5.4%, respectively. Survival was associated with receipt of hematopoietic stem cell transplantation (HSCT) (hazard ratio [HR], 0.28), duration of first complete remission (CR1) (HR, 0.31 for > 12 months) and attainment of remission after relapse (HR, 4.03). Patients who achieved complete remission (CR) before HSCT, had an improved OS and EFS of 56.0 ± 11.8% and 50.5 ± 11.9%, respectively compared to those who underwent HSCT without CR (3-year OS and EFS of 10.0 ± 9.5%). Treatment failure after HSCT was predominantly because of disease recurrence (52%) followed by treatment-related mortality (10%). The prognosis of r/r ML-DS remains dismal even in the current treatment period and serve as a reference point for current prognostication and future interventional studies. Clinical trials aimed at improving the survival of patients with r/r ML-DS are needed.
Collapse
Affiliation(s)
- Nikhil Raghuram
- Division of Hematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Kentaro Nakashima
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Syaza Rahman
- Division of Paediatric Haematology-Oncology and BM Transplantation, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Evangelia Antoniou
- Department of Pediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Torjus Skajaa
- Department of Haematology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Pietro Merli
- Department of Pediatric Hematology/Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Sapienza University of Rome, Rome, Italy
| | - Anupam Verma
- Division of Hematology/Oncology, Department of Pediatrics, University of Utah and Primary Children's Hospital, Salt Lake City, UT
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Karen R. Rabin
- Pediatric Hematology-Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
| | - Catherine Aftandilian
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Rishi S. Kotecha
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, University of WA, Perth, WA, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
| | - Daniel Cheuk
- Department of Paediatrics and Adolescent Medicine, the University of Hong Kong and Hong Kong Children's Hospital, Hong Kong, China
| | - Kirsi Jahnukainen
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Walentyna Balwierz
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Alice Norton
- Department of Haematology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
| | - Maureen O’Brien
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Sonia Cellot
- Division of Hematology, Department of Pediatrics, Ste-Justine Hospital, Montréal, Université de Montréal, Montréal, QC, Canada
| | - Ashley Chopek
- Pediatric Blood and Marrow Transplant Program, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Nira Arad-Cohen
- Pediatric Hematology-Oncology Department, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Bianca Goemans
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marta Rojas-Vasquez
- Department of Pediatric Hematology-Oncology, Stollery Children's Hospital, University of Alberta, Edmonton, Canada
| | - Hany Ariffin
- Division of Paediatric Haematology-Oncology and BM Transplantation, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Jack Bartram
- Department of Haematology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - E. Anders Kolb
- Nemours Center for Cancer and Blood Disorders/Alfred I. DuPont Hospital for Children, Wilmington, DE
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Sapienza University of Rome, Rome, Italy
| | | | - Henrik Hasle
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Bryan McGuire
- Division of Hematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Afia Hasnain
- Division of Genome Diagnostics, Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Lillian Sung
- Division of Hematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Johann Hitzler
- Division of Hematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| |
Collapse
|
29
|
Li J, Gao J, Liu A, Liu W, Xiong H, Liang C, Fang Y, Dai Y, Shao J, Yu H, Wang L, Wang L, Yang L, Yan M, Zhai X, Shi X, Tian X, Ju X, Chen Y, Wang J, Zhang L, Liang H, Chen S, Zhang J, Cao H, Jin J, Hu Q, Wang J, Wang Y, Zhou M, Han Y, Zhang R, Zhao W, Wang X, Lin L, Zhang R, Gao C, Xu L, Zhang Y, Fan J, Wu Y, Lin W, Yu J, Qi P, Huang P, Peng X, Peng Y, Wang T, Zheng H. Homoharringtonine-Based Induction Regimen Improved the Remission Rate and Survival Rate in Chinese Childhood AML: A Report From the CCLG-AML 2015 Protocol Study. J Clin Oncol 2023; 41:4881-4892. [PMID: 37531592 PMCID: PMC10617822 DOI: 10.1200/jco.22.02836] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/26/2023] [Accepted: 06/14/2023] [Indexed: 08/04/2023] Open
Abstract
PURPOSE Homoharringtonine (HHT) is commonly used for the treatment of Chinese adult AML, and all-trans retinoic acid (ATRA) has been verified in acute promyelocytic leukemia (APL). However, the efficacy and safety of HHT-based induction therapy have not been confirmed for childhood AML, and ATRA-based treatment has not been evaluated among patients with non-APL AML. PATIENTS AND METHODS This open-label, multicenter, randomized Chinese Children's Leukemia Group-AML 2015 study was performed across 35 centers in China. Patients with newly diagnosed childhood AML were first randomly assigned to receive an HHT-based (H arm) or etoposide-based (E arm) induction regimen and then randomly allocated to receive cytarabine-based (AC arm) or ATRA-based (AT arm) maintenance therapy. The primary end points were the complete remission (CR) rate after induction therapy, and the secondary end points were the overall survival (OS) and event-free survival (EFS) at 3 years. RESULTS We enrolled 1,258 patients, of whom 1,253 were included in the intent-to-treat analysis. The overall CR rate was significantly higher in the H arm than in the E arm (79.9% v 73.9%, P = .014). According to the intention-to-treat analysis, the 3-year OS was 69.2% (95% CI, 65.1 to 72.9) in the H arm and 62.8% (95% CI, 58.7 to 66.6) in the E arm (P = .025); the 3-year EFS was 61.1% (95% CI, 56.8 to 65.0) in the H arm and 53.4% (95% CI, 49.2 to 57.3) in the E arm (P = .022). Among the per-protocol population, who received maintenance therapy, the 3-year EFS did not differ significantly across the four arms (H + AT arm: 70.7%, 95% CI, 61.1 to 78.3; H + AC arm: 74.8%, 95% CI, 67.0 to 81.0, P = .933; E + AC arm: 72.9%, 95% CI, 65.1 to 79.2, P = .789; E + AT arm: 66.2%, 95% CI, 56.8 to 74.0, P = .336). CONCLUSION HHT is an alternative combination regimen for childhood AML. The effects of ATRA-based maintenance are comparable with those of cytarabine-based maintenance therapy.
Collapse
Affiliation(s)
- Jing Li
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Ju Gao
- West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, China
| | | | - Wei Liu
- Children's Hospital of Henan Province, Zhengzhou, China
| | - Hao Xiong
- Wuhan Children's Hospital, Wuhan, China
| | - Changda Liang
- Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Yongjun Fang
- Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yunpeng Dai
- Shandong First Medical University Affiliated Shandong Provincial Hospital, Jinan, China
| | - Jingbo Shao
- Shanghai Children's Hospital, Shanghai, China
| | - Hui Yu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingzhen Wang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Wang
- Children's Hospital of Hebei Province, Shijiazhuang, China
| | - Liangchun Yang
- Department of Pediatrics, Xiangya Hospital Central South University, Changsha, China
| | - Mei Yan
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaowen Zhai
- Children's Hospital of Fudan University, Shanghai, China
| | - Xiaodong Shi
- Capital Institute of Pediatrics' Children's Hospital, Beijing, China
| | - Xin Tian
- Kunming Children's Hospital, Kunming, China
| | - Xiuli Ju
- Qilu Hospital of Shandong University, Jinan, China
| | - Yan Chen
- Children's Hospital of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jing Wang
- Children's Hospital of Shanxi Province, Taiyuan, China
| | - Leping Zhang
- Peking University People's Hospital, Beijing, China
| | - Hui Liang
- Women and Children's Hospital, Qingdao University, Qingdao, China
| | - Sen Chen
- Tianjin Children's Hospital, Tianjin, China
| | | | - Haixia Cao
- Qinghai Women's and Children's Hospital, Xining, China
| | - Jiao Jin
- The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qun Hu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junlan Wang
- Northwest Women's and Children's Hospital, Xian, China
| | | | - Min Zhou
- Chengdu Women's and Children's Central Hospital, Chengdu, China
| | - Yueqin Han
- Children's Hospital of Liaocheng, Liaocheng, China
| | - Rong Zhang
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Weihong Zhao
- First Hospital, Peking University, Beijing, China
| | | | - Limin Lin
- Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Ruidong Zhang
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Chao Gao
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute, Beijing, China
| | - Liting Xu
- Children's Hospital of Zhejiang University School of Medicine, the Pediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuanyuan Zhang
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Jia Fan
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Ying Wu
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Wei Lin
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Jiaole Yu
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Peijing Qi
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Pengli Huang
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Xiaoxia Peng
- Center for Clinical Epidemiology and Evidence-Based Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yaguang Peng
- Center for Clinical Epidemiology and Evidence-Based Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Tianyou Wang
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Huyong Zheng
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology Oncology, Beijing, China
- National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University), Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| |
Collapse
|
30
|
Bansal D, Davidson A, Supriyadi E, Njuguna F, Ribeiro RC, Kaspers GJL. SIOP PODC adapted risk stratification and treatment guidelines: Recommendations for acute myeloid leukemia in resource-limited settings. Pediatr Blood Cancer 2023; 70:e28087. [PMID: 31774234 DOI: 10.1002/pbc.28087] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/21/2019] [Accepted: 10/30/2019] [Indexed: 01/09/2023]
Abstract
In low- and middle-income countries (LMICs), limited resources, suboptimal risk stratification, and disproportionate patient-to-infrastructure ratio result in low survival of patients with acute myeloid leukemia (AML). A high incidence of relapse, inherent to the biology, renders management arduous. The challenge of treating AML in LMICs is of balancing the intensity of myelosuppressive chemotherapy, which appears necessary for cure, with available supportive care, which influences treatment-related mortality. The recommendations outlined in this paper are based on published evidence and expert opinion. The principle of this adapted protocol is to tailor treatment to available resources, reduce preventable toxic death, and direct limited resources toward those children who are most likely to be cured.
Collapse
Affiliation(s)
- Deepak Bansal
- Department of Pediatrics, Hematology-Oncology Unit, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alan Davidson
- Haematology-Oncology Service, Red Cross Children's Hospital, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Eddy Supriyadi
- Pediatric Hematology-Oncology Division, Department of Pediatrics, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Festus Njuguna
- Department of Child Health and Paediatrics, Moi University, Eldoret, Kenya
| | - Raul C Ribeiro
- St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Gertjan J L Kaspers
- Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
31
|
Zhao Y, Guo H, Chang Y. MRD-directed and risk-adapted individualized stratified treatment of AML. Chin J Cancer Res 2023; 35:451-469. [PMID: 37969959 PMCID: PMC10643342 DOI: 10.21147/j.issn.1000-9604.2023.05.04] [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: 10/10/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023] Open
Abstract
Measurable residual disease (MRD) has been widely recognized as a biomarker for deeply evaluating complete remission (CR), predicting relapse, guiding pre-emptive interventions, and serving as an endpoint surrogate for drug testing. However, despite the emergence of new technologies, there remains a lack of comprehensive understanding regarding the proper techniques, sample materials, and optimal time points for MRD assessment. In this review, we summarized the MRD methods, sample sources, and evaluation frequency according to the risk category of the European Leukemia Net (ELN) 2022. Additionally, we emphasize the importance of properly utilizing and combining these technologies. We have also refined the flowchart outlining each time point for pre-emptive interventions and intervention paths. The evaluation of MRD in acute myeloid leukemia (AML) is sophisticated, clinically applicable, and technology-dependent, and necessitates standardized approaches and further research.
Collapse
Affiliation(s)
- Yijing Zhao
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Hanfei Guo
- Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto 94304, USA
- the First Hospital of Jilin University, Cancer Center, Changchun 133021, China
| | - Yingjun Chang
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| |
Collapse
|
32
|
Ni J, Chen M, Su Y, Gao Q, Liu L, Lu X. Right femoral vein and right dorsal artery thrombosis in childhood acute myeloid leukemia: A case report. Medicine (Baltimore) 2023; 102:e35121. [PMID: 37832057 PMCID: PMC10578772 DOI: 10.1097/md.0000000000035121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/17/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND It is rare for newly diagnosed (de novo) or newly treated acute myeloid leukemia (AML) complicated with thrombotic complications, especially combined arterial and venous thrombosis. METHODS We reported a 13-year-old boy diagnosed with AML and leukocytosis, who developed right femoral vein and right dorsal artery thrombosis during chemotherapy. After treatment with low molecular weight heparin, diosmin, and alprostadil, symptoms were relieved. Unfortunately, the child suffered from coagulopathy afterward, which was unexpectedly caused by vitamin K deficiency. RESULTS After supplementation with vitamin K and prothrombin complex concentrate, coagulation function recovered. CONCLUSION For childhood AML patients with high thrombotic risks, close monitoring during anticoagulant treatment was necessary. Concomitantly, we should be alert to past medication history and combined medication use, especially those that may lead to vitamin K deficiency, secondary bleeding, and coagulation disorders. Rational use of antibiotics, anticoagulants, and antitumor drugs must be guaranteed.
Collapse
Affiliation(s)
- Jiaqi Ni
- Department of Pharmacy/Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects & Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Min Chen
- Department of Pharmacy/Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects & Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yali Su
- Key Laboratory of Birth Defects & Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qianqian Gao
- Key Laboratory of Birth Defects & Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Ultrasound, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lingjun Liu
- Key Laboratory of Birth Defects & Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Lu
- Key Laboratory of Birth Defects & Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Tigu AB, Constantinescu CS, Teodorescu P, Kegyes D, Munteanu R, Feder R, Peters M, Pralea I, Iuga C, Cenariu D, Marcu A, Tanase A, Colita A, Drula R, Bergthorsson JT, Greiff V, Dima D, Selicean C, Rus I, Zdrenghea M, Gulei D, Ghiaur G, Tomuleasa C. Design and preclinical testing of an anti-CD41 CAR T cell for the treatment of acute megakaryoblastic leukaemia. J Cell Mol Med 2023; 27:2864-2875. [PMID: 37667538 PMCID: PMC10538266 DOI: 10.1111/jcmm.17810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/28/2023] [Accepted: 05/26/2023] [Indexed: 09/06/2023] Open
Abstract
Acute megakaryoblastic leukaemia (AMkL) is a rare subtype of acute myeloid leukaemia (AML) representing 5% of all reported cases, and frequently diagnosed in children with Down syndrome. Patients diagnosed with AMkL have low overall survival and have poor outcome to treatment, thus novel therapies such as CAR T cell therapy could represent an alternative in treating AMkL. We investigated the effect of a new CAR T cell which targets CD41, a specific surface antigen for M7-AMkL, against an in vitro model for AMkL, DAMI Luc2 cell line. The performed flow cytometry evaluation highlighted a percentage of 93.8% CAR T cells eGFP-positive and a limited acute effect on lowering the target cell population. However, the interaction between effector and target (E:T) cells, at a low ratio, lowered the cell membrane integrity, and reduced the M7-AMkL cell population after 24 h of co-culture, while the cytotoxic effect was not significant in groups with higher E:T ratio. Our findings suggest that the anti-CD41 CAR T cells are efficient for a limited time spawn and the cytotoxic effect is visible in all experimental groups with low E:T ratio.
Collapse
Affiliation(s)
- Adrian Bogdan Tigu
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Catalin Sorin Constantinescu
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Intensive Care UnitEmergency Clinical HospitalCluj‐NapocaRomania
| | - Patric Teodorescu
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns HopkinsJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - David Kegyes
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Richard Feder
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Mareike Peters
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Ioana Pralea
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Cristina Iuga
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Drug AnalysisSchool of PharmacyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Diana Cenariu
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Andra Marcu
- Department of PediatricsCarol Davila University of Medicine and PharmacyBucharestRomania
- Department of Stem Cell TransplantationFundeni Clinical InstituteBucharestRomania
| | - Alina Tanase
- Department of PediatricsCarol Davila University of Medicine and PharmacyBucharestRomania
- Department of Stem Cell TransplantationFundeni Clinical InstituteBucharestRomania
| | - Anca Colita
- Department of PediatricsCarol Davila University of Medicine and PharmacyBucharestRomania
- Department of Stem Cell TransplantationFundeni Clinical InstituteBucharestRomania
| | - Rares Drula
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Jon Thor Bergthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health SciencesUniversity of IcelandReykjavíkIceland
- Department of Laboratory HematologyLandspitali University HospitalReykjavíkIceland
| | - Victor Greiff
- Department of ImmunologyUniversity of Oslo and Oslo University HospitalOsloNorway
| | - Delia Dima
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| | - Cristina Selicean
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| | - Ioana Rus
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| | - Mihnea Zdrenghea
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| | - Diana Gulei
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns HopkinsJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| |
Collapse
|
34
|
Kirkham JK, Liu YC, Foy SG, Ma J, Gheorghe G, Furtado LV, Popescu MI, Klco JM, Karol SE, Blackburn PR. Clinical and genomic characterization of an ATRA-insensitive acute promyelocytic leukemia variant with a FNDC3B::RARB fusion. Genes Chromosomes Cancer 2023; 62:617-623. [PMID: 37283355 DOI: 10.1002/gcc.23180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/08/2023] Open
Abstract
The promyelocytic leukemia-retinoic acid receptor-α (PML::RARA) fusion is the hallmark of acute promyelocytic leukemia (APL) and is observed in over 95% of APL cases. RARA and homologous receptors RARB and RARG are occasionally fused to other gene partners, which differentially affect sensitivity to targeted therapies. Most APLs without RARA fusions have rearrangements involving RARG or RARB, both of which frequently show resistance to all-trans-retinoic acid (ATRA) and/or multiagent chemotherapy for acute myeloid leukemia (AML). We present a 13-year-old male diagnosed with variant APL with a novel FNDC3B::RARB in-frame fusion that showed no response to ATRA but responded well to conventional AML therapy. While FNDC3B has been identified as a rare RARA translocation partner in ATRA-sensitive variant APL, it has never been reported as a fusion partner with RARB and it is only the second known fusion partner with RARB in variant APL. We also show that this novel fusion confers an RNA expression signature that is similar to APL, despite clinical resistance to ATRA monotherapy.
Collapse
MESH Headings
- Male
- Humans
- Adolescent
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/metabolism
- Translocation, Genetic
- Tretinoin/therapeutic use
- Leukemia, Myeloid, Acute/genetics
- Retinoic Acid Receptor alpha/genetics
- Genomics
- Oncogene Proteins, Fusion/genetics
- Fibronectins/genetics
Collapse
Affiliation(s)
- Justin K Kirkham
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yen-Chun Liu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Scott G Foy
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jing Ma
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Gabriela Gheorghe
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Larissa V Furtado
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Marcela I Popescu
- Department of Pediatric Hematology and Oncology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Patrick R Blackburn
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
35
|
Egan G, Tasian SK. Relapsed pediatric acute myeloid leukaemia: state-of-the-art in 2023. Haematologica 2023; 108:2275-2288. [PMID: 36861399 PMCID: PMC10483345 DOI: 10.3324/haematol.2022.281106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Although outcomes of children and adolescents with newly diagnosed acute myeloid leukemia (AML) have improved significantly over the past two decades, more than one-third of patients continue to relapse and experience suboptimal long-term outcomes. Given the small numbers of patients with relapsed AML and historical logistical barriers to international collaboration including poor trial funding and drug availability, the management of AML relapse has varied among pediatric oncology cooperative groups with several salvage regimens utilized and a lack of universally defined response criteria. The landscape of relapsed pediatric AML treatment is changing rapidly, however, as the international AML community harnesses collective knowledge and resources to characterize the genetic and immunophenotypic heterogeneity of relapsed disease, identify biological targets of interest within specific AML subtypes, develop new precision medicine approaches for collaborative investigation in early-phase clinical trials, and tackle challenges of universal drug access across the globe. This review provides a comprehensive overview of progress achieved to date in the treatment of pediatric patients with relapsed AML and highlights modern, state-of-the-art therapeutic approaches under active and emerging clinical investigation that have been facilitated by international collaboration among academic pediatric oncologists, laboratory scientists, regulatory agencies, pharmaceutical partners, cancer research sponsors, and patient advocates.
Collapse
Affiliation(s)
- Grace Egan
- Division of Haematology/Oncology, The Hospital for Sick Children, Department of Paediatrics, University of Toronto; Toronto, Ontario
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Pennsylvania, United States; University of Pennsylvania Perelman School of Medicine and Abramson Cancer Center; Philadelphia, Pennsylvania.
| |
Collapse
|
36
|
Tomizawa D, Tsujimoto SI. Risk-Stratified Therapy for Pediatric Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:4171. [PMID: 37627199 PMCID: PMC10452723 DOI: 10.3390/cancers15164171] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/08/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is the second most common type of leukemia in children. Recent advances in high-resolution genomic profiling techniques have uncovered the mutational landscape of pediatric AML as distinct from adult AML. Overall survival rates of children with AML have dramatically improved in the past 40 years, currently reaching 70% to 80% in developed countries. This was accomplished by the intensification of conventional chemotherapy, improvement in risk stratification using leukemia-specific cytogenetics/molecular genetics and measurable residual disease, appropriate use of allogeneic hematopoietic stem cell transplantation, and improvement in supportive care. However, the principle therapeutic approach for pediatric AML has not changed substantially for decades and improvement in event-free survival is rather modest. Further refinements in risk stratification and the introduction of emerging novel therapies to contemporary therapy, through international collaboration, would be key solutions for further improvements in outcomes.
Collapse
Affiliation(s)
- Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children’s Cancer Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Shin-Ichi Tsujimoto
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
| |
Collapse
|
37
|
Li SQ, Chen M, Huang XY, Wang H, Chang YJ. Challenges facing minimal residual disease testing for acute myeloid leukemia and promising strategies to overcome them. Expert Rev Hematol 2023; 16:981-990. [PMID: 37978882 DOI: 10.1080/17474086.2023.2285985] [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: 08/24/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Minimal residual disease (MRD) has been an important biomarker for relapse prediction and treatment choice in patients with acute myeloid leukemia (AML). False-positive or false-negative MRD results due to the low specificity and sensitivity of techniques such as multiparameter flow cytometry (MFC), real-time quantitative polymerase chain reaction, and next-generation sequencing, as well as the biological characteristics of residual leukemia cells, including antigen shift, clone involution, heterogeneous genome of the blast cells, and lack of specific targets, all restrict the clinical use of MRD. AREAS COVERED We summarized the challenges of the techniques for MRD detection, and their application in the clinical setting. We also discussed strategies to overcome these challenges, such as the MFC MRD method based on leukemia stem cells, single-cell DNA sequencing or single-cell RNA sequencing for the investigation of biological characteristics of residual leukemia cells, and the potential of omics techniques for MRD detection. We further noted out that prospective clinical trials are needed to answer clinical questions related to MRD in patients with AML. EXPERT OPINION MRD is an important biomarker for individual therapy of patients with AML. In the future, it is important to increase the specificity and sensitivity of the detection techniques.
Collapse
Affiliation(s)
- Si-Qi Li
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, Xicheng District, P.R.C
| | - Man Chen
- Department of Laboratory Medicine, Hebei Yanda Ludaopei Hospital, Langfang, Hebei, P.R.C
| | - Xi-Yi Huang
- Department of Experimental Medicine, School of Public Health, Xiamen University, Xiamen, P.R.C
| | - Hui Wang
- Department of Laboratory Medicine, Hebei Yanda Ludaopei Hospital, Langfang, Hebei, P.R.C
| | - Ying-Jun Chang
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Peking University Institute of Hematology, Beijing, Xicheng District, P.R.C
| |
Collapse
|
38
|
Marrero RJ, Cao X, Wu H, Elsayed AH, Klco JM, Ribeiro RC, Rubnitz JE, Ma X, Meshinchi S, Aplenc R, Kolb EA, Ries RE, Alonzo TA, Pounds SB, Lamba JK. SAMHD1 single nucleotide polymorphisms impact outcome in children with newly diagnosed acute myeloid leukemia. Blood Adv 2023; 7:2538-2550. [PMID: 36689724 PMCID: PMC10242642 DOI: 10.1182/bloodadvances.2022009088] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/08/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Cytarabine arabinoside (Ara-C) has been the cornerstone of acute myeloid leukemia (AML) chemotherapy for decades. After cellular uptake, it is phosphorylated into its active triphosphate form (Ara-CTP), which primarily exerts its cytotoxic effects by inhibiting DNA synthesis in proliferating cells. Interpatient variation in the enzymes involved in the Ara-C metabolic pathway has been shown to affect intracellular abundance of Ara-CTP and, thus, its therapeutic benefit. Recently, SAMHD1 (SAM and HD domain-containing deoxynucleoside triphosphate triphosphohydrolase 1) has emerged to play a role in Ara-CTP inactivation, development of drug resistance, and, consequently, clinical response in AML. Despite this, the impact of genetic variations in SAMHD1 on outcome in AML has not been investigated in depth. In this study, we evaluated 25 single nucleotide polymorphisms (SNPs) within the SAMHD1 gene for association with clinical outcome in 400 pediatric patients with newly diagnosed AML from 2 clinical trials, AML02 and AML08. Three SNPs, rs1291128, rs1291141, and rs7265241 located in the 3' region of SAMHD1 were significantly associated with at least 1 clinical outcome: minimal residual disease after induction I, event-free survival (EFS), or overall survival (OS) in the 2 cohorts. In an independent cohort of patients from the COG-AAML1031 trial (n = 854), rs7265241 A>G remained significantly associated with EFS and OS. In multivariable analysis, all the SNPs remained independent predictors of clinical outcome. These results highlight the relevance of the SAMHD1 pharmacogenomics in context of response to Ara-C in AML and warrants the need for further validation in expanded patient cohorts.
Collapse
Affiliation(s)
- Richard J. Marrero
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL
| | - Xueyuan Cao
- Department of Health Promotion and Disease Prevention, University of Tennessee Health Science Center, Memphis, TN
| | - Huiyun Wu
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Abdelrahman H. Elsayed
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL
| | - Jeffery M. Klco
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Raul C. Ribeiro
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jeffrey E. Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Richard Aplenc
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - E. Anders Kolb
- Nemours Center for Cancer and Blood Disorders, Alfred I. DuPont Hospital for Children, Wilmington, DE
| | - Rhonda E. Ries
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Todd A. Alonzo
- Children's Oncology Group Statistics and Data Center, Monrovia, CA
- Biostatistics Division, University of Southern California, Los Angeles, CA
| | - Stanley B. Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL
- Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| |
Collapse
|
39
|
Nguyen NHK, Rafiee R, Tagmount A, Sobh A, Loguinov A, de Jesus Sosa AK, Elsayed AH, Gbadamosi M, Seligson N, Cogle CR, Rubnitz J, Ribeiro R, Downing J, Cao X, Pounds SB, Vulpe CD, Lamba JK. Genome-wide CRISPR/Cas9 screen identifies etoposide response modulators associated with clinical outcomes in pediatric AML. Blood Adv 2023; 7:1769-1783. [PMID: 36111891 PMCID: PMC10182178 DOI: 10.1182/bloodadvances.2022007934] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/06/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022] Open
Abstract
Etoposide is used to treat a wide range of malignant cancers, including acute myeloid leukemia (AML) in children. Despite the use of intensive chemotherapeutic regimens containing etoposide, a significant proportion of pediatric patients with AML become resistant to treatment and relapse, leading to poor survival. This poses a pressing clinical challenge to identify mechanisms underlying drug resistance to enable effective pharmacologic targeting. We performed a genome-wide CRISPR/Cas9 synthetic-lethal screening to identify functional modulators of etoposide response in leukemic cell line and integrated results from CRISPR-screen with gene expression and clinical outcomes in pediatric patients with AML treated with etoposide-containing regimen. Our results confirmed the involvement of well-characterized genes, including TOP2A and ABCC1, as well as identified novel genes such as RAD54L2, PRKDC, and ZNF451 that have potential to be novel drug targets. This study demonstrates the ability for leveraging CRISPR/Cas9 screening in conjunction with clinically relevant endpoints to make meaningful discoveries for the identification of prognostic biomarkers and novel therapeutic targets to overcome treatment resistance.
Collapse
Affiliation(s)
- Nam H. K. Nguyen
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| | - Roya Rafiee
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| | - Abderrahmane Tagmount
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL
| | - Amin Sobh
- UF Health Cancer Center, Gainesville, FL
| | - Alex Loguinov
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL
| | - Angelica K. de Jesus Sosa
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| | - Abdelrahman H. Elsayed
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| | - Mohammed Gbadamosi
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| | - Nathan Seligson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL
| | - Christopher R. Cogle
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL
| | - Jeffery Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Raul Ribeiro
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - James Downing
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Xueyuan Cao
- University of Tennessee Health Science Center, Memphis, TN
| | - Stanley B. Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL
| |
Collapse
|
40
|
[Chinese consensus on minimal residual disease detection and interpretation of patients with acute lymphoblastic leukemia (2023)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2023; 44:267-275. [PMID: 37356994 PMCID: PMC10282871 DOI: 10.3760/cma.j.issn.0253-2727.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Indexed: 06/27/2023]
|
41
|
Messori A, Hyeraci G. Meta-analysis on allogeneic transplant for treating pediatric patients with acute myeloid leukemia in first remission: reanalysis of primary data. Ann Hematol 2023; 102:1603-1605. [PMID: 36877260 DOI: 10.1007/s00277-023-05101-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/09/2023] [Indexed: 03/07/2023]
Affiliation(s)
- Andrea Messori
- Centro Operativo and HTA Committee, Regione Toscana, Viale Alderotti 26/N, 50136, Florence, Italy.
| | - Giulia Hyeraci
- Centro Operativo and HTA Committee, Regione Toscana, Viale Alderotti 26/N, 50136, Florence, Italy
| |
Collapse
|
42
|
Singh N, Gupta A, Kumar S, Mawalankar G, Gupta B, Dhole N, Kori R, Singh A. Flow cytometric measurable residual disease in adult acute myeloid leukemia: a preliminary report from Eastern India. J Hematop 2023; 16:17-25. [PMID: 38175369 DOI: 10.1007/s12308-022-00527-2] [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: 10/15/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Presence of measurable residual disease (MRD) in acute myeloid leukemia (AML) is considered to be an independent predictor of relapse and poorer survival outcomes. MRD can be measured by flow cytometric, quantitative PCR, and NGS-based assays at varying sensitivities. There is scant Indian data on different aspects of MFC-MRD in AML including analysis strategies as well as molecular spectrum, clinical correlation, etc. This retrospective observational study included all newly diagnosed patients of acute myeloid leukemia in whom complete baseline diagnostic workup was available including flow cytometry and cytogenetic and molecular studies. Among patients with cytogenetic abnormalities (n = 25), no statistically significant correlation was observed between flow cytometric MRD positivity and presence of ≥ 3 mutations as well as relapsed disease. However, in AML patients with normal karyotype (n = 32), MRD positivity correlated strongly with relapsed status (p = 0.02), although no significant correlation was found with respect to FLT3 mutation, IDH mutation, NPM1 mutation, or complex genotype. Interestingly, 90.5% of MRD-positive patients belonged to ELN (2017) intermediate to high-risk category unlike only 9.5% in the good risk category (p = 0.0002). Median relapse-free survival was 8.5 months with a follow-up range of 3-24 months. On the basis of the observations of the present study, it can be clearly inferred that MRD status affects relapse status in the normal karyotype subgroup and can delineate patients who require stem cell transplantation in addition to molecular signatures.
Collapse
Affiliation(s)
- Neha Singh
- Hematopathology, Tata Memorial Center, Varanasi, India.
| | - Avinash Gupta
- Hematopathology, Tata Memorial Center, Varanasi, India
| | - Sujeet Kumar
- Adult Hematolymphoid Unit, Tata Memorial Center, Varanasi, India
| | | | - Bhumika Gupta
- Hematopathology, Tata Memorial Center, Varanasi, India
| | - Nilesh Dhole
- Hematopathology, Tata Memorial Center, Varanasi, India
| | | | - Anil Singh
- Adult Hematolymphoid Unit, Tata Memorial Center, Varanasi, India
| |
Collapse
|
43
|
Karlsson L, Cheuk D, De Moerloose B, Hasle H, Jahnukainen K, Juul-Dam KL, Kaspers G, Kovalova Z, Lausen B, Nyström UN, Palle J, Pronk CJ, Saks K, Tierens A, Zeller B, Abrahamsson J. Characteristics and outcome of primary resistant disease in paediatric acute myeloid leukaemia. Br J Haematol 2023; 201:757-765. [PMID: 36762836 DOI: 10.1111/bjh.18685] [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: 11/29/2022] [Revised: 01/16/2023] [Accepted: 01/25/2023] [Indexed: 02/11/2023]
Abstract
A significant proportion of events in paediatric acute myeloid leukaemia (AML) are caused by resistant disease (RD). We investigated clinical and biological characteristics in 66 patients with RD from 1013 children with AML registered and treated according to the NOPHO-AML 93, NOPHO-AML 2004, DB AML-01 and NOPHO-DBH AML 2012 protocols. Risk factors for RD were age10 years or older and a white-blood-cell count (WBC) of 100 × 109 /L or more at diagnosis. The five-year overall survival (OS) was 38% (95% confidence interval [CI]: 28%-52%). Of the 63 children that received salvage therapy with chemotherapy, 59% (N = 37) achieved complete remission (CR) with OS 57% (95% CI: 42%-75%) compared to 12% (95% CI: 4%-35%) for children that did not achieve CR. Giving more than two salvage chemotherapy courses did not increase CR rates. OS for all 43 patients receiving allogeneic haematopoietic stem cell transplantation (HSCT) was 49% (95% CI: 36%-66%). Those achieving CR and proceeding to HSCT had an OS of 56% (95% CI: 41%-77%, N = 30). This study showed that almost 40% of children with primary resistant AML can be cured with salvage therapy followed by HSCT. Children that did not achieve CR after two salvage courses with chemotherapy did not benefit from additional chemotherapy.
Collapse
Affiliation(s)
- Lene Karlsson
- Department of Pediatrics, Institution for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Cheuk
- Department of Pediatrics and Adolescent Medicine, Hong Kong Children's Hospital and Hong Kong Pediatric Hematology and Oncology Study Group (HKPHOSG), Hong Kong, China
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology, Ghent University Hospital, Ghent, Belgium
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Kirsi Jahnukainen
- New Children's hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Gertjan Kaspers
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
| | - Zanna Kovalova
- Department of Paediatric Oncology/Haematology, Children's Clinical University Hospital, Riga, Latvia
| | - Birgitte Lausen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Josefine Palle
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | | | - Kadri Saks
- Department of Paediatrics, SA Tallinna Lastehaigla, Tallinn, Estonia
| | - Anne Tierens
- Department of Pathobiology and Laboratory Medicine, University Health Network, Toronto General Hospital, Toronto, Ontario, Canada
| | - Bernward Zeller
- Department of Pediatrics, Oslo University Hospital, Oslo, Norway
| | - Jonas Abrahamsson
- Department of Pediatrics, Institution for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
44
|
Shiba N. Comprehensive molecular understanding of pediatric acute myeloid leukemia. Int J Hematol 2023; 117:173-181. [PMID: 36653696 DOI: 10.1007/s12185-023-03533-x] [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: 10/31/2022] [Revised: 12/28/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
Pediatric acute myeloid leukemia (AML) is a heterogeneous disease with various genetic abnormalities. Recent advances in genetic analysis have enabled the identification of causative genes in > 90% of pediatric AML cases. Fusion genes such as RUNX1::RUNX1T1, CBFB::MYH11, and KMT2A::MLLT3 are frequently detected in > 70% of pediatric AML cases, whereas FLT3-internal tandem duplication, CEBPA-bZip, and NPM1 mutations are detected in approximately 5-15% of cases, respectively. Conversely, mutations in DNMT3A, TET2, and IDH, which are common in adults, are extremely rare in pediatric AML. The genetic characteristics of pediatric AML are slightly different from those of adult AML. For accurate risk stratification and treatment intensity, genome analysis should be performed in a simple, fast, and inexpensive manner and the results should be returned to patients in real time. As with acute lymphoblastic leukemia, the presence or absence of minimal residual disease is an important factor in determining the success of treatment against AML, and it is important to predict prognosis and formulate treatment strategies considering the genetic abnormalities. For the development and clinical application of new molecularly targeted therapies based on identified genetic abnormalities, it is necessary to explore when and in which combinations drugs will be most effective.
Collapse
Affiliation(s)
- Norio Shiba
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
45
|
Fujikawa T, Kishimoto K, Inoue S, Nishimura A, Tojo R, Uemura S, Nakamura S, Saito A, Kozaki A, Ishida T, Mori T, Higashino M, Koyama J, Kawamura A, Hasegawa D, Kosaka Y. Epidural Spinal Cord Compression as the Presenting Manifestation of Acute Myeloid Leukemia: A Case Report and Literature Review. Intern Med 2023; 62:453-457. [PMID: 35793962 PMCID: PMC9970809 DOI: 10.2169/internalmedicine.9580-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We herein report a rare case of spinal cord compression due to epidural involvement of acute myeloid leukemia (AML). A 14-year-old boy presented with a 7-day history of back pain, paraplegia and hypoesthesia. Contrast-enhanced computed tomography revealed an epidural mass. Emergency laminectomy and resection of the mass were performed. Histopathologically, the resected mass was comparable to an extramedullary mass of AML. Chemotherapy was initiated, and complete remission was achieved. Neurological sequelae remained after the treatment. Based on the present and previous reports, spinal cord compression from epidural AML involvement may progress rapidly.
Collapse
Affiliation(s)
- Tomoko Fujikawa
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Kenji Kishimoto
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Shotaro Inoue
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Akihiro Nishimura
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Ryunosuke Tojo
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Suguru Uemura
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Sayaka Nakamura
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Atsuro Saito
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Aiko Kozaki
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Toshiaki Ishida
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | - Takeshi Mori
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| | | | - Junji Koyama
- Department of Neurosurgery, Kobe Children's Hospital, Japan
| | | | | | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Japan
| |
Collapse
|
46
|
Ramos Elbal E, Fuster JL, Campillo JA, Galera AM, Cortés MB, Llinares ME, Jiménez I, Plaza M, Banaclocha HM, Galián JA, Blanquer Blanquer M, Martínez Sánchez MV, Muro M, Minguela A. Measurable residual disease study through three different methods can anticipate relapse and guide pre-emptive therapy in childhood acute myeloid leukemia. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:1446-1454. [PMID: 36598635 DOI: 10.1007/s12094-022-03042-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/04/2022] [Indexed: 01/05/2023]
Abstract
PURPOSE Although outcomes of children with acute myeloid leukemia (AML) have improved over the last decades, around one-third of patients relapse. Measurable (or minimal) residual disease (MRD) monitoring may guide therapy adjustments or pre-emptive treatments before overt hematological relapse. METHODS In this study, we review 297 bone marrow samples from 20 real-life pediatric AML patients using three MRD monitoring methods: multiparametric flow cytometry (MFC), fluorescent in situ hybridization (FISH) and polymerase chain reaction (PCR). RESULTS Patients showed a 3-year overall survival of 73% and a 3-year event-free survival of 68%. Global relapse rate was of 25%. All relapses were preceded by the reappearance of MRD detection by: (1) MFC (p = 0.001), (2) PCR and/or FISH in patients with an identifiable chromosomal translocation (p = 0.03) and/or (3) one log increase of Wilms tumor gene 1 (WT1) expression in two consecutive samples (p = 0.02). The median times from MRD detection to relapse were 26, 111, and 140 days for MFC, specific PCR and FISH, and a one log increment of WT1, respectively. CONCLUSIONS MFC, FISH and PCR are complementary methods that can anticipate relapse of childhood AML by weeks to several months. However, in our series, pre-emptive therapies were not able to prevent disease progression. Therefore, more sensitive MRD monitoring methods that further anticipate relapse and more effective pre-emptive therapies are needed.
Collapse
Affiliation(s)
- Eduardo Ramos Elbal
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - José Luis Fuster
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - José Antonio Campillo
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Ana María Galera
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Mar Bermúdez Cortés
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - María Esther Llinares
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Irene Jiménez
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Mercedes Plaza
- Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Helios Martínez Banaclocha
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - José Antonio Galián
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Miguel Blanquer Blanquer
- Haematology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - María Victoria Martínez Sánchez
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Manuel Muro
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain
| | - Alfredo Minguela
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120, Murcia, Spain.
| |
Collapse
|
47
|
Wang B, Wen X, Zhang R, Zhu G, Wu Y, Zhang Y, Lin W, Yu J, Fan J, Li J, Yang J, Qin M, Zheng H. Homoharringtonine-Based Induction Therapy Reduces the Recurrence Rate of Pediatric Acute Myeloid Leukemia After Allogeneic Hematopoietic Stem Cell Transplantation. Cell Transplant 2023; 32:9636897231183559. [PMID: 37470325 PMCID: PMC10363872 DOI: 10.1177/09636897231183559] [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: 03/17/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 07/21/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is an effective treatment for acute myeloid leukemia (AML). Pediatric patients with AML who relapse after HSCT have an extremely poor prognosis. We performed a retrospective study of pediatric patients diagnosed with AML from August 2015 to October 2019 who were treated with HSCT. Kaplan-Meier analyses were used to evaluate overall survival (OS), event-free survival (EFS), and cumulative recurrence rate (CRR). Cox regression analysis was used to determine the association between the baseline characteristics and relapse. A total of 37 pediatric patients met the inclusion criteria. Twenty-eight (75.7%) patients survived, and 9 (24.3%) patients died. The OS rates of AML patients treated with HSCT were 89.2% ± 5.1%, 75.7% ± 7.1%, and 75.7% ± 7.1% at 1, 3, and 5 years, respectively, and the CRRs were 11.4% ± 5.4%, 24.7% ± 7.7%, and 33.1% ± 10.4% at 1, 3, and 5 years after HSCT, respectively; four of nine children who relapsed after transplantation died. Induction with etoposide rather than homoharringtonine and fungal infections could be high-risk factors for recurrence after transplantation. The association between homoharringtonine-based induction therapy and a low recurrence rate persisted after adjusting for age, sex, risk stratification, fusion genes, and fungal infections. This study clarifies the clinical features and poor prognosis of post-transplant relapse in pediatric AML and indicates the urgent need for effective therapy for patients who relapse after HSCT.
Collapse
Affiliation(s)
- Bin Wang
- Transplantation Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xiaojia Wen
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Ruidong Zhang
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Guanghua Zhu
- Transplantation Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Ying Wu
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Yuanyuan Zhang
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Wei Lin
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jiaole Yu
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jia Fan
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jing Li
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jun Yang
- Transplantation Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Maoquan Qin
- Transplantation Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Huyong Zheng
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| |
Collapse
|
48
|
Wijnen NE, Koedijk JB, Klein K, Luesink M, Goemans BF, Zwaan CM, Kaspers GJL. Treating CD33-Positive de novo Acute Myeloid Leukemia in Pediatric Patients: Focus on the Clinical Value of Gemtuzumab Ozogamicin. Onco Targets Ther 2023; 16:297-308. [PMID: 37153641 PMCID: PMC10155714 DOI: 10.2147/ott.s263829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/20/2023] [Indexed: 05/10/2023] Open
Abstract
Although survival in pediatric acute myeloid leukemia (AML) has increased considerably over the past decades, refractory disease and relapse rates remain high. Refractory and relapsed disease are difficult to treat, with overall survival rates less than 40-50%. Preventing relapse should, therefore, be one of the highest priorities. Current conventional chemotherapy regimens are hard to intensify due to associated toxic complications, hence more effective therapies that do not increase toxicity are needed. A promising targeted agent is the CD33-directed antibody-drug conjugate gemtuzumab ozogamicin (GO). Because CD33 is highly expressed on leukemic cells in the majority of AML patients, GO can be useful for a broad range of patients. Better relapse-free survival (RFS) after therapy including GO has been reported in several pediatric clinical trials; however, ambiguity about the clinical value of GO in newly diagnosed children remains. Treatment with GO in de novo AML patients aged ≥1 month, in combination with standard chemotherapy is approved in the United States, whereas in Europe, GO is only approved for newly diagnosed patients aged ≥15 years. In this review, we aimed to clarify the clinical value of GO for treatment of newly diagnosed pediatric AML patients. Based on current literature, GO seems to have additional value, in terms of RFS, and acceptable toxicity when used in addition to chemotherapy during initial treatment. Moreover, in KMT2A-rearranged patients, the clinical value of GO was even more evident. Also, we addressed predictors of response, being CD33 expression and SNPs, PgP-1 and Annexin A5. The near finalized intent-to-file clinical trial in the MyeChild consortium investigates whether fractionated dosing has additional value for pediatric AML, which may pave the way for a broader application of GO in pediatric AML.
Collapse
Affiliation(s)
- Noa E Wijnen
- Pediatric Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pediatric Oncology, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
- Correspondence: Noa E Wijnen, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht, 3584 CS, the Netherlands, Tel +31(0)889727272, Email
| | - Joost B Koedijk
- Pediatric Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pediatric Oncology, Erasmus MC-Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Kim Klein
- Pediatric Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Wilhelmina Children’s Hospital/University Medical Center, Utrecht, the Netherlands
| | - Maaike Luesink
- Pediatric Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Bianca F Goemans
- Pediatric Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - C Michel Zwaan
- Pediatric Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pediatric Oncology, Erasmus MC-Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Gertjan J L Kaspers
- Pediatric Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pediatric Oncology, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| |
Collapse
|
49
|
Meena JP, Makkar H, Gupta AK, Bakhshi S, Gupta R, Thakral D, Chopra A, Tanwar P, Upadhyay AD, Pathak N, Seth R. A comprehensive analysis of cytogenetics, molecular profile, and survival among pediatric acute myeloid leukemia: a prospective study from a tertiary referral center. AMERICAN JOURNAL OF BLOOD RESEARCH 2022; 12:177-189. [PMID: 36742278 PMCID: PMC9890188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/15/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS The objectives of this study were to investigate the cyto-molecular profile and survival of pediatric acute myeloid leukemia (AML). METHODS This prospective study was carried out in a tertiary care hospital from October 2018 to December 2020. Karyotype and cytogenetics analyses were done to identify chromosomal aberrations in pediatric AML. The targeted molecular panel utilized the polymerase chain reaction (PCR), reverse transcription-polymerase chain reaction (RT-PCR), and fragment analysis. RESULTS A total of 70 patients of AML with aged ≤18 years were enrolled in this study. The cytogenetic analyses revealed abnormal/recurrent cytogenetic abnormalities (CA) in 64.3% of patients and normal cytogenetics (CN) in 35.7% of patients. FAB M2 subtype showed frequent aberrant expression of the CD19 marker. CD7, CD11b, and CD36a were significantly present in the absence of molecular markers. Common chromosomal abnormalities were t(translocation) (8;21) (55%), monosomy/deletion 7 (13%), monosomal karyotype (5%) and complex karyotype (3%). The fusion transcripts RUNX1-RUNX1T1 [t(8;21)] (41%) and CBFB-MYH11 [t(16;16)] (3%) were detected by RT-PCR and FLT3-TKD D835 mutation (1.5%) by allele-specific oligo PCR. Fragment analysis revealed NPM1 (8%) mutation and FLT-ITD (9.5%) mutations. Complete remission was achieved in all evaluable patients. The median follow-up period of our patients was 225 days (IQR 28; 426 days). The median event-free survival (EFS) in all patients was 11.9 months (95% CI, 5-12.6 months). The forty months overall survival probability (pOS) was 58% in all patients. CONCLUSION The majority of patients had abnormal/recurrent cytogenetics abnormalities. FAB M2 subtype showed frequent aberrant expression of the CD19. The absence of molecular markers may suggest the presence of CD7, CD11b, and CD36a expression. The overall survival has increased considerably in LMIC.
Collapse
Affiliation(s)
- Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Harshita Makkar
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Aditya Kumar Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B.R.A. IRCH, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr. B.R.A. IRCH, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Deepshi Thakral
- Laboratory Oncology Unit, Dr. B.R.A. IRCH, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr. B.R.A. IRCH, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Pranay Tanwar
- Laboratory Oncology Unit, Dr. B.R.A. IRCH, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ashish Datt Upadhyay
- Department of Biostatistics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Nivedita Pathak
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Rachna Seth
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| |
Collapse
|
50
|
Ganzel C, Sun Z, Baslan T, Zhang Y, Gönen M, Abdel-Wahab OI, Racevskis J, Garrett-Bakelman F, Lowe SW, Fernandez HF, Ketterling R, Luger SM, Litzow M, Lazarus HM, Rowe JM, Tallman MS, Levine RL, Paietta E. Measurable residual disease by flow cytometry in acute myeloid leukemia is prognostic, independent of genomic profiling. Leuk Res 2022; 123:106971. [PMID: 36332294 PMCID: PMC9789386 DOI: 10.1016/j.leukres.2022.106971] [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: 05/02/2022] [Revised: 10/04/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
Abstract
Measurable residual disease (MRD) assessment provides a potent indicator of the efficacy of anti-leukemic therapy. It is unknown, however, whether integrating MRD with molecular profiling better identifies patients at risk of relapse. To investigate the clinical relevance of MRD in relation to a molecular-based prognostic schema, we measured MRD by flow cytometry in 189 AML patients enrolled in ECOG-ACRIN E1900 trial (NCT00049517) in morphologic complete remission (CR) (28.8 % of the original cohort) representing 44.4 % of CR patients. MRD positivity was defined as ≥ 0.1 % of leukemic bone marrow cells. Risk classification was based on standard cytogenetics, fluorescence-in-situ-hybridization, somatic gene analysis, and sparse whole genome sequencing for copy number ascertainment. At 84.6 months median follow-up of patients still alive at the time of analysis (range 47.0-120 months), multivariate analysis demonstrated that MRD status at CR (p = 0.001) and integrated molecular risk (p = 0.0004) independently predicted overall survival (OS). Among risk classes, MRD status significantly affected OS only in the favorable risk group (p = 0.002). Expression of CD25 (α-chain of the interleukin-2 receptor) by leukemic myeloblasts at diagnosis negatively affected OS independent of post-treatment MRD levels. These data suggest that integrating MRD with genetic profiling and pre-treatment CD25 expression may improve prognostication in AML.
Collapse
Affiliation(s)
- Chezi Ganzel
- Hematology Department, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| | - Zhuoxin Sun
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar I Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Janis Racevskis
- Department of Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Francine Garrett-Bakelman
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Departments of Medicine and Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hugo F Fernandez
- Malignant Hematology and Cellular Therapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Rhett Ketterling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Selina M Luger
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Litzow
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jacob M Rowe
- Hematology Department, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Martin S Tallman
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|