1
|
Péterffy B, Krizsán S, Egyed B, Bedics G, Benard-Slagter A, Palit S, Erdélyi DJ, Müller J, Nagy T, Hegyi LL, Bekő A, Kenéz LA, Jakab Z, Péter G, Zombori M, Csanádi K, Ottóffy G, Csernus K, Vojcek Á, Tiszlavicz LG, Gábor KM, Kelemen Á, Hauser P, Kállay K, Kertész G, Gaál Z, Szegedi I, Barna G, Márk Á, Haltrich I, Hevessy Z, Ujfalusi A, Kajtár B, Timár B, Kiss C, Kriván G, Matolcsy A, Savola S, Kovács G, Bödör C, Alpár D. Molecular Profiling Reveals Novel Gene Fusions and Genetic Markers for Refined Patient Stratification in Pediatric Acute Lymphoblastic Leukemia. Mod Pathol 2025; 38:100741. [PMID: 40010436 DOI: 10.1016/j.modpat.2025.100741] [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: 12/04/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/28/2025]
Abstract
Risk-adapted treatment protocols conferred remarkable improvement in the survival rates of pediatric acute lymphoblastic leukemia/lymphoma (ALL/LBL). Nevertheless, clinical management is still challenging in certain molecular subgroups and in the presence of alterations associated with an increased rate of relapse. In this study, disease-relevant genomic and transcriptomic profiles were established in a prospective, multicenter, real-world cohort involving 192 children diagnosed with ALL/LBL. Gene fusions were detected in 34.9% of B-ALL and 46.4% of T-ALL patients, with novel chimeric genes involving JAK2, KMT2A, PAX5, RUNX1, and NOTCH1, and with KMT2A-rearranged patients displaying the worst 3-year event-free survival (P = .019). Nonsynonymous mutations were uncovered in 74.9% of the analyzed patients, and pairwise scrutiny of genetic lesions revealed recurrent clonal selection mechanisms commonly converging on the same pathway (eg, Ras, JAK/STAT, and Notch) in individual patients. Investigation of matched diagnostic and relapse samples unraveled complex subclonal variegation, and mutations affecting the NT5C2, TP53, CDKN2A, and PIK3R1 genes, emerging at the time of relapse. TP53 and CREBBP mutations, even as subclonal aberrations, were associated with shorter 3-year event-free survival among all patients with B-ALL (TP53 mutant vs wild-type: P = .008, CREBBP mutant vs wild-type: P = .010), and notably, B-ALL patients showing no measurable residual disease on day 33 could be further stratified based on TP53 mutational status (P < .001). Our in-depth molecular characterization performed across all risk groups identified novel opportunities for molecularly targeted therapy in 55.9% of high-risk and 31.6% of standard/intermediate-risk patients.
Collapse
Affiliation(s)
- Borbála Péterffy
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Szilvia Krizsán
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Bálint Egyed
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Gábor Bedics
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | - Sander Palit
- MRC Holland, Department of Oncogenetics, Amsterdam, The Netherlands
| | | | - Judit Müller
- Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Tibor Nagy
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Lajos László Hegyi
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Anna Bekő
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Lili Anna Kenéz
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Jakab
- Hungarian Childhood Cancer Registry, Hungarian Pediatric Oncology Network, Budapest, Hungary
| | - György Péter
- Hemato-Oncology Unit, Heim Pál Children's Hospital, Budapest, Hungary
| | - Marianna Zombori
- Hemato-Oncology Unit, Heim Pál Children's Hospital, Budapest, Hungary
| | - Krisztina Csanádi
- Hemato-Oncology Unit, Heim Pál Children's Hospital, Budapest, Hungary
| | - Gábor Ottóffy
- Department of Pediatrics, Oncohaematology Division, University of Pécs Medical School, Pécs, Hungary
| | - Katalin Csernus
- Department of Pediatrics, Oncohaematology Division, University of Pécs Medical School, Pécs, Hungary
| | - Ágnes Vojcek
- Department of Pediatrics, Oncohaematology Division, University of Pécs Medical School, Pécs, Hungary
| | - Lilla Györgyi Tiszlavicz
- Department of Pediatrics and Pediatric Health Care Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Krisztina Mita Gábor
- Department of Pediatrics and Pediatric Health Care Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Ágnes Kelemen
- Velkey László Child's Health Center, Borsod-Abaúj-Zemplén County Central Hospital and University Teaching Hospital, Miskolc, Hungary
| | - Péter Hauser
- Velkey László Child's Health Center, Borsod-Abaúj-Zemplén County Central Hospital and University Teaching Hospital, Miskolc, Hungary
| | - Krisztián Kállay
- Pediatric Hematology and Stem Cell Transplantation Department, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Gabriella Kertész
- Pediatric Hematology and Stem Cell Transplantation Department, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Zsuzsanna Gaál
- Division of Pediatric Hematology-Oncology, Institute of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Szegedi
- Division of Pediatric Hematology-Oncology, Institute of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Barna
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ágnes Márk
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Irén Haltrich
- Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Hevessy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Hungary
| | - Anikó Ujfalusi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Hungary
| | - Béla Kajtár
- Department of Pathology, University of Pécs Medical School, Pécs, Hungary
| | - Botond Timár
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Csongor Kiss
- Division of Pediatric Hematology-Oncology, Institute of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gergely Kriván
- Pediatric Hematology and Stem Cell Transplantation Department, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - András Matolcsy
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Suvi Savola
- MRC Holland, Department of Oncogenetics, Amsterdam, The Netherlands
| | - Gábor Kovács
- Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Csaba Bödör
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Donát Alpár
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE "Lendület" Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
2
|
Bartram J, Ancliff P, Vora A. How I treat infant acute lymphoblastic leukemia. Blood 2025; 145:35-42. [PMID: 38905593 DOI: 10.1182/blood.2023023154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024] Open
Abstract
ABSTRACT Infant acute lymphoblastic leukemia (ALL) is an aggressive malignancy that has historically been associated with a very poor prognosis. Despite large cooperative international trials and incremental increases in intensity of therapy, there has been no significant improvement in outcome over the last 3 decades. Using representative cases, we highlight the key differences between KMT2A-rearranged and KMT2A-germ line infant ALL, and how advances in molecular diagnostics are unpicking KMT2A-germ line genetics and guiding treatment reduction. We focus on KM2TA-rearranged infant B-cell ALL for which the last few years have seen the emergence of novel therapies that both are more effective and less toxic than conventional chemotherapy. Of these, there is promising early data on the efficacy and tolerability of the bispecific T-cell engager monoclonal antibody, blinatumomab, as well as the use of autologous and allogeneic chimeric antigen receptor T-cell therapy. We discuss how we can improve risk stratification and incorporate these new agents to replace the most toxic elements of currently deployed intensive chemotherapy schedules with their associated unacceptable toxicity.
Collapse
Affiliation(s)
- Jack Bartram
- Department of Haematology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Philip Ancliff
- Department of Haematology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Ajay Vora
- Department of Haematology, Great Ormond Street Hospital for Children, London, United Kingdom
| |
Collapse
|
3
|
Guest EM, Kairalla JA, Devidas M, Hibbitts E, Carroll AJ, Heerema NA, Kubaney HR, August MA, Ramesh S, Yoo B, Farooqi MS, Pauly MG, Wechsler DS, Miles RR, Reid JM, Kihei CD, Gore L, Raetz EA, Hunger SP, Loh ML, Brown PA. Azacitidine as epigenetic priming for chemotherapy is safe and well-tolerated in infants with newly diagnosed KMT2A-rearranged acute lymphoblastic leukemia: Children's Oncology Group trial AALL15P1. Haematologica 2024; 109:3918-3927. [PMID: 38867582 PMCID: PMC11609799 DOI: 10.3324/haematol.2024.285158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024] Open
Abstract
Infants less than 1 year old diagnosed with KMT2A-rearranged (KMT2A-r) acute lymphoblastic leukemia (ALL) are at high risk of failure to achieve remission, relapse, and death due to leukemia, despite intensive therapies. Infant KMT2A-r ALL blasts are characterized by DNA hypermethylation. Epigenetic priming with DNA methyltransferase inhibitors increases the cytotoxicity of chemotherapy in preclinical studies. The Children's Oncology Group trial AALL15P1 tested the safety and tolerability of 5 days of azacitidine treatment immediately prior to the start of chemotherapy on day 6, in four post-induction chemotherapy courses for infants with newly diagnosed KMT2A-r ALL. The treatment was well-tolerated, with only two of 31 evaluable patients (6.5%) experiencing dose-limiting toxicity. Whole genome bisulfite sequencing of peripheral blood mononuclear cells demonstrated decreased DNA methylation in 87% of samples tested following 5 days of azacitidine treatment. Event-free survival was similar to that in prior studies of newly diagnosed infant ALL. Azacitidine is safe and results in decreased DNA methylation of peripheral blood mononuclear cells in infants with KMT2A-r ALL, but the incorporation of azacitidine to enhance cytotoxicity did not impact survival. Clinicaltrials.gov identifier: NCT02828358.
Collapse
Affiliation(s)
- Erin M Guest
- Division of Hematology, Oncology, Bone Marrow Transplant, Children's Mercy Kansas City, University of Missouri-Kansas City School of Medicine, Kansas City, MO.
| | - John A Kairalla
- Department of Biostatistics, University of Florida, Gainesville, FL
| | | | - Emily Hibbitts
- Department of Biostatistics, University of Florida, Gainesville, FL
| | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL
| | - Nyla A Heerema
- Department of Pathology, The Ohio State University, Columbus, OH
| | | | - Margaret A August
- Health Informatics and Technology, Children's Mercy Kansas City, Kansas City, MO
| | | | - Byunggil Yoo
- Research Informatics, Children's Mercy Kansas City, Kansas City, MO
| | - Midhat S Farooqi
- Department of Pathology and Laboratory Medicine, Children's Mercy Kansas City, University of Missouri-Kansas City School of Medicine, Kansas City, MO
| | - Melinda G Pauly
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta and Emory University, Atlanta, GA
| | - Daniel S Wechsler
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta and Emory University, Atlanta, GA
| | - Rodney R Miles
- University of Utah, Department of Pathology, Salt Lake City, UT
| | | | | | - Lia Gore
- Children's Hospital Colorado, Center for Cancer and Blood Disorders, Denver, CO
| | | | - Stephen P Hunger
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Mignon L Loh
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute and the Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, 98105
| | | |
Collapse
|
4
|
Cuglievan B, Kantarjian H, Rubnitz JE, Cooper TM, Zwaan CM, Pollard JA, DiNardo CD, Kadia TM, Guest E, Short NJ, McCall D, Daver N, Nunez C, Haddad FG, Garcia M, Bhalla KN, Maiti A, Catueno S, Fiskus W, Carter BZ, Gibson A, Roth M, Khazal S, Tewari P, Abbas HA, Bourgeois W, Andreeff M, Shukla NN, Truong DD, Connors J, Ludwig JA, Stutterheim J, Salzer E, Juul-Dam KL, Sasaki K, Mahadeo KM, Tasian SK, Borthakur G, Dickson S, Jain N, Jabbour E, Meshinchi S, Garcia-Manero G, Ravandi F, Stein EM, Kolb EA, Issa GC. Menin inhibitors in pediatric acute leukemia: a comprehensive review and recommendations to accelerate progress in collaboration with adult leukemia and the international community. Leukemia 2024; 38:2073-2084. [PMID: 39179671 PMCID: PMC11436367 DOI: 10.1038/s41375-024-02368-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/26/2024]
Abstract
Aberrant expression of HOX and MEIS1 family genes, as seen in KMT2A-rearranged, NUP98-rearranged, or NPM1-mutated leukemias leads to arrested differentiation and leukemia development. HOX family genes are essential gatekeepers of physiologic hematopoiesis, and their expression is regulated by the interaction between KMT2A and menin. Menin inhibitors block this interaction, downregulate the abnormal expression of MEIS1 and other transcription factors and thereby release the differentiation block. Menin inhibitors show significant clinical efficacy against KMT2A-rearranged and NPM1-mutated acute leukemias, with promising potential to address unmet needs in various pediatric leukemia subtypes. In this collaborative initiative, pediatric and adult hematologists/oncologists, and stem cell transplant physicians have united their expertise to explore the potential of menin inhibitors in pediatric leukemia treatment internationally. Our efforts aim to provide a comprehensive clinical overview of menin inhibitors, integrating preclinical evidence and insights from ongoing global clinical trials. Additionally, we propose future international, inclusive, and efficient clinical trial designs, integrating pediatric populations in adult trials, to ensure broad access to this promising therapy for all children and adolescents with menin-dependent leukemias.
Collapse
Affiliation(s)
- Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey E Rubnitz
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Todd M Cooper
- Cancer and Blood Disorders Center, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - C Michel Zwaan
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands; Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands; The Innovative Therapies for Children with Cancer Consortium, Paris, France
| | | | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erin Guest
- Department of Pediatric Oncology, Children's Mercy, Kansas City, MO, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David McCall
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cesar Nunez
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fadi G Haddad
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Miriam Garcia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kapil N Bhalla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samanta Catueno
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Warren Fiskus
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bing Z Carter
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amber Gibson
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Roth
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sajad Khazal
- Division of Transplant and Cellular Therapy, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Priti Tewari
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Hussein A Abbas
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neerav N Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Danh D Truong
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeremy Connors
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Joseph A Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Elisabeth Salzer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Kristian L Juul-Dam
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kris M Mahadeo
- Division of Pediatric Transplantation and Cellular Therapy, Duke University, Durham, NC, USA
| | - Sarah K Tasian
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samantha Dickson
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eytan M Stein
- Department of Leukemia, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Anders Kolb
- Moseley Institute for Cancer and Blood Disorders, Nemours Children's Health, Wilmington, DE, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
5
|
Burmeister T, Ströh AS, Kehden B, Trautmann H, Meyer C, Marschalek R, Larghero P, Schwartz S, Steffen B, Spriewald B, Heinicke T, Jäkel N, Westermann J, Nachtkamp K, Viardot A, Topp MS, Neumann M, Baldus CD, Gökbuget N, Brüggemann M. Measurable residual disease quantification in adult patients with KMT2A-rearranged acute lymphoblastic leukemia. Leukemia 2024; 38:1600-1603. [PMID: 38519799 PMCID: PMC11216977 DOI: 10.1038/s41375-024-02209-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/25/2024]
Affiliation(s)
- Thomas Burmeister
- Department of Hematology, Oncology and Tumor Immunology, CVK, Charité-Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Aeint-Steffen Ströh
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Britta Kehden
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Heiko Trautmann
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Claus Meyer
- Diagnostic Center of Acute Leukemia (DCAL), Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Rolf Marschalek
- Diagnostic Center of Acute Leukemia (DCAL), Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Patrizia Larghero
- Diagnostic Center of Acute Leukemia (DCAL), Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Stefan Schwartz
- Department of Hematology, Oncology and Tumor Immunology, CBF, Charité Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Björn Steffen
- Department of Medicine II, Goethe University, University Hospital, Frankfurt, Germany
| | - Bernd Spriewald
- University Hospital Erlangen, Department of Internal Medicine 5, Hematology and Oncology, Erlangen, Germany
| | - Thomas Heinicke
- Department of Hematology and Oncology, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Nadja Jäkel
- Department of Hematology, Oncology, University Hospital, Halle/Saale, Germany
| | - Jörg Westermann
- Department of Hematology, Oncology and Tumor Immunology, CVK, Charité-Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kathrin Nachtkamp
- Department of Hematology, Oncology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Andreas Viardot
- Department of Hematology, Oncology, University Hospital Ulm, Ulm, Germany
| | - Max S Topp
- Department of Hematology, Oncology, University Hospital Würzburg, Würzburg, Germany
| | - Martin Neumann
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Claudia D Baldus
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nicola Gökbuget
- Department of Medicine II, Goethe University, University Hospital, Frankfurt, Germany
| | - Monika Brüggemann
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
6
|
Tang M, Antić Ž, Fardzadeh P, Pietzsch S, Schröder C, Eberhardt A, van Bömmel A, Escherich G, Hofmann W, Horstmann MA, Illig T, McCrary JM, Lentes J, Metzler M, Nejdl W, Schlegelberger B, Schrappe M, Zimmermann M, Miarka-Walczyk K, Pastorczak A, Cario G, Renard BY, Stanulla M, Bergmann AK. An artificial intelligence-assisted clinical framework to facilitate diagnostics and translational discovery in hematologic neoplasia. EBioMedicine 2024; 104:105171. [PMID: 38810562 PMCID: PMC11154115 DOI: 10.1016/j.ebiom.2024.105171] [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: 09/11/2023] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND The increasing volume and intricacy of sequencing data, along with other clinical and diagnostic data, like drug responses and measurable residual disease, creates challenges for efficient clinical comprehension and interpretation. Using paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) as a use case, we present an artificial intelligence (AI)-assisted clinical framework clinALL that integrates genomic and clinical data into a user-friendly interface to support routine diagnostics and reveal translational insights for hematologic neoplasia. METHODS We performed targeted RNA sequencing in 1365 cases with haematological neoplasms, primarily paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) from the AIEOP-BFM ALL study. We carried out fluorescence in situ hybridization (FISH), karyotyping and arrayCGH as part of the routine diagnostics. The analysis results of these assays as well as additional clinical information were integrated into an interactive web interface using Bokeh, where the main graph is based on Uniform Manifold Approximation and Projection (UMAP) analysis of the gene expression data. At the backend of the clinALL, we built both shallow machine learning models and a deep neural network using Scikit-learn and PyTorch respectively. FINDINGS By applying clinALL, 78% of undetermined patients under the current diagnostic protocol were stratified, and ambiguous cases were investigated. Translational insights were discovered, including IKZF1plus status dependent subpopulations of BCR::ABL1 positive patients, and a subpopulation within ETV6::RUNX1 positive patients that has a high relapse frequency. Our best machine learning models, LDA and PASNET-like neural network models, achieve F1 scores above 97% in predicting patients' subgroups. INTERPRETATION An AI-assisted clinical framework that integrates both genomic and clinical data can take full advantage of the available data, improve point-of-care decision-making and reveal clinically relevant insights promptly. Such a lightweight and easily transferable framework works for both whole transcriptome data as well as the cost-effective targeted RNA-seq, enabling efficient and equitable delivery of personalized medicine in small clinics in developing countries. FUNDING German Ministry of Education and Research (BMBF), German Research Foundation (DFG) and Foundation for Polish Science.
Collapse
Affiliation(s)
- Ming Tang
- Department of Human Genetics, Hannover Medical School, Hannover, Germany; L3S Research Centre, Leibniz University Hannover, Germany
| | - Željko Antić
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | | | - Stefan Pietzsch
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Charlotte Schröder
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | | | - Alena van Bömmel
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Gabriele Escherich
- Clinic of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Winfried Hofmann
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Martin A Horstmann
- Clinic of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany; Research Institute Children's Cancer Centre Hamburg, Hamburg, Germany
| | - Thomas Illig
- Hannover Unified Bio Bank, Hannover Medical School, Hannover, Germany
| | - J Matt McCrary
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Jana Lentes
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Markus Metzler
- Department of Paediatrics, University Hospital Erlangen, Erlangen, Germany
| | - Wolfgang Nejdl
- L3S Research Centre, Leibniz University Hannover, Germany
| | | | - Martin Schrappe
- Department of Paediatrics, University Medical Centre Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Martin Zimmermann
- Department of Paediatric Haematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Karolina Miarka-Walczyk
- Department of Paediatrics, Oncology and Haematology, Medical University of Lodz, Lodz, Poland
| | - Agata Pastorczak
- Department of Paediatrics, Oncology and Haematology, Medical University of Lodz, Lodz, Poland
| | - Gunnar Cario
- Department of Paediatrics, University Medical Centre Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Bernhard Y Renard
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
| | - Martin Stanulla
- Department of Paediatric Haematology and Oncology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
7
|
Maurer-Granofszky M, Kohrer S, Fischer S, Schumich A, Nebral K, Larghero P, Meyer C, Mecklenbrauker A, Muhlegger N, Marschalek R, Haas OA, Panzer-Grumayer R, Dworzak MN. Genomic breakpoint-specific monitoring of measurable residual disease in pediatric non-standard-risk acute myeloid leukemia. Haematologica 2024; 109:740-750. [PMID: 37345487 PMCID: PMC10910191 DOI: 10.3324/haematol.2022.282424] [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: 11/16/2022] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
Pediatric acute myeloid leukemia (AML) is a highly heterogeneous disease making standardized measurable residual disease (MRD) assessment challenging. Currently, patient-specific DNA-based assays are only rarely applied for MRD assessment in pediatric AML. We tested whether quantification of genomic breakpoint-specific sequences via quantitative polymerase chain reaction (gDNA-PCR) provides a reliable means of MRD quantification in children with non-standardrisk AML and compared its results to those obtained with state-of-the-art ten-color flow cytometry (FCM). Breakpointspecific gDNA-PCR assays were established according to Euro-MRD consortium guidelines. FCM-MRD assessment was performed according to the European Leukemia Network guidelines with adaptations for pediatric AML. Of 77 consecutively recruited non-standard-risk pediatric AML cases, 49 (64%) carried a chromosomal translocation potentially suitable for MRD quantification. Genomic breakpoint analysis returned a specific DNA sequence in 100% (41/41) of the cases submitted for investigation. MRD levels were evaluated using gDNA-PCR in 243 follow-up samples from 36 patients, achieving a quantitative range of at least 10-4 in 231/243 (95%) of samples. Comparing gDNA-PCR with FCM-MRD data for 183 bone marrow follow-up samples at various therapy timepoints showed a high concordance of 90.2%, considering a cut-off of ≥0.1%. Both methodologies outperformed morphological assessment. We conclude that MRD monitoring by gDNA-PCR is feasible in pediatric AML with traceable genetic rearrangements and correlates well with FCM-MRD in the currently applied clinically relevant range, while being more sensitive below that. The methodology should be evaluated in larger cohorts to pave the way for clinical application.
Collapse
Affiliation(s)
| | - Stefan Kohrer
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna
| | - Susanna Fischer
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna
| | - Angela Schumich
- St. Anna Children's Cancer Research Institute (CCRI), Vienna
| | - Karin Nebral
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna
| | - Patrizia Larghero
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt/Main
| | - Claus Meyer
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt/Main
| | - Astrid Mecklenbrauker
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna
| | - Nora Muhlegger
- St. Anna Children's Cancer Research Institute (CCRI), Vienna
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt/Main
| | - Oskar A Haas
- St. Anna Children's Cancer Research Institute (CCRI), Vienna
| | | | - Michael N Dworzak
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna, Austria; St. Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, Vienna.
| |
Collapse
|
8
|
He J, Munir F, Catueno S, Connors JS, Gibson A, Robusto L, McCall D, Nunez C, Roth M, Tewari P, Garces S, Cuglievan B, Garcia MB. Biological Markers of High-Risk Childhood Acute Lymphoblastic Leukemia. Cancers (Basel) 2024; 16:858. [PMID: 38473221 PMCID: PMC10930495 DOI: 10.3390/cancers16050858] [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: 01/21/2024] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024] Open
Abstract
Childhood acute lymphoblastic leukemia (ALL) has witnessed substantial improvements in prognosis; however, a subset of patients classified as high-risk continues to face higher rates of relapse and increased mortality. While the National Cancer Institute (NCI) criteria have traditionally guided risk stratification based on initial clinical information, recent advances highlight the pivotal role of biological markers in shaping the prognosis of childhood ALL. This review delves into the emerging understanding of high-risk childhood ALL, focusing on molecular, cytogenetic, and immunophenotypic markers. These markers not only contribute to unraveling the underlying mechanisms of the disease, but also shed light on specific clinical patterns that dictate prognosis. The paradigm shift in treatment strategies, exemplified by the success of tyrosine kinase inhibitors in Philadelphia chromosome-positive leukemia, underscores the importance of recognizing and targeting precise risk factors. Through a comprehensive exploration of high-risk childhood ALL characteristics, this review aims to enhance our comprehension of the disease, offering insights into its molecular landscape and clinical intricacies in the hope of contributing to future targeted and tailored therapies.
Collapse
Affiliation(s)
- Jiasen He
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Faryal Munir
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Samanta Catueno
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Jeremy S. Connors
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Amber Gibson
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Lindsay Robusto
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - David McCall
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Cesar Nunez
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Michael Roth
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Priti Tewari
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Sofia Garces
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| | - Miriam B. Garcia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA (D.M.)
| |
Collapse
|
9
|
Pieters R, Mullighan CG, Hunger SP. Advancing Diagnostics and Therapy to Reach Universal Cure in Childhood ALL. J Clin Oncol 2023; 41:5579-5591. [PMID: 37820294 PMCID: PMC10730082 DOI: 10.1200/jco.23.01286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/24/2023] [Accepted: 08/10/2023] [Indexed: 10/13/2023] Open
Abstract
Systemic combination chemotherapy and intrathecal chemotherapy markedly increased the survival rate of children with ALL. In the past two decades, the use of minimal (measurable) residual disease (MRD) measurements early in therapy improved risk group stratification with subsequent treatment intensifications for patients at high risk of relapse, and enabled a reduction of treatment for low-risk patients. The recent development of more sensitive MRD technologies may further affect risk stratification. Molecular genetic profiling has led to the discovery of many new subtypes and their driver genetic alterations. This increased our understanding of the biological basis of ALL, improved risk classification, and enabled implementation of precision medicine. In the past decade, immunotherapies, including bispecific antibodies, antibody-drug conjugates, and cellular therapies directed against surface proteins, led to more effective and less toxic therapies, replacing intensive chemotherapy courses and allogeneic stem-cell transplantation in patients with relapsed and refractory ALL, and are now being tested in newly diagnosed patients. It has taken 50-60 years to increase the cure rate in childhood ALL from 0% to 90% by stepwise improvements in chemotherapy. This review provides an overview of how the developments over the past 10-15 years mentioned above have significantly changed the diagnostic and treatment approach in ALL, and discusses how the integrated use of molecular and immunotherapeutic insights will very likely direct efforts to cure those children with ALL who are not cured today, and improve the quality of life for survivors who should have decades of life ahead. Future efforts must focus on making effective, yet very expensive, new technologies and therapies available to children with ALL worldwide.
Collapse
Affiliation(s)
- Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Charles G. Mullighan
- Department of Pathology and Hematological Malignancies Program, Comprehensive Cancer Center, St Jude Children's Research Hospital, Memphis, TN
| | - Stephen P. Hunger
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
10
|
Kim R, Bergugnat H, Pastoret C, Pasquier F, Raffoux E, Larcher L, Passet M, Grardel N, Delabesse E, Kubetzko S, Caye-Eude A, Meyer C, Marschalek R, Lafage-Pochitaloff M, Thiebaut-Bertrand A, Balsat M, Escoffre-Barbe M, Blum S, Baumann M, Banos A, Straetmans N, Gallego-Hernanz MP, Chalandon Y, Graux C, Soulier J, Leguay T, Hunault M, Huguet F, Lhéritier V, Dombret H, Boissel N, Clappier E. Genetic alterations and MRD refine risk assessment for KMT2A-rearranged B-cell precursor ALL in adults: a GRAALL study. Blood 2023; 142:1806-1817. [PMID: 37595275 DOI: 10.1182/blood.2023021501] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023] Open
Abstract
KMT2A-rearranged (KMT2A-r) B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is widely recognized as a high-risk leukemia in both children and adults. However, there is a paucity of data on adults treated in recent protocols, and the optimal treatment strategy for these patients is still a matter of debate. In this study, we set out to refine the prognosis of adult KMT2A-r BCP-ALL treated with modern chemotherapy regimen and investigate the prognostic impact of comutations and minimal residual disease (MRD). Of 1091 adult patients with Philadelphia-negative BCP-ALL enrolled in 3 consecutive trials from the Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL), 141 (12.9%) had KMT2A-r, with 5-year cumulative incidence of relapse (CIR) and overall survival (OS) rates of 40.7% and 53.3%, respectively. Molecular profiling highlighted a low mutational burden in this subtype, reminiscent of infant BCP-ALL. However, the presence of TP53 and/or IKZF1 alterations defined a subset of patients with significantly poorer CIR (69.3% vs 36.2%; P = .001) and OS (28.1% vs 60.7%; P = .006) rates. Next, we analyzed the prognostic implication of MRD measured after induction and first consolidation, using both immunoglobulin (IG) or T-cell receptor (TR) gene rearrangements and KMT2A genomic fusion as markers. In approximately one-third of patients, IG/TR rearrangements were absent or displayed clonal evolution during the disease course, compromising MRD monitoring. In contrast, KMT2A-based MRD was highly reliable and strongly associated with outcome, with early good responders having an excellent outcome (3-year CIR, 7.1%; OS, 92.9%). Altogether, our study reveals striking heterogeneity in outcomes within adults with KMT2A-r BCP-ALL and provides new biomarkers to guide risk-based therapeutic stratification.
Collapse
Affiliation(s)
- Rathana Kim
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Hugo Bergugnat
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Cédric Pastoret
- Hematology Laboratory, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Florence Pasquier
- Department of Hematology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Emmanuel Raffoux
- Hematology Department, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Lise Larcher
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Marie Passet
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nathalie Grardel
- Hematology Laboratory, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Eric Delabesse
- Hematology Laboratory, Institut Universitaire de Cancer Toulouse-Oncopole, INSERM 1037, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Susanne Kubetzko
- Department of Hematology, University Hospital of Zürich, Zürich, Switzerland
| | - Aurélie Caye-Eude
- Genetics Department, Molecular Genetics Unit, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Claus Meyer
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia, Goethe University, Frankfurt/Main, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia, Goethe University, Frankfurt/Main, Germany
| | - Marine Lafage-Pochitaloff
- Laboratoire de Cytogénétique Hématologique, Hôpital Timone Enfant, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille Université, Marseille, France
| | | | - Marie Balsat
- Department of Hematology, Hôpital Lyon Sud, Pierre Benite, France
| | | | - Sabine Blum
- Department of Hematology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Michael Baumann
- Klinik für Medizinische Onkologie und Hämatologie, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Anne Banos
- Department of Hematology, Centre Hospitalier de la Côte Basque, Bayonne, France
| | - Nicole Straetmans
- Department of Hematology, University Hospital Saint-Luc, Brussels, Belgium
| | | | - Yves Chalandon
- Division of Hematology, Department of Oncology, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland, for the Swiss Group for Clinical Cancer Research
- Swiss Group for Clinical Cancer Research
| | - Carlos Graux
- Department of Hematology, Université Catholique de Louvain, Centre Hospitalier Universitaire UCLouvain Namur-Godinne, Yvoir, Belgium
| | - Jean Soulier
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Thibaut Leguay
- Department of Hematology, Centre Hospitalier Universitaire de Bordeaux, Hôpital du Haut-Levêque, Pessac, France
| | - Mathilde Hunault
- Département des Maladies du Sang, Centre Hospitalier Universitaire Angers, INSERM, CNRS, CRCI2NA, Fédération Hospitalo-Universitaire Grand Ouest Against Leukemia, Université d'Angers, Université de Nantes, Angers, France
| | - Françoise Huguet
- Department of Hematology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire de Cancer Toulouse-Oncopole, Toulouse, France
| | - Véronique Lhéritier
- Coordination du Groupe Group for Research on Adult Acute Lymphoblastic Leukemia, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre Benite, France
| | - Hervé Dombret
- Hematology Department, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Nicolas Boissel
- Hematology Department, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Emmanuelle Clappier
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| |
Collapse
|
11
|
Sperlazza J, Galeotti J, Hucks G, Alexander TB. Multiple lineage switches in KMT2A rearranged infant leukemia, responsive to combination therapy with CPX-351 and inotuzumab. Pediatr Blood Cancer 2023; 70:e30645. [PMID: 37638847 DOI: 10.1002/pbc.30645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/29/2023]
Affiliation(s)
- Justin Sperlazza
- Division of Pediatric Hematology-Oncology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Jonathan Galeotti
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - George Hucks
- Division of Pediatric Hematology-Oncology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Thomas B Alexander
- Division of Pediatric Hematology-Oncology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
12
|
Schneider P, Crump NT, Arentsen-Peters STCJM, Smith AL, Hagelaar R, Adriaanse FRS, Bos RS, de Jong A, Nierkens S, Koopmans B, Milne TA, Pieters R, Stam RW. Modelling acquired resistance to DOT1L inhibition exhibits the adaptive potential of KMT2A-rearranged acute lymphoblastic leukemia. Exp Hematol Oncol 2023; 12:81. [PMID: 37740239 PMCID: PMC10517487 DOI: 10.1186/s40164-023-00445-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/11/2023] [Indexed: 09/24/2023] Open
Abstract
In KMT2A-rearranged acute lymphoblastic leukemia (ALL), an aggressive malignancy, oncogenic KMT2A-fusion proteins inappropriately recruit DOT1L to promote leukemogenesis, highlighting DOT1L as an attractive therapeutic target. Unfortunately, treatment with the first-in-class DOT1L inhibitor pinometostat eventually leads to non-responsiveness. To understand this we established acquired pinometostat resistance in pediatric KMT2A::AFF1+ B-ALL cells. Interestingly, these cells became mostly independent of DOT1L-mediated H3K79 methylation, but still relied on the physical presence of DOT1L, HOXA9 and the KMT2A::AFF1 fusion. Moreover, these cells selectively lost the epigenetic regulation and expression of various KMT2A-fusion target genes such as PROM1/CD133, while other KMT2A::AFF1 target genes, including HOXA9 and CDK6 remained unaffected. Concomitantly, these pinometostat-resistant cells showed upregulation of several myeloid-associated genes, including CD33 and LILRB4/CD85k. Taken together, this model comprehensively shows the adaptive potential of KMT2A-rearranged ALL cells upon losing dependency on one of its main oncogenic properties.
Collapse
Affiliation(s)
- Pauline Schneider
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Nicholas T Crump
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | | | - Alastair L Smith
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Rico Hagelaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | | | - Romy S Bos
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Anja de Jong
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Bianca Koopmans
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Thomas A Milne
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Ronald W Stam
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
13
|
Faulk KE, Kairalla JA, Dreyer ZE, Carroll AJ, Heerema NA, Devidas M, Carroll WL, Raetz EA, Loh ML, Hunger SP, Borowitz M, Wang C, Guest E, Brown PA. Minimal residual disease predicts outcomes in KMT2A-rearranged but not KMT2A-germline infant acute lymphoblastic leukemia: Report from Children's Oncology Group study AALL0631. Pediatr Blood Cancer 2023; 70:e30467. [PMID: 37259259 PMCID: PMC10687300 DOI: 10.1002/pbc.30467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/02/2023] [Accepted: 05/17/2023] [Indexed: 06/02/2023]
Abstract
We measured minimal residual disease (MRD) by multiparameter flow cytometry at three time points (TP) in 117 infants with KMT2A (lysine [K]-specific methyltransferase 2A)-rearranged and 58 with KMT2A-germline acute lymphoblastic leukemia (ALL) on Children's Oncology Group AALL0631 study. For KMT2A-rearranged patients, 3-year event-free survival (EFS) by MRD-positive (≥0.01%) versus MRD-negative (<0.01%) was: TP1: 25% (±6%) versus 49% (±7%; p = .0009); TP2: 21% (±8%) versus 47% (±7%; p < .0001); and TP3: 22% (±14%) versus 51% (±6%; p = .0178). For KMT2A-germline patients, 3-year EFS was: TP1: 88% (±12%) versus 87% (±5%; p = .73); TP2: 100% versus 88% (±5%; p = .24); and TP3: 100% versus 87% (±5%; p = .53). MRD was a strong independent outcome predictor in KMT2A-rearranged, but not KMT2A-germline infant ALL.
Collapse
Affiliation(s)
- Kelly E. Faulk
- University of Colorado Anschutz Medical Campus, Pediatric Oncology, Denver, CO, USA
| | | | - ZoAnn E. Dreyer
- Texas Children’s Hospital, Pediatric Oncology, Houston, TX, USA
| | | | | | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - William L. Carroll
- Perlmutter Cancer Center, and the New York University Grossman School of Medicine, Division of Pediatric Hematology/Oncology, New York, NY, USA
| | - Elizabeth A. Raetz
- Perlmutter Cancer Center, and the New York University Grossman School of Medicine, Division of Pediatric Hematology/Oncology, New York, NY, USA
| | - Mignon L. Loh
- University of Washington, Pediatric Oncology, Seattle, WA, USA
| | - Stephen P. Hunger
- Division of Oncology and the Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Cindy Wang
- University of Florida, Biostatistics, Gainesville, FL, USA
| | - Erin Guest
- Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Mercy Kansas City, Kansas City, MO, USA
| | | |
Collapse
|
14
|
Chen JZ, Wang LN, Luo XQ, Tang YL. The genomic landscape of sensitivity to arsenic trioxide uncovered by genome-wide CRISPR-Cas9 screening. Front Oncol 2023; 13:1178686. [PMID: 37251921 PMCID: PMC10214836 DOI: 10.3389/fonc.2023.1178686] [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: 03/03/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Arsenic trioxide (ATO) is a promising anticancer drug for hematological malignancy. Given the dramatic efficacy of acute promyelocytic leukemia (APL), ATO has been utilized in other types of cancers, including solid tumors. Unfortunately, the results were not comparable with the effects on APL, and the resistance mechanism has not been clarified yet. This study intends to identify relevant genes and pathways affecting ATO drug sensitivity through genome-wide CRISPR-Cas9 knockdown screening to provide a panoramic view for further study of ATO targets and improved clinical outcomes. Methods A genome-wide CRISPR-Cas9 knockdown screening system was constructed for ATO screening. The screening results were processed with MAGeCK, and the results were subjected to pathway enrichment analysis using WebGestalt and KOBAS. We also performed protein-protein interaction (PPI) network analysis using String and Cytoscape, followed by expression profiling and survival curve analysis of critical genes. Virtual screening was used to recognize drugs that may interact with the hub gene. Results We applied enrichment analysis and identified vital ATO-related pathways such as metabolism, chemokines and cytokines production and signaling, and immune system responses. In addition, we identified KEAP1 as the top gene relating to ATO resistance. We found that KEAP1 expression was higher in the pan-cancer, including ALL, than in normal tissue. Patients with acute myeloid leukemia (AML) with higher KEAP1 expression had worse overall survival (OS). A virtual screen showed that etoposide and eltrombopag could bind to KEAP1 and potentially interact with ATO. Discussion ATO is a multi-target anticancer drug, and the key pathways regulating its sensitivity include oxidative stress, metabolism, chemokines and cytokines, and the immune system. KEAP1 is the most critical gene regulating ATO drug sensitivity, which is related to AML prognosis and may bind to some clinical drugs leading to an interaction with ATO. These integrated results provided new insights into the pharmacological mechanism of ATO and potentiate for further applications in cancer treatments.
Collapse
Affiliation(s)
- Jun-Zhu Chen
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li-Na Wang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xue-Qun Luo
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan-Lai Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Mizuki K, Honda Y, Asai H, Higuchi N, Morita H, Yabe H, Kusuhara K. Successful Retransplantation With Killer Cell Immunoglobulin-like Receptor Ligand-mismatched Cord Blood in Infant Acute Lymphoblastic Leukemia That Relapsed After Transplantation. J Pediatr Hematol Oncol 2023; 45:e547-e550. [PMID: 36706271 DOI: 10.1097/mph.0000000000002614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 12/05/2022] [Indexed: 01/29/2023]
Abstract
The prognosis of children with KMT2A -rearranged ( KMT2A -r) acute lymphoblastic leukemia (ALL) remains dismal. This report describes the successful retransplantation of a patient with infant ALL who relapsed both bone marrow and central nervous system. The patient received HLA-matched cord blood transplantation (CBT) and relapsed 18 months later. After achieving the second remission, the patient received a killer cell immunoglobulin-like receptor ligand-mismatched CBT with a reduced-intensity conditioning regimen and has been in remission for 52 months. Thus, killer cell immunoglobulin-like receptor ligand-mismatched CBT with reduced-intensity conditioning might be a treatment option for patients with KMT2A- r ALL who relapsed after transplantation, even with extramedullary relapse.
Collapse
Affiliation(s)
- Kazuyoshi Mizuki
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu
| | - Yuko Honda
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu
| | - Hiroshi Asai
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu
| | - Naoko Higuchi
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu
| | - Hiromi Morita
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu
| | - Hiromasa Yabe
- Department of Pediatrics, Tokai University School of Medicine, Isehara, Japan
| | - Koichi Kusuhara
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu
| |
Collapse
|
16
|
van der Sluis IM, de Lorenzo P, Kotecha RS, Attarbaschi A, Escherich G, Nysom K, Stary J, Ferster A, Brethon B, Locatelli F, Schrappe M, Scholte-van Houtem PE, Valsecchi MG, Pieters R. Blinatumomab Added to Chemotherapy in Infant Lymphoblastic Leukemia. N Engl J Med 2023; 388:1572-1581. [PMID: 37099340 DOI: 10.1056/nejmoa2214171] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
BACKGROUND KMT2A-rearranged acute lymphoblastic leukemia (ALL) in infants is an aggressive disease with 3-year event-free survival below 40%. Most relapses occur during treatment, with two thirds occurring within 1 year and 90% within 2 years after diagnosis. Outcomes have not improved in recent decades despite intensification of chemotherapy. METHODS We studied the safety and efficacy of blinatumomab, a bispecific T-cell engager molecule targeting CD19, in infants with KMT2A-rearranged ALL. Thirty patients younger than 1 year of age with newly diagnosed KMT2A-rearranged ALL were given the chemotherapy used in the Interfant-06 trial with the addition of one postinduction course of blinatumomab (15 μg per square meter of body-surface area per day; 28-day continuous infusion). The primary end point was clinically relevant toxic effects, defined as any toxic effect that was possibly or definitely attributable to blinatumomab and resulted in permanent discontinuation of blinatumomab or death. Minimal residual disease (MRD) was measured by polymerase chain reaction. Data on adverse events were collected. Outcome data were compared with historical control data from the Interfant-06 trial. RESULTS The median follow-up was 26.3 months (range, 3.9 to 48.2). All 30 patients received the full course of blinatumomab. No toxic effects meeting the definition of the primary end point occurred. Ten serious adverse events were reported (fever [4 events], infection [4], hypertension [1], and vomiting [1]). The toxic-effects profile was consistent with that reported in older patients. A total of 28 patients (93%) either were MRD-negative (16 patients) or had low levels of MRD (<5×10-4 [i.e., <5 leukemic cells per 10,000 normal cells], 12 patients) after the blinatumomab infusion. All the patients who continued chemotherapy became MRD-negative during further treatment. Two-year disease-free survival was 81.6% in our study (95% confidence interval [CI], 60.8 to 92.0), as compared with 49.4% (95% CI, 42.5 to 56.0) in the Interfant-06 trial; the corresponding values for overall survival were 93.3% (95% CI, 75.9 to 98.3) and 65.8% (95% CI, 58.9 to 71.8). CONCLUSIONS Blinatumomab added to Interfant-06 chemotherapy appeared to be safe and had a high level of efficacy in infants with newly diagnosed KMT2A-rearranged ALL as compared with historical controls from the Interfant-06 trial. (Funded by the Princess Máxima Center Foundation and others; EudraCT number, 2016-004674-17.).
Collapse
Affiliation(s)
- Inge M van der Sluis
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Paola de Lorenzo
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Rishi S Kotecha
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Andishe Attarbaschi
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Gabriele Escherich
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Karsten Nysom
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Jan Stary
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Alina Ferster
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Benoit Brethon
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Franco Locatelli
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Martin Schrappe
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Peggy E Scholte-van Houtem
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Maria G Valsecchi
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| | - Rob Pieters
- From the Princess Máxima Center for Pediatric Oncology (I.M.S., P.E.S.-H., R.P.), and the Dutch Childhood Oncology Group (I.M.S., R.P.) - both in Utrecht, the Netherlands; Tettamanti Center (P.L.) and Biostatistics and Clinical Epidemiology (M.G.V.), Fondazione IRCCS San Gerardo dei Tintori, Monza, the School of Medicine and Surgery, University of Milano-Bicocca, Milan (M.G.V.), and the Department of Pediatric Hematology-Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome (F.L.) - all in Italy; Australian and New Zealand Children's Hematology and Oncology Group, Perth Children's Hospital (R.S.K.), Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia (R.S.K.), and Curtin Medical School, Curtin University (R.S.K.) - all in Perth, WA, Australia; St. Anna Children's Hospital, Department of Pediatric Hematology and Oncology, Medical University of Vienna, and St. Anna Children's Cancer Research Institute - both in Vienna (A.A.); the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg (G.E.) the Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg (G.E.), and the ALL-Berlin-Frankfurt-Münster (BFM) Group, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel (M.S.) - all in Germany; the Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University Hospital, Copenhagen (K.N.); Czech Working Group for Pediatric Hematology (J.S.) and CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol (J.S.) - all in Prague, Czech Republic; Hôpital Universitaire des Enfants Reine Fabiola, Brussels (A.F.); and the Department of Pediatric Hematology, University Robert Debre Hospital, Assistance Publique-Hôpitaux de Paris, Paris (B.B.)
| |
Collapse
|
17
|
Popov A, Tsaur G, Permikin Z, Henze G, Verzhbitskaya T, Plekhanova O, Nokhrina E, Valochnik A, Sibiryakov P, Zerkalenkova E, Olshanskaya Y, Gindina T, Movchan L, Shorikov E, Streneva O, Khlebnikova O, Makarova O, Arakaev O, Boichenko E, Kondratchik K, Ponomareva N, Lapotentova E, Aleinikova O, Miakova N, Novichkova G, Karachunskiy A, Fechina L. Genetic characteristics and treatment outcome in infants with KMT2A germline B-cell precursor acute lymphoblastic leukemia: Results of MLL-Baby protocol. Pediatr Blood Cancer 2023; 70:e30204. [PMID: 36715125 DOI: 10.1002/pbc.30204] [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: 08/26/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 01/31/2023]
Abstract
The aim of this study was to present the diagnostic and outcome characteristics of infants with germline status of KMT2A gene (KMT2A-g) B-cell precursor acute lymphoblastic leukemia (BCP-ALL) treated consistently according to the MLL-Baby protocol, a moderate-intensity protocol. Of the 139 patients enrolled in the MLL-Baby study, 100 (71.9%) carried different types of rearranged KMT2A (KMT2A-r), while the remaining 39 infants (28.1%) had KMT2A-g. KMT2A-g patients were generally older (77% older than 6 months), less likely to have a very high white blood cell count (greater than 100 × 109 /L), less likely to be central nervous system (CNS)-positive, and more likely to be CD10-positive. The 6-year event-free survival and overall survival rates for all 39 patients were 0.74 (standard error [SE] 0.07) and 0.80 (SE 0.07), respectively. Relapse was the most common adverse event (n = 5), with a cumulative incidence of relapse (CIR) of 0.13 (SE 0.06), while the incidence of a second malignancy (n = 1) and death in remission (n = 3) was 0.03 (SE 0.04) and 0.08 (SE 0.04), respectively. None of the initial parameters, including genetics and the presence of recently described fusions of NUTM1 and PAX5 genes, was able to distinguish patients with different outcomes. Only rapidity of response, measured as minimal residual disease (MRD) by flow cytometry, showed a statistically significant impact. Moderate-intensity therapy, as used in the MLL-Baby protocol in infants with KMT2A-g BCP-ALL, yields results comparable to other infant studies. Patients with a slow multicolor flow cytometry (MFC)-MRD response should be subjected to advanced therapies, such as targeted or immunotherapies.
Collapse
Affiliation(s)
- Alexander Popov
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Grigory Tsaur
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Ural State Medical University, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Zhan Permikin
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Ural State Medical University, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Guenter Henze
- Department of Pediatric Oncology Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tatiana Verzhbitskaya
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Olga Plekhanova
- Regional Children's Hospital, Ekaterinburg, Russian Federation
| | | | - Alena Valochnik
- Belarussian Research Centre for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Petr Sibiryakov
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Elena Zerkalenkova
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Yulia Olshanskaya
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Tatiana Gindina
- R.M. Gorbacheva Research Institute of Pediatric Oncology, Hematology and Transplantation, Pavlov University of Saint Petersburg, Saint Petersburg, Russian Federation
| | - Liudmila Movchan
- Belarussian Research Centre for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Egor Shorikov
- PET-Technology Centre of Nuclear Medicine, Ekaterinburg, Russian Federation
| | - Olga Streneva
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | | | - Olga Makarova
- Regional Children's Hospital, Ekaterinburg, Russian Federation
| | - Oleg Arakaev
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Elmira Boichenko
- City Children's Hospital No. 1, Saint Petersburg, Russian Federation
| | | | | | - Elena Lapotentova
- Belarussian Research Centre for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Olga Aleinikova
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation.,Belarussian Research Centre for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Natalia Miakova
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Galina Novichkova
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexander Karachunskiy
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Larisa Fechina
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| |
Collapse
|
18
|
Górecki M, Kozioł I, Kopystecka A, Budzyńska J, Zawitkowska J, Lejman M. Updates in KMT2A Gene Rearrangement in Pediatric Acute Lymphoblastic Leukemia. Biomedicines 2023; 11:biomedicines11030821. [PMID: 36979800 PMCID: PMC10045821 DOI: 10.3390/biomedicines11030821] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/10/2023] Open
Abstract
The KMT2A (formerly MLL) encodes the histone lysine-specific N-methyltransferase 2A and is mapped on chromosome 11q23. KMT2A is a frequent target for recurrent translocations in acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), or mixed lineage (biphenotypic) leukemia (MLL). Over 90 KMT2A fusion partners have been identified until now, including the most recurring ones—AFF1, MLLT1, and MLLT3—which encode proteins regulating epigenetic mechanisms. The presence of distinct KMT2A rearrangements is an independent dismal prognostic factor, while very few KMT2A rearrangements display either a good or intermediate outcome. KMT2A-rearranged (KMT2A-r) ALL affects more than 70% of new ALL diagnoses in infants (<1 year of age), 5–6% of pediatric cases, and 15% of adult cases. KMT2A-rearranged (KMT2A-r) ALL is characterized by hyperleukocytosis, a relatively high incidence of central nervous system (CNS) involvement, an aggressive course with early relapse, and early relapses resulting in poor prognosis. The exact pathways of fusions and the effects on the final phenotypic activity of the disease are still subjects of much research. Future trials could consider the inclusion of targeted immunotherapeutic agents and prioritize the identification of prognostic factors, allowing for the less intensive treatment of some infants with KMT2A ALL. The aim of this review is to summarize our knowledge and present current insight into the mechanisms of KMT2A-r ALL, portray their characteristics, discuss the clinical outcome along with risk stratification, and present novel therapeutic strategies.
Collapse
Affiliation(s)
- Mateusz Górecki
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Ilona Kozioł
- Student Scientific Society of the Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Agnieszka Kopystecka
- Student Scientific Society of the Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Julia Budzyńska
- Student Scientific Society of the Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Joanna Zawitkowska
- Department of Paediatric Haematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| |
Collapse
|
19
|
Niswander LM, Graff ZT, Chien CD, Chukinas JA, Meadows CA, Leach LC, Loftus JP, Kohler ME, Tasian SK, Fry TJ. Potent preclinical activity of FLT3-directed chimeric antigen receptor T-cell immunotherapy against FLT3- mutant acute myeloid leukemia and KMT2A-rearranged acute lymphoblastic leukemia. Haematologica 2023; 108:457-471. [PMID: 35950535 PMCID: PMC9890025 DOI: 10.3324/haematol.2022.281456] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/03/2022] [Indexed: 02/03/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell immunotherapies targeting CD19 or CD22 induce remissions in the majority of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (ALL), although relapse due to target antigen loss or downregulation has emerged as a major clinical dilemma. Accordingly, great interest exists in developing CAR T cells directed against alternative leukemia cell surface antigens that may help to overcome immunotherapeutic resistance. The fms-like tyrosine kinase 3 receptor (FLT3) is constitutively activated via FLT3 mutation in acute myeloid leukemia (AML) or wild-type FLT3 overexpression in KMT2A (lysine-specific methyltransferase 2A)-rearranged ALL, which are associated with poor clinical outcomes in children and adults. We developed monovalent FLT3-targeted CAR T cells (FLT3CART) and bispecific CD19xFLT3CART and assessed their anti-leukemia activity in preclinical models of FLT3-mutant AML and KMT2A-rearranged infant ALL. We report robust in vitro FLT3CART-induced cytokine production and cytotoxicity against AML and ALL cell lines with minimal cross-reactivity against normal hematopoietic and non-hematopoietic tissues. We also observed potent in vivo inhibition of leukemia proliferation in xenograft models of both FLT3-mutant AML and KMT2A-rearranged ALL, including a post-tisagenlecleucel ALL-to-AML lineage switch patient-derived xenograft model pairing. We further demonstrate significant in vitro and in vivo activity of bispecific CD19xFLT3CART against KMT2Arearranged ALL and posit that this additional approach might also diminish potential antigen escape in these high-risk leukemias. Our preclinical data credential FLT3CART as a highly effective immunotherapeutic strategy for both FLT3- mutant AML and KMT2A-rearranged ALL which is poised for further investigation and clinical translation.
Collapse
Affiliation(s)
- Lisa M Niswander
- Children's Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research; Philadelphia PA
| | - Zachary T Graff
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA; Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO
| | - Christopher D Chien
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, MD
| | - John A Chukinas
- Children's Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research; Philadelphia PA
| | - Christina A Meadows
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO
| | - Lillie C Leach
- Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO
| | - Joseph P Loftus
- Children's Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research; Philadelphia, PA
| | - M Eric Kohler
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA; Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO
| | - Sarah K Tasian
- Children's Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research; Philadelphia PA, USA; University of Pennsylvania Perelman School of Medicine and Abramson Cancer Center; Philadelphia PA.
| | - Terry J Fry
- Center for Cancer and Blood Disorders, Children's Hospital Colorado; Aurora, CO, USA; Department of Pediatrics, University of Colorado Anschutz Medical Campus; Aurora, CO.
| |
Collapse
|
20
|
Tomizawa D. Evolution and optimization of therapies for acute lymphoblastic leukemia in infants. Int J Hematol 2023; 117:162-172. [PMID: 36441356 DOI: 10.1007/s12185-022-03502-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
Acute lymphoblastic leukemia (ALL) in infants accounts for less than 5% of pediatric ALL and is biologically and clinically unique. Approximately 70% to 80% of cases present as an aggressive leukemia with KMT2A gene rearrangement (KMT2A-r), which is one of the most difficult-to-cure forms of pediatric leukemia. Owing to continuing global efforts through multicenter clinical trials since the mid-1990s, a standard of care for infant KMT2A-r ALL, including minimal residual disease-based risk stratifications, "hybrid chemotherapy" incorporating myeloid leukemia-like drugs (e.g., cytarabine) into the ALL chemotherapy backbone, and selective use of allogeneic hematopoietic stem cell transplantation, has now been established. However, there are still many concerns regarding treatment of infants with KMT2A-r ALL, including insufficient efficacy of the current standard therapies, limited pharmacokinetic/pharmacodynamic data on drugs in infants, and management of both acute and late toxicities. Refinements in risk stratification based on leukemia biology, as well as the introduction of emerging novel immunotherapies and molecular-targeted drugs to contemporary therapy, through international collaboration would provide key solutions for further improvement in outcomes.
Collapse
Affiliation(s)
- Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan.
| |
Collapse
|
21
|
Cheung LC, Aya-Bonilla C, Cruickshank MN, Chiu SK, Kuek V, Anderson D, Chua GA, Singh S, Oommen J, Ferrari E, Hughes AM, Ford J, Kunold E, Hesselman MC, Post F, Faulk KE, Breese EH, Guest EM, Brown PA, Loh ML, Lock RB, Kees UR, Jafari R, Malinge S, Kotecha RS. Preclinical efficacy of azacitidine and venetoclax for infant KMT2A-rearranged acute lymphoblastic leukemia reveals a new therapeutic strategy. Leukemia 2023; 37:61-71. [PMID: 36380143 PMCID: PMC9883157 DOI: 10.1038/s41375-022-01746-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/20/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022]
Abstract
Infants with KMT2A-rearranged B-cell acute lymphoblastic leukemia (ALL) have a dismal prognosis. Survival outcomes have remained static in recent decades despite treatment intensification and novel therapies are urgently required. KMT2A-rearranged infant ALL cells are characterized by an abundance of promoter hypermethylation and exhibit high BCL-2 expression, highlighting potential for therapeutic targeting. Here, we show that hypomethylating agents exhibit in vitro additivity when combined with most conventional chemotherapeutic agents. However, in a subset of samples an antagonistic effect was seen between several agents. This was most evident when hypomethylating agents were combined with methotrexate, with upregulation of ATP-binding cassette transporters identified as a potential mechanism. Single agent treatment with azacitidine and decitabine significantly prolonged in vivo survival in KMT2A-rearranged infant ALL xenografts. Treatment of KMT2A-rearranged infant ALL cell lines with azacitidine and decitabine led to differential genome-wide DNA methylation, changes in gene expression and thermal proteome profiling revealed the target protein-binding landscape of these agents. The selective BCL-2 inhibitor, venetoclax, exhibited in vitro additivity in combination with hypomethylating or conventional chemotherapeutic agents. The addition of venetoclax to azacitidine resulted in a significant in vivo survival advantage indicating the therapeutic potential of this combination to improve outcome for infants with KMT2A-rearranged ALL.
Collapse
Affiliation(s)
- Laurence C Cheung
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Carlos Aya-Bonilla
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
- The University of Western Australia, Perth, WA, Australia
| | | | - Sung K Chiu
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Vincent Kuek
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
- The University of Western Australia, Perth, WA, Australia
| | - Denise Anderson
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Grace-Alyssa Chua
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Sajla Singh
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Joyce Oommen
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Emanuela Ferrari
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Anastasia M Hughes
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Jette Ford
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Elena Kunold
- Department of Oncology-Pathology, Clinical Proteomics Mass Spectrometry, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Maria C Hesselman
- Department of Oncology-Pathology, Clinical Proteomics Mass Spectrometry, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Frederik Post
- Department of Oncology-Pathology, Clinical Proteomics Mass Spectrometry, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Kelly E Faulk
- University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO, USA
| | - Erin H Breese
- Cancer and Blood Diseases Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Erin M Guest
- Division of Hematology, Oncology, Blood and Marrow Transplantation, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Patrick A Brown
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, John Hopkins University, Baltimore, MD, USA
| | - Mignon L Loh
- Division of Pediatric Hematology, Oncology, Bone Marrow Transplant and Cellular Therapy, Seattle Children's Hospital, Seattle, WA, USA
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre/School of Women's and Children's Health/UNSW Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | - Ursula R Kees
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
- The University of Western Australia, Perth, WA, Australia
| | - Rozbeh Jafari
- Department of Oncology-Pathology, Clinical Proteomics Mass Spectrometry, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Sébastien Malinge
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
- The University of Western Australia, Perth, WA, Australia
| | - Rishi S Kotecha
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia.
- Curtin Medical School, Curtin University, Perth, WA, Australia.
- The University of Western Australia, Perth, WA, Australia.
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, WA, Australia.
| |
Collapse
|
22
|
Darzentas F, Szczepanowski M, Kotrová M, Hartmann A, Beder T, Gökbuget N, Schwartz S, Bastian L, Baldus CD, Pál K, Darzentas N, Brüggemann M. Insights into IGH clonal evolution in BCP-ALL: frequency, mechanisms, associations, and diagnostic implications. Front Immunol 2023; 14:1125017. [PMID: 37143651 PMCID: PMC10151743 DOI: 10.3389/fimmu.2023.1125017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction The malignant transformation leading to a maturation arrest in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) occurs early in B-cell development, in a pro-B or pre-B cell, when somatic recombination of variable (V), diversity (D), and joining (J) segment immunoglobulin (IG) genes and the B-cell rescue mechanism of VH replacement might be ongoing or fully active, driving clonal evolution. In this study of newly diagnosed BCP-ALL, we sought to understand the mechanistic details of oligoclonal composition of the leukemia at diagnosis, clonal evolution during follow-up, and clonal distribution in different hematopoietic compartments. Methods Utilizing high-throughput sequencing assays and bespoke bioinformatics we identified BCP-ALL-derived clonally-related IGH sequences by their shared 'DNJ-stem'. Results We introduce the concept of 'marker DNJ-stem' to cover the entirety of, even lowly abundant, clonally-related family members. In a cohort of 280 adult patients with BCP-ALL, IGH clonal evolution at diagnosis was identified in one-third of patients. The phenomenon was linked to contemporaneous recombinant and editing activity driven by aberrant ongoing DH/VH-DJH recombination and VH replacement, and we share insights and examples for both. Furthermore, in a subset of 167 patients with molecular subtype allocation, high prevalence and high degree of clonal evolution driven by ongoing DH/VH-DJH recombination were associated with the presence of KMT2A gene rearrangements, while VH replacements occurred more frequently in Ph-like and DUX4 BCP-ALL. Analysis of 46 matched diagnostic bone marrow and peripheral blood samples showed a comparable clonal and clonotypic distribution in both hematopoietic compartments, but the clonotypic composition markedly changed in longitudinal follow-up analysis in select cases. Thus, finally, we present cases where the specific dynamics of clonal evolution have implications for both the initial marker identification and the MRD monitoring in follow-up samples. Discussion Consequently, we suggest to follow the marker DNJ-stem (capturing all family members) rather than specific clonotypes as the MRD target, as well as to follow both VDJH and DJH family members since their respective kinetics are not always parallel. Our study further highlights the intricacy, importance, and present and future challenges of IGH clonal evolution in BCP-ALL.
Collapse
Affiliation(s)
- Franziska Darzentas
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Monika Szczepanowski
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Michaela Kotrová
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Alina Hartmann
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Clinical Research Unit “CATCH-ALL” (KFO 5010/1), funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation), Bonn, Germany
| | - Thomas Beder
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nicola Gökbuget
- Department of Medicine II, Hematology/Oncology, Goethe University Hospital, Frankfurt/M, Germany
| | - Stefan Schwartz
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lorenz Bastian
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Clinical Research Unit “CATCH-ALL” (KFO 5010/1), funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation), Bonn, Germany
| | - Claudia Dorothea Baldus
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Clinical Research Unit “CATCH-ALL” (KFO 5010/1), funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation), Bonn, Germany
| | - Karol Pál
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Nikos Darzentas
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
- *Correspondence: Nikos Darzentas,
| | - Monika Brüggemann
- Medical Department II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Clinical Research Unit “CATCH-ALL” (KFO 5010/1), funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation), Bonn, Germany
| |
Collapse
|
23
|
Updates in infant acute lymphoblastic leukemia and the potential for targeted therapy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:611-617. [PMID: 36485124 PMCID: PMC9821252 DOI: 10.1182/hematology.2022000359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Outcomes for infants diagnosed under 1 year of age with KMT2A-rearranged acute lymphoblastic leukemia (ALL) have remained stagnant over the past 20 years. Successive treatment protocols have previously focused on intensification of conventional chemotherapy, but increased treatment-related toxicity and chemoresistance have led to a plateau in survival. We have now entered an era of immunotherapy with integration of agents, such as blinatumomab or chimeric antigen receptor T-cell therapy, into the standard chemotherapy backbone, showing significant promise for improving the dismal outcomes for this disease. There remains much optimism for the future as a wealth of preclinical studies have identified additional novel targeted agents, such as venetoclax or menin inhibitors, ready for incorporation into treatment, providing further ammunition to combat this aggressive disease. In contrast, infants with KMT2A-germline ALL have demonstrated excellent survival outcomes with current therapy, but there remains a high burden of treatment-related morbidity. Greater understanding of the underlying blast genetics for infants with KMT2A-germline ALL and incorporation of immunotherapeutic approaches may enable a reduction in the intensity of chemotherapy while maintaining the excellent outcomes.
Collapse
|
24
|
Popov A, Tsaur G, Permikin Z, Fominikh V, Verzhbitskaya T, Riger T, Demina A, Shorikov E, Kustanovich A, Movchan L, Streneva O, Khlebnikova O, Makarova O, Arakaev O, Solodovnikov A, Boichenko E, Kondratchik K, Ponomareva N, Lapotentova E, Aleinikova O, Miakova N, Novichkova G, Karachunskiy A, Fechina L. Incidence and prognostic value of central nervous system involvement in infants with B-cell precursor acute lymphoblastic leukemia treated according to the MLL-Baby protocol. Pediatr Blood Cancer 2022; 69:e29860. [PMID: 35713168 DOI: 10.1002/pbc.29860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/04/2022] [Accepted: 06/08/2022] [Indexed: 11/12/2022]
Abstract
AIM The aim of the study was to evaluate the incidence and prognostic impact of central nervous system (CNS) involvement in infants with B-cell precursor acute lymphoblastic leukemia (BCP-ALL), as well as its relation with minimal residual disease (MRD) data. METHODS A total of 139 consecutive infants with BCP-ALL from the MLL-Baby trial were studied. Cerebrospinal fluid (CSF) samples were investigated by microscopy of cytospin slides. MRD was evaluated according to the protocol schedule by flow cytometry and PCR for fusion gene transcripts (FGT). RESULTS Involvement of the CNS at any level was found in 50 infants (36.0%). The incidence of CNS involvement was higher in patients with KMT2A gene rearrangements (44.0% for KMT2A-r vs. 15.4% for KMT2A-g, p = .003). The outcome of CNS-positive infants was significantly worse than that of CNS-negative infants, although this prognostic impact was limited to the KMT2A-r group (event-free survival 0.21 for CNS-positive vs. 0.48 for CNS-negative infants, p = .044). CNS-positive infants could not be treated successfully by conventional chemotherapy alone, irrespective of the rapidity of MRD response. In contrast, the combination of initial CNS negativity and FGT-MRD negativity identified a group comprising up to one-third of infants with KMT2A-r ALL who can be treated with chemotherapy and achieve very good outcomes (disease-free survival above 95%), and remaining patients should be allocated to receive other types of treatment. CONCLUSION We can conclude that this combination of initial CNS involvement and MRD data can significantly improve risk-group allocation in future clinical trials enrolling infants with ALL.
Collapse
Affiliation(s)
- Alexander Popov
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Grigory Tsaur
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Ural State Medical University, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Zhan Permikin
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Ural State Medical University, Ekaterinburg, Russian Federation
| | - Veronika Fominikh
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Tatiana Verzhbitskaya
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Tatiana Riger
- Regional Children's Hospital, Ekaterinburg, Russian Federation
| | - Anna Demina
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Egor Shorikov
- PET-Technology Centre of Nuclear Medicine, Ekaterinburg, Russian Federation
| | - Anatoly Kustanovich
- The Sharett Institute of Oncology, Hadassah Medical Centre, Jerusalem, Israel
| | - Liudmila Movchan
- Belarussian Research Centre for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Olga Streneva
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | | | - Olga Makarova
- Regional Children's Hospital, Ekaterinburg, Russian Federation
| | - Oleg Arakaev
- Regional Children's Hospital, Ekaterinburg, Russian Federation
| | - Alexander Solodovnikov
- Ural State Medical University, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Elmira Boichenko
- City Children's Hospital No 1, Saint Petersburg, Russian Federation
| | | | | | - Elena Lapotentova
- Belarussian Research Centre for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Olga Aleinikova
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation.,Belarussian Research Centre for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Natalia Miakova
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Galina Novichkova
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexander Karachunskiy
- National Research and Clinical Centre for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Larisa Fechina
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| |
Collapse
|
25
|
Wen J, Zhou M, Shen Y, Long Y, Guo Y, Song L, Xiao J. Poor treatment responses were related to poor outcomes in pediatric B cell acute lymphoblastic leukemia with KMT2A rearrangements. BMC Cancer 2022; 22:859. [PMID: 35933338 PMCID: PMC9357304 DOI: 10.1186/s12885-022-09804-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/16/2022] [Indexed: 11/21/2022] Open
Abstract
Background The KMT2A gene, formerly named the MLL gene, is rearranged (KMT2Ar) in 70–75% of infants, 5–6% of children and 10–15% of adult patients with B cell acute lymphoblastic leukemia (B-ALL). The outcome after chemotherapy of pediatric cases remains poor, and only a few studies have investigated the clinical and laboratory features, treatment response and prognosis in Chinese populations. Methods A total of 48 B-ALL children with KMT2Ar were enrolled in the study, and clinical and laboratory data were collected and analyzed by age group. The relationship between prognosis and traditional risk factors and treatment response was investigated for these patients who received chemotherapy. Results The 48 enrolled patients included 28 males and 20 females; 18 (37.50%) or 30 (62.50%) patients were an age of < 12 m (infant B-ALL) or of > 12 m at onset. An initial WBC count of 300 × 109/L was detected in 7 (14.58%) patients; testicular leukemia (TL) or central nervous system involvement was found in 5 (10.41%) or 3 (6.25%) patients, respectively. Statistical differences were not found in the age groups of sex or initial WBC count, whereas TL was more common in the infant group (P < 0.05). 11q23 was detected in 18 patients; KMT2Ar was detected in 46 (95.83%) or 45 (93.75%) patients by FISH or multiplex RT–PCR technology, respectively; RNA-seq data were obtained for 18 patients, and 3 patients with uncommon KMT2Ar were identified. KMT2A-AFF1, KMT2A-MLLT3 and KMT2A-MLLT1 were the most common transcripts. Statistical differences were not found in treatment response by age groups, including dexamethasone induction, bone marrow (BM) smear status and minimal residual disease (MRD) level at different time points (TP), treatment-related mortality (TRM), or complete remission (CR) rate (P > 0.05); MRD levels monitored by FCM or PCR were unequal at the same TP. Four patients died of treatment, and TRM was 8.33%; 40 patients achieved CR, and the CR rate for the cohort was 83.33%. Seven patients quit, 15 patients relapsed, and the 5 yr cumulative relapse rate was 59.16 ± 9.16%; the 5 yr prospective EFS (pEFS) for patients who were included or excluded from the TRM group was 36.86 ± 8.48% or 40.84 ± 9.16%, respectively. Multivariate analysis for prognosis and hazard ratio was performed for 37 patients without TRM and revealed that an initial WBC count of > 300 × 109/L and a positive level of FCM-MRD were strongly related to a poor outcome for B-ALL patients with KMT2Ar (P < 0.05). Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09804-w.
Collapse
Affiliation(s)
- Jinquan Wen
- Department of Pediatric Hematology, Caihong Hospital of Xianyang, Xi'an, People's Republic of China
| | - Min Zhou
- Department of Hematology, Chengdu Women and Children's Central Hospital, Chengdu, People's Republic of China
| | - Yali Shen
- Department of Hematology, Children's Hospital of Chongqing Medical University, Yuzhong District, Zhongshan 2nd Road, Chongqing, 400014, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China
| | - Yueting Long
- Department of Pediatrics, The Second Affiliated Hospital of Guizhou Medical University, Kaili, People's Republic of China
| | - Yuxia Guo
- Department of Hematology, Children's Hospital of Chongqing Medical University, Yuzhong District, Zhongshan 2nd Road, Chongqing, 400014, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, People's Republic of China
| | - Lin Song
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Jianwen Xiao
- Department of Hematology, Children's Hospital of Chongqing Medical University, Yuzhong District, Zhongshan 2nd Road, Chongqing, 400014, People's Republic of China.
| |
Collapse
|
26
|
Immunoglobulin/T-Cell Receptor Gene Rearrangement Analysis Using RNA-Seq. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2453:61-77. [PMID: 35622320 DOI: 10.1007/978-1-0716-2115-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Identification of immunoglobulin (IG) and T-cell receptor (TR) gene rearrangements in acute lymphoblastic leukemia (ALL) patients at initial presentation are crucial for monitoring of minimal residual disease (MRD) during subsequent follow-up and thereby for appropriate risk-group stratification. Here we describe how RNA-Seq data can be generated and subsequently analyzed with ARResT/Interrogate to identify possible MRD markers. In addition to the procedures, possible pitfalls will be discussed. Similar strategies can be employed for other lymphoid malignancies, such as lymphoma and myeloma.
Collapse
|
27
|
Chen C, Yu W, Alikarami F, Qiu Q, Chen CH, Flournoy J, Gao P, Uzun Y, Fang L, Davenport JW, Hu Y, Zhu Q, Wang K, Libbrecht C, Felmeister A, Rozich I, Ding YY, Hunger SP, Felix CA, Wu H, Brown PA, Guest EM, Barrett DM, Bernt KM, Tan K. Single-cell multiomics reveals increased plasticity, resistant populations, and stem-cell-like blasts in KMT2A-rearranged leukemia. Blood 2022; 139:2198-2211. [PMID: 34864916 PMCID: PMC8990373 DOI: 10.1182/blood.2021013442] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
KMT2A-rearranged (KMT2A-r) infant acute lymphoblastic leukemia (ALL) is a devastating malignancy with a dismal outcome, and younger age at diagnosis is associated with increased risk of relapse. To discover age-specific differences and critical drivers that mediate poor outcome in KMT2A-r ALL, we subjected KMT2A-r leukemias and normal hematopoietic cells from patients of different ages to single-cell multiomics analyses. We uncovered the following critical new insights: leukemia cells from patients <6 months have significantly increased lineage plasticity. Steroid response pathways are downregulated in the most immature blasts from younger patients. We identify a hematopoietic stem and progenitor-like (HSPC-like) population in the blood of younger patients that contains leukemic blasts and form an immunosuppressive signaling circuit with cytotoxic lymphocytes. These observations offer a compelling explanation for the ability of leukemias in young patients to evade chemotherapy and immune-mediated control. Our analysis also revealed preexisting lymphomyeloid primed progenitors and myeloid blasts at initial diagnosis of B-ALL. Tracking of leukemic clones in 2 patients whose leukemia underwent a lineage switch documented the evolution of such clones into frank acute myeloid leukemia (AML). These findings provide critical insights into KMT2A-r ALL and have clinical implications for molecularly targeted and immunotherapy approaches. Beyond infant ALL, our study demonstrates the power of single-cell multiomics to detect tumor intrinsic and extrinsic factors affecting rare but critical subpopulations within a malignant population that ultimately determines patient outcome.
Collapse
Affiliation(s)
- Changya Chen
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Wenbao Yu
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine
| | | | - Qi Qiu
- Department of Genetics, Perelman School of Medicine
- Penn Epigenetics Institute, and
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA
| | - Chia-Hui Chen
- Division of Oncology and Center for Childhood Cancer Research and
| | - Jennifer Flournoy
- Department of Genetics, Perelman School of Medicine
- Penn Epigenetics Institute, and
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA
| | - Peng Gao
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Yasin Uzun
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Li Fang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Yuxuan Hu
- School of Computer Science and Technology, Xidian University, Xi'an, China
| | - Qin Zhu
- Graduate Group in Genomics and Computational Biology
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, and
| | - Clara Libbrecht
- Division of Oncology and Center for Childhood Cancer Research and
| | - Alex Felmeister
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Isaiah Rozich
- Graduate Group in Immunology, University of Pennsylvania, Philadelphia, PA
| | - Yang-Yang Ding
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Pediatrics, Perelman School of Medicine
| | - Stephen P Hunger
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Pediatrics, Perelman School of Medicine
| | - Carolyn A Felix
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Pediatrics, Perelman School of Medicine
| | - Hao Wu
- Department of Genetics, Perelman School of Medicine
- Penn Epigenetics Institute, and
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA
| | - Patrick A Brown
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, MD; and
| | - Erin M Guest
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, MO
| | - David M Barrett
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Pediatrics, Perelman School of Medicine
| | - Kathrin M Bernt
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Pediatrics, Perelman School of Medicine
| | - Kai Tan
- Division of Oncology and Center for Childhood Cancer Research and
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine
- Department of Genetics, Perelman School of Medicine
- Penn Epigenetics Institute, and
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
28
|
Syed S, Song A, Hussaini M. Expanding diagnostic criteria: Multiorgan T-Cell/myeloid mixed phenotype acute leukemia with t(v;11q23) KMT2A-rearrangement successfully treated by allogeneic stem cell transplant. Leuk Res Rep 2022; 17:100306. [PMID: 35356416 PMCID: PMC8958533 DOI: 10.1016/j.lrr.2022.100306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 11/26/2022] Open
Abstract
Mixed phenotype acute leukemia (MPAL) consists of a leukemia of two different lineages (myeloid, T, and/or B) co-occurring in the same tissue. KMT2A-rearrangement is rare and usually seen in B/myeloid MPAL. We report a unique case of T/myeloid MPAL with a t(v;11q23) KMT2A-rearrangement, with acute myeloid leukemia (AML) in the bone marrow but concurrent T-cell acute lymphoblastic leukemia (T-ALL) in lymph node and skin. Genomic interrogation suggests an undifferentiated stem cells with KMT2A rearrangement as the founder mutation that acquired additional lineage-specific mutations resulting in AML in the marrow and T-ALL in other sites.
Collapse
Affiliation(s)
- Suhayb Syed
- University of South Florida, Tampa, FL, United States
| | - Amy Song
- Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Mohammad Hussaini
- Department of Pathology and Lab Medicines, H. Lee Moffitt Cancer Center, United States
| |
Collapse
|
29
|
Tomizawa D, Miyamura T, Koh K, Ishii E. Acute lymphoblastic leukemia in infants: A quarter century of nationwide efforts in Japan. Pediatr Int 2022; 64:e14935. [PMID: 34324764 DOI: 10.1111/ped.14935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/16/2021] [Accepted: 07/27/2021] [Indexed: 12/01/2022]
Abstract
Acute lymphoblastic leukemia (ALL) with KMT2A gene rearrangement (KMT2A-r) in infants is a biologically and clinically unique disease and one of the most difficult to cure forms of pediatric leukemia. Multicenter clinical trials have been carried out in Japan since the mid-1990s by introducing allogeneic hematopoietic stem cell transplantation (HSCT) in first remission, which led to a modest improvement in outcome of infants with KMT2A-r ALL. Because of the emerging evidence that HSCT does not benefit every infant with KMT2A-r ALL, the Japanese Pediatric Leukemia/Lymphoma Study Group trial MLL-10 introduced risk stratification using age and presence of central nervous system leukemia, and introduced intensive chemotherapy, including high-dose cytarabine in early consolidation; indication of HSCT was restricted to the patients with high-risk features. The trial resulted in excellent 3-year event-free survival of 66.2% (standard error, 5.6%) and overall survival of 83.9% (standard error, 4.3%) for 75 patients with KMT2A-r ALL recruited between 2011 and 2015. This Japanese experience and the results of the infant ALL trials worldwide suggest the importance of introducing effective therapy in the early phase of therapy, thus clearing minimal residual disease as rapidly as possible. However, further improvement in outcome is unlikely with conventional treatment approaches. Introduction of biology-driven novel agents and/or immunotherapies through international collaboration would be key solutions to overcome the disease.
Collapse
Affiliation(s)
- Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Takako Miyamura
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Eiichi Ishii
- Department of Pediatrics, Ehime University Graduate School of Medicine, Toon, Japan
| |
Collapse
|
30
|
Popov A, Tsaur G, Verzhbitskaya T, Riger T, Permikin Z, Demina A, Mikhailova E, Shorikov E, Arakaev O, Streneva O, Khlebnikova O, Makarova O, Miakova N, Fominikh V, Boichenko E, Kondratchik K, Ponomareva N, Novichkova G, Karachunskiy A, Fechina L. Comparison of minimal residual disease measurement by multicolour flow cytometry and PCR for fusion gene transcripts in infants with acute lymphoblastic leukaemia with KMT2A gene rearrangements. Br J Haematol 2021; 201:510-519. [PMID: 34970734 DOI: 10.1111/bjh.18021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022]
Abstract
This study aimed to evaluate the concordance between minimal residual disease (MRD) results obtained by multicolour flow cytometry (MFC) and polymerase chain reaction for fusion gene transcripts (FGTs) in infants with acute lymphoblastic leukaemia (ALL) associated with rearrangement of the KMT2A gene (KMT2A-r). A total of 942 bone marrow (BM) samples from 123 infants were studied for MFC-MRD and FGT-MRD. In total, 383 samples (40.7%) were concordantly MRD-negative. MRD was detected by the two methods in 441 cases (46.8%); 99 samples (10.5%) were only FGT-MRD-positive and 19 (2.0%) were only MFC-MRD-positive. A final concordance rate of 87.4% was established. Most discordance occurred if residual leukaemia was present at levels close to the sensitivity limits. Neither the type of KMT2A fusion nor a new type of treatment hampering MFC methodology had an influence on the concordance rate. The prognostic value of MFC-MRD and FGT-MRD differed. MFC-MRD was able to identify a rapid response at early time-points, whereas FGT-MRD was a reliable relapse predictor at later treatment stages. Additionally, the most precise risk definition was obtained when combining the two methods. Because of the high comparability in results, these two rather simple and inexpensive approaches could be good options of high clinical value.
Collapse
Affiliation(s)
- Alexander Popov
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Grigory Tsaur
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation.,Ural State Medical University, Ekaterinburg, Russian Federation
| | - Tatiana Verzhbitskaya
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Tatiana Riger
- Regional Children's Hospital, Ekaterinburg, Russian Federation
| | - Zhan Permikin
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Ural State Medical University, Ekaterinburg, Russian Federation
| | - Anna Demina
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Ekaterina Mikhailova
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Egor Shorikov
- PET-Technology Center of Nuclear Medicine, Ekaterinburg, Russian Federation
| | - Oleg Arakaev
- Regional Children's Hospital, Ekaterinburg, Russian Federation
| | - Olga Streneva
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | | | - Olga Makarova
- Regional Children's Hospital, Ekaterinburg, Russian Federation
| | - Natalia Miakova
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Veronika Fominikh
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Elmira Boichenko
- City Children's Hospital №1, Saint-Petersburg, Russian Federation
| | | | | | - Galina Novichkova
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexander Karachunskiy
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Larisa Fechina
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| |
Collapse
|
31
|
Stutterheim J, de Lorenzo P, van der Sluin IM, Alten J, Ancliffe P, Attarbaschi A, Aversa L, Boer JM, Biondi A, Brethon B, Diaz P, Cazzaniga G, Escherich G, Ferster A, Kotecha RS, Lausen B, Leung AW, Locatelli F, Silverman L, Stary J, Szczepanski T, van der Velden VHJ, Vora A, Zuna J, Schrappe M, Valsecchi MG, Pieters R. Minimal residual disease and outcome characteristics in infant KMT2A-germline acute lymphoblastic leukaemia treated on the Interfant-06 protocol. Eur J Cancer 2021; 160:72-79. [PMID: 34785111 DOI: 10.1016/j.ejca.2021.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND The outcome of infants with KMT2A-germline acute lymphoblastic leukaemia (ALL) is superior to that of infants with KMT2A-rearranged ALL but has been inferior to non-infant ALL patients. Here, we describe the outcome and prognostic factors for 167 infants with KMT2A-germline ALL enrolled in the Interfant-06 study. METHODS Univariate analysis on prognostic factors (age, white blood cell count at diagnosis, prednisolone response and CD10 expression) was performed on KMT2A-germline infants in complete remission at the end of induction (EOI; n = 163). Bone marrow minimal residual disease (MRD) was measured in 73 patients by real-time quantitative polymerase chain reaction at various time points (EOI, n = 68; end of consolidation, n = 56; and before OCTADAD, n = 57). MRD results were classified as negative, intermediate (<5∗10-4), and high (≥5∗10-4). RESULTS The 6-year event-free and overall survival was 73.9% (standard error [SE] = 3.6) and 87.2% (SE = 2.7). Relapses occurred early, within 36 months from diagnosis in 28 of 31 (90%) infants. Treatment-related mortality was 3.6%. Age <6 months was a favourable prognostic factor with a 6-year disease-free survival (DFS) of 91% (SE = 9.0) compared with 71.7% (SE = 4.2) in infants >6 months of age (P = 0.04). Patients with high EOI MRD ≥5 × 10-4 had a worse outcome (6-year DFS 61.4% [SE = 12.4], n = 16), compared with patients with undetectable EOI MRD (6-year DFS 87.9% [SE = 6.6], n = 28) or intermediate EOI MRD <5 × 10-4 (6-year DFS 76.4% [SE = 11.3], n = 24; P = 0.02). CONCLUSION We conclude that young age at diagnosis and low EOI MRD seem favourable prognostic factors in infants with KMT2A-germline ALL and should be considered for risk stratification in future clinical trials.
Collapse
Affiliation(s)
- J Stutterheim
- Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - P de Lorenzo
- Center of Bioinformatics, Biostatistics and Bioimaging, University of Milano-Bicocca, Monza, Italy; Pediatrics, School of Medicine and Surgery, University of Milano- Bicocca, Fondazione MBBM/San Gerardo Hospital, Monza, Italy
| | - I M van der Sluin
- Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - J Alten
- Pediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University of Kiel, Germany
| | - P Ancliffe
- United Kingdom Children Cancer Study Group, London, United Kingdom
| | - A Attarbaschi
- St Anna Children's Hospital, Pediatric Hematology and Oncology, Austria
| | | | - J M Boer
- Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - A Biondi
- Pediatrics, School of Medicine and Surgery, University of Milano- Bicocca, Fondazione MBBM/San Gerardo Hospital, Monza, Italy
| | - B Brethon
- Department of Pediatric Hematology, University Robert Debre Hospital, APHP, Paris, France
| | - P Diaz
- Chilean National Pediatric Oncology Group, Santiago, Chile
| | - G Cazzaniga
- Tettamanti Research Center, Pediatrics, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - G Escherich
- German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg, Germany
| | - A Ferster
- European Organisation for Research and Treatment of Cancer Children Leukemia Group, Brussels, Belgium
| | - R S Kotecha
- Australian and New Zealand Children's Haematology/Oncology Group, Perth Children's Hospital, Perth, Australia; Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - B Lausen
- Rigshospitalet, University Hospital, Department of Pediatrics, Copenhagen, Denmark
| | - Alex Wk Leung
- The Chinese University of Hong Kong, Shatin, Hong Kong, Special Administrative Region, People's Republic of China
| | - F Locatelli
- Department of Pediatric Haematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza University of Rome, Rome, Italy
| | - L Silverman
- Dana-Farber Cancer Institute, Pediatric Oncology, Boston, MA, USA
| | - J Stary
- Czech Working Group for Pediatric Hematology, Prague, Czech Republic
| | - T Szczepanski
- Polish Pediatric Leukemia/Lymphoma Study Group, Department of Pediatric Hematology and Oncology, Medical University of Silesia, Zabrze, Katowice, Poland
| | | | - A Vora
- United Kingdom Children Cancer Study Group, London, United Kingdom
| | - J Zuna
- CLIP, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - M Schrappe
- Berlin-Frankfurt-Miu (¨)nster Group Germany, Kiel, Germany
| | - M G Valsecchi
- Center of Bioinformatics, Biostatistics and Bioimaging, University of Milano-Bicocca, Monza, Italy
| | - R Pieters
- Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Dutch Childhood Oncology Group, Utrecht, the Netherlands
| |
Collapse
|
32
|
Neonatal acute lymphoblastic leukemia (MLL-AF9) with leukemia cutis. Pediatr Neonatol 2021; 62:676-678. [PMID: 34217651 DOI: 10.1016/j.pedneo.2021.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/25/2021] [Accepted: 06/01/2021] [Indexed: 11/20/2022] Open
|
33
|
Hematopoietic stem cell transplantation for infants with high-risk KMT2A gene-rearranged acute lymphoblastic leukemia. Blood Adv 2021; 5:3891-3899. [PMID: 34500465 DOI: 10.1182/bloodadvances.2020004157] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/07/2021] [Indexed: 12/27/2022] Open
Abstract
The role of allogeneic hematopoietic stem cell transplantation (HSCT) for infants with acute lymphoblastic leukemia (ALL) and KMT2A gene rearrangement (KMT2A-r) is controversial in terms of both its efficacy and potential for acute and late toxicities. In Japanese Pediatric Leukemia/Lymphoma Study Group trial MLL-10, by introducing intensive chemotherapy, indication of HSCT was restricted to patients with high-risk (HR) features only (KMT2A-r and either age <180 days or presence of central nervous system leukemia). Of the 56 HR patients, 49 achieved complete remission. Forty-three patients received HSCT in first remission including 38 patients receiving protocol-specified HSCT with conditioning consisting of individualized targeted doses of busulfan, etoposide, and cyclophosphamide. Three-year event-free survival (EFS) of 56.8% (95% confidence interval [CI], 42.4% to 68.8%) and overall survival of 80.2% (95% CI, 67.1% to 88.5%) were accomplished. Univariable analysis showed that Interfant-HR criteria and flow cytometric minimal residual disease (MRD; ≥0.01%), both at the end of induction and at the end of consolidation (EOC), were significantly associated with poorer EFS. In the multivariable analysis, positive MRD at EOC was solely associated with poor EFS (P < .001). Rapid pretransplant MRD clearance and tailored HSCT strategy in the MLL-10 trial resulted in a favorable outcome for infants with HR KMT2A-r ALL. However, considering the high rate of potentially life-threatening toxicities and the risk of late effects, its indication should be further restricted or even eliminated in the future by introducing more effective therapeutic modalities with minimal toxicities. This trial was registered at the University Hospital Medical Information Network Clinical Trials Registry (UMIN-CTR) as #UMIN000004801.
Collapse
|
34
|
Comparison of the blood, bone marrow, and cerebrospinal fluid metabolomes in children with b-cell acute lymphoblastic leukemia. Sci Rep 2021; 11:19613. [PMID: 34608220 PMCID: PMC8490393 DOI: 10.1038/s41598-021-99147-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
Metabolomics may shed light on treatment response in childhood acute lymphoblastic leukemia (ALL), however, most assessments have analyzed bone marrow or cerebrospinal fluid (CSF), which are not collected during all phases of therapy. Blood is collected frequently and with fewer risks, but it is unclear whether findings from marrow or CSF biomarker studies may translate. We profiled end-induction plasma, marrow, and CSF from N = 10 children with B-ALL using liquid chromatography-mass spectrometry. We estimated correlations between plasma and marrow/CSF metabolite abundances detected in ≥ 3 patients using Spearman rank correlation coefficients (rs). Most marrow metabolites were detected in plasma (N = 661; 81%), and we observed moderate-to-strong correlations (median rs 0.62, interquartile range [IQR] 0.29–0.83). We detected 328 CSF metabolites in plasma (90%); plasma-CSF correlations were weaker (median rs 0.37, IQR 0.07–0.70). We observed plasma-marrow correlations for metabolites in pathways associated with end-induction residual disease (pyruvate, asparagine) and plasma-CSF correlations for a biomarker of fatigue (gamma-glutamylglutamine). There is considerable overlap between the plasma, marrow, and CSF metabolomes, and we observed strong correlations for biomarkers of clinically relevant phenotypes. Plasma may be suitable for biomarker studies in B-ALL.
Collapse
|
35
|
Boer JM, Valsecchi MG, Hormann FM, Antić Ž, Zaliova M, Schwab C, Cazzaniga G, Arfeuille C, Cavé H, Attarbaschi A, Strehl S, Escherich G, Imamura T, Ohki K, Grüber TA, Sutton R, Pastorczak A, Lammens T, Lambert F, Li CK, Carrillo de Santa Pau E, Hoffmann S, Möricke A, Harrison CJ, Den Boer ML, De Lorenzo P, Stam RW, Bergmann AK, Pieters R. Favorable outcome of NUTM1-rearranged infant and pediatric B cell precursor acute lymphoblastic leukemia in a collaborative international study. Leukemia 2021; 35:2978-2982. [PMID: 34211097 PMCID: PMC8478641 DOI: 10.1038/s41375-021-01333-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Judith M Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.
- Oncode Institute, Utrecht, Netherlands.
| | - Maria Grazia Valsecchi
- Interfant Trial Data Center, School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Femke M Hormann
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Željko Antić
- Institute of Human Genetics, Medical School Hannover, Hannover, Germany
| | - Marketa Zaliova
- CLIP, Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Claire Schwab
- Leukaemia Research Cytogenetics Group, Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Giovanni Cazzaniga
- Tettamanti Research Center, Pediatric Clinic, University of Milan-Bicocca, Monza, Italy
| | - Chloé Arfeuille
- Department of Genetics, Robert Debré Hospital and University of Paris, Paris, France
| | - Hélène Cavé
- Department of Genetics, Robert Debré Hospital and University of Paris, Paris, France
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Sabine Strehl
- CCRI, St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Hamburg, Germany
| | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kentaro Ohki
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Tanja A Grüber
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rosemary Sutton
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of NSW, Randwick, NSW, Australia
| | - Agata Pastorczak
- Department of Pediatric Oncology and Hematology, Medical University of Lodz, Lodz, CA, Poland
| | - Tim Lammens
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Pediatric Hemato-oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | | | - Chi Kong Li
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | - Steve Hoffmann
- Computational Biology, Leibniz Institute on Ageing-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Anja Möricke
- Department of Pediatrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Christine J Harrison
- Leukaemia Research Cytogenetics Group, Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Monique L Den Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
- Erasmus MC-Sophia Children's Hospital, Department of Pediatric Oncology/Hematology, Rotterdam, Netherlands
| | - Paola De Lorenzo
- Interfant Trial Data Center, School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Tettamanti Research Center, Pediatric Clinic, University of Milan-Bicocca, Monza, Italy
| | - Ronald W Stam
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Anke K Bergmann
- Institute of Human Genetics, Medical School Hannover, Hannover, Germany
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
36
|
Ceppi F, Rizzati F, Colombini A, Conter V, Cazzaniga G. Utilizing the prognostic impact of minimal residual disease in treatment decisions for pediatric acute lymphoblastic leukemia. Expert Rev Hematol 2021; 14:795-807. [PMID: 34374613 DOI: 10.1080/17474086.2021.1967137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Acute lymphoblastic leukemia (ALL) is the first pediatric cancer where the assessment of early response to therapy by minimal residual disease (MRD) monitoring has demonstrated its importance to improve risk-based treatment approaches. The most standardized tools to study MRD in ALL are multiparametric flow cytometry and realtime-quantitative polymerase chain reaction amplification-based methods. In recent years, MRD measurement has reached greater levels of sensitivity and standardization through international laboratory networks collaboration. AREAS COVERED We herewith describe how to assess and apply the prognostic impact of MRD in treatment decisions, with specific focus on pediatric ALL. We also highlight the role of MRD monitoring in the context of genetically homogeneous subgroups of pediatric ALL. However, some queries remain to be addressed and emerging technologies hold the promise of improving MRD detection in ALL patients. EXPERT OPINION Emerging technologies, like next generation flow cytometry, droplet digital PCR, and next generation sequencing appear to be important methods for assessing MRD in pediatric ALL. These more specific and/or sensitive MRD monitoring methods may help to predict relapse with greater accuracy, and are currently being used in clinical trials to improve pediatric ALL outcome by optimizing patient stratification and earlier MRD-based interventional therapy.
Collapse
Affiliation(s)
- Francesco Ceppi
- Pediatric Hematology-Oncology Unit, Division of Pediatrics, Woman-Mother-Child Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Frida Rizzati
- Pediatric Hematology-Oncology Unit, Division of Pediatrics, Woman-Mother-Child Department, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Antonella Colombini
- Pediatric Hematology-Oncology, University Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Valentino Conter
- Pediatric Hematology-Oncology, University Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Giovanni Cazzaniga
- Centro Ricerca Tettamanti, Pediatrics, School of Medicine, University of Milano Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy.,Medical Genetics, School of Medicine, University of Milano Bicocca, Monza, Italy
| |
Collapse
|
37
|
Liao W, Kohler ME, Fry T, Ernst P. Does lineage plasticity enable escape from CAR-T cell therapy? Lessons from MLL-r leukemia. Exp Hematol 2021; 100:1-11. [PMID: 34298117 PMCID: PMC8611617 DOI: 10.1016/j.exphem.2021.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 01/20/2023]
Abstract
The clinical success of engineered, CD19-directed chimeric antigen receptor (CAR) T cells in relapsed, refractory B-cell acute lymphoblastic leukemia (B-ALL) has generated great enthusiasm for the use of CAR T cells in patients with cytogenetics that portend a poor prognosis with conventional cytotoxic therapies. One such group includes infants and children with mixed lineage leukemia (MLL1, KMT2A) rearrangements (MLL-r), who fare much worse than patients with low- or standard-risk B-ALL. Although early clinical trials using CD19 CAR T cells for MLL-r B-ALL produced complete remission in most patients, relapse with CD19-negative disease was a common mechanism of treatment failure. Whereas CD19neg relapse has been observed across a broad spectrum of B-ALL patients treated with CD19-directed therapy, patients with MLL-r have manifested the emergence of AML, often clonally related to the B-ALL, suggesting that the inherent heterogeneity or lineage plasticity of MLL-r B-ALL may predispose patients to a myeloid relapse. Understanding the factors that enable and drive myeloid relapse may be important to devise strategies to improve durability of remissions. In this review, we summarize clinical observations to date with MLL-r B-ALL and generally discuss lineage plasticity as a mechanism of escape from immunotherapy.
Collapse
Affiliation(s)
- Wenjuan Liao
- Department of Pediatrics, Section of Hematology/Oncology/BMT, Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO
| | - M Eric Kohler
- Department of Pediatrics, Section of Hematology/Oncology/BMT, Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO
| | - Terry Fry
- Department of Pediatrics, Section of Hematology/Oncology/BMT, Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO; Immunology Department and HI3 Initiative, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO
| | - Patricia Ernst
- Department of Pediatrics, Section of Hematology/Oncology/BMT, Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO; Pharmacology Department, University of Colorado, Denver/Anschutz Medical Campus. Aurora, CO.
| |
Collapse
|
38
|
Bartram J, Balasch-Carulla M, Bhojaraja S, Adams S, Cheng D, Inglott S, Kulkarni N, Mahendrayogam A, O'Connor O, Pavasovic V, Vora A. Blinatumomab for paediatric mixed phenotype acute leukaemia. Br J Haematol 2021; 195:289-292. [PMID: 34254287 DOI: 10.1111/bjh.17707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jack Bartram
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK.,SIHMDS-Haematology, Great Ormond Street Hospital for Children, London, UK
| | | | - Shashank Bhojaraja
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Stuart Adams
- SIHMDS-Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Danny Cheng
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Sarah Inglott
- SIHMDS-Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Nimish Kulkarni
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| | | | - Olya O'Connor
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Vesna Pavasovic
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Ajay Vora
- Department of Haematology, Great Ormond Street Hospital for Children, London, UK
| |
Collapse
|
39
|
Cheung LC, de Kraa R, Oommen J, Chua GA, Singh S, Hughes AM, Ferrari E, Ford J, Chiu SK, Stam RW, Kees UR, Malinge S, Kotecha RS. Preclinical Evaluation of Carfilzomib for Infant KMT2A-Rearranged Acute Lymphoblastic Leukemia. Front Oncol 2021; 11:631594. [PMID: 33937032 PMCID: PMC8082024 DOI: 10.3389/fonc.2021.631594] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Infants with KMT2A-rearranged B-cell precursor acute lymphoblastic leukemia (ALL) have poor outcomes. There is an urgent need to identify novel agents to improve survival. Proteasome inhibition has emerged as a promising therapeutic strategy for several hematological malignancies. The aim of this study was to determine the preclinical efficacy of the selective proteasome inhibitor carfilzomib, for infants with KMT2A-rearranged ALL. METHODS Eight infant ALL cell lines were extensively characterized for immunophenotypic and cytogenetic features. In vitro cytotoxicity to carfilzomib was assessed using a modified Alamar Blue assay with cells in logarithmic growth. The Bliss Independence model was applied to determine synergy between carfilzomib and the nine conventional chemotherapeutic agents used to treat infants with ALL. Established xenograft models were used to identify the maximal tolerated dose of carfilzomib and determine in vivo efficacy. RESULTS Carfilzomib demonstrated low IC50 concentrations within the nanomolar range (6.0-15.8 nm) across the panel of cell lines. Combination drug testing indicated in vitro synergy between carfilzomib and several conventional chemotherapeutic agents including vincristine, daunorubicin, dexamethasone, L-asparaginase, and 4-hydroperoxycyclophosphamide. In vivo assessment did not lead to a survival advantage for either carfilzomib monotherapy, when used to treat both low or high disease burden, or for carfilzomib in combination with multi-agent induction chemotherapy comprising of vincristine, dexamethasone, and L-asparaginase. CONCLUSIONS Our study highlights that in vitro efficacy does not necessarily translate to benefit in vivo and emphasizes the importance of in vivo validation prior to suggesting an agent for clinical use. Whilst proteasome inhibitors have an important role to play in several hematological malignancies, our findings guard against prioritization of carfilzomib for treatment of KMT2A-rearranged infant ALL in the clinical setting.
Collapse
Affiliation(s)
- Laurence C. Cheung
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
| | | | - Joyce Oommen
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Grace-Alyssa Chua
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Sajla Singh
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Anastasia M. Hughes
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Emanuela Ferrari
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Jette Ford
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Sung K. Chiu
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Ronald W. Stam
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Ursula R. Kees
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Sébastien Malinge
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Rishi S. Kotecha
- Division of Children’s Leukaemia and Cancer Research, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children’s Hospital, Perth, WA, Australia
| |
Collapse
|
40
|
Popov A, Fominikh V, Mikhailova E, Shelikhova L, Tsaur G, Abugova Y, Zerkalenkova E, Olshanskaya Y, Balashov D, Novichkova G, Maschan A, Miakova N. Blinatumomab following haematopoietic stem cell transplantation - a novel approach for the treatment of acute lymphoblastic leukaemia in infants. Br J Haematol 2021; 194:174-178. [PMID: 33843056 DOI: 10.1111/bjh.17466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022]
Abstract
Blinatumomab with subsequent haematopoietic stem cell transplantation was applied in 13 infants with acute lymphoblastic leukaemia (ALL). Eight patients were treated in first remission due to slow clearance of minimal residual disease (MRD); one for MRD-reappearance after long MRD negativity, one for primary refractory disease and three during relapse treatment. In slow MRD responders, complete MRD response was achieved prior to transplantation, with an 18-month event-free survival of 75%. In contrast, only one of five patients with relapsed/refractory ALL is still in complete remission. These data provide a basis for future studies of immunotherapy in very high-risk infant ALL.
Collapse
Affiliation(s)
- Alexander Popov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Veronika Fominikh
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Ekaterina Mikhailova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Larisa Shelikhova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Grigory Tsaur
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Yulia Abugova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Elena Zerkalenkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Yulia Olshanskaya
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Dmitry Balashov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Galina Novichkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexey Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Natalia Miakova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| |
Collapse
|