1
|
Becht DC, Selvam K, Lachance C, Côté V, Li K, Nguyen MC, Pareek A, Shi X, Wen H, Blanco MA, Côté J, Kutateladze TG. A multivalent engagement of ENL with MOZ. Nat Struct Mol Biol 2025; 32:709-718. [PMID: 39794553 DOI: 10.1038/s41594-024-01455-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 11/19/2024] [Indexed: 01/13/2025]
Abstract
The epigenetic cofactor ENL (eleven-nineteen-leukemia) and the acetyltransferase MOZ (monocytic leukemia zinc finger) have vital roles in transcriptional regulation and are implicated in aggressive forms of leukemia. Here, we describe the mechanistic basis for the intertwined association of ENL and MOZ. Genomic analysis shows that ENL and MOZ co-occupy active promoters and that MOZ recruits ENL to its gene targets. Structural studies reveal a multivalent assembly of ENL at the intrinsically disordered region (IDR) of MOZ. While the extraterminal (ET) domain of ENL recognizes the canonical ET-binding motif in IDR, the YEATS domains of ENL and homologous AF9 bind to a set of acetylation sites in the MOZ IDR that are generated by the acetyltransferase CBP (CREB-binding protein). Our findings suggest a multifaceted acetylation-dependent and independent coupling of ENL, MOZ and CBP/p300, which may contribute to leukemogenic activities of the ENL-MOZ assembly and chromosomal translocations of ENL, MOZ and CBP/p300.
Collapse
Affiliation(s)
- Dustin C Becht
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Karthik Selvam
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Catherine Lachance
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Valérie Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Kuai Li
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Minh Chau Nguyen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Akshay Pareek
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Hong Wen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - M Andres Blanco
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA, USA
| | - Jacques Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
2
|
Jassinskaja M, Ghosh S, Watral J, Davoudi M, Claesson Stern M, Daher U, Eldeeb M, Zhang Q, Bryder D, Hansson J. A complex interplay of intra- and extracellular factors regulates the outcome of fetal- and adult-derived MLL-rearranged leukemia. Leukemia 2024; 38:1115-1130. [PMID: 38555405 PMCID: PMC11073998 DOI: 10.1038/s41375-024-02235-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
Infant and adult MLL1/KMT2A-rearranged (MLLr) leukemia represents a disease with a dismal prognosis. Here, we present a functional and proteomic characterization of in utero-initiated and adult-onset MLLr leukemia. We reveal that fetal MLL::ENL-expressing lymphomyeloid multipotent progenitors (LMPPs) are intrinsically programmed towards a lymphoid fate but give rise to myeloid leukemia in vivo, highlighting a complex interplay of intra- and extracellular factors in determining disease subtype. We characterize early proteomic events of MLL::ENL-mediated transformation in fetal and adult blood progenitors and reveal that whereas adult pre-leukemic cells are mainly characterized by retained myeloid features and downregulation of ribosomal and metabolic proteins, expression of MLL::ENL in fetal LMPPs leads to enrichment of translation-associated and histone deacetylases signaling proteins, and decreased expression of inflammation and myeloid differentiation proteins. Integrating the proteome of pre-leukemic cells with their secretome and the proteomic composition of the extracellular environment of normal progenitors highlights differential regulation of Igf2 bioavailability, as well as of VLA-4 dimer and its ligandome, upon initiation of fetal- and adult-origin leukemia, with implications for human MLLr leukemia cells' ability to communicate with their environment through granule proteins. Our study has uncovered opportunities for targeting ontogeny-specific proteomic vulnerabilities in in utero-initiated and adult-onset MLLr leukemia.
Collapse
Affiliation(s)
- Maria Jassinskaja
- Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
- York Biomedical Research Institute, Department of Biology, University of York, YO10 5DD, York, UK
| | - Sudip Ghosh
- Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Joanna Watral
- Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Mina Davoudi
- Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Melina Claesson Stern
- Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Ugarit Daher
- Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Mohamed Eldeeb
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
| | - Qinyu Zhang
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
| | - David Bryder
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, SE-221 84, Lund, Sweden
| | - Jenny Hansson
- Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.
| |
Collapse
|
3
|
Mendoza-Castrejon J, Magee JA. Layered immunity and layered leukemogenicity: Developmentally restricted mechanisms of pediatric leukemia initiation. Immunol Rev 2023; 315:197-215. [PMID: 36588481 PMCID: PMC10301262 DOI: 10.1111/imr.13180] [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] [Indexed: 01/03/2023]
Abstract
Hematopoietic stem cells (HSCs) and multipotent progenitor cells (MPPs) arise in successive waves during ontogeny, and their properties change significantly throughout life. Ontological changes in HSCs/MPPs underlie corresponding changes in mechanisms of pediatric leukemia initiation. As HSCs and MPPs progress from fetal to neonatal, juvenile and adult stages of life, they undergo transcriptional and epigenetic reprogramming that modifies immune output to meet age-specific pathogenic challenges. Some immune cells arise exclusively from fetal HSCs/MPPs. We propose that this layered immunity instructs cell fates that underlie a parallel layered leukemogenicity. Indeed, some pediatric leukemias, such as juvenile myelomonocytic leukemia, myeloid leukemia of Down syndrome, and infant pre-B-cell acute lymphoblastic leukemia, are age-restricted. They only present during infancy or early childhood. These leukemias likely arise from fetal progenitors that lose competence for transformation as they age. Other childhood leukemias, such as non-infant pre-B-cell acute lymphoblastic leukemia and acute myeloid leukemia, have mutation profiles that are common in childhood but rare in morphologically similar adult leukemias. These differences could reflect temporal changes in mechanisms of mutagenesis or changes in how progenitors respond to a given mutation at different ages. Interactions between leukemogenic mutations and normal developmental switches offer potential targets for therapy.
Collapse
Affiliation(s)
- Jonny Mendoza-Castrejon
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
| | - Jeffrey A. Magee
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
| |
Collapse
|
4
|
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: 5] [Impact Index Per Article: 2.5] [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
|
5
|
Balduzzi A, Buechner J, Ifversen M, Dalle JH, Colita AM, Bierings M. Acute Lymphoblastic Leukaemia in the Youngest: Haematopoietic Stem Cell Transplantation and Beyond. Front Pediatr 2022; 10:807992. [PMID: 35281233 PMCID: PMC8911028 DOI: 10.3389/fped.2022.807992] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022] Open
Abstract
The ALL SCTped 2012 FORUM (For Omitting Radiation Under Majority age) trial compared outcomes for children ≥4 years of age transplanted for acute lymphoblastic leukaemia (ALL) who were randomised to myeloablation with a total body irradiation (TBI)-based or chemotherapy-based conditioning regimen. The TBI-based preparation was associated with a lower rate of relapse compared with chemoconditioning. Nevertheless, the age considered suitable for TBI was progressively raised over time to spare the most fragile youngest patients from irradiation-related complications. The best approach to use for children <4 years of age remains unclear. Children diagnosed with ALL in their first year of life, defined as infants, have a remarkably poorer prognosis compared with older children. This is largely explained by the biology of their ALL, with infants often carrying a KMT2A gene rearrangement, as well as by their fragility. In contrast, the clinical presentations and biological features of ALL in children >1 year but <4 years often resemble those presented by older children. In this review, we explore the state of the art regarding haematopoietic stem cell transplantation (HSCT) in children <4 years, the preparative regimens available, and new developments in the field that may influence treatment decisions.
Collapse
Affiliation(s)
- Adriana Balduzzi
- Clinica Pediatrica Università degli Studi di Milano-Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma, Monza, Italy
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | | | - Jean-Hugues Dalle
- Hôpital Robert Debré, GH AP-HP. Nord Université de Paris, Paris, France
| | - Anca M Colita
- Department of Pediatric Hematology and BMT, Fundeni Clinical Institute, "Carol Davila" University of Medicine, Bucharest, Romania
| | | |
Collapse
|
6
|
Ibrahimova A, Winestone LE, Miller TP, Kettler K, Seif AE, Huang YS, Elgarten CW, Myers RM, Fisher BT, Aplenc R, Getz KD. Presentation acuity, induction mortality, and resource utilization in infants with acute leukemia. Pediatr Blood Cancer 2021; 68:e28940. [PMID: 33704911 PMCID: PMC8283996 DOI: 10.1002/pbc.28940] [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: 05/14/2020] [Revised: 12/28/2020] [Accepted: 01/11/2021] [Indexed: 11/11/2022]
Abstract
BACKGROUND Treatment of infants with acute leukemia remains challenging, especially for acute lymphocytic leukemia (ALL). Infants have shown markedly higher rates of induction mortality compared with noninfants. There are limited data on presentation acuity and supportive care utilization in this age group. METHODS In retrospective analyses of patients treated for new onset ALL or acute myeloid leukemia (AML) at pediatric hospitals contributing to the Pediatric Health Information System, we compared presentation acuity, induction mortality, and resource utilization in infants relative to noninfants less than 10 years at diagnosis. RESULTS Analyses included 10 359 children with ALL (405 infants, 9954 noninfants) and 871 AML (189 infants, 682 noninfants). Infants were more likely to present with multisystem organ failure compared to noninfants for both ALL (12% and 1%, PR = 10.8, 95% CI: 7.4, 15.7) and AML (6% vs. 3%; PR = 2.0, 95% CI: 1.0, 3.7). Infants with ALL had higher induction mortality compared to noninfants, even after accounting for differences in anthracycline exposure and presentation acuity (2.7% vs. 0.5%, HR = 2.1, 95% CI: 1.0, 4.8). Conversely, infants and noninfants with AML had similar rates of induction mortality (3.2% vs. 2.1%, HR = 1.2, 95% CI: 0.3, 3.9), which were comparable to rates among infants with ALL. Infants with ALL and AML had greater requirements for blood products, diuretics, supplemental oxygen, and ventilation during induction relative to noninfants. CONCLUSIONS Infants with leukemia present with higher acuity compared with noninfants. Induction mortality and supportive care requirements for infants with ALL were similar to all children with AML, and significantly higher than those for noninfants with ALL.
Collapse
Affiliation(s)
- Azada Ibrahimova
- Department of Pediatrics and Adolescent Medicine, Einstein Healthcare Network Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lena E. Winestone
- Division of AIBMT, Department of Pediatrics, UCSF Benioff Children’s Hospital, San Francisco, California, USA
| | - Tamara P. Miller
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA,Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kyle Kettler
- Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Alix E. Seif
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Yuan-Shung Huang
- Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Caitlin W. Elgarten
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Regina M. Myers
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brian T. Fisher
- Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Perelman School of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA,Division of Infectious Diseases, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Richard Aplenc
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Perelman School of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kelly D. Getz
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Perelman School of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Abstract
PURPOSE OF THE REVIEW Infant leukemia is a rare, distinct subgroup of pediatric acute leukemias diagnosed in children under 1 year of age and characterized by unique, aggressive biology. Here, we review its clinical presentation, underlying molecular biology, current treatment strategies, and novel therapeutic approaches. RECENT FINDINGS Infant leukemias are associated with high-risk molecular features and high rates of chemotherapy resistance. International collaborative clinical trials have led to better understanding of the underlying molecular biology, refined risk-based stratification, and investigated the use of hematopoietic stem cell transplantation. However, intensification of chemotherapy has failed to improve outcomes, and current regimens are associated with significant treatment-related and long-term toxicities. Infants with leukemia remain a challenging group to treat. We must continue collaborative efforts to move beyond traditional cytotoxic chemotherapy, incorporate molecularly targeted strategies and immunotherapy, and increase access to clinical trials to improve outcomes for this high-risk group of patients.
Collapse
|
8
|
Brown P, Inaba H, Annesley C, Beck J, Colace S, Dallas M, DeSantes K, Kelly K, Kitko C, Lacayo N, Larrier N, Maese L, Mahadeo K, Nanda R, Nardi V, Rodriguez V, Rossoff J, Schuettpelz L, Silverman L, Sun J, Sun W, Teachey D, Wong V, Yanik G, Johnson-Chilla A, Ogba N. Pediatric Acute Lymphoblastic Leukemia, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 18:81-112. [PMID: 31910389 DOI: 10.6004/jnccn.2020.0001] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy. Advancements in technology that enhance our understanding of the biology of the disease, risk-adapted therapy, and enhanced supportive care have contributed to improved survival rates. However, additional clinical management is needed to improve outcomes for patients classified as high risk at presentation (eg, T-ALL, infant ALL) and who experience relapse. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for pediatric ALL provide recommendations on the workup, diagnostic evaluation, and treatment of the disease, including guidance on supportive care, hematopoietic stem cell transplantation, and pharmacogenomics. This portion of the NCCN Guidelines focuses on the frontline and relapsed/refractory management of pediatric ALL.
Collapse
Affiliation(s)
- Patrick Brown
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Hiroto Inaba
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Colleen Annesley
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Susan Colace
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Mari Dallas
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Kara Kelly
- Roswell Park Comprehensive Cancer Center
| | | | | | | | - Luke Maese
- Huntsman Cancer Institute at the University of Utah
| | - Kris Mahadeo
- The University of Texas MD Anderson Cancer Center
| | | | | | | | - Jenna Rossoff
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - Laura Schuettpelz
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | - Weili Sun
- City of Hope National Medical Center
| | - David Teachey
- Abramson Cancer Center at the University of Pennsylvania
| | | | | | | | | |
Collapse
|
9
|
Li K, Xiong H, Li Y, Zhou P, Li J, Li H, Tao F, Wang Z, Chen Z. WITHDRAWN: Clinical features and outcomes of infant acute lymphoblastic leukemia from a single center in China. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2021. [DOI: 10.1016/j.phoj.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
10
|
Pennella CL, Deu MA, Rossi JG, Baialardo EM, Alonso CN, Rubio P, Guitter MR, La Rosa CGS, Alfaro EM, Zubizarreta PA, Felice MS. No benefit of Interfant protocols compared to BFM-based protocols for infants with acute lymphoblastic leukemia. Results from an institution in Argentina. Pediatr Blood Cancer 2020; 67:e28624. [PMID: 32729239 DOI: 10.1002/pbc.28624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 07/05/2020] [Accepted: 07/15/2020] [Indexed: 11/06/2022]
Abstract
BACKGROUND Infant acute lymphoblastic leukemia (ALL) is an infrequent disease characterized by clinical and biological features related to poor prognosis. Adapted therapies were designed without a clear consensus regarding the best treatment options. We aimed to compare the outcome between infant ALL cases receiving Interfant versus BFM-based protocols. PROCEDURE This is a retrospective observational study. From April 1990 to June 2018, infant ALL cases were enrolled in one of the five consecutive treatment protocols. Clinical, demographic, and biological features and outcome were evaluated. A comparative analysis was performed between Interfant protocols and BFM-based protocols. RESULTS During the studied period, 1913 ALL patients were admitted and 116 (6%) were infants. Treatment administered was: ALL-BFM'90 (n = 16), 1-ALL96-BFM/HPG (n = 7), Interfant-99 (n = 39), Interfant-06 (n = 35), and ALLIC-BFM'2009 (n = 19). The 5-year event-free survival probability (EFSp) was 31.9(standard error [SE] 4.6)% for the entire population, with a significant difference among risk groups according to Interfant-06 criteria (P = .0029). KMT2A-rearrangement status was the strongest prognostic factor (P = .048), independently of the protocol strategy. The median time for relapse was 24.1 months for patients with minimal residual disease (MRD)-negative versus 11.5 months for those with MRD-positive (P = .0386). EFSp and cumulative relapse risk probability (CRRp) were similar. Interfant protocols showed comparable induction (8.1% vs 7.1%, P = .852) and complete remission mortality (21.6% vs 28.6%, P = .438), failing to reduce the relapse rate (48.5% vs 30.7%, P = .149). CONCLUSIONS Interfant protocols and BFM-based protocols presented comparable results. The risk group stratification proposed by Interfant-06 was validated by our results, and MRD seems useful to identify patients with an increased risk of early relapse.
Collapse
Affiliation(s)
- Carla L Pennella
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - María A Deu
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Jorge G Rossi
- Department of Immunology and Rheumatology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Edgardo M Baialardo
- Department of Genetics, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Cristina N Alonso
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Patricia Rubio
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Myriam R Guitter
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Cristian G Sánchez La Rosa
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Elizabeth M Alfaro
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - Pedro A Zubizarreta
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| | - María S Felice
- Department of Hematology-Oncology, Hospital de Pediatría S.A.M.I.C Prof. Dr. Juan P. Garrahan, Buenos Aires, Argentina
| |
Collapse
|
11
|
The efficiency of murine MLL-ENL-driven leukemia initiation changes with age and peaks during neonatal development. Blood Adv 2020; 3:2388-2399. [PMID: 31405949 DOI: 10.1182/bloodadvances.2019000554] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022] Open
Abstract
MLL rearrangements are translocation mutations that cause both acute lymphoblastic leukemia and acute myeloid leukemia (AML). These translocations can occur as sole clonal driver mutations in infant leukemias, suggesting that fetal or neonatal hematopoietic progenitors may be exquisitely sensitive to transformation by MLL fusion proteins. To test this possibility, we used transgenic mice to induce one translocation product, MLL-ENL, during fetal, neonatal, juvenile and adult stages of life. When MLL-ENL was induced in fetal or neonatal mice, almost all died of AML. In contrast, when MLL-ENL was induced in adult mice, most survived for >1 year despite sustained transgene expression. AML initiation was most efficient when MLL-ENL was induced in neonates, and even transient suppression of MLL-ENL in neonates could prevent AML in most mice. MLL-ENL target genes were induced more efficiently in neonatal progenitors than in adult progenitors, consistent with the distinct AML initiation efficiencies. Interestingly, transplantation stress mitigated the developmental barrier to leukemogenesis. Since fetal/neonatal progenitors were highly competent to initiate MLL-ENL-driven AML, we tested whether Lin28b, a fetal master regulator, could accelerate leukemogenesis. Surprisingly, Lin28b suppressed AML initiation rather than accelerating it. This may explain why MLL rearrangements often occur before birth in human infant leukemia patients, but transformation usually does not occur until after birth, when Lin28b levels decline. Our findings show that the efficiency of MLL-ENL-driven AML initiation changes through the course of pre- and postnatal development, and developmental programs can be manipulated to impede transformation.
Collapse
|
12
|
Cherungonath A, Appaji L, Padma M, Arunakumari BS, Arunkumar AR, Avinash T, Vijay CR, Madhumati DS. Profile of Acute Lymphoblastic Leukemia in Children Under 2 Years of Age. Indian J Med Paediatr Oncol 2018. [DOI: 10.4103/ijmpo.ijmpo_10_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Abstract
Context: Acute lymphoblastic leukemia (ALL) shows substantial differences in clinical and laboratory features and treatment responsiveness in different subgroups. Pediatric ALL <2 years is associated with worse outcome. Infantile ALL is characterized by high white blood cell (WBC) counts, bulky extramedullary disease, translocations of 11q23 locus. There is paucity of data on ALL under 2 years of age. Aims: This study aims to analyze clinical, hematological, biochemical, immunophenotypical parameters, and treatment responsiveness of ALL under 2 years. Settings and Designs: It is a retrospective data analysis conducted at a Tertiary Care Cancer Centre in South India. The study population includes all pediatric ALL, registered at this institute during January 2009 to December 2013, who were under 2 years at the time of presentation. Materials and Methods: There were 122 cases of ALL under 2 years of age of whom 48 refused treatment, and four were lost to follow-up. Thus, 70 cases are eligible for analysis. Details on clinical, hematological, biochemical, radiological, immunophenotypical, and cytogenetic features were collected from case records, hospital cancer registry, and analysis done using SPSS version 20. Kaplan–Meier curves plotted for survival analysis. Results:: Male:female ratio was 1.26:1 (infants - 1:1; 1–2 years group - 1.3:1). The most common clinical features were hepatomegaly (95%) and fever (89%). Hemoglobin level >11 gm% was seen in 12%, WBC counts above 50,000 in 26% and platelets below 20,000 in 20%. Elevated lactate dehydrogenase presents in 64% and uric acid in 12%. Immunophenotype done in 44 children; precursor-B ALL-41, precursor-T ALL-3. Central nervous system -positive disease and 11q23 translocation were noted in one infant each. Conclusions: Infants with ALL are associated with poor prognosis. Children in the 1–2 years are associated with significantly better outcome. WBC counts >50,000 are associated with poor prognosis among infants. Treatment refusal/abandonment rate is 43% in the study population which is comparable to that in literature.
Collapse
Affiliation(s)
- Anoop Cherungonath
- Department of Pediatric Oncology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - L Appaji
- Department of Pediatric Oncology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - M Padma
- Department of Pediatric Oncology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - BS Arunakumari
- Department of Pediatric Oncology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - AR Arunkumar
- Department of Pediatric Oncology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - T Avinash
- Department of Pediatric Oncology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - CR Vijay
- Department of Epidemiology and Biostatistics, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - DS Madhumati
- Department of Pathology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| |
Collapse
|
13
|
Hudson BF, Oostendorp LJM, Candy B, Vickerstaff V, Jones L, Lakhanpaul M, Bluebond-Langner M, Stone P. The under reporting of recruitment strategies in research with children with life-threatening illnesses: A systematic review. Palliat Med 2017; 31:419-436. [PMID: 27609607 PMCID: PMC5405809 DOI: 10.1177/0269216316663856] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Researchers report difficulties in conducting research with children and young people with life-limiting conditions or life-threatening illnesses and their families. Recruitment is challenged by barriers including ethical, logistical and clinical considerations. AIM To explore how children and young people (aged 0-25 years) with life-limiting conditions or life-threatening illnesses and their families were identified, invited and consented to research published in the last 5 years. DESIGN Systematic review. DATA SOURCES MEDLINE, PsycINFO, Web of Science, Sciences Citation Index and SCOPUS were searched for original English language research published between 2009 and 2014, recruiting children and young people with life-limiting conditions or life-threatening illness and their families. RESULTS A total of 215 studies - 152 qualitative, 54 quantitative and 9 mixed methods - were included. Limited recruitment information but a range of strategies and difficulties were provided. The proportion of eligible participants from those screened could not be calculated in 80% of studies. Recruitment rates could not be calculated in 77%. A total of 31% of studies recruited less than 50% of eligible participants. Reasons given for non-invitation included missing clinical or contact data, or clinician judgements of participant unsuitability. Reasons for non-participation included lack of interest and participants' perceptions of potential burdens. CONCLUSION All stages of recruitment were under reported. Transparency in reporting of participant identification, invitation and consent is needed to enable researchers to understand research implications, bias risk and to whom results apply. Research is needed to explore why consenting participants decide to take part or not and their experiences of research recruitment.
Collapse
Affiliation(s)
- Briony F Hudson
- Louis Dundas Centre for Children’s Palliative Care, UCL Institute of Child Health, London, UK
- Marie Curie Palliative Care Research Department, UCL Division of Psychiatry, London, UK
| | - Linda JM Oostendorp
- Louis Dundas Centre for Children’s Palliative Care, UCL Institute of Child Health, London, UK
| | - Bridget Candy
- Marie Curie Palliative Care Research Department, UCL Division of Psychiatry, London, UK
| | - Victoria Vickerstaff
- Marie Curie Palliative Care Research Department, UCL Division of Psychiatry, London, UK
| | - Louise Jones
- Marie Curie Palliative Care Research Department, UCL Division of Psychiatry, London, UK
| | - Monica Lakhanpaul
- Population, Policy and Practice Programme, UCL Institute of Child Health, London, UK
| | - Myra Bluebond-Langner
- Louis Dundas Centre for Children’s Palliative Care, UCL Institute of Child Health, London, UK
| | - Paddy Stone
- Marie Curie Palliative Care Research Department, UCL Division of Psychiatry, London, UK
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW The prognosis for infants less than 12 months of age who are diagnosed with acute lymphoblastic leukemia (ALL) remains poor despite overall advances in the treatment of childhood ALL. In this review, we highlight the recent advances in the understanding of the pathogenesis of infant ALL and discuss opportunities for translating these findings into clinical trials. RECENT FINDINGS Infant ALL can be divided into two major disease types, defined by the presence or absence of KMT2A (MLL) rearrangement (KMT2A-R). Recent molecular profiling studies have found that infant ALL with KMT2A-R is an epigenomic disease that lacks other somatic driver mutations. Strategies to intensify therapy have not improved survival for infants with KMT2A-R ALL. In contrast, infant ALL without KMT2A-R is more similar to ALL of older children and survival has improved modestly with intensification of chemotherapy. Discovery of clonal molecular markers that predict chemoresistance will allow further risk classification and development of novel treatment strategies. Modern clinical trials are integrating molecularly targeted therapies into the treatment of infant ALL. SUMMARY Advances in molecular profiling and integration of targeted therapy have the potential to reduce toxicity and improve survival for infants with ALL.
Collapse
|
15
|
Geerlinks AV, Issekutz T, Wahlstrom JT, Sullivan KE, Cowan MJ, Dvorak CC, Fernandez CV. Severe, persistent, and fatal T-cell immunodeficiency following therapy for infantile leukemia. Pediatr Blood Cancer 2016; 63:2046-9. [PMID: 27354010 PMCID: PMC7168093 DOI: 10.1002/pbc.26108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/16/2016] [Accepted: 05/26/2016] [Indexed: 01/31/2023]
Abstract
We describe five cases of children who completed chemotherapy for infantile acute lymphoblastic leukemia (ALL) and soon after were diagnosed with severe T-cell, non-HIV immunodeficiency, with varying B-cell and NK-cell depletion. There was near absence of CD3(+) , CD4(+) , and CD8(+) cells. All patients developed multiple, primarily opportunistic infections. Unfortunately, four patients died, although one was successfully treated by hematopoietic stem cell transplantation. These immunodeficiencies appeared to be secondary to intensive infant ALL chemotherapy. Our report highlights the importance of the early consideration of this life-threatening immune complication in patients who received chemotherapy for infantile ALL.
Collapse
Affiliation(s)
- Ashley V. Geerlinks
- Department of PediatricsIWK Health Centre, Dalhousie UniversityHalifax, NSCanada
| | - Thomas Issekutz
- Department of PediatricsIWK Health Centre, Dalhousie UniversityHalifax, NSCanada
| | - Justin T. Wahlstrom
- Department of PediatricsBenioff Children's Hospital, University of California San FranciscoSan Francisco, CA
| | - Kathleen E. Sullivan
- Department of PediatricsChildren's Hospital of Philadelphia, University of Pennsylvania Perelman School of MedicinePhiladelphiaCA
| | - Morton J. Cowan
- Department of PediatricsBenioff Children's Hospital, University of California San FranciscoSan Francisco, CA
| | - Christopher C. Dvorak
- Department of PediatricsBenioff Children's Hospital, University of California San FranciscoSan Francisco, CA
| | - Conrad V. Fernandez
- Department of PediatricsIWK Health Centre, Dalhousie UniversityHalifax, NSCanada
| |
Collapse
|
16
|
Revisiting the biology of infant t(4;11)/MLL-AF4+ B-cell acute lymphoblastic leukemia. Blood 2015; 126:2676-85. [PMID: 26463423 DOI: 10.1182/blood-2015-09-667378] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Infant B-cell acute lymphoblastic leukemia (B-ALL) accounts for 10% of childhood ALL. The genetic hallmark of most infant B-ALL is chromosomal rearrangements of the mixed-lineage leukemia (MLL) gene. Despite improvement in the clinical management and survival (∼85-90%) of childhood B-ALL, the outcome of infants with MLL-rearranged (MLL-r) B-ALL remains dismal, with overall survival <35%. Among MLL-r infant B-ALL, t(4;11)+ patients harboring the fusion MLL-AF4 (MA4) display a particularly poor prognosis and a pro-B/mixed phenotype. Studies in monozygotic twins and archived blood spots have provided compelling evidence of a single cell of prenatal origin as the target for MA4 fusion, explaining the brief leukemia latency. Despite its aggressiveness and short latency, current progress on its etiology, pathogenesis, and cellular origin is limited as evidenced by the lack of mouse/human models recapitulating the disease phenotype/latency. We propose this is because infant cancer is from an etiologic and pathogenesis standpoint distinct from adult cancer and should be seen as a developmental disease. This is supported by whole-genome sequencing studies suggesting that opposite to the view of cancer as a "multiple-and-sequential-hit" model, t(4;11) alone might be sufficient to spawn leukemia. The stable genome of these patients suggests that, in infant developmental cancer, one "big-hit" might be sufficient for overt disease and supports a key contribution of epigenetics and a prenatal cell of origin during a critical developmental window of stem cell vulnerability in the leukemia pathogenesis. Here, we revisit the biology of t(4;11)+ infant B-ALL with an emphasis on its origin, genetics, and disease models.
Collapse
|
17
|
Abstract
Treatment of infants with acute lymphoblastic leukemia (ALL), especially those with mixed lineage leukemia (MLL) rearrangement (MLL-r), which account for approximately 80% of cases, is still a major challenge for pediatric hematologists and oncologists worldwide. Continuing efforts by collaborative clinical study groups in Europe, North America, and Japan have rescued approximately half of the MLL-r ALL patients with intensive chemotherapy with or without allogeneic hematopoietic stem cell transplantation. Recent progress has clarified the unique mechanism of MLL-r ALL: the aberrant methylation and histone modifications via DOT1L and other related molecules by MLL fusion proteins lead to leukemogenetic gene expression, thus to overt leukemia. In order to overcome this dismal subtype of ALL, novel targeted therapy based on leukemia biology is urgently needed. Due to the extreme rarity of the disease, collaboration between the study groups in Europe (Interfant), North America (Children's Oncology Group), and Japan (Japanese Pediatric Leukemia/Lymphoma Study Group) is under way.
Collapse
Affiliation(s)
- Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
18
|
Dreyer ZE, Hilden JM, Jones TL, Devidas M, Winick NJ, Willman CL, Harvey RC, Chen IM, Behm FG, Pullen J, Wood BL, Carroll AJ, Heerema NA, Felix CA, Robinson B, Reaman GH, Salzer WL, Hunger SP, Carroll WL, Camitta BM. Intensified chemotherapy without SCT in infant ALL: results from COG P9407 (Cohort 3). Pediatr Blood Cancer 2015; 62:419-26. [PMID: 25399948 PMCID: PMC5145261 DOI: 10.1002/pbc.25322] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 09/23/2014] [Indexed: 11/12/2022]
Abstract
BACKGROUND Infants with acute lymphoblastic leukemia (ALL) present with aggressive disease and a poor prognosis. Early relapse within 6-9 months of diagnosis is common. Approximately 75% of infants have MLL-rearranged (MLL-R) ALL with event free survival (EFS) ranging from 20% to 30%. Children's Oncology Group (COG) P9407 used shortened (46 weeks), intensified therapy to address early relapse and poor EFS. PROCEDURE P9407 therapy was modified three times for induction toxicity resulting in three cohorts of therapy. One hundred forty-seven infants were enrolled in the third cohort. RESULTS We report an overall 5-year EFS and OS of 42.3 ± 6% and 52.9 ± 6.5% respectively. Poor prognostic factors included age ≤90 days at diagnosis, MLL-R ALL and white cell count ≥50,000/μl. For infants ≤90 days of age, the 5-year EFS was 15.5 ± 10.1% and 48.5 ± 6.7% for those >90 days (P < 0.0001). Among infants >90 days of age, 5-year EFS rates were 43.8 ± 8% for MLL-R versus 69.1 ± 13.6% for MLL-germline ALL (P < 0.0001). CONCLUSIONS Age ≤90 days at diagnosis was the most important prognostic factor. Despite shortened therapy with early intensification, EFS remained less than 50% overall in MLL-R ALL.
Collapse
Affiliation(s)
- ZoAnn E. Dreyer
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX
| | - Joanne M. Hilden
- University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO
| | - Tamekia L. Jones
- Department of Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, FL
| | - Meenakshi Devidas
- Department of Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, FL
| | - Naomi J. Winick
- Pediatric Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Cheryl L. Willman
- Department of Pathology and UMN Cancer Center, University of New Mexico Health Services, Albuquerque, NM
| | - Richard C. Harvey
- Department of Pathology and UMN Cancer Center, University of New Mexico Health Services, Albuquerque, NM
| | - I-Ming Chen
- Department of Pathology and UMN Cancer Center, University of New Mexico Health Services, Albuquerque, NM
| | | | - Jeanette Pullen
- Pediatric Hematology/Oncology, University of Mississippi Medical Center, Jackson, MS
| | - Brent L. Wood
- Laboratory Medicine, Seattle Children’s Hospital, Seattle, WA
| | - 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
| | - Carolyn A. Felix
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Blaine Robinson
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Gregory H. Reaman
- Hematology-Oncology, Children’s National Medical Center, Washington, DC
| | - Wanda L. Salzer
- Mark O Hatfield-Warren Grant Magnuson Clinical Canter, Pediatric Oncology, Silver Spring, MD
| | - Stephen P. Hunger
- University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO
| | - William L. Carroll
- NYU Cancer Institute, New York University Langone Medical Center, New York, NY
| | - Bruce M. Camitta
- Midwest Center for Cancer and Blood Disorders, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
19
|
Salzer WL, Jones TL, Devidas M, Dreyer ZE, Gore L, Winick NJ, Sung L, Raetz E, Loh ML, Wang CY, De Lorenzo P, Valsecchi MG, Pieters R, Carroll WL, Hunger SP, Hilden JM, Brown P. Decreased induction morbidity and mortality following modification to induction therapy in infants with acute lymphoblastic leukemia enrolled on AALL0631: a report from the Children's Oncology Group. Pediatr Blood Cancer 2015; 62:414-8. [PMID: 25407157 PMCID: PMC4480675 DOI: 10.1002/pbc.25311] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/22/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Infants with acute lymphoblastic leukemia (ALL) have a poor prognosis. Intensification of therapy has resulted in fewer relapses but increased early deaths, resulting in failure to improve survival. PROCEDURE AALL0631 is a Phase 3 study for infants (<366 days of age) with newly diagnosed ALL. Induction initially (Cohort 1) consisted of 3 weeks of therapy based on COG P9407. Due to excessive early mortality, induction was amended to a less intensive 5 weeks of therapy based on Interfant-99. Additionally, enhanced supportive care guidelines were incorporated with hospitalization during induction until evidence of marrow recovery and recommendations for prevention/treatment of infections (Cohort 2). RESULTS Induction mortality was significantly lower for patients in Cohort 2 (2/123, 1.6%) versus Cohort 1 (4/26, 15.4%; P = 0.009). All induction deaths were infection related except one due to progressive disease (Cohort 2). Sterile site infections were lower for patients in Cohort 2 (24/123, 19.5%) versus Cohort 1 (15/26, 57.7%; P = 0.0002), with a significantly lower rate of Gram positive infections during induction for patients in Cohort 2, P = 0.0002. No clinically significant differences in grades 3-5 non-infectious toxicities were observed between the two cohorts. Higher complete response rates were observed at end induction intensification for Cohort 2 (week 9, 94/100, 94%) versus Cohort 1 (week 7, 17/25, 68%; P = 0.0.0012). CONCLUSION De-intensification of induction therapy and enhanced supportive care guidelines significantly decreased induction mortality and sterile site infections, without decreasing complete remission rates.
Collapse
Affiliation(s)
- Wanda L. Salzer
- U.S. Army Medical Research and Materiel Command, Fort Detrick, MD
| | - Tamekia L. Jones
- Department of Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, FL
| | - Meenakshi Devidas
- Department of Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, FL
| | - ZoAnn E. Dreyer
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX
| | - Lia Gore
- Childrens Hospital Colorado and the University of Colorado School of Medicine, Aurora, CO
| | - Naomi J. Winick
- Division of Pediatric Hematology/Oncology, University of Texas Southwestern School of Medicine, Dallas, TX
| | - Lillian Sung
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elizabeth Raetz
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah
| | - Mignon L. Loh
- Department of Pediatrics, University of California at San Francisco, CA
| | - Cindy Y. Wang
- Children’s Oncology Group Statistics and Data Center University of Florida, Gainesville, FL
| | - Paola De Lorenzo
- Interfant Trial Data Center, Pediatric Clinic, University of Milano-Bicocca, Monza, Italy,Department of Health Sciences, University of Milano Bicocca, Monza, Italy
| | | | - Rob Pieters
- Princess Maxima Center for Pediatric Oncology, Utrecht, Pediatric Oncology, Erasmus MC - Sophia Children’s Hospital, Rotterdam, Netherlands
| | | | - Stephen P Hunger
- Childrens Hospital Colorado and the University of Colorado School of Medicine, Aurora, CO
| | - Joanne M. Hilden
- Childrens Hospital Colorado and the University of Colorado School of Medicine, Aurora, CO
| | | |
Collapse
|
20
|
Early use of allogeneic hematopoietic stem cell transplantation for infants with MLL gene-rearrangement-positive acute lymphoblastic leukemia. Leukemia 2014; 29:290-6. [DOI: 10.1038/leu.2014.172] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/14/2014] [Accepted: 05/28/2014] [Indexed: 11/08/2022]
|
21
|
Kotecha RS, Gottardo NG, Kees UR, Cole CH. The evolution of clinical trials for infant acute lymphoblastic leukemia. Blood Cancer J 2014; 4:e200. [PMID: 24727996 PMCID: PMC4003413 DOI: 10.1038/bcj.2014.17] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) in infants has a significantly inferior outcome in comparison with older children. Despite initial improvements in survival of infants with ALL since establishment of the first pediatric cooperative group ALL trials, the poor outcome has plateaued in recent years. Historically, infants were treated on risk-adapted childhood ALL protocols. These studies were pivotal in identifying the need for infant-specific protocols, delineating prognostic categories and the requirement for a more unified approach between study groups to overcome limitations in accrual because of low incidence. This subsequently led to the development of collaborative infant-specific studies. Landmark outcomes have included the elimination of cranial radiotherapy following the discovery of intrathecal and high-dose systemic therapy as a superior and effective treatment strategy for central nervous system disease prophylaxis, with improved neurodevelopmental outcome. Universal prospective identification of independent adverse prognostic factors, including presence of a mixed lineage leukemia rearrangement and young age, has established the basis for risk stratification within current trials. The infant-specific trials have defined limits to which conventional chemotherapeutic agents can be intensified to optimize the balance between treatment efficacy and toxicity. Despite variations in therapeutic intensity, there has been no recent improvement in survival due to the equilibrium between relapse and toxicity. Ultimately, to improve the outcome for infants with ALL, key areas still to be addressed include identification and adaptation of novel prognostic markers and innovative therapies, establishing the role of hematopoietic stem cell transplantation in first complete remission, treatment strategies for relapsed/refractory disease and monitoring and timely intervention of late effects in survivors. This would be best achieved through a single unified international trial.
Collapse
Affiliation(s)
- R S Kotecha
- 1] Department of Haematology and Oncology, Princess Margaret Hospital for Children, Perth, Western Australia, Australia [2] Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia [3] School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | - N G Gottardo
- 1] Department of Haematology and Oncology, Princess Margaret Hospital for Children, Perth, Western Australia, Australia [2] Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia [3] School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | - U R Kees
- Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia
| | - C H Cole
- 1] Department of Haematology and Oncology, Princess Margaret Hospital for Children, Perth, Western Australia, Australia [2] Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia [3] School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
22
|
Cohen IJ, Wolff JE. How long can folinic acid rescue be delayed after high-dose methotrexate without toxicity? Pediatr Blood Cancer 2014; 61:7-10. [PMID: 24038885 DOI: 10.1002/pbc.24770] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/20/2013] [Indexed: 11/11/2022]
Abstract
To determine the optimal time of folinic acid rescue after methotrexate (MTX) treatment in patients with ALL, we selected and evaluated relevant studies that included doses, rescue delay, and side effects. Rescue at 42-48 hours resulted in considerable toxicity, except when low doses of MTX were used (1 g/m(2)) or serum MTX levels remained consistently low at 24, 30, and 36 hours. Rescue started at 30-36 hours was safe. In the absence of evidence that later rescue improves prognosis, we suggest that folinic acid rescue (105 mg/m(2)) be started no later than 36 hours from the start of MTX (5-6 g/m(2)).
Collapse
Affiliation(s)
- Ian Joseph Cohen
- The Raphael Recanati Genetic Institute, Rabin Medical Center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
23
|
Cytogenetics and outcome of infants with acute lymphoblastic leukemia and absence of MLL rearrangements. Leukemia 2013; 28:428-30. [PMID: 24072099 DOI: 10.1038/leu.2013.280] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
24
|
Potent obatoclax cytotoxicity and activation of triple death mode killing across infant acute lymphoblastic leukemia. Blood 2013; 121:2689-703. [PMID: 23393050 DOI: 10.1182/blood-2012-04-425033] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Survival in infants younger than 1 year who have acute lymphoblastic leukemia (ALL) is inferior whether MLL is rearranged (R) or germline (G). MLL translocations confer chemotherapy resistance, and infants experience excess complications. We characterized in vitro sensitivity to the pan-antiapoptotic BCL-2 family inhibitor obatoclax mesylate in diagnostic leukemia cells from 54 infants with ALL/bilineal acute leukemia because of the role of prosurvival BCL-2 proteins in resistance, their imbalanced expression in infant ALL, and evidence of obatoclax activity with a favorable toxicity profile in early adult leukemia trials. Overall, half maximal effective concentrations (EC50s) were lower than 176 nM (the maximal plasma concentration [Cmax] with recommended adult dose) in 76% of samples, whether in MLL-AF4, MLL-ENL, or other MLL-R or MLL-G subsets, and regardless of patients' poor prognostic features. However, MLL status and partner genes correlated with EC50. Combined approaches including flow cytometry, Western blot, obatoclax treatment with death pathway inhibition, microarray analyses, and/or electron microscopy indicated a unique killing mechanism involving apoptosis, necroptosis, and autophagy in MLL-AF4 ALL cell lines and primary MLL-R and MLL-G infant ALL cells. This in vitro obatoclax activity and its multiple killing mechanisms across molecular cytogenetic subsets provide a rationale to incorporate a similarly acting compound into combination strategies to combat infant ALL.
Collapse
|
25
|
Brown P. Treatment of infant leukemias: challenge and promise. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2013; 2013:596-600. [PMID: 24319237 PMCID: PMC4729208 DOI: 10.1182/asheducation-2013.1.596] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Leukemia in infants is rare but generates tremendous interest due to its aggressive clinical presentation in a uniquely vulnerable host, its poor response to current therapies, and its unique biology that is increasingly pointing the way toward novel therapeutic approaches. This review highlights the key clinical, pathologic, and epidemiologic features of infant leukemia, including the high frequency of mixed lineage leukemia (MLL) gene rearrangements. The state of the art with regard to current approaches to risk stratified treatment of infant leukemia in the major international cooperative groups is discussed. Finally, exciting recent discoveries elucidating the molecular biology of infant leukemia are reviewed and novel targeted therapeutic strategies, including FLT3 inhibition and modulation of aberrant epigenetic programs, are suggested.
Collapse
Affiliation(s)
- Patrick Brown
- Departments of Oncology and Pediatrics, Johns Hopkins University School of Medicine
| |
Collapse
|