51
|
Yoshimura S, Panetta JC, Hu J, Li L, Gocho Y, Du G, Umezawa A, Karol SE, Pui CH, Mullighan CG, Konopleva M, Stock W, Teachey DT, Jain N, Yang JJ. Preclinical pharmacokinetic and pharmacodynamic evaluation of dasatinib and ponatinib for the treatment of T-cell acute lymphoblastic leukemia. Leukemia 2023; 37:1194-1203. [PMID: 37076694 PMCID: PMC10347458 DOI: 10.1038/s41375-023-01900-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023]
Abstract
LCK is a novel therapeutic target in ~40% of T-cell acute lymphoblastic leukemia (T-ALL), and dasatinib and ponatinib can act as LCK inhibitors with therapeutic effects. We herein report a comprehensive preclinical pharmacokinetic and pharmacodynamic evaluation of dasatinib and ponatinib in LCK-activated T-ALL. In 51 human T-ALL cases, these two drugs showed similar patterns of cytotoxic activity, with ponatinib being slightly more potent. Given orally in mice, ponatinib was associated with slower clearance with a longer Tmax and higher AUC0-24 h, although maximum pLCK inhibition was comparable between the two drugs. After establishing the exposure-to-response models, we simulated the steady-state pLCK inhibitory effects of each drug at currently approved dosages in humans: dasatinib at 140 mg and ponatinib at 45 mg once daily are both sufficient to achieve >50% pLCK inhibition for 13.0 and 13.9 h/day, respectively, comparable to pharmacodynamic profiles of these agents in BCR::ABL1 leukemias. Moreover, we developed a dasatinib-resistant T-ALL cell line model with LCK T316I mutation, in which ponatinib retained partial activity against LCK. In conclusion, we described the pharmacokinetic and pharmacodynamic profiles of dasatinib and ponatinib as LCK inhibitors in T-ALL, providing critical data for the development of human trials of these agents.
Collapse
Affiliation(s)
- Satoshi Yoshimura
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Advanced Pediatric Medicine, Tohoku University School of Medicine, Tokyo, Japan
| | - John C Panetta
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jianzhong Hu
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
- Amgen, 1 Amgen Center Drive, Thousand Oaks, CA, USA
| | - Lie Li
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yoshihiro Gocho
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Guoqing Du
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Akihiro Umezawa
- Department of Advanced Pediatric Medicine, Tohoku University School of Medicine, Tokyo, Japan
| | - Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marina Konopleva
- Department of Oncology and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wendy Stock
- Department of Medicine Section of Hematology-Oncology, University of Chicago, Chicago, IL, USA
| | - David T Teachey
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
| | - Nitin Jain
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
52
|
Angot L, Schneider P, Vannier JP, Abdoul-Azize S. Beyond Corticoresistance, A Paradoxical Corticosensitivity Induced by Corticosteroid Therapy in Pediatric Acute Lymphoblastic Leukemias. Cancers (Basel) 2023; 15:2812. [PMID: 37345151 DOI: 10.3390/cancers15102812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Known as a key effector in relapse of acute lymphoblastic leukemia (ALL), resistance to drug-induced apoptosis, is tightly considered one of the main prognostic factors for the disease. ALL cells are constantly developing cellular strategies to survive and resist therapeutic drugs. Glucocorticoids (GCs) are one of the most important agents used in the treatment of ALL due to their ability to induce cell death. The mechanisms of GC resistance of ALL cells are largely unknown and intense research is currently focused on this topic. Such resistance can involve different cellular and molecular mechanisms, including the modulation of signaling pathways involved in the regulation of proliferation, apoptosis, autophagy, metabolism, epigenetic modifications and tumor suppressors. Recently, several studies point to the paradoxical role of GCs in many survival processes that may lead to therapy-induced resistance in ALL cells, which we called "paradoxical corticosensitivity". In this review, we aim to summarize all findings on cell survival pathways paradoxically activated by GCs with an emphasis on previous and current knowledge on gene expression and signaling pathways.
Collapse
Affiliation(s)
- Laure Angot
- Normandie University, UNIROUEN, IRIB, Inserm, U1234, 76183 Rouen, France
| | - Pascale Schneider
- Normandie University, UNIROUEN, IRIB, Inserm, U1234, 76183 Rouen, France
- Department of Pediatric Immuno-Hemato-Oncology, Rouen University Hospital, 76038 Rouen, France
| | | | | |
Collapse
|
53
|
Sato A, Hatta Y, Imai C, Oshima K, Okamoto Y, Deguchi T, Hashii Y, Fukushima T, Hori T, Kiyokawa N, Kato M, Saito S, Anami K, Sakamoto T, Kosaka Y, Suenobu S, Imamura T, Kada A, Saito AM, Manabe A, Kiyoi H, Matsumura I, Koh K, Watanabe A, Miyazaki Y, Horibe K. Nelarabine, intensive L-asparaginase, and protracted intrathecal therapy for newly diagnosed T-cell acute lymphoblastic leukaemia in children and young adults (ALL-T11): a nationwide, multicenter, phase 2 trial including randomisation in the very high-risk group. Lancet Haematol 2023:S2352-3026(23)00072-8. [PMID: 37167992 DOI: 10.1016/s2352-3026(23)00072-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND T-cell acute lymphoblastic leukaemia has distinct biological characteristics and a poorer prognosis than B-cell precursor acute lymphoblastic leukaemia. This trial aimed to reduce the rate of radiation and haematopoietic stem-cell transplantation (HSCT) while improving outcomes by adding nelarabine, intensified L-asparaginase, and protracted intrathecal therapy in the Berlin-Frankfurt-Münster (BFM)-type treatment. METHODS In this nationwide, multicenter, phase 2 trial, we enrolled patients with newly diagnosed T-cell acute lymphoblastic leukaemia (age <25 years at diagnosis) conducted by Japan Children's Cancer Group and Japan Adult Leukemia Study Group. Patients were stratified into standard-risk, high-risk, and very-high-risk groups according to prednisolone response, CNS status, and end-of-consolidation minimal residual disease. We used the Associazione Italiana di Ematologia Oncologia Pediatrica (AIEOP)-BFM-ALL 2000-backbone chemotherapy. Nelarabine (650 mg/m2 per day for 5 days) was given to high-risk and very high-risk patients. All patients received, until the measurement of end-of-consolidation minimal residual disease, an identical therapy schedule, which included the prednisolone pre-phase remission induction therapy with dexamethasone (10 mg/m2 per day, for 3 weeks [for patients <10 years] or for 2 weeks including a 7-day off interval [for patients ≥10 years]) instead of prednisolone, and consolidation therapy added with Escherichia coli-derived L-asparaginase. On the basis of the stratification, patients received different intensities of treatment; L-asparaginase-intensified standard BFM-type therapy for standard risk and nelarabine-added high risk BFM-type therapy for high risk. In the very high-risk group, patients were randomly assigned (1:1) to group A (BFM-based block therapy) and group B (another block therapy, including high-dose dexamethasone) stratified by hospital, age (≥18 years or <18 years), and end-of-induction bone marrow blast percentage of M1 (<5%) or M2 (≥5%, <25%)+M3 (≥25%). Cranial radiotherapy was limited to patients with overt CNS disease at diagnosis (CNS3; >5 white blood cells per μL with blasts) and patients with no evidence of CNS disease received protracted triple intrathecal therapy. Only very high-risk patients were scheduled to receive HSCT. The primary endpoint was 3-year event-free survival for the entire cohort and the proportion of patients with disappearance of minimal residual disease between randomly assigned groups A and B in the very high-risk group. Secondary endpoints were overall survival, remission induction rate, and occurrence of adverse events. 3 years after the completion of patient accrual, a primary efficacy analysis was performed in the full analysis set and the per-protocol set. This study is registered with the Japan Registry of Clinical Trials, jRCTs041180145. FINDINGS Between Dec 1, 2011, and Nov 30, 2017, of 349 eligible patients (median age 9 years [IQR 6-13]), 238 (68%) were male, and 28 (8%) patients had CNS3 status. 168 (48%) patients were stratified as standard risk, 103 (30%) as high risk, 39 (11%) as very high risk, and 39 (11%) as no risk (patients who had off protocol treatment before risk assessment. The composite complete remission (complete remission plus complete remission in suppression) rate after remission induction therapy was 89% (298 of 335 patients). HSCT was performed in 35 (10%) of 333 patients. With a median follow-up of 5·2 years (IQR 3·6-6·7), 3-year event-free survival was 86·4% (95% CI 82·3-89·7%) and 3-year overall survival was 91·3% (87·7-93·8%). The proportion of minimal residual disease disappearance was 0·86 (12 of 14 patients; 95% CI 0·57-0·98) in group A and 0·50 (6 of 12 patients, 0·21-0·79) in group B. Grade 3 peripheral motor neuropathy was seen in 11 (3%) of 349 patients and sensory neuropathy was seen in 6 (2%) patients. The most common grade 3 or worse adverse event was febrile neutropenia (294 [84%] of 349 patients). Treatment-related death occurred in three patients due to sepsis, gastric perforation, or intracranial haemorrhage during remission induction. INTERPRETATION The ALL-T11 protocol produced encouraging outcomes with acceptable toxicities despite limited cranial radiotherapy and HSCT use. FUNDING Ministry of Health, Labor and Welfare of Japan, and Japan Agency for Medical Research and Development. TRANSLATION For the Japanese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Hematology and Oncology, Miyagi Children's Hospital, Sendai, Japan.
| | - Yoshihiro Hatta
- Department of Hematology and Rheumatology, Nihon University Itabashi Hospital, Tokyo, Japan
| | - Chihaya Imai
- Department of Pediatrics, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Koichi Oshima
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Yasuhiro Okamoto
- Department of Pediatrics, Kagoshima University Hospital, Kagoshima, Japan
| | - Takao Deguchi
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University, Osaka, Japan
| | - Takashi Fukushima
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Japan
| | - Toshinori Hori
- Department of Pediatrics, Aichi Medical University Hospital, Aich, Japan
| | - Nobutaka Kiyokawa
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Motohiro Kato
- Department of Pediatrics, The University of Tokyo, Tokyo, Japan
| | - Shoji Saito
- Department of Pediatrics, Shinshu University Hospital, Matsumoto, Japan
| | - Kenichi Anami
- Department of Medical Oncology, Hematology, and Infectious Diseases, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Tatsuhiro Sakamoto
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology/Oncology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Souichi Suenobu
- Department of Pediatrics, Oita University Hospital, Oita, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, University Hospital Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akiko Kada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University Hospital, Sapporo, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Arata Watanabe
- Department of Pediatrics, Nakadori General Hospital, Akita, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Keizo Horibe
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
54
|
Öfverholm I, Rezayee F, Heyman M, Harila A, Arvidsson L, Schmiegelow K, Norén-Nyström U, Barbany G. The prognostic impact of IKZF1 deletions and UKALL genetic classifiers in paediatric B-cell precursor acute lymphoblastic leukaemia treated according to NOPHO 2008 protocols. Br J Haematol 2023. [PMID: 37156607 DOI: 10.1111/bjh.18852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
We investigated 390 paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) patients treated according to NOPHO ALL 2008, regarding copy number alterations (CNA) of eight loci associated with adverse prognosis, including IKZF1. The impact on outcome was investigated for each locus individually, combined as CNA profiles and together with cytogenetic information. The presence of IKZF1 deletion or a poor-risk CNA profile was associated with poor outcome in the whole cohort. In the standard-risk group, IKZF1-deleted cases had an inferior probability of relapse-free survival (pRFS) (p ≤ 0.001) and overall survival (pOS) (p ≤ 0.001). Additionally, among B-other patients, IKZF1 deletion correlated with poor pRFS (60% vs. 90%) and pOS (65% vs. 89%). Both IKZF1 deletion and a poor-risk CNA profile were independent factors for relapse and death in multivariable analyses adjusting for known risk factors including measurable residual disease. Our data indicate that BCP-ALL patients with high-risk CNA or IKZF1 deletion have worse prognosis despite otherwise low-risk features. Conversely, patients with both a good CNA and cytogenetic profile had a superior relapse-free (p ≤ 0.001) and overall survival (p ≤ 0.001) in the cohort, across all risk groups. Taken together, our findings highlight the potential of CNA assessment to refine stratification in ALL.
Collapse
Affiliation(s)
- Ingegerd Öfverholm
- Department of Molecular Medicine and Surgery and Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Fatemah Rezayee
- Department of Molecular Medicine and Surgery and Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Mats Heyman
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Arja Harila
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Linda Arvidsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kjeld Schmiegelow
- Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
- Faculty of Medicine, Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Gisela Barbany
- Department of Molecular Medicine and Surgery and Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
55
|
Campbell M, Kiss C, Zimmermann M, Riccheri C, Kowalczyk J, Felice MS, Kuzmanovic M, Kovacs G, Kosmidis H, Gonzalez A, Bilic E, Castillo L, Kolenova A, Jazbec J, Popa A, Konstantinov D, Kappelmayer J, Szczepanski T, Dworzak M, Buldini B, Gaipa G, Marinov N, Rossi J, Nagy A, Gaspar I, Stary J, Schrappe M. Childhood Acute Lymphoblastic Leukemia: Results of the Randomized Acute Lymphoblastic Leukemia Intercontinental-Berlin-Frankfurt-Münster 2009 Trial. J Clin Oncol 2023:JCO2201760. [PMID: 37141547 DOI: 10.1200/jco.22.01760] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
PURPOSE The International Berlin-Frankfurt-Münster (BFM) study group conducted a study on pediatric acute lymphoblastic leukemia (ALL). Minimal residual disease (MRD) was assessed using flow cytometry (FCM), and the impact of early intensification and methotrexate (MTX) dose on survival was evaluated. PATIENTS AND METHODS We included 6,187 patients younger than 19 years. MRD by FCM refined the risk group definition previously used in the ALL intercontinental-BFM 2002 study on the basis of age, WBC count, unfavorable genetic aberrations, and treatment response measured morphologically. Patients at intermediate risk (IR) and high risk (HR) were randomly assigned to protocol augmented protocol I phase B (IB) versus IB regimen. MTX doses of 2 versus 5 g/m2 every 2 weeks, four times, were evaluated in precursor B-cell-ALL (pcB-ALL) IR. RESULTS The 5-year event-free survival (EFS ± SE) and overall survival (OS ± SE) rates were 75.2% ± 0.6% and 82.6% ± 0.5%, respectively. Their values in risk groups were standard risk (n = 624), 90.7% ± 1.4% and 94.7% ± 1.1%; IR (n = 4,111), 77.9% ± 0.7% and 85.7% ± 0.6%; and HR (n = 1,452), 60.8% ± 1.5% and 68.4% ± 1.4%, respectively. MRD by FCM was available in 82.6% of cases. The 5-year EFS rates in patients randomly assigned to protocol IB (n = 1,669) and augmented IB (n = 1,620) were 73.6% ± 1.2% and 72.8% ± 1.2%, respectively (P = .55), while those in patients receiving MTX doses of 2 g/m2 (n = 1,056) and MTX 5 g/m2 (n = 1,027) were 78.8% ± 1.4% and 78.9% ± 1.4%, respectively (P = .84). CONCLUSION The MRDs were successfully assessed using FCM. An MTX dose of 2 g/m2 was effective in preventing relapse in non-HR pcB-ALL. Augmented IB showed no advantages over the standard IB.
Collapse
Affiliation(s)
- Myriam Campbell
- Department of Pediatric Hematology and Oncology, Hospital Roberto del Rio, Universidad de Chile, Chilean National Pediatric Oncology Group, PINDA, Santiago, Chile
| | - Csongor Kiss
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Martin Zimmermann
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Cecilia Riccheri
- Argentine Group for the Treatment of Acute Leukemia, GATLA, Buenos Aires, Argentina
| | - Jerzy Kowalczyk
- Department of Pediatric, Hematology, Oncology, and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Maria S Felice
- Hematology and Oncology Department, Hospital de Pediatría Prof. Dr Juan P. Garrahan, SAHOP, Buenos Aires, Argentina
| | - Milos Kuzmanovic
- Mother and Child Health Care Institute of Serbia "Dr Vukan Cupic", Faculty of Medicine, Belgrade, Serbia
| | - Gabor Kovacs
- 2nd Department of Pediatrics Semmelweis University, Budapest, Hungary
| | - Helen Kosmidis
- Pediatric and Adolescent Oncology Clinic, Children's Hospital MITERA, Athens, Greece
| | | | - Ernest Bilic
- School of Medicine Division of Pediatric Hematology and Oncology, University Hospital Center, University of Zagreb, Zagreb, Croatia
| | - Luis Castillo
- Pediatric Hemato-Oncology Department, Hospital Pereira Rossell, Pérez Scremini Foundation, Montevideo, Uruguay
| | - Alexandra Kolenova
- Department of Pediatric Hematology and Oncology, National Institute of Children's Diseases and Medical School, Comenius University, Bratislava, Slovakia
| | - Janez Jazbec
- University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Alexander Popa
- Pediatric Oncology and Hematology Research Institute of N.N.Blokhin National Cancer Research Center, Center, Moscow, Russia
| | - Dobrin Konstantinov
- Pediatric Hematology & Oncology Department, University Hospital "Tsaritsa Johanna-ISUL", Sofia, Bulgaria
| | - Janos Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia, Katowice, Poland
| | - Michael Dworzak
- St Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Barbara Buldini
- Mother and Child's Health Department, Division of Pediatric Hematology, Oncology and Stem Cell Transplant, University of Padova, Padova, Veneto, Italy
| | - Giuseppe Gaipa
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Neda Marinov
- Chilean National Pediatric Oncology Group, PINDA, Hospital Roberto del Rio/Universidad de Chile, Santiago, Chile
- Hospital del Salvador, Universidad de Chile, Santiago, Chile
| | - Jorge Rossi
- Immunology and Rheumatology Department, Hospital de Pediatría Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina
| | - Attila Nagy
- Department of Interventional Epidemiology, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Imre Gaspar
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jan Stary
- Department of Pediatric Hematology and Oncology Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Martin Schrappe
- Department of Pediatric and Adolescent Medicine, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
56
|
Ribeiro RC, Conter V. Optimizing Pediatric Leukemia Care in Countries With Limited Resources. J Clin Oncol 2023:JCO2300451. [PMID: 37141548 DOI: 10.1200/jco.23.00451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Affiliation(s)
- Raul C Ribeiro
- Division of Leukemia and Lymphoma, Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| | - Valentino Conter
- Pediatric Hemato-Oncology, Fondazione MBBM, University Milano Bicocca, Ospedale San Gerardo, Monza, Italy
| |
Collapse
|
57
|
Muggeo P, Grassi M, D'Ascanio V, Brescia V, Fontana A, Piacente L, Di Serio F, Giordano P, Faienza MF, Santoro N. Bone Remodeling Markers in Children with Acute Lymphoblastic Leukemia after Intensive Chemotherapy: The Screenshot of a Biochemical Signature. Cancers (Basel) 2023; 15:cancers15092554. [PMID: 37174020 PMCID: PMC10177249 DOI: 10.3390/cancers15092554] [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: 03/08/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
PURPOSE to investigate the effects of intensive chemotherapy and glucocorticoid (GC) treatment on bone remodeling markers in children with acute lymphoblastic leukemia (ALL). METHODS A cross-sectional study was carried out in 39 ALL children (aged 7.64 ± 4.47) and 49 controls (aged 8.7 ± 4.7 years). Osteoprotegerin (OPG), receptor activator of NF-κB ligand (RANKL), osteocalcin (OC), C-terminal telopeptide of type I collagen (CTX), bone alkaline phosphatase (bALP), tartrate-resistant acid phosphatase 5b (TRACP5b), procollagen type I N-terminal propeptide (P1NP), Dickkopf-1 (DKK-1), and sclerostin were assessed. Statistical analysis was conducted using the principal component analysis (PCA) to study patterns of associations in bone markers. RESULTS ALL patients showed significantly higher OPG, RANKL, OC, CTX, and TRACP5b than the controls (p ≤ 0.02). Considering ALL group, we found a strong positive correlation among OC, TRACP5b, P1NP, CTX, and PTH (r = 0.43-0.69; p < 0.001); between CTX and P1NP (r = 0.5; p = 0.001); and between P1NP and TRAcP (r = 0.63; p < 0.001). The PCA revealed OC, CTX, and P1NP as the main markers explaining the variability of the ALL cohort. CONCLUSIONS Children with ALL showed a signature of bone resorption. The assessment of bone biomarkers could help identify ALL individuals who are most at risk of developing bone damage and who need preventive interventions.
Collapse
Affiliation(s)
- Paola Muggeo
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinic, 70124 Bari, Italy
| | - Massimo Grassi
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinic, 70124 Bari, Italy
| | - Vito D'Ascanio
- Institute of Sciences of Food Production (ISPA), Italian National Research Council (CNR), 70126 Bari, Italy
| | - Vincenzo Brescia
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy
| | - Antonietta Fontana
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy
| | - Laura Piacente
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University "A. Moro", 70124 Bari, Italy
| | - Francesca Di Serio
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, 70124 Bari, Italy
| | - Paola Giordano
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University "A. Moro", 70124 Bari, Italy
| | - Nicola Santoro
- Department of Pediatric Oncology and Hematology, University Hospital of Policlinic, 70124 Bari, Italy
| |
Collapse
|
58
|
Ampatzidou M, Papadhimitriou SI, Paisiou A, Paterakis G, Tzanoudaki M, Papadakis V, Florentin L, Polychronopoulou S. The Prognostic Effect of CDKN2A/2B Gene Deletions in Pediatric Acute Lymphoblastic Leukemia (ALL): Independent Prognostic Significance in BFM-Based Protocols. Diagnostics (Basel) 2023; 13:diagnostics13091589. [PMID: 37174980 PMCID: PMC10178600 DOI: 10.3390/diagnostics13091589] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
One of the most frequent genes affected in pediatric ALL is the CDKN2A/2B gene, acting as a secondary cooperating event and playing an important role in cell-cycle regulation and chemosensitivity. Despite its inclusion in combined CNA (copy-number alterations) classifiers, like the IKZF1plus entity and the UKALL CNA profile, the prognostic impact of the individual gene deletions outside the context of a combined CNA evaluation remains controversial. Addressing the CDKN2A/2B deletions' additive prognostic effect in current risk-stratification algorithms, we present a retrospective study of a Greek pediatric ALL cohort comprising 247 patients studied over a 24-year period (2000-2023). Herein, we provide insight regarding the correlation with disease features, MRD clearance, and independent prognostic significance for this ALL cohort treated with contemporary BFM-based treatment protocols. Within an extended follow-up time of 135 months, the presence of the CDKN2A/2B deletions (biallelic or monoallelic) was associated with inferior EFS rates (65.1% compared to 91.8% for the gene non-deleted subgroup, p < 0.001), with the relapse rate accounting for 22.2% and 5.9%, respectively (p < 0.001). The presence of the biallelic deletion was associated with the worst outcomes (EFS 57.2% vs. 89.6% in the case of any other status, monoallelic or non-deleted, p < 0.001). Survival differences were demonstrated for B-ALL cases (EFS 65.3% vs. 93.6% for the non-deleted B-ALL subgroup, p < 0.001), but the prognostic effect was not statistically significant within the T-ALL cohort (EFS 64.3 vs. 69.2, p = 0.947). The presence of the CDKN2A/2B deletions clearly correlated with inferior outcomes within all protocol-defined risk groups (standard risk (SR): EFS 66.7% vs. 100%, p < 0.001, intermediate risk (IR): EFS 77.1% vs. 97.9%, p < 0.001, high risk (HR): EFS 42.1% vs. 70.5% p < 0.001 for deleted vs non-deleted cases in each patient risk group); additionally, in this study, the presence of the deletion differentiated prognosis within both MRD-positive and -negative subgroups on days 15 and 33 of induction. In multivariate analysis, the presence of the CDKN2A/2B deletions was the most important prognostic factor for relapse and overall survival, yielding a hazard ratio of 5.2 (95% confidence interval: 2.59-10.41, p < 0.001) and 5.96 (95% confidence interval: 2.97-11.95, p < 0.001), respectively, designating the alteration's independent prognostic significance in the context of modern risk stratification. The results of our study demonstrate that the presence of the CDKN2A/2B deletions can further stratify all existing risk groups, identifying patient subgroups with different outcomes. The above biallelic deletions could be incorporated into future risk-stratification algorithms, refining MRD-based stratification. In the era of targeted therapies, future prospective controlled clinical trials will further explore the possible use of cyclin-dependent kinase inhibitors (CDKIs) in CDKN2A/2B-affected ALL pediatric subgroups.
Collapse
Affiliation(s)
- Mirella Ampatzidou
- Department of Pediatric Hematology-Oncology (TAO), "Aghia Sophia" Children's Hospital, 11527 Athens, Greece
| | - Stefanos I Papadhimitriou
- Laboratory of Hematology, Unit of Molecular Cytogenetics, "G. Gennimatas" General Hospital, 11527 Athens, Greece
| | - Anna Paisiou
- Bone Marrow Transplantation Unit, "Aghia Sophia" Children's Hospital, 11527 Athens, Greece
| | - Georgios Paterakis
- Laboratory of Flow Cytometry, Department of Immunology, "G. Gennimatas" General Hospital, 11527 Athens, Greece
| | - Marianna Tzanoudaki
- Department of Immunology, "Aghia Sophia" Children's Hospital, 11527 Athens, Greece
| | - Vassilios Papadakis
- Department of Pediatric Hematology-Oncology (TAO), "Aghia Sophia" Children's Hospital, 11527 Athens, Greece
| | - Lina Florentin
- Alfa Laboratory Diagnostic Center, YGEIA Hospital, 11524 Athens, Greece
| | - Sophia Polychronopoulou
- Department of Pediatric Hematology-Oncology (TAO), "Aghia Sophia" Children's Hospital, 11527 Athens, Greece
| |
Collapse
|
59
|
Imai C, Sato A, Hiwatari M, Shimomura Y, Hori T, Suenobu S, Imamura T, Hara J, Hasegawa D, Takahashi H, Moriya K, Katayama S, Tomizawa D, Moritake H, Taga T, Horibe K, Koh K, Manabe A, Okamoto Y. Outcomes following induction failure in Japanese children with acute lymphoblastic leukemia. Int J Hematol 2023:10.1007/s12185-023-03600-3. [PMID: 37037958 DOI: 10.1007/s12185-023-03600-3] [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: 01/23/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/12/2023]
Abstract
The characteristics and prognosis of Japanese children with acute lymphoblastic leukemia (ALL) who fail to achieve complete remission after remission induction chemotherapy (i.e., experience induction failure) are poorly understood. Therefore, we retrospectively analyzed data of patients enrolled in Japanese clinical trials for newly diagnosed ALL between 1996 and 2009. Among 4956 participants, 89 (1.8%) experienced induction failure. With a 6.0-year median follow-up, the 5-year overall survival rate of the entire cohort was 43.0% ± 5.5%. Survival rates did not differ between patients with B-cell precursor ALL (BCP-ALL) and T-cell ALL (T-ALL). In multivariate analysis, day 15 M3 marrow (bone marrow blast count ≥ 25%) was significantly correlated with poorer survival in the whole or BCP-ALL cohorts. In T-ALL, age < 6 years was significantly associated with poor survival. However, due to the small sample size, this correlation must be further investigated. Most T-ALL and BCR-ABL-positive BCP-ALL patients underwent allogeneic stem cell transplantation (allo-SCT). Survival rates did not differ between BCR-ABL-negative BCP-ALL patients who did and did not undergo allo-SCT, possibly due to the inclusion of lower-risk patients in the latter group. In conclusion, the induction failure rate and survival after diagnosis of induction failure in our study were comparable to previously reported figures.
Collapse
Affiliation(s)
- Chihaya Imai
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-Dori, Chuou-Ku, Niigata City, Niigata, 951-8510, Japan.
| | - Atsushi Sato
- Department of Hematology and Oncology, Miyagi Children's Hospital, Miyagi, Japan
| | - Mitsuteru Hiwatari
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Yasuto Shimomura
- Department of Pediatrics, Aichi Medical University, Aichi, Japan
| | - Toshinori Hori
- Department of Pediatrics, Aichi Medical University, Aichi, Japan
| | | | - Toshihiko Imamura
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junichi Hara
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Hiroyuki Takahashi
- Department of Pediatrics, Toho University Omori Medical Center, Tokyo, Japan
| | - Kunihiko Moriya
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Saori Katayama
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Daisuke Tomizawa
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Hiroshi Moritake
- Department of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Keizo Horibe
- Clinical Research Center, Nagoya Medical Center, Nagoya, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Yasuhiro Okamoto
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
60
|
Mishra V, Jain S, Anand V, Malhotra P, Tejwani N, Kapoor G. Impact of minimal residual disease on relapse in childhood acute lymphoblastic leukemia: Lessons learnt from a tertiary cancer center in India. Pediatr Hematol Oncol 2023; 40:517-528. [PMID: 36930957 DOI: 10.1080/08880018.2023.2186553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 02/13/2023] [Accepted: 02/25/2023] [Indexed: 03/19/2023]
Abstract
Prognostic predictive value of end of induction minimal residual disease (EOI-MRD) is well established in acute lymphoblastic leukemia (ALL). We evaluated the factors likely to affect EOI-MRD positivity (>0.01%) by flow cytometry and relapse in different BFM-95 (Berlin-Frankfurt-Munich) risk groups among children and adolescents. In this retrospective study, data of 223 newly diagnosed patients with ALL was analyzed. Association between demographic and pretreatment characteristics with EOI-MRD was assessed. Risk factors for relapse were analyzed using univariate and multivariate Cox regression. Proportion of the SR (standard risk), MR (moderate risk), and HR (high risk) patients was 18.8%, 60.9%, 20.3%, respectively. Positive EOI-MRD among these risk groups was observed in 11.9%, 18.3%, and 55.5% patients respectively (p value <.01%). MRD positivity was more likely to be associated with older age (>10 years) and BFM-HR patients (p value .0008 and <.0001). Thirty-four (15.2%) patients relapsed in the whole cohort. On univariate analysis, statistically significant factors for RFS (relapse-free survival) included hyperleukocytosis, high-risk cytogenetics, NCI (National Cancer Institute) high risk, poor day-8 prednisolone response, BFM-HR and positive EOI-MRD status. Of all these only EOI-MRD retained its impact by multivariate analysis. Positive EOI-MRD significantly predicted relapse in BFM-MR with 5-year RFS of 88.0% and 68.4% (p value .02). Five-year RFS of EOI-MRD negative and positive groups were 86.4% and 65.5%, respectively (p value .004). EOI-MRD is a powerful tool to predict relapse in children and adolescent with ALL especially in BFM-MR. Application of MRD in HR patients needs to be redefined in conjunction with other variables.
Collapse
Affiliation(s)
- Varsha Mishra
- Department of Pediatric Hematology Oncology, Rajiv Gandhi Cancer Institute, New Delhi, India
| | - Sandeep Jain
- Department of Pediatric Hematology Oncology, Rajiv Gandhi Cancer Institute, New Delhi, India
| | - Vaneet Anand
- Department of Pediatric Hematology Oncology, Rajiv Gandhi Cancer Institute, New Delhi, India
| | - Payal Malhotra
- Department of Pediatric Hematology Oncology, Rajiv Gandhi Cancer Institute, New Delhi, India
| | - Narender Tejwani
- Department of Pathology, Pediatric Oncology, Rajiv Gandhi Cancer Institute, New Delhi, India
| | - Gauri Kapoor
- Department of Pediatric Hematology Oncology, Rajiv Gandhi Cancer Institute, New Delhi, India
| |
Collapse
|
61
|
Attarbaschi A, Möricke A, Harrison CJ, Mann G, Baruchel A, De Moerloose B, Conter V, Devidas M, Elitzur S, Escherich G, Hunger SP, Horibe K, Manabe A, Loh ML, Pieters R, Schmiegelow K, Silverman LB, Stary J, Vora A, Pui CH, Schrappe M, Zimmermann M. Outcomes of Childhood Noninfant Acute Lymphoblastic Leukemia With 11q23/ KMT2A Rearrangements in a Modern Therapy Era: A Retrospective International Study. J Clin Oncol 2023; 41:1404-1422. [PMID: 36256911 PMCID: PMC9995095 DOI: 10.1200/jco.22.01297] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/01/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE We aimed to study prognostic factors and efficacy of allogeneic hematopoietic stem-cell transplantation (allo-HSCT) in first remission of patients with noninfant childhood acute lymphoblastic leukemia (ALL) with 11q23/KMT2A rearrangements treated with chemotherapy regimens between 1995 and 2010. PATIENTS AND METHODS Data were retrospectively retrieved from 629 patients with 11q23/KMT2A-rearranged ALL from 17 members of the Ponte-di-Legno Childhood ALL Working Group. Clinical and biologic characteristics, early response assessed by minimal residual disease at the end of induction (EOI) therapy, and allo-HSCT were analyzed for their impact on outcomes. RESULTS A specific 11q23/KMT2A translocation partner gene was identified in 84.3% of patients, with the most frequent translocations being t(4;11)(q21;q23) (n = 273; 51.5%), t(11;19)(q23;p13.3) (n = 106; 20.0%), t(9;11)(p21_22;q23) (n = 76; 14.3%), t(6;11)(q27;q23) (n = 20; 3.8%), and t(10;11)(p12;q23) (n = 14; 2.6%); 41 patients (7.7%) had less frequently identified translocation partner genes. Patient characteristics and early response varied among subgroups, indicating large biologic heterogeneity and diversity in therapy sensitivity among 11q23/KMT2A-rearranged ALL. The EOI remission rate was 93.2%, and the 5-year event-free survival (EFS) for the entire cohort was 69.1% ± 1.9%, with a range from 41.7% ± 17.3% for patients with t(9;11)-positive T-ALL (n = 9) and 64.8% ± 3.0% for patients with t(4;11)-positive B-ALL (n = 266) to 91.2% ± 4.9% for patients with t(11;19)-positive T-ALL (n = 34). Low EOI minimal residual disease was associated with favorable EFS, and induction failure was particularly predictive of nonresponse to further therapy and relapse and poor EFS. In addition, EFS was not improved by allo-HSCT compared with chemotherapy only in patients with both t(4;11)-positive B-ALL (n = 64 v 51; P = .10) and 11q23/KMT2A-rearranged T-ALL (n = 16 v 10; P = .69). CONCLUSION Compared with historical data, prognosis of patients with noninfant 11q23/KMT2A-rearranged ALL has improved, but allo-HSCT failed to affect outcome. Targeted therapies are needed to reduce relapse and treatment-related mortality rates.
Collapse
Affiliation(s)
- Andishe Attarbaschi
- St Anna Children's Hospital and St Anna Children's Cancer Research Institute, Medical University of Vienna, Vienna, Austria
| | - Anja Möricke
- Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Christine J. Harrison
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | - Georg Mann
- St Anna Children's Hospital and St Anna Children's Cancer Research Institute, Medical University of Vienna, Vienna, Austria
| | - André Baruchel
- Robert Debré University Hospital (APHP), Université Paris Cité, Paris, France
| | | | - Valentino Conter
- University of Milano-Bicocca, MBBM Foundation/ASST Monza, Monza, Italy
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| | - Sarah Elitzur
- Schneider Children's Medical Center, Tel Aviv, Israel
- Tel Aviv University, Tel Aviv, Israel
| | - Gabriele Escherich
- Clinic of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | | | - Keizo Horibe
- National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Atsushi Manabe
- Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Mignon L. Loh
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA
| | - Rob Pieters
- Princess Máxima Centre for Pediatric Oncology, Utrecht, the Netherlands
| | - Kjeld Schmiegelow
- Rigshospitalet and University Hospital Copenhagen, Copenhagen, Denmark
- Faculty of Medicine, Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Jan Stary
- University Hospital Motol and Charles University, Prague, Czech Republic
| | - Ajay Vora
- Great Ormond Street Hospital, London, United Kingdom
| | - Ching-Hon Pui
- St Jude Children's Research Hospital, Memphis, TN
- University of Tennessee, Memphis, TN
| | - Martin Schrappe
- Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | | |
Collapse
|
62
|
Choi JK, Mead PE. Laboratory Aspects of Minimal / Measurable Residual Disease Testing in B-Lymphoblastic Leukemia. Clin Lab Med 2023; 43:115-125. [PMID: 36764804 DOI: 10.1016/j.cll.2022.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Minimal residual disease detection provides critical prognostic predictor of treatment outcome and is the standard of care for B lymphoblastic leukemia. Flow cytometry-based minimal residual disease detection is the most common test modality and has high sensitivity (0.01%) and a rapid turnaround time (24 hours). This article details the leukemia associated immunophenotype analysis approach for flow cytometry-based minimal residual disease detection used at St. Jude Children's Research Hospital and importance of using guide gates and back-gating.
Collapse
Affiliation(s)
- John Kim Choi
- Division of Laboratory Medicine, The University of Alabama at Birmingham, WP P230N, 619 19th Street South, Birmingham, AL 35249-7331, USA.
| | - Paul E Mead
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, D4026G, Mailstop 342, Memphis, TN 38105, USA
| |
Collapse
|
63
|
Gidl A, Füreder A, Benesch M, Dworzak M, Engstler G, Jones N, Kropshofer G, Pötschger U, Poyer F, Tamesberger M, Witt V, Mann G, Attarbaschi A. Incidence and Risk Factors of Venous Thromboembolism in Childhood Acute Lymphoblastic Leukaemia - a Population-Based Analysis of the Austrian Berlin-Frankfurt-Münster (BFM) Study Group. Pediatr Hematol Oncol 2023; 40:181-191. [PMID: 35848787 DOI: 10.1080/08880018.2022.2089791] [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] [Indexed: 02/07/2023]
Abstract
Venous thromboembolism (VTE) is a well-known complication of the treatment of pediatric acute lymphoblastic leukemia (ALL). We analyzed 1026 ALL patients 1-18-years-old, who were enrolled into the AIEOP-BFM ALL 2000 or 2009 studies in Austria, with regard to the incidence and risk factors of VTE. The 2.5-year cumulative incidence (CI) of VTE ≥ grade 2 was 4%±1% (n = 36/1026). Twenty VTE (56%) were found in the central nervous system (19 cerebral venous sinus and 1 cortical vein thrombosis), and 16 (44%) at other sites (7 deep vein thromboses (DVT) of the lower extremity, 4 DVT of the upper extremity, 4 central venous line-thromboses, 1 pulmonary embolism). Most VTE occurred during induction and early consolidation therapy (81%) and were associated with L-asparaginase within 4 and corticosteroids withing 1 week(s) preceding the event (89 and 86%, respectively). In multivariable analysis, two independent risk factors were found. Patients 10-18-years-old had an increased (hazard-ratio: 2.156, p = 0.0389), whereas treatments in trial AIEOP-BFM ALL 2009 had a lower risk for VTE (hazard-ratio: 0.349, p = 0.0270). In conclusion, the 2.5-year CI of VTE among our pediatric patient cohort was <5% and adolescent age was the main patient-related risk factor. This older age group might benefit from primary prophylactic measures.
Collapse
Affiliation(s)
- Anna Gidl
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Anna Füreder
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Martin Benesch
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Michael Dworzak
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria.,St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Gernot Engstler
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Neil Jones
- Department of Pediatrics and Adolescent Medicine, University Clinics Salzburg, Salzburg, Austria
| | - Gabriele Kropshofer
- Division of Pediatric Hematology and Oncology and Stem Cell Transplantation, Department of Pediatrics and Adolescent Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Fiona Poyer
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Melanie Tamesberger
- Department of Pediatrics and Adolescent Medicine, Kepler University Hospital Linz, Linz, Austria
| | - Volker Witt
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Georg Mann
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria.,St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
64
|
Sawaisorn P, Atjanasuppat K, Uaesoontrachoon K, Rattananon P, Treesuppharat W, Hongeng S, Anurathapan U. Comparison of the efficacy of second and third generation lentiviral vector transduced CAR CD19 T cells for use in the treatment of acute lymphoblastic leukemia both in vitro and in vivo models. PLoS One 2023; 18:e0281735. [PMID: 36780428 PMCID: PMC9925013 DOI: 10.1371/journal.pone.0281735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
T cells genetically engineered to express a chimeric antigen receptor (CAR) specifically binding to a CD19 antigen has become the frontline of hematological malignancies immunotherapy. Their remarkable antitumor effect has exerted complete remission in treating B-cell malignancies. Although successful patient treatment has been shown, improvement to the structure of CAR to enhance its safety and efficacy profile is warranted. Transduction with a lentiviral vector (LVV) leading to the expression of CARs is also a critical step in redirecting T cells to target specific tumor antigens. To improve the efficacy of CD19 CARs in this study, the transduction ability of second and third generations LVV were compared. Ex vivo expansion of CD19 CARs T cells from healthy donors' peripheral blood mononuclear cells was performed after transduction of T cells with second and third generations LVV. Transduction efficacy of transduced T cells was determined to show a higher percentage in the third generations LVV transduced cells, with no changes in viability and identity of cells characterized by immunophenotyping. Testing the cytotoxic capacity of third generations LVV-transduced T cells against target cells showed higher reactivity against control cells. Cytokine expression was detected on the CD19 CARs T cells, suggesting that these cells limit in vitro growth of B-cell leukemia via secretion of the pro-inflammatory cytokine IFN γ. To investigate whether the third generation LVV transduced T cells can limit CD19 lymphoma growth in vivo, an analysis of tumor burden in a mouse model assessed by bioluminescence imaging was performed. We found that, in the presence of CD19 CARs T cells, the level of tumor burden was markedly reduced. In addition, an increase in the length of survival in mice receiving CAR-CD19 T cells was also observed. This suggests that transduction with third generations LVV generate a functional CAR-CD19 T cells, which may provide a safer and effective therapy for B-cell malignancies.
Collapse
Affiliation(s)
- Piamsiri Sawaisorn
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Korakot Atjanasuppat
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | | | - Worapapar Treesuppharat
- Thammasat University Research Unit in Mechanisms of Drug Action and Molecular Imaging, Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani, Thailand
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Usanarat Anurathapan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
65
|
HLA-haploidentical hematopoietic stem cells transplantation with regulatory and conventional T-cell adoptive immunotherapy in pediatric patients with very high-risk acute leukemia. Bone Marrow Transplant 2023; 58:526-533. [PMID: 36774432 PMCID: PMC9919737 DOI: 10.1038/s41409-023-01911-x] [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: 08/10/2022] [Revised: 12/16/2022] [Accepted: 01/04/2023] [Indexed: 02/13/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is still needed for many children with very high-risk acute leukemia. An HLA-haploidentical family donor is a suitable option for those without an HLA-matched donor. Here we present outcomes of a novel HLA-haploidentical HSCT (haplo-HSCT) strategy with adoptive immunotherapy with thymic-derived CD4+CD25+ FoxP3+ regulatory T cells (Tregs) and conventional T cells (Tcons) performed between January 2017 and July 2021 in 20 children with high-risk leukemia. Median age was 14.5 years (range, 4-21), 15 had acute lymphoblastic leukemia, 5 acute myeloid leukemia. The conditioning regimen included total body irradiation (TBI), thiotepa, fludarabine, cyclophosphamide. Grafts contained a megadose of CD34+ cells (mean 12.4 × 106/Kg), Tregs (2 × 106/Kg) and Tcons (0.5-1 × 106/Kg). All patients achieved primary, sustained full-donor engraftment. Only one patient relapsed (5%). The incidence of non-relapse mortality was 15% (3/20 patients). Five/20 patients developed ≥ grade 2 acute Graft versus Host Disease (aGvHD). It resolved in 4 who are alive and disease-free; 1 patient developed chronic GvHD (cGvHD). The probability of GRFS was 60 ± 0.5% (95% CI: 2.1-4.2) (Fig. 6), CRFS was 79 ± 0.9% (95% CI: 3.2-4.9) as 16/20 patients are alive and leukemia-free. The median follow-up was 2.1 years (range 0.5 months-5.1 years). This innovative approach was associated with very promising outcomes of HSCT strategy in pediatric patients.
Collapse
|
66
|
Popov A, Henze G, Roumiantseva J, Budanov O, Verzhbitskaya T, Boyakova E, Tsaur G, Fadeeva M, Lagoyko S, Zharikova L, Miakova N, Litvinov D, Khlebnikova O, Streneva O, Ponomareva N, Novichkova G, Fechina L, Karachunskiy A. Flow cytometric MRD at the end of consolidation in childhood B-lineage acute lymphoblastic leukemia has significant prognostic value but limited clinical implications: Results of study ALL-MB 2008. Leuk Res 2023; 125:106998. [PMID: 36566537 DOI: 10.1016/j.leukres.2022.106998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/27/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Alexander Popov
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation.
| | - Guenter Henze
- Department of Pediatric Oncology Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Roumiantseva
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Oleg Budanov
- National Research and Clinical Center 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
| | - Elena Boyakova
- Moscow City Blood Center named after OK Gavrilov, Moscow, Russian Federation
| | - Grigory Tsaur
- Regional Children's Hospital, Ekaterinburg, Russian Federation; Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Maria Fadeeva
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Svetlana Lagoyko
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Liudmila Zharikova
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Natalia Miakova
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Dmitry Litvinov
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | | | - Olga Streneva
- Regional Children's Hospital, Ekaterinburg, Russian Federation; Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | | | - Galina Novichkova
- 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
| | - Alexander Karachunskiy
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| |
Collapse
|
67
|
Kato M. Recent progress in pediatric lymphoblastic leukemia. Int J Hematol 2023; 117:155-161. [PMID: 36456860 DOI: 10.1007/s12185-022-03501-x] [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: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
The probability of long-term survival for children with lymphoblastic leukemia has improved dramatically over recent decades, mainly owing to advances in genomic analysis techniques, which have improved our understanding of the nature of leukemic cells and prognostic prediction based on the evaluation of precise treatment response. Risk-adjusted chemotherapy based on these advances has simultaneously reduced relapse rates and minimized complications. In addition, recent genomic analyses have deepened our understanding of the pathogenesis of leukemia and revealed the involvement of germline variations in the clinical course of leukemia treatment. Additionally, advances in minimal residual disease assays and the introduction of immunotherapy are expected to further improve therapeutic analyses. Further advances in clinical and translational research are anticipated to improve survival to 100% in a healthy state.
Collapse
Affiliation(s)
- Motohiro Kato
- Department of Pediatrics, The University of Tokyo, 7-3-1, Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
68
|
Yafour N, Hamzy F, Elkababri M, Yakoub-Agha I, Bekadja MA. [Acute lymphoblastic leukemia in developing countries: Management from the transplant indication (allo/auto) until post-transplant follow-up. Guidelines from the SFGM-TC]. Bull Cancer 2023; 110:S30-S38. [PMID: 35562231 DOI: 10.1016/j.bulcan.2022.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/26/2022]
Abstract
Management of acute lymphoblastic leukemia (ALL) patients in countries with limited resources depends on the means of prognostic stratification, available treatment and logistics. During the 12th annual harmonization workshops of the francophone Society of bone marrow transplantation and cellular therapy (SFGM-TC), a designated working group reviewed the literature in order to elaborate unified guidelines for allogeneic hematopoietic cell transplantation (Allo-HCT) in this disease. Conventional poor prognostic factors can be used to determine the indication of allo-HCT in first remission. Patients lacking a HLA-matched related donor can be allografted with a haploidentical donor allo-HCT if available. Chemotherapy based conditioning regimen can be used if TBI is not available, because the probability to find a radiotherapy department with the capacity for total body irradiation is low. For patients with Philadelphia chromosome positive (Phi+) ALL, post-transplantation tyrosine kinase inhibitors as a systematic maintenance strategy is recommended. Autologous HCT is optional for Phi+ ALL patients with negative minimal residual disease, who not eligible for allo-HCT. Patients with refractory/relapsed disease have a poor prognosis which highlights the importance of acquiring in the future new therapies such as: blinatumumab, inotuzumab, and CAR-T cells.
Collapse
Affiliation(s)
- Nabil Yafour
- Université d'Oran 1, Ahmed-Ben-Bella, établissement hospitalier et universitaire 1(er) Novembre 1954, faculté de médecine, service d'hématologie et de thérapie cellulaire, BP 4166 Ibn-Rochd, 31000 Oran, Algérie.
| | - Faty Hamzy
- Hôpital Cheikh-Zaïd universitaire international, service d'hématologie et greffe, cité Al-Irfane-Hay Ryad avenue Allal-al-Fassi, 10000 Rabat, Maroc
| | - Maria Elkababri
- Hôpital d'enfants de Rabat, université Mohammed V de Rabat, service d'hématologie et oncologie pédiatrique, Rabat, Maroc
| | | | - Mohamed Amine Bekadja
- Université d'Oran 1, Ahmed-Ben-Bella, établissement hospitalier et universitaire 1(er) Novembre 1954, faculté de médecine, service d'hématologie et de thérapie cellulaire, BP 4166 Ibn-Rochd, 31000 Oran, Algérie
| |
Collapse
|
69
|
Dargenio M, Bonifacio M, Chiaretti S, Vitale A, Fracchiolla NS, Papayannidis C, Giglio F, Salutari P, Audisio E, Scappini B, Zappasodi P, Defina M, Forghieri F, Scattolin AM, Todisco E, Lunghi M, Guolo F, Del Principe MI, Annunziata M, Lazzarotto D, Cedrone M, Pasciolla C, Imovilli A, Tanasi I, Trappolini S, Cerrano M, La Starza R, Krampera M, Di Renzo N, Candoni A, Pizzolo G, Ferrara F, Foà R. Incidence, treatment and outcome of central nervous system relapse in adult acute lymphoblastic leukaemia patients treated front-line with paediatric-inspired regimens: A retrospective multicentre Campus ALL study. Br J Haematol 2023; 200:440-450. [PMID: 36335916 PMCID: PMC10098932 DOI: 10.1111/bjh.18537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/08/2022]
Abstract
Within the Campus ALL network we analyzed the incidence, characteristics, treatment and outcome of a central nervous system (CNS) relapse in 1035 consecutive adult acute lymphoblastic leukemia (ALL) patients treated frontline with pediatric-inspired protocols between 2009 and 2020. Seventy-one patients (6.8%) experienced a CNS recurrence, more frequently in T- (28/278; 10%) than in B-ALL (43/757; 5.7%) (p = 0.017). An early CNS relapse-< 12 months from diagnosis-was observed in 41 patients. In multivariate analysis, risk factors for early CNS relapse included T-cell phenotype (p = <0.001), hyperleucocytosis >100 × 109 /L (p<0.001) and male gender (p = 0.015). Treatment was heterogeneous, including chemotherapy, radiotherapy, intrathecal therapy and novel agents. A complete remission (CR) was obtained in 39 patients (55%) with no differences among strategies. After CR, 26 patients underwent an allogenic transplant, with a significant overall survival benefit compared to non-transplanted patients (p = 0.012). After a median observation of 8 months from CNS relapse, 23 patients (32%) were alive. In multivariate analysis, the time to CNS relapse was the strongest predictor of a lower 2-year post-relapse survival (p<0.001). In conclusion, in adult ALL the outcome after a CNS relapse remains very poor. Effective CNS prophylaxis remains the best approach and allogenic transplant should be pursued when possible.
Collapse
Affiliation(s)
- Michelina Dargenio
- Ospedale Vito Fazzi, Unità Operativa di Ematologia e Trapianto, Lecce, Italy
| | - Massimiliano Bonifacio
- Dipartimento di Medicina, Sezione di Ematologia, Università e Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Sabina Chiaretti
- Divisione di Ematologia, Dipartimento di Medicina Traslazionale e di Precisione, Università Sapienza, Rome, Italy
| | - Antonella Vitale
- Divisione di Ematologia, Dipartimento di Medicina Traslazionale e di Precisione, Università Sapienza, Rome, Italy
| | | | - Cristina Papayannidis
- IRCCS Azienda Ospedaliero-Universitaria, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Fabio Giglio
- Ospedale San Raffaele, Unità Operativa di Ematologia, Milan, Italy
| | - Prassede Salutari
- Unità Operativa Complessa di Ematologia, Ospedale Civile Spirito Santo, Pescara, Italy
| | - Ernesta Audisio
- Dipartimento di Ematologia e Oncologia, AO Città della Salute e della Scienza, Torino, Italy
| | | | - Patrizia Zappasodi
- Unità Operativa di Ematologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marzia Defina
- Unità Operativa Complessa di Ematologia, AOUS, Università di Siena, Siena, Italy
| | - Fabio Forghieri
- Unità Operativa Complessa di Ematologia, AOU Policlinico, Modena, Italy
| | | | - Elisabetta Todisco
- Unità Operativa Ematologia, Istituto Europeo di Oncologia (IEO), Milan, Italy
| | - Monia Lunghi
- Divisione di Ematologia, Dipartimento di Medicina Traslazionale, Università del Piemonte Orientale, Novara, Italy
| | - Fabio Guolo
- Clinica Ematologica, Dipartimento di Medicina Interna, Università degli Studi di Genova, Genoa, Italy.,Dipartimento di Oncologia e Ematologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Mario Annunziata
- Unità Operativa Complessa di Ematologia, AORN Cardarelli, Naples, Italy
| | - Davide Lazzarotto
- Clinica ematologica e centro trapianti, Azienda sanitaria universitaria Friuli Centrale, Udine, Italy
| | - Michele Cedrone
- Unità Operativa Complessa di Ematologia, Ospedale San Giovanni Addolorata, Rome, Italy
| | - Crescenza Pasciolla
- Unità Operativa di Ematologia, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | | | - Ilaria Tanasi
- Dipartimento di Medicina, Sezione di Ematologia, Università e Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | - Marco Cerrano
- Unità Operativa di Ematologia, Presidio Molinette-AOU Città della Salute e della Scienza, Torino, Italy
| | - Roberta La Starza
- Sezione di Ematologia e Trapianto, Università degli Studi di Perugia, A.O. Perugia, Italy
| | - Mauro Krampera
- Dipartimento di Medicina, Sezione di Ematologia, Università e Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Nicola Di Renzo
- Ospedale Vito Fazzi, Unità Operativa di Ematologia e Trapianto, Lecce, Italy
| | - Anna Candoni
- Unità Operativa Complessa di Ematologia, AORN Cardarelli, Naples, Italy
| | - Giovanni Pizzolo
- Dipartimento di Medicina, Sezione di Ematologia, Università e Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Felicetto Ferrara
- Dipartimento di Biomedicina e Prevenzione, Università degli studi Tor Vergata, Rome, Italy
| | - Robin Foà
- Divisione di Ematologia, Dipartimento di Medicina Traslazionale e di Precisione, Università Sapienza, Rome, Italy
| |
Collapse
|
70
|
Kaczmarska A, Derebas J, Pinkosz M, Niedźwiecki M, Lejman M. The Landscape of Secondary Genetic Rearrangements in Pediatric Patients with B-Cell Acute Lymphoblastic Leukemia with t(12;21). Cells 2023; 12:cells12030357. [PMID: 36766699 PMCID: PMC9913634 DOI: 10.3390/cells12030357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
The most frequent chromosomal rearrangement in childhood B-cell acute lymphoblastic leukemia (B-ALL) is translocation t(12;21)(p13;q22). It results in the fusion of the ETV6::RUNX1 gene, which is active in the regulation of multiple crucial cellular pathways. Recent studies hypothesize that many translocations are influenced by RAG-initiated deletions, as well as defects in the RAS and NRAS pathways. According to a "two-hit" model for the molecular pathogenesis of pediatric ETV6::RUNX1-positive B-ALL, the t(12;21) translocation requires leukemia-causing secondary mutations. Patients with ETV6::RUNX1 express up to 60 different aberrations, which highlights the heterogeneity of this B-ALL subtype and is reflected in differences in patient response to treatment and chances of relapse. Most studies of secondary genetic changes have concentrated on deletions of the normal, non-rearranged ETV6 allele. Other predominant structural changes included deletions of chromosomes 6q and 9p, loss of entire chromosomes X, 8, and 13, duplications of chromosome 4q, or trisomy of chromosomes 21 and 16, but the impact of these changes on overall survival remains unclarified. An equally genetically diverse group is the recently identified new B-ALL subtype ETV6::RUNX1-like ALL. In our review, we provide a comprehensive description of recurrent secondary mutations in pediatric B-ALL with t(12;21) to emphasize the value of investigating detailed molecular mechanisms in ETV6::RUNX1-positive B-ALL, both for our understanding of the etiology of the disease and for future clinical advances in patient treatment and management.
Collapse
Affiliation(s)
- Agnieszka Kaczmarska
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
| | - Justyna Derebas
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
| | - Michalina Pinkosz
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
| | - Maciej Niedźwiecki
- Department of Pediatrics, Hematology and Oncology Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
- Correspondence:
| |
Collapse
|
71
|
Junk SV, Schaeffeler E, Zimmermann M, Möricke A, Beier R, Schütte P, Fedders B, Alten J, Hinze L, Klein N, Kulozik A, Muckenthaler MU, Koehler R, Borkhardt A, Vijayakrishnan J, Ellinghaus D, Forster M, Franke A, Wintering A, Kratz CP, Schrappe M, Schwab M, Houlston RS, Cario G, Stanulla M. Chemotherapy-related hyperbilirubinemia in pediatric acute lymphoblastic leukemia: a genome-wide association study from the AIEOP-BFM ALL study group. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:21. [PMID: 36639636 PMCID: PMC9838013 DOI: 10.1186/s13046-022-02585-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/26/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Characterization of clinical phenotypes in context with tumor and host genomic information can aid in the development of more effective and less toxic risk-adapted and targeted treatment strategies. To analyze the impact of therapy-related hyperbilirubinemia on treatment outcome and to identify contributing genetic risk factors of this well-recognized adverse effect we evaluated serum bilirubin levels in 1547 pediatric patients with acute lymphoblastic leukemia (ALL) and conducted a genome-wide association study (GWAS). PATIENTS AND METHODS Patients were treated in multicenter trial AIEOP-BFM ALL 2000 for pediatric ALL. Bilirubin toxicity was graded 0 to 4 according to the Common Toxicity Criteria (CTC) of the National Cancer Institute. In the GWAS discovery cohort, including 650 of the 1547 individuals, genotype frequencies of 745,895 single nucleotide variants were compared between 435 patients with hyperbilirubinemia (CTC grades 1-4) during induction/consolidation treatment and 215 patients without it (grade 0). Replication analyses included 224 patients from the same trial. RESULTS Compared to patients with no (grade 0) or moderate hyperbilirubinemia (grades 1-2) during induction/consolidation, patients with grades 3-4 had a poorer 5-year event free survival (76.6 ± 3% versus 87.7 ± 1% for grades 1-2, P = 0.003; 85.2 ± 2% for grade 0, P < 0.001) and a higher cumulative incidence of relapse (15.6 ± 3% versus 9.0 ± 1% for grades 1-2, P = 0.08; 11.1 ± 1% for grade 0, P = 0.007). GWAS identified a strong association of the rs6744284 variant T allele in the UGT1A gene cluster with risk of hyperbilirubinemia (allelic odds ratio (OR) = 2.1, P = 7 × 10- 8). TT-homozygotes had a 6.5-fold increased risk of hyperbilirubinemia (grades 1-4; 95% confidence interval (CI) = 2.9-14.6, P = 7 × 10- 6) and a 16.4-fold higher risk of grade 3-4 hyperbilirubinemia (95% CI 6.1-43.8, P = 2 × 10- 8). Replication analyses confirmed these associations with joint analysis yielding genome-wide significance (allelic OR = 2.1, P = 6 × 10- 11; 95% CI 1.7-2.7). Moreover, rs6744284 genotypes were strongly linked to the Gilbert's syndrome-associated UGT1A1*28/*37 allele (r2 = 0.70), providing functional support for study findings. Of clinical importance, the rs6744284 TT genotype counterbalanced the adverse prognostic impact of high hyperbilirubinemia on therapy outcome. CONCLUSIONS Chemotherapy-related hyperbilirubinemia is a prognostic factor for treatment outcome in pediatric ALL and genetic variation in UGT1A aids in predicting the clinical impact of hyperbilirubinemia. TRIAL REGISTRATION http://www. CLINICALTRIALS gov ; #NCT00430118.
Collapse
Affiliation(s)
- Stefanie V. Junk
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Elke Schaeffeler
- Margarete-Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Martin Zimmermann
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Anja Möricke
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Rita Beier
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Peter Schütte
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Birthe Fedders
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Julia Alten
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Laura Hinze
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Norman Klein
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Andreas Kulozik
- grid.7700.00000 0001 2190 4373Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Martina U. Muckenthaler
- grid.7700.00000 0001 2190 4373Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Rolf Koehler
- grid.7700.00000 0001 2190 4373Department of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Arndt Borkhardt
- grid.411327.20000 0001 2176 9917Clinic for Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jayaram Vijayakrishnan
- grid.18886.3fDivision of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, UK
| | - David Ellinghaus
- grid.9764.c0000 0001 2153 9986Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Michael Forster
- grid.9764.c0000 0001 2153 9986Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Andre Franke
- grid.9764.c0000 0001 2153 9986Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Astrid Wintering
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Christian P. Kratz
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Martin Schrappe
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Schwab
- Margarete-Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany ,grid.10392.390000 0001 2190 1447Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University of Tuebingen, Tuebingen, Germany ,grid.10392.390000 0001 2190 1447Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany ,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany
| | - Richard S. Houlston
- grid.18886.3fDivision of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Gunnar Cario
- grid.412468.d0000 0004 0646 2097Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Martin Stanulla
- grid.10423.340000 0000 9529 9877Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
72
|
Della Starza I, De Novi LA, Elia L, Bellomarino V, Beldinanzi M, Soscia R, Cardinali D, Chiaretti S, Guarini A, Foà R. Optimizing Molecular Minimal Residual Disease Analysis in Adult Acute Lymphoblastic Leukemia. Cancers (Basel) 2023; 15:374. [PMID: 36672325 PMCID: PMC9856386 DOI: 10.3390/cancers15020374] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Minimal/measurable residual disease (MRD) evaluation has resulted in a fundamental instrument to guide patient management in acute lymphoblastic leukemia (ALL). From a methodological standpoint, MRD is defined as any approach aimed at detecting and possibly quantifying residual neoplastic cells beyond the sensitivity level of cytomorphology. The molecular methods to study MRD in ALL are polymerase chain reaction (PCR) amplification-based approaches and are the most standardized techniques. However, there are some limitations, and emerging technologies, such as digital droplet PCR (ddPCR) and next-generation sequencing (NGS), seem to have advantages that could improve MRD analysis in ALL patients. Furthermore, other blood components, namely cell-free DNA (cfDNA), appear promising and are also being investigated for their potential role in monitoring tumor burden and response to treatment in hematologic malignancies. Based on the review of the literature and on our own data, we hereby discuss how emerging molecular technologies are helping to refine the molecular monitoring of MRD in ALL and may help to overcome some of the limitations of standard approaches, providing a benefit for the care of patients.
Collapse
Affiliation(s)
- Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
- GIMEMA Foundation, 00182 Rome, Italy
| | - Lucia Anna De Novi
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Loredana Elia
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Vittorio Bellomarino
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Marco Beldinanzi
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Deborah Cardinali
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Sabina Chiaretti
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Anna Guarini
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Via Benevento 6, 00161 Rome, Italy
| |
Collapse
|
73
|
Muacevic A, Adler JR, Bin Shuayl MK, Farwana FM, Alhassan AT, Alsaif FA. The Successful Implementation of a Modified Pediatric Acute Lymphoblastic Leukemia Protocol to Treat an Elderly Patient With T-cell Lymphoblastic Leukemia: A Case Report. Cureus 2023; 15:e33729. [PMID: 36655154 PMCID: PMC9840415 DOI: 10.7759/cureus.33729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a group of hematological malignancies most commonly seen in pediatrics. The disease process localizes in lymphoid organs, the central nervous system, the mediastinum, and bone marrow (BM). The clinical features of T-cell acute lymphoblastic leukemia (T-ALL) in adults include evidence of generalized lymphadenopathy, hepatosplenomegaly, immunosuppression, and hypercalcemia. There is limited research on the efficacy of using modified pediatric treatment regimens in the elderly over the age of 60 with ALL; this case report aims to illustrate the successful treatment of a 67-year-old male patient diagnosed with T-ALL, using a modified Children's Oncology Group (COG) protocol. Through this, it has been shown to be an effective, safe, and efficacious treatment option for our patient.
Collapse
|
74
|
Elitzur S, Izraeli S, Ben-Yehuda D, Gatt ME. Lymphoid Leukemias. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00077-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
75
|
A single flow cytometric MRD measurement in children with B-lineage acute lymphocytic leukemia and hyperleukocytosis redefines the requirements of high-risk treatment: Results of the study ALL-MB 2008. Leuk Res 2022; 123:106982. [DOI: 10.1016/j.leukres.2022.106982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/03/2022] [Accepted: 10/21/2022] [Indexed: 11/07/2022]
|
76
|
Cai Z, Liu Y, Tang B, Wu Z, Wang Z, Lin R, Xu X, Huang Z, Ou J, Li X, Liu X, Liu Q, Zhou H. Dynamics of minimal residual disease defines a novel risk-classification and the role of allo-HSCT in adult Ph-negative B-cell acute lymphoblastic leukemia. Leuk Lymphoma 2022; 63:3181-3190. [PMID: 36098226 DOI: 10.1080/10428194.2022.2115841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The prognosis of minimal residual disease (MRD) in acute lymphoblastic leukemia (ALL) patients is well established. However, the implementation of dynamic MRD for risk classification and decision-making for allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains vague. In this study, we collected multiparameter flow cytometry (MFC)-MRD data of Ph-negative B-ALL patients (n = 134) from the Precision-Classification-Directed-Target-Total-Therapy-ALL-2016 (PDT-ALL-2016) cohort and stratified it into high-(HR), medium-(MR), and standard-risk (SR) groups. With a median of 3.65 years follow-up (95% CI: 3.037-4.263), 3-year OS rate was 51.8 ± 8.3% in HR, compared with MR 61.5 ± 10.8% (p = 0.472), and SR 73.3 ± 5.9% (p = 0.006). Multivariate analysis shows that integrated dynamic MRD is an independent factor for overall survival. Compared to pediatric-inspired chemotherapy, allo-HSCT significantly improves the survival of the HR cohort (p < 0.001), but not in MR and SR. Finally, our study suggests that integrated dynamic MRD defines a novel risk-classification criteria and highlights the benefits of allo-HSCT in adult patients with Ph-negative ALL.
Collapse
Affiliation(s)
- Zihong Cai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiqian Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingqing Tang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhengwei Wu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhixiang Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ren Lin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuli Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zicong Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiawang Ou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaofang Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoli Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
77
|
Thastrup M, Duguid A, Mirian C, Schmiegelow K, Halsey C. Central nervous system involvement in childhood acute lymphoblastic leukemia: challenges and solutions. Leukemia 2022; 36:2751-2768. [PMID: 36266325 PMCID: PMC9712093 DOI: 10.1038/s41375-022-01714-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022]
Abstract
Delivery of effective anti-leukemic agents to the central nervous system (CNS) is considered essential for cure of childhood acute lymphoblastic leukemia. Current CNS-directed therapy comprises systemic therapy with good CNS-penetration accompanied by repeated intrathecal treatments up to 26 times over 2-3 years. This approach prevents most CNS relapses, but is associated with significant short and long term neurotoxicity. Despite this burdensome therapy, there have been no new drugs licensed for CNS-leukemia since the 1960s, when very limited anti-leukemic agents were available and there was no mechanistic understanding of leukemia survival in the CNS. Another major barrier to improved treatment is that we cannot accurately identify children at risk of CNS relapse, or monitor response to treatment, due to a lack of sensitive biomarkers. A paradigm shift in treating the CNS is needed. The challenges are clear - we cannot measure CNS leukemic load, trials have been unable to establish the most effective CNS treatment regimens, and non-toxic approaches for relapsed, refractory, or intolerant patients are lacking. In this review we discuss these challenges and highlight research advances aiming to provide solutions. Unlocking the potential of risk-adapted non-toxic CNS-directed therapy requires; (1) discovery of robust diagnostic, prognostic and response biomarkers for CNS-leukemia, (2) identification of novel therapeutic targets combined with associated investment in drug development and early-phase trials and (3) engineering of immunotherapies to overcome the unique challenges of the CNS microenvironment. Fortunately, research into CNS-ALL is now making progress in addressing these unmet needs: biomarkers, such as CSF-flow cytometry, are now being tested in prospective trials, novel drugs are being tested in Phase I/II trials, and immunotherapies are increasingly available to patients with CNS relapses. The future is hopeful for improved management of the CNS over the next decade.
Collapse
Affiliation(s)
- Maria Thastrup
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Alasdair Duguid
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Christian Mirian
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Halsey
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
78
|
Zuna J, Hovorkova L, Krotka J, Koehrmann A, Bardini M, Winkowska L, Fronkova E, Alten J, Koehler R, Eckert C, Brizzolara L, Trkova M, Stuchly J, Zimmermann M, De Lorenzo P, Valsecchi MG, Conter V, Stary J, Schrappe M, Biondi A, Trka J, Zaliova M, Cazzaniga G, Cario G. Minimal residual disease in BCR::ABL1-positive acute lymphoblastic leukemia: different significance in typical ALL and in CML-like disease. Leukemia 2022; 36:2793-2801. [PMID: 35933523 DOI: 10.1038/s41375-022-01668-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/09/2022]
Abstract
Recently, we defined "CML-like" subtype of BCR::ABL1-positive acute lymphoblastic leukemia (ALL), resembling lymphoid blast crisis of chronic myeloid leukemia (CML). Here we retrospectively analyzed prognostic relevance of minimal residual disease (MRD) and other features in 147 children with BCR::ABL1-positive ALL (diagnosed I/2000-IV/2021, treated according to EsPhALL (n = 133) or other (n = 14) protocols), using DNA-based monitoring of BCR::ABL1 genomic breakpoint and clonal immunoglobulin/T-cell receptor gene rearrangements. Although overall prognosis of CML-like (n = 48) and typical ALL (n = 99) was similar (5-year-EFS 60% and 49%, respectively; 5-year-OS 75% and 73%, respectively), typical ALL presented more relapses while CML-like patients more often died in the first remission. Prognostic role of MRD was significant in the typical ALL (p = 0.0005 in multivariate analysis for EFS). In contrast, in CML-like patients MRD was not significant (p values > 0.2) and inapplicable for therapy adjustment. Moreover, in the typical ALL, risk-prediction could be further improved by considering initial hyperleukocytosis. Early distinguishing typical BCR::ABL1-positive ALL and CML-like patients is essential to enable optimal treatment approach in upcoming protocols. For the typical ALL, tyrosine-kinase inhibitors and concurrent chemotherapy with risk-directed intensity should be recommended; in the CML-like disease, no relevant prognostic feature applicable for therapy tailoring was found so far.
Collapse
Affiliation(s)
- Jan Zuna
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic.
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
- University Hospital Motol, Prague, Czech Republic.
| | - Lenka Hovorkova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Justina Krotka
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Amelie Koehrmann
- Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Michela Bardini
- Tettamanti Research Center, Pediatrics, University of Milano-Bicocca/Fondazione Tettamanti, Monza, Italy
| | - Lucie Winkowska
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eva Fronkova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Julia Alten
- Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Rolf Koehler
- Department of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Lisa Brizzolara
- Tettamanti Research Center, Pediatrics, University of Milano-Bicocca/Fondazione Tettamanti, Monza, Italy
| | - Marie Trkova
- Centre for Medical Genetics and Reproductive Medicine GENNET, Prague, Czech Republic
| | - Jan Stuchly
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Zimmermann
- Department of Pediatric Hematology and Oncology, Medical School Hannover, Hannover, Germany
| | - Paola De Lorenzo
- EsPhALL Trial Data Center, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Maria Grazia Valsecchi
- EsPhALL Trial Data Center, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Valentino Conter
- Pediatric Hemato-Oncolgy, Fondazione MBBM/ASST-Monza, University of Milano-Bicocca, Monza, Italy
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Martin Schrappe
- Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Andrea Biondi
- Pediatric Hemato-Oncolgy, Fondazione MBBM/ASST-Monza, University of Milano-Bicocca, Monza, Italy
| | - Jan Trka
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Marketa Zaliova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Giovanni Cazzaniga
- Tettamanti Research Center, Pediatrics, University of Milano-Bicocca/Fondazione Tettamanti, Monza, Italy
- Medical Genetics, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Gunnar Cario
- Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
79
|
Yu CH, Wu G, Chang CC, Jou ST, Lu MY, Lin KH, Chen SH, Wu KH, Huang FL, Cheng CN, Chang HH, Hedges D, Wang JL, Yen HJ, Li MJ, Chou SW, Hung CT, Lin ZS, Lin CY, Chen HY, Ni YL, Hsu YC, Lin DT, Lin SW, Yang JJ, Pui CH, Yu SL, Yang YL. Sequential Approach to Improve the Molecular Classification of Childhood Acute Lymphoblastic Leukemia. J Mol Diagn 2022; 24:1195-1206. [PMID: 35963521 PMCID: PMC9667711 DOI: 10.1016/j.jmoldx.2022.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/27/2022] [Accepted: 08/03/2022] [Indexed: 10/15/2022] Open
Abstract
Identification of specific leukemia subtypes is a key to successful risk-directed therapy in childhood acute lymphoblastic leukemia (ALL). Although RNA sequencing (RNA-seq) is the best approach to identify virtually all specific leukemia subtypes, the routine use of this method is too costly for patients in resource-limited countries. This study enrolled 295 patients with pediatric ALL from 2010 to 2020. Routine screening could identify major cytogenetic alterations in approximately 69% of B-cell ALL (B-ALL) cases by RT-PCR, DNA index, and multiplex ligation-dependent probe amplification. STIL-TAL1 was present in 33% of T-cell ALL (T-ALL) cases. The remaining samples were submitted for RNA-seq. More than 96% of B-ALL cases and 74% of T-ALL cases could be identified based on the current molecular classification using this sequential approach. Patients with Philadelphia chromosome-like ALL constituted only 2.4% of the entire cohort, a rate even lower than those with ZNF384-rearranged (4.8%), DUX4-rearranged (6%), and Philadelphia chromosome-positive (4.4%) ALL. Patients with ETV6-RUNX1, high hyperdiploidy, PAX5 alteration, and DUX4 rearrangement had favorable prognosis, whereas those with hypodiploid and KMT2A and MEF2D rearrangement ALL had unfavorable outcomes. With the use of multiplex ligation-dependent probe amplification, DNA index, and RT-PCR in B-ALL and RT-PCR in T-ALL followed by RNA-seq, childhood ALL can be better classified to improve clinical assessments.
Collapse
Affiliation(s)
- Chih-Hsiang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Gang Wu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Chia-Ching Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shiann-Tarng Jou
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Yao Lu
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kai-Hsin Lin
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Shu-Huey Chen
- Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital and School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Fang-Liang Huang
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chao-Neng Cheng
- Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Hsiu-Hao Chang
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dale Hedges
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jinn-Li Wang
- Division of Hematology Oncology, Department of Pediatrics, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Ju Yen
- Department of Pediatrics, Taipei Veterans General Hospital and National Yang-Ming Chiao-Tung University School of Medicine, Taipei, Taiwan
| | - Meng-Ju Li
- Department of Pediatrics, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Shu-Wei Chou
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Chen-Ting Hung
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ze-Shiang Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Yu Lin
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Hsuan-Yu Chen
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Yu-Ling Ni
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yin-Chen Hsu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dong-Tsamn Lin
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jun J Yang
- Department of Pharmacology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan; Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Yung-Li Yang
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
80
|
Pawinska-Wasikowska K, Bukowska-Strakova K, Surman M, Rygielska M, Sadowska B, Ksiazek T, Klekawka T, Wieczorek A, Skoczen S, Balwierz W. Go with the Flow—Early Assessment of Measurable Residual Disease in Children with Acute Lymphoblastic Leukemia Treated According to ALL IC-BFM2009. Cancers (Basel) 2022; 14:cancers14215359. [PMID: 36358778 PMCID: PMC9653819 DOI: 10.3390/cancers14215359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Monitoring of residual disease is a very important aspect of modern treatment approaches in many types of cancer. In acute leukemias in both children and adults, molecular and cytometric methods are used to assess the burden of leukemia at different points during therapy. Residual disease measured at the end of induction was shown to be the strongest predictor of outcome. Analyzing the outcomes of children with acute lymphoblastic leukemia (ALL), we aimed to establish the most informative cut-off and time point of assessment. Applying only the measurement of residual disease by flow cytometry along with genotypic findings, we managed to identify patients with a poor prognosis. Although new precise, molecular techniques as the next generation sequencing strategy are approaching daily clinical practice, flow cytometry is still a reliable, standardized method of residual disease detection. We may say ‘go with the flow’; thus, the assessment of residual disease by multiparametric flow cytometry is a proper method for the management of ALL patients according to risk-adapted therapies. Abstract Measurable residual disease (MRD) is a well-known tool for the evaluation of the early response to treatment in patients with acute lymphoblastic leukemia (ALL). In respect to predicting the relapse the most informative cut-off and time point of MRD measurement during therapy were evaluated in our study. Between 1 January 2013 and 31 December 2019, multiparametric flow cytometry (MFC) MRD was measured in the bone marrow of 140 children with ALL treated according to the ALL IC-BFM2009 protocol. The MRD cut-off of 0.1% and day 33, end of induction, were the most discriminatory for all patients. Patients with negative MRD on day 15 and 33 had a higher 5-year overall survival—OS (100%) and a higher relapse-free survival—RFS rate (97.6%) than those with positive levels of MRD (≥0.01%) at both time points (77.8% and 55.6%, p = 0.002 and 0.001, respectively). Most patients with residual disease below 0.1% on day 15 exhibit hyperdiploidy or ETV6-RUNX1 in ALL cells. Measurement of MRD at early time points can be used with simplified genetic analysis to better identify low and high-risk patients, allowing personalized therapies and further improvement in outcomes in pediatric ALL.
Collapse
Affiliation(s)
- Katarzyna Pawinska-Wasikowska
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children’s Hospital, 30-663 Krakow, Poland
| | - Karolina Bukowska-Strakova
- Department of Clinical Immunology and Transplantation, Faculty of Medicine, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Correspondence:
| | - Marta Surman
- Department of Clinical Immunology and Transplantation, Faculty of Medicine, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
| | - Monika Rygielska
- Hematology Laboratory, Department of Pediatric Oncology and Hematology, University Children’s Hospital, 30-663 Krakow, Poland
| | - Beata Sadowska
- Department of Pediatric Oncology and Hematology, Cytogenetics and Molecular Genetics Laboratory, University Children’s Hospital, 30-663 Krakow, Poland
| | - Teofila Ksiazek
- Department of Pediatric Oncology and Hematology, Cytogenetics and Molecular Genetics Laboratory, University Children’s Hospital, 30-663 Krakow, Poland
- Department of Medical Genetics, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland
| | - Tomasz Klekawka
- Department of Pediatric Oncology and Hematology, University Children’s Hospital, 30-663 Krakow, Poland
| | - Aleksandra Wieczorek
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children’s Hospital, 30-663 Krakow, Poland
| | - Szymon Skoczen
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children’s Hospital, 30-663 Krakow, Poland
| | - Walentyna Balwierz
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children’s Hospital, 30-663 Krakow, Poland
| |
Collapse
|
81
|
Vigliotta I, Armuzzi S, Barone M, Solli V, Pistis I, Borsi E, Taurisano B, Mazzocchetti G, Martello M, Poletti A, Sartor C, Rizzello I, Pantani L, Tacchetti P, Papayannidis C, Mancuso K, Rocchi S, Zamagni E, Curti A, Arpinati M, Cavo M, Terragna C. The ALLgorithMM: How to define the hemodilution of bone marrow samples in lymphoproliferative diseases. Front Oncol 2022; 12:1001048. [PMID: 36276072 PMCID: PMC9582597 DOI: 10.3389/fonc.2022.1001048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Minimal residual disease (MRD) is commonly assessed in bone marrow (BM) aspirate. However, sample quality can impair the MRD measurement, leading to underestimated residual cells and to false negative results. To define a reliable and reproducible method for the assessment of BM hemodilution, several flow cytometry (FC) strategies for hemodilution evaluation have been compared. Methods For each BM sample, cells populations with a well-known distribution in BM and peripheral blood - e.g., mast cells (MC), immature (IG) and mature granulocytes (N) – have been studied by FC and quantified alongside the BM differential count. Results The frequencies of cells’ populations were correlated to the IG/N ratio, highlighting a mild correlation with MCs and erythroblasts (R=0.25 and R=0.38 respectively, with p-value=0.0006 and 0.0000052), whereas no significant correlation was found with B or T-cells. The mild correlation between IG/N, erythroblasts and MCs supported the combined use of these parameters to evaluate BM hemodilution, hence the optimization of the ALLgorithMM. Once validated, the ALLgorithMM was employed to evaluate the dilution status of BM samples in the context of MRD assessment. Overall, we found that 32% of FC and 52% of Next Generation Sequencing (NGS) analyses were MRD negative in samples resulted hemodiluted (HD) or at least mildly hemodiluted (mHD). Conclusions The high frequency of MRD-negative results in both HD and mHD samples implies the presence of possible false negative MRD measurements, impairing the correct assessment of patients’ response to therapy and highlighs the importance to evaluate BM hemodilution.
Collapse
Affiliation(s)
- Ilaria Vigliotta
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
- *Correspondence: Ilaria Vigliotta, ; Carolina Terragna,
| | - Silvia Armuzzi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Martina Barone
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Vincenza Solli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Ignazia Pistis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
| | - Enrica Borsi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Barbara Taurisano
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Gaia Mazzocchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Marina Martello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Andrea Poletti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Chiara Sartor
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Ilaria Rizzello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Lucia Pantani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
| | - Paola Tacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
| | - Cristina Papayannidis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
| | - Katia Mancuso
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Serena Rocchi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Mario Arpinati
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Michele Cavo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Carolina Terragna
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- *Correspondence: Ilaria Vigliotta, ; Carolina Terragna,
| |
Collapse
|
82
|
Gore L. What are the Long-Term Complications of Pediatric ALL Treatments and How Can They Be Mitigated? Perspectives on Long-term Complications of Curative Treatment in Childhood ALL. Best Pract Res Clin Haematol 2022; 35:101403. [DOI: 10.1016/j.beha.2022.101403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
83
|
Lee JW, Kim Y, Ahn A, Lee JM, Yoo JW, Kim S, Cho B, Chung NG, Kim M. Clinical implication of minimal residual disease assessment by next-generation sequencing-based immunoglobulin clonality assay in pediatric B-acute lymphoblastic leukemia. Front Oncol 2022; 12:957743. [PMID: 36185293 PMCID: PMC9521036 DOI: 10.3389/fonc.2022.957743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Measuring minimal residual disease (MRD) during treatment is valuable to identify acute lymphoblastic leukemia (ALL) patients who require intensified treatment to avert relapse. We performed the next-generation sequencing (NGS)-based immunoglobulin gene (Ig) clonality assay and evaluated its clinical implication in pediatric B-ALL patients to assess MRD. Fifty-five patients who were diagnosed and treated with de novo (n = 44) or relapsed/refractory B-ALL (n = 11) were enrolled. MRD assessment was performed using the LymphoTrack® Dx IGH and IGK assay panels. The percentage of the clonal sequences per total read count was calculated as MRD (% of B cells). The data were normalized as the proportion of total nucleated cells (TNC) by LymphoQuant™ Internal control or the B-cell proportion in each sample estimated by flow cytometry or immunohistochemistry. Clonal Ig rearrangement was identified in all patients. The normalized MRD value was significantly lower than the unnormalized MRD value (p < 0.001). When categorizing patients, 27 of 50 patients (54%) achieved normalized MRD <0.01%, while 6 of them did not achieve MRD <0.01% when applying the unnormalized value. The normalized post-induction MRD value of 0.01% proved to be a significant threshold value for both 3-year event-free survival (100% for MRD <0.01% vs. 60.9% ± 10.2% for MRD ≥0.01%, p = 0.007) and 3-year overall survival (100% for MRD <0.01% vs. 78.3% ± 8.6% for MRD ≥0.01%, p = 0.011). However, unnormalized MRD was not a significant factor for outcome in this cohort. Our study demonstrated that MRD assessment by NGS-based Ig clonality assay could be applied in most pediatric B-ALL patients. Normalized post-induction MRD <0.01% was a significant prognostic indicator.
Collapse
Affiliation(s)
- Jae Wook Lee
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yonggoo Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ari Ahn
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jong Mi Lee
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jae Won Yoo
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seongkoo Kim
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Bin Cho
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Nack-Gyun Chung
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- *Correspondence: Nack-Gyun Chung, ; Myungshin Kim,
| | - Myungshin Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- *Correspondence: Nack-Gyun Chung, ; Myungshin Kim,
| |
Collapse
|
84
|
Schilstra CE, McCleary K, Fardell JE, Donoghoe MW, McCormack E, Kotecha RS, Lourenco RDA, Ramachandran S, Cockcroft R, Conyers R, Cross S, Dalla-Pozza L, Downie P, Revesz T, Osborn M, Alvaro F, Wakefield CE, Marshall GM, Mateos MK, Trahair TN. Prospective longitudinal evaluation of treatment-related toxicity and health-related quality of life during the first year of treatment for pediatric acute lymphoblastic leukemia. BMC Cancer 2022; 22:985. [PMID: 36109702 PMCID: PMC9479356 DOI: 10.1186/s12885-022-10072-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 09/09/2022] [Indexed: 01/19/2023] Open
Abstract
Background Pediatric acute lymphoblastic leukemia (ALL) therapy is accompanied by treatment-related toxicities (TRTs) and impaired quality of life. In Australia and New Zealand, children with ALL are treated with either Children’s Oncology Group (COG) or international Berlin-Frankfurt-Munster (iBFM) Study Group-based therapy. We conducted a prospective registry study to document symptomatic TRTs (venous thrombosis, neurotoxicity, pancreatitis and bone toxicity), compare TRT outcomes to retrospective TRT data, and measure the impact of TRTs on children’s general and cancer-related health-related quality of life (HRQoL) and parents’ emotional well-being. Methods Parents of children with newly diagnosed ALL were invited to participate in the ASSET (Acute Lymphoblastic Leukaemia Subtypes and Side Effects from Treatment) study and a prospective, longitudinal HRQoL study. TRTs were reported prospectively and families completed questionnaires for general (Healthy Utility Index Mark 3) and cancer specific (Pediatric Quality of Life Inventory (PedsQL)-Cancer Module) health related quality of life as well the Emotion Thermometer to assess emotional well-being. Results Beginning in 2016, 260 pediatric patients with ALL were enrolled on the TRT registry with a median age at diagnosis of 59 months (range 1–213 months), 144 males (55.4%), majority with Pre-B cell immunophenotype, n = 226 (86.9%), 173 patients (66.5%) treated according to COG platform with relatively equal distribution across risk classification sub-groups. From 2018, 79 families participated in the HRQoL study through the first year of treatment. There were 74 TRT recorded, reflecting a 28.5% risk of developing a TRT. Individual TRT incidence was consistent with previous studies, being 7.7% for symptomatic VTE, 11.9% neurotoxicity, 5.4% bone toxicity and 5.0% pancreatitis. Children’s HRQoL was significantly lower than population norms throughout the first year of treatment. An improvement in general HRQoL, measured by the HUI3, contrasted with the lack of improvement in cancer-related HRQoL measured by the PedsQL Cancer Module over the first 12 months. There were no persisting differences in the HRQoL impact of COG compared to iBFM therapy. Conclusions It is feasible to prospectively monitor TRT incidence and longitudinal HRQoL impacts during ALL therapy. Early phases of ALL therapy, regardless of treatment platform, result in prolonged reductions in cancer-related HRQoL. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10072-x.
Collapse
|
85
|
Fazio G, Bresolin S, Silvestri D, Quadri M, Saitta C, Vendramini E, Buldini B, Palmi C, Bardini M, Grioni A, Rigamonti S, Galbiati M, Mecca S, Savino AM, Peloso A, Tu JW, Bhatia S, Borkhardt A, Micalizzi C, Lo Nigro L, Locatelli F, Conter V, Rizzari C, Valsecchi MG, te Kronnie G, Biondi A, Cazzaniga G. PAX5 fusion genes are frequent in poor risk childhood acute lymphoblastic leukaemia and can be targeted with BIBF1120. EBioMedicine 2022; 83:104224. [PMID: 35985167 PMCID: PMC9403348 DOI: 10.1016/j.ebiom.2022.104224] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/07/2022] [Accepted: 07/30/2022] [Indexed: 10/31/2022] Open
Abstract
Background Methods Findings Interpretation Funding
Collapse
|
86
|
Kroeze E, Arias Padilla L, Bakker M, Boer JM, Hagleitner MM, Burkhardt B, Mori T, Attarbaschi A, Verdú-Amorós J, Pillon M, Anderzhanova L, Kabíčková E, Chiang AKS, Kebudi R, Mellgren K, Lazic J, Jazbec J, Meijerink JPP, Beishuizen A, Loeffen JLC. Pediatric Precursor B-Cell Lymphoblastic Malignancies: From Extramedullary to Medullary Involvement. Cancers (Basel) 2022; 14:cancers14163895. [PMID: 36010889 PMCID: PMC9405801 DOI: 10.3390/cancers14163895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/22/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
B-cell lymphoblastic lymphoma (BCP-LBL) and B-cell acute lymphoblastic leukemia (BCP-ALL) are the malignant counterparts of immature B-cells. BCP-ALL is the most common hematological malignancy in childhood, while BCP-LBL accounts for only 1% of all hematological malignancies in children. Therefore, BCP-ALL has been well studied and treatment protocols have changed over the last decades, whereas treatment for BCP-LBL has stayed roughly the same. Clinical characteristics of 364 pediatric patients with precursor B-cell malignancies were studied, consisting of BCP-LBL (n = 210) and BCP-ALL (n = 154) patients. Our results indicate that based on the clinical presentation of disease, B-cell malignancies probably represent a spectrum ranging from complete isolated medullary disease to apparent complete extramedullary disease. Hepatosplenomegaly and peripheral blood involvement are the most important discriminators, as both seen in 80% and 95% of the BCP-ALL patients and in 2% of the BCP-LBL patients, respectively. In addition, we show that the overall survival rates in this cohort differ significantly between BCP-LBL and BCP-ALL patients aged 1−18 years (p = 0.0080), and that the outcome for infants (0−1 years) with BCP-LBL is significantly decreased compared to BCP-LBL patients of all other pediatric ages (p < 0.0001).
Collapse
Affiliation(s)
- Emma Kroeze
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Laura Arias Padilla
- NHL-BFM Study Center and Pediatric Hematology and Oncology, University Hospital Muenster, 48149 Muenster, Germany
| | - Max Bakker
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Judith M. Boer
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | | | - Birgit Burkhardt
- NHL-BFM Study Center and Pediatric Hematology and Oncology, University Hospital Muenster, 48149 Muenster, Germany
| | - Takeshi Mori
- Department of Hematology and Oncology, Hyogo Prefectural Kobe Children’s Hospital, Kobe 650-0047, Japan
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children’s Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Jaime Verdú-Amorós
- Pediatric Oncology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Marta Pillon
- Clinic of Pediatric Hemato-Oncology, Department of Women’s and Children’s Health, University of Padova, 46010 Padova, Italy
| | - Liliya Anderzhanova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117198 Moscow, Russia
| | - Edita Kabíčková
- Department of Pediatric Hematology and Oncology, Charles University, 2nd Medical School and University Hospital Motol, 150 06 Prague, Czech Republic
| | - Alan K. S. Chiang
- Department of Pediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Rejin Kebudi
- Division of Pediatric Hematology-Oncology, Istanbul University, Oncology Institute, 34452 İstanbul, Turkey
| | - Karin Mellgren
- Department of Pediatric Oncology, Sahlgrenska University Hospital, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Jelena Lazic
- Department for Hematology and Oncology, University Children’s Hospital, University of Belgrade, 11000 Belgrade, Serbia
| | - Janez Jazbec
- Division of Pediatrics, Hematology and Oncology, University Medical Center Ljubljana, SI-1000 Ljubljana, Slovenia
| | | | - Auke Beishuizen
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Erasmus Medical Center, Sophia Children’s Hospital, 3015 CN Rotterdam, The Netherlands
| | - Jan L. C. Loeffen
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Correspondence:
| |
Collapse
|
87
|
Dai Q, Liu R, Wang Y, Ye L, Peng L, Shi R, Guo S, He J, Yang H, Zhang G, Jiang Y. Longer Time Intervals From Symptom Onset to Diagnosis Affect the Overall Survival in Children With Acute Lymphoblastic Leukemia. J Pediatr Hematol Oncol 2022; 44:285-292. [PMID: 34699460 DOI: 10.1097/mph.0000000000002344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/08/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Early diagnosis and timely treatment are essential for effective cancer control and have been widely analyzed in childhood cancer. However, few studies have described the time to diagnosis and treatment in children with ALL. This study investigated delays in diagnosis and treatment initiation and their impact on survival. METHODS This retrospective cohort study included 419 patients 0 to 14 years old at a tertiary hospital between 2011 and 2015. The optimal cutoff values for delays were determined by X-tile software. The Kaplan-Meier method and Cox regression models were used to evaluate the impact of delays on survival. RESULTS The median diagnosis, treatment, and total delays were 21 (interquartile range [IQR]: 11-35), 4 (IQR: 2-7), and 26 (IQR: 16-43) days, respectively. The results of multivariate analyses showed that diagnosis delay, risk stratification, and minimal residual disease level were independent predictors for treatment outcome in childhood ALL. CONCLUSIONS These findings suggested that a longer time to diagnosis negatively affected the clinical outcome of childhood ALL. Reducing the time to diagnosis could help to improve survival in these patients.
Collapse
Affiliation(s)
- Qingkai Dai
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Rui Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Yuefang Wang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Lei Ye
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Luyun Peng
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Rui Shi
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Siqi Guo
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Jiajing He
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Hao Yang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Ge Zhang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Yongmei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
88
|
Poyer F, Dieckmann K, Dworzak M, Tamesberger M, Haas O, Jones N, Nebral K, Köhrer S, Moser R, Kropshofer G, Peters C, Urban C, Mann G, Pötschger U, Attarbaschi A. Second malignant neoplasms after treatment of 1487 children and adolescents with acute lymphoblastic leukemia-A population-based analysis of the Austrian ALL-BFM Study Group. EJHAEM 2022; 3:940-948. [PMID: 36051012 PMCID: PMC9421960 DOI: 10.1002/jha2.488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 11/07/2022]
Abstract
Second malignant neoplasms (SMN) after primary childhood acute lymphoblastic leukemia (ALL) are rare. Among 1487 ALL patients diagnosed between 1981 and 2010 in Austria, the 10-year cumulative incidence of an SMN was 1.1% ± 0.3%. There was no difference in the 10-year incidence of SMNs with regard to diagnostic-, response- and therapy-related ALL characteristics except for a significantly higher incidence in patients with leukocytes ≥50.0 G/L at ALL diagnosis (2.1% ± 1.0% vs. 0% for 20.0-50.0 G/L, and 1.0% ± 0.3% for < 20.0 G/L; p = 0.033). Notably, there was no significant difference in the incidence of SMNs between patients with or without cranial radiotherapy (1.2% ± 0.5% vs. 0.8% ± 0.3%; p = 0.295). Future strategies must decrease the incidence of SMNs, as this event still leads to death in one-third (7/19) of the patients.
Collapse
Affiliation(s)
- Fiona Poyer
- Department of Pediatric Hematology and Oncology, St. Anna Children's HospitalMedical University of ViennaViennaAustria
| | - Karin Dieckmann
- Department of RadiotherapyMedical University of ViennaViennaAustria
| | - Michael Dworzak
- Department of Pediatric Hematology and Oncology, St. Anna Children's HospitalMedical University of ViennaViennaAustria
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
| | - Melanie Tamesberger
- Department of Pediatrics and Adolescent MedicineKepler University Hospital LinzLinzAustria
| | - Oskar Haas
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
- Labdia DiagnosticsViennaAustria
| | - Neil Jones
- Department of Pediatrics and Adolescent MedicineUniversity Clinics SalzburgSalzburgAustria
| | | | - Stefan Köhrer
- Department of Pediatric Hematology and Oncology, St. Anna Children's HospitalMedical University of ViennaViennaAustria
- Labdia DiagnosticsViennaAustria
| | - Reinhard Moser
- Department of Pediatrics and Adolescent MedicineState Hospital LeobenLeobenAustria
| | - Gabriele Kropshofer
- Division of Pediatric Hematology and Oncology and Stem Cell Transplantation, Department of Pediatrics and Adolescent MedicineMedical University of InnsbruckInnsbruckAustria
| | - Christina Peters
- Department of Pediatric Hematology and Oncology, St. Anna Children's HospitalMedical University of ViennaViennaAustria
| | - Christian Urban
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent MedicineMedical University of GrazGrazAustria
| | - Georg Mann
- Department of Pediatric Hematology and Oncology, St. Anna Children's HospitalMedical University of ViennaViennaAustria
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
| | - Ulrike Pötschger
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's HospitalMedical University of ViennaViennaAustria
| | | |
Collapse
|
89
|
Ulrikh E, Kalinina E, Dikareva E, Komlichenko E, Li O, Zhamborova O, Rizhinashvili I, Dzharbaeva A, Govorov I, Artemenko V, Bezrukikh V, Salogub G, Pervunina T, Urmancheeva A. Personalized treatment of malignant tumors during pregnancy. Medicine (Baltimore) 2022; 101:e29803. [PMID: 35777052 PMCID: PMC9239643 DOI: 10.1097/md.0000000000029803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The combination of pregnancy and cancer is a challenge for the patient and a problematic clinical dilemma for the doctor. In this retrospective observational cohort study, we have tried to analyze our experience in the management of such patients. This review includes 41 patients with malignant neoplasms detected during pregnancy who received treatment at the Almazov National Medical Research Centre from 2015-2021. The majority of patients received treatment during pregnancy (n=26, 63.4%): chemotherapy - 19 (46.3%) (in 2 cases in combination with surgery), surgical treatment - 7 (17, 1%) patients. In most cases, delivery was at term (n=28, 68.3%). All children born at term were mature and had no growth restriction, regardless of whether the mothers received treatment during pregnancy or not. When detecting cancer during pregnancy, an immediate follow-up examination is required to assess the extent of the tumor and current fetal state. If pregnancy prolongation is requested, the treatment should not be postponed, except for systemic chemotherapy in the first trimester of pregnancy, pelvic radiation at any term.
Collapse
Affiliation(s)
- E. Ulrikh
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
- North-Western State Medical University named after I.I. Mechnikov, St. Petersburg, Russia
- *Correspondence: Elena Ulrikh, Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia (e-mail: )
| | - E. Kalinina
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - E. Dikareva
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - E. Komlichenko
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - O. Li
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - O. Zhamborova
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - I. Rizhinashvili
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - A. Dzharbaeva
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - I. Govorov
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - V. Artemenko
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - V. Bezrukikh
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - G. Salogub
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - T. Pervunina
- Personalised medicine center, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - A. Urmancheeva
- North-Western State Medical University named after I.I. Mechnikov, St. Petersburg, Russia
- National Medical Research Center of Oncology named after N.N. Petrov, St. Petersburg, Russia
| |
Collapse
|
90
|
Bone Marrow Stromal Cell Regeneration Profile in Treated B-Cell Precursor Acute Lymphoblastic Leukemia Patients: Association with MRD Status and Patient Outcome. Cancers (Basel) 2022; 14:cancers14133088. [PMID: 35804860 PMCID: PMC9265080 DOI: 10.3390/cancers14133088] [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] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 12/10/2022] Open
Abstract
For the last two decades, measurable residual disease (MRD) has become one of the most powerful independent prognostic factors in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). However, the effect of therapy on the bone marrow (BM) microenvironment and its potential relationship with the MRD status and disease free survival (DFS) still remain to be investigated. Here we analyzed the distribution of mesenchymal stem cells (MSC) and endothelial cells (EC) in the BM of treated BCP-ALL patients, and its relationship with the BM MRD status and patient outcome. For this purpose, the BM MRD status and EC/MSC regeneration profile were analyzed by multiparameter flow cytometry (MFC) in 16 control BM (10 children; 6 adults) and 1204 BM samples from 347 children and 100 adult BCP-ALL patients studied at diagnosis (129 children; 100 adults) and follow-up (824 childhood samples; 151 adult samples). Patients were grouped into a discovery cohort (116 pediatric BCP-ALL patients; 338 samples) and two validation cohorts (74 pediatric BCP-ALL, 211 samples; and 74 adult BCP-ALL patients; 134 samples). Stromal cells (i.e., EC and MSC) were detected at relatively low frequencies in all control BM (16/16; 100%) and in most BCP-ALL follow-up samples (874/975; 90%), while they were undetected in BCP-ALL BM at diagnosis. In control BM samples, the overall percentage of EC plus MSC was higher in children than adults (p = 0.011), but with a similar EC/MSC ratio in both groups. According to the MRD status similar frequencies of both types of BM stromal cells were detected in BCP-ALL BM studied at different time points during the follow-up. Univariate analysis (including all relevant prognostic factors together with the percentage of stromal cells) performed in the discovery cohort was used to select covariates for a multivariate Cox regression model for predicting patient DFS. Of note, an increased percentage of EC (>32%) within the BCP-ALL BM stromal cell compartment at day +78 of therapy emerged as an independent unfavorable prognostic factor for DFS in childhood BCP-ALL in the discovery cohort—hazard ratio (95% confidence interval) of 2.50 (1−9.66); p = 0.05—together with the BM MRD status (p = 0.031). Further investigation of the predictive value of the combination of these two variables (%EC within stromal cells and MRD status at day +78) allowed classification of BCP-ALL into three risk groups with median DFS of: 3.9, 3.1 and 1.1 years, respectively (p = 0.001). These results were confirmed in two validation cohorts of childhood BCP-ALL (n = 74) (p = 0.001) and adult BCP-ALL (n = 40) (p = 0.004) treated at different centers. In summary, our findings suggest that an imbalanced EC/MSC ratio in BM at day +78 of therapy is associated with a shorter DFS of BCP-ALL patients, independently of their MRD status. Further prospective studies are needed to better understand the pathogenic mechanisms involved.
Collapse
|
91
|
Lu Y, Li Z, Lim EH, Huan PT, Kham SKY, Yeoh AEJ. Digital PCR for Minimal Residual Disease Quantitation Using Immunoglobulin/T-Cell Receptor Gene Rearrangements in Acute Lymphoblastic Leukemia: A Proposed Analytic Algorithm. J Mol Diagn 2022; 24:655-665. [PMID: 35390515 DOI: 10.1016/j.jmoldx.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/11/2022] [Accepted: 03/04/2022] [Indexed: 10/18/2022] Open
Abstract
In minimal residual disease (MRD), where there are exceedingly low target copy numbers, digital PCR (dPCR) can improve MRD quantitation. However, standards for dPCR MRD interpretation in acute lymphoblastic leukemia are lacking. Here, for immunoglobulin/T-cell receptor-based MRD, we propose an objective, statistics-based analytic algorithm. In 161 postinduction samples from 79 children with acute lymphoblastic leukemia, MRD was performed by dPCR and real-time quantitative PCR (qPCR) using the same markers and primer-probe sets. The dPCR raw data were analyzed by using an automated algorithm. dPCR and qPCR results were highly concordant (P < 0.0001): 98% (50 of 51) of qPCR positive were positive by dPCR, whereas 95% (61 of 64) of qPCR negative results were also negative by dPCR. For MRD quantitation, both qPCR and dPCR were tightly correlated (R2 = 0.94). Using more DNA (1 μg × 7 versus 630 ng × 3), dPCR improved sensitivity of MRD quantitation by one log10 (median MRD positive cutoff 1.6 × 10-5). With dPCR, 83% (29 of 35) of positive-not-quantifiable results by qPCR could be assigned positive/negative MRD status. Seven replicates of tested samples and negative controls were optimal. Compared with qPCR, dPCR could improve MRD sensitivity by one log10. We proposed an automatable, statistics-based algorithm that minimized interoperator variance for dPCR MRD.
Collapse
Affiliation(s)
- Yi Lu
- VIVA-NUS Centre for Translational Research in Acute Leukemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zhenhua Li
- VIVA-NUS Centre for Translational Research in Acute Leukemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Evelyn Huizi Lim
- VIVA-NUS Centre for Translational Research in Acute Leukemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Pei Tee Huan
- VIVA-NUS Centre for Translational Research in Acute Leukemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shirley Kow Yin Kham
- VIVA-NUS Centre for Translational Research in Acute Leukemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Allen Eng-Juh Yeoh
- VIVA-NUS Centre for Translational Research in Acute Leukemia, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; VIVA-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| |
Collapse
|
92
|
Starza ID, Eckert C, Drandi D, Cazzaniga G. Minimal Residual Disease Analysis by Monitoring Immunoglobulin and T-Cell Receptor Gene Rearrangements by Quantitative PCR and Droplet Digital PCR. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2453:79-89. [PMID: 35622321 DOI: 10.1007/978-1-0716-2115-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Analysis of immunoglobulin and T-cell receptor gene rearrangements by real-time quantitative polymerase chain reaction (RQ-PCR) is the gold standard for sensitive and accurate minimal residual disease (MRD) monitoring; it has been extensively standardized and guidelines have been developed within the EuroMRD consortium ( www.euromrd.org ). However, new generations of PCR-based methods are standing out as potential alternatives to RQ-PCR, such as digital PCR technology (dPCR), the third-generation implementation of conventional PCR, which has the potential to overcome some of the limitations of RQ-PCR such as allowing the absolute quantification of nucleic acid targets without the need for a calibration curve. During the last years, droplet digital PCR (ddPCR) technology has been compared to RQ-PCR in several hematologic malignancies showing its proficiency for MRD analysis. So far, no established guidelines for ddPCR MRD analysis and data interpretation have been defined and its potential is still under investigation. However, a major standardization effort is underway within the EuroMRD consortium ( www.euromrd.org ) for future application of ddPCR in standard clinical practice.
Collapse
Affiliation(s)
- Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy.,GIMEMA Foundation, Rome, Italy
| | - Cornelia Eckert
- Department of Pediatric Oncology Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium, and German Cancer Research Center, Heidelberg, Germany
| | - Daniela Drandi
- Hematology Division, Department of Molecular Biotechnology and Health sciences, University of Torino, Torino, Italy
| | - Giovanni Cazzaniga
- Centro Ricerca Tettamanti, Fondazione Tettamanti, Centro Maria Letizia Verga, Monza, Italy. .,Genetics, Department of Medicine and Surgery, University of Milan Bicocca, Monza, Italy.
| | | |
Collapse
|
93
|
Piktel D, Nair RR, Rellick SL, Geldenhuys WJ, Martin KH, Craig MD, Gibson LF. Pitavastatin Is Anti-Leukemic in a Bone Marrow Microenvironment Model of B-Lineage Acute Lymphoblastic Leukemia. Cancers (Basel) 2022; 14:cancers14112681. [PMID: 35681662 PMCID: PMC9179467 DOI: 10.3390/cancers14112681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Chemoresistance after chemotherapy is a negative prognostic indicator for B-cell acute lymphoblastic leukemia (ALL), necessitating the search for novel therapies. By growing ALL cells together with bone marrow stromal cells, we developed a chemoresistant ALL model. Using this model, we found that the lipid lowering drug pitavastatin had antileukemic activity in this chemoresistant co-culture model. Our data suggests that pitavastatin may be a novel treatment option for repurposing in chemoresistant, relapse ALL. Abstract The lack of complete therapeutic success in the treatment of B-cell acute lymphoblastic leukemia (ALL) has been attributed, in part, to a subset of cells within the bone marrow microenvironment that are drug resistant. Recently, the cholesterol synthesis inhibitor, pitavastatin (PIT), was shown to be active in acute myeloid leukemia, prompting us to evaluate it in our in vitro co-culture model, which supports a chemo-resistant ALL population. We used phospho-protein profiling to evaluate the use of lipid metabolic active compounds in these chemo-resistant cells, due to the up-regulation of multiple active survival signals. In a co-culture with stromal cells, a shift towards anabolic processes occurred, which was further confirmed by assays showing increased lipid content. The treatment of REH leukemia cells with pitavastatin in the co-culture model resulted in significantly higher leukemic cell death than exposure to the standard-of-care chemotherapeutic agent, cytarabine (Ara-C). Our data demonstrates the use of pitavastatin as a possible alternative treatment strategy to improve patient outcomes in chemo-resistant, relapsed ALL.
Collapse
Affiliation(s)
- Debbie Piktel
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
| | - Rajesh R. Nair
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
| | - Stephanie L. Rellick
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
| | - Werner J. Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA;
| | - Karen H. Martin
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | | | - Laura F. Gibson
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Correspondence: ; Tel.: +1-304-293-7206
| |
Collapse
|
94
|
Kośmider K, Karska K, Kozakiewicz A, Lejman M, Zawitkowska J. Overcoming Steroid Resistance in Pediatric Acute Lymphoblastic Leukemia-The State-of-the-Art Knowledge and Future Prospects. Int J Mol Sci 2022; 23:ijms23073795. [PMID: 35409154 PMCID: PMC8999045 DOI: 10.3390/ijms23073795] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/20/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy among children. Despite the enormous progress in ALL therapy, resulting in achieving a 5-year survival rate of up to 90%, the ambitious goal of reaching a 100% survival rate is still being pursued. A typical ALL treatment includes three phases: remission induction and consolidation and maintenance, preceded by a prednisone prephase. Poor prednisone response (PPR) is defined as the presence of ≥1.0 × 109 blasts/L in the peripheral blood on day eight of therapy and results in significantly frequent relapses and worse outcomes. Hence, identifying risk factors of steroid resistance and finding methods of overcoming that resistance may significantly improve patients' outcomes. A mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK-ERK) pathway seems to be a particularly attractive target, as its activation leads to steroid resistance via a phosphorylating Bcl-2-interacting mediator of cell death (BIM), which is crucial in the steroid-induced cell death. Several mutations causing activation of MAPK-ERK were discovered, notably the interleukin-7 receptor (IL-7R) pathway mutations in T-cell ALL and rat sarcoma virus (Ras) pathway mutations in precursor B-cell ALL. MAPK-ERK pathway inhibitors were demonstrated to enhance the results of dexamethasone therapy in preclinical ALL studies. This report summarizes steroids' mechanism of action, resistance to treatment, and prospects of steroids therapy in pediatric ALL.
Collapse
Affiliation(s)
- Kamil Kośmider
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland; (K.K.); (A.K.)
| | - Katarzyna Karska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland;
| | - Agata Kozakiewicz
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland; (K.K.); (A.K.)
| | - Monika Lejman
- Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland;
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland;
- Correspondence:
| |
Collapse
|
95
|
Quantification of Minimal Disease by Digital PCR in ALK-Positive Anaplastic Large Cell Lymphoma: A Step towards Risk Stratification in International Trials? Cancers (Basel) 2022; 14:cancers14071703. [PMID: 35406475 PMCID: PMC8996924 DOI: 10.3390/cancers14071703] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 02/04/2023] Open
Abstract
Minimal disseminated and residual disease (MDD/MRD) analyzed by qualitative PCR for NPM-ALK fusion transcripts are validated prognostic factors in pediatric ALK-positive anaplastic large cell lymphoma (ALCL). Although potentially promising, MDD quantification by quantitative real-time PCR in international trials is technically challenging. Quantification of early MRD might further improve risk stratification. We aimed to assess droplet digital PCR for quantification of minimal disease in an inter-laboratory setting in a large cohort of 208 uniformly treated ALCL patients. Inter-laboratory quality control showed high concordance. Using a previously described cut-off of 30 copies NPM-ALK/104 copies ABL1 (NCN) in bone marrow and peripheral blood, MDD quantification allowed identification of very high-risk patients (5-year PFS% 34 ± 5 for patients with ≥30 NCN compared to 74 ± 6 and 76 ± 5 for patients with negative or <30 NCN, respectively, p < 0.0001). While MRD positivity was confirmed as a prognostic marker for the detection of very high-risk patients in this large study, quantification of MRD fusion transcripts did not improve stratification. PFS% was 80 ± 5 and 73 ± 6 for MDD- and MRD-negative patients, respectively, versus 35 ± 10 and 16 ± 8 for MRD-positive patients with <30 and ≥30 NCN, p < 0.0001. Our results suggest that MDD quantification by dPCR enables improved patient stratification in international clinical studies and patient selection for early clinical trials already at diagnosis.
Collapse
|
96
|
Popov A, Henze G, Roumiantseva J, Budanov O, Belevtsev M, Verzhbitskaya T, Boyakova E, Movchan L, Tsaur G, Fadeeva M, Lagoyko S, Zharikova L, Miakova N, Litvinov D, Khlebnikova O, Streneva O, Stolyarova E, Ponomareva N, Novichkova G, Fechina L, Aleinikova O, Karachunskiy A. A simple algorithm with one flow cytometric MRD measurement identifies more than 40% of children with ALL who can be cured with low-intensity therapy. The ALL-MB 2008 trial results. Leukemia 2022; 36:1382-1385. [PMID: 35322171 DOI: 10.1038/s41375-022-01542-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/28/2022] [Accepted: 03/08/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Alexander Popov
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation.
| | - Guenter Henze
- Department of Pediatric Oncology Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Roumiantseva
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Oleg Budanov
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation.,Belarussian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Mikhail Belevtsev
- Belarussian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Tatiana Verzhbitskaya
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Elena Boyakova
- Moscow City Blood Center named after OK Gavrilov, Moscow, Russian Federation
| | - Liudmila Movchan
- Belarussian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Grigory Tsaur
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Maria Fadeeva
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Svetlana Lagoyko
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Liudmila Zharikova
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Natalia Miakova
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Dmitry Litvinov
- National Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | | | - Olga Streneva
- Regional Children's Hospital, Ekaterinburg, Russian Federation.,Research Institute of Medical Cell Technologies, Ekaterinburg, Russian Federation
| | - Elena Stolyarova
- Belarussian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | | | - Galina Novichkova
- 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
| | - Olga Aleinikova
- Belarussian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus.,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
| |
Collapse
|
97
|
Jensen KS, Oskarsson T, Lähteenmäki PM, Flaegstad T, Jónsson ÓG, Svenberg P, Schmiegelow K, Heyman M, Norén-Nyström U, Schrøder H, Albertsen BK. Temporal changes in incidence of relapse and outcome after relapse of childhood acute lymphoblastic leukemia over three decades; a Nordic population-based cohort study. Leukemia 2022; 36:1274-1282. [PMID: 35314777 DOI: 10.1038/s41375-022-01540-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/11/2022] [Accepted: 03/01/2022] [Indexed: 11/09/2022]
Abstract
Relapse remains the main obstacle to curing childhood acute lymphoblastic leukemia (ALL). The aims of this study were to compare incidence of relapse, prognostic factors, and survival after relapse between three consecutive Nordic Society of Pediatric Hematology and Oncology trials. Relapse occurred as a primary event in 638 of 4 458 children (1.0-14.9 years) diagnosed with Ph-negative ALL between 1992 and 2018. The 5-year cumulative incidence of relapse was 17.3% (95% CI 15.4-19.2%) and 16.5% (95% CI 14.3-18.8%) for patients in the ALL1992 and ALL2000 trials, respectively, but decreased to 8.4% (95% CI 7.0-10.1%) for patients in the ALL2008 trial. No changes in duration of first complete remission and site of relapse were observed over time; however, high hyperdiploidy, and t(12;21) decreased in the ALL2008 trial. The 4-year overall survival after relapse was 56.6% (95% CI 52.5-60.5%) and no statistically significant temporal improvements were observed. Age ≥10 years, T-cell immunophenotype, bone-marrow involvement, early and very early relapse, hypodiploidy, and Down syndrome all independently predicted worse outcome after relapse. Improvements in the primary treatment of childhood ALL has resulted in fewer relapses. However, failure to improve outcome of remaining relapses suggests a selection of harder-to-cure relapses and calls for new therapeutic strategies.
Collapse
Affiliation(s)
- Karen Schow Jensen
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Trausti Oskarsson
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden.,Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Päivi M Lähteenmäki
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden.,Department of Pediatric and Adolescent Hematology/Oncology, Turku University Hospital, FICAN-west, and Turku University, Turku, Finland
| | - Trond Flaegstad
- Department of Pediatrics, Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway.,Department of Pediatrics, University Hospital of North Norway, Tromsø, Norway
| | | | - Petter Svenberg
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mats Heyman
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Henrik Schrøder
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitte Klug Albertsen
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark. .,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
98
|
Takahashi Y, Ishida H, Imamura T, Tamefusa K, Suenobu S, Usami I, Yumura-Yagi K, Hasegawa D, Nishimura S, Suzuki N, Hashii Y, Deguchi T, Moriya-Saito A, Kosaka Y, Kato K, Kobayashi R, Kawasaki H, Hori H, Sato A, Kudo T, Nakahata T, Oda M, Hara J, Horibe K. JACLS ALL-02 SR protocol reduced-intensity chemotherapy produces excellent outcomes in patients with low-risk childhood acute lymphoblastic leukemia. Int J Hematol 2022; 115:890-897. [PMID: 35258855 DOI: 10.1007/s12185-022-03315-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 10/18/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. As overall cure rates of childhood ALL have improved, reduction of overall treatment intensity while still ensuring excellent outcomes is imperative for low-risk patients. We report the outcomes of patients treated following the standard-risk protocol from the prospective Japan Association of Childhood Leukemia Study (JACLS) ALL-02 study, which was conducted between 2002 and 2008 for patients with newly diagnosed ALL aged 1-18 years. Of 1138 patients with B-cell precursor ALL, 388 (34.1%) were allocated to this protocol. Excellent outcomes were achieved despite the overall treatment intensity being lower than that of most contemporary protocols: 4 years event-free survival (EFS) was 92.3% and 4 years overall survival 98.2%. Patients with high hyperdiploidy (HHD) involving triple trisomy (trisomy of chromosomes 4, 10, and 17) or ETV6-RUNX1 had even better outcomes (4 years EFS 97.6% and 100%, respectively). Unique characteristics of this protocol include a selection of low-risk patients with a low initial WBC count and good early treatment response and reduction of cumulative doses of chemotherapeutic agents while maintaining dose density. In Japan, we are currently investigating the feasibility of this protocol while incorporating minimal residual disease into the patient stratification strategy.
Collapse
Affiliation(s)
- Yoshihiro Takahashi
- Department of Pediatrics, Aomori Prefectural Central Hospital, Aomori, Japan
| | - Hisashi Ishida
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465, Kajiichou, Hirokouji Kawaramachidori, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Kosuke Tamefusa
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Souichi Suenobu
- Division of General Pediatrics and Emergency Medicine, Department of Pediatrics, Oita University Faculty of Medicine, Oita, Japan
| | - Ikuya Usami
- Department of Pediatric Hematology and Oncology, Hyogo Prefectural Amagasaki General Medical Center, Hyogo, Japan
| | | | - Daiichiro Hasegawa
- Department of Hematology/Oncology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | | | - Nobuhiro Suzuki
- Department of Pediatrics, Hokkaido Medical Center for Child Health and Rehabilitation, Sapporo, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University, Suita, Japan
| | - Takao Deguchi
- Department of Pediatrics, Mie University, Tsu, Japan
| | - Akiko Moriya-Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology/Oncology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Koji Kato
- Department of Hematology Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Ryoji Kobayashi
- Department of Pediatrics, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Hirohide Kawasaki
- Department of Pediatrics, Kansai Medical University, Hirakata, Japan
| | - Hiroki Hori
- Department of Pediatrics, Mie University, Tsu, Japan
| | - Atsushi Sato
- Department of Hematology/Oncology, Miyagi Children's Hospital, Sendai, Japan
| | - Toru Kudo
- Saiseikai Nishiotaru Hospital, Otaru, Japan
| | - Tatsutoshi Nakahata
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Megumi Oda
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan
| | - Junichi Hara
- Department of Pediatric Hematology/Oncology, Osaka City General Hospital, Osaka, Japan
| | - Keizo Horibe
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
99
|
Successful Salvage of Very Early Relapse in Pediatric Acute Lymphoblastic Leukemia With Inotuzumab Ozogamicin and HLA-haploidentical Peripheral Blood Stem Cell Transplantation With Posttransplant Cyclophosphamide. J Pediatr Hematol Oncol 2022; 44:62-64. [PMID: 33512872 DOI: 10.1097/mph.0000000000002079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/20/2020] [Indexed: 11/25/2022]
Abstract
Herein, we describe a 14-year-old female patient with B-cell precursor acute lymphoblastic leukemia who relapsed in early consolidation. Minimal residual disease-negative complete remission was obtained after 1 cycle of inotuzumab ozogamicin therapy. She underwent HLA-haploidentical peripheral blood stem cell transplantation after a myeloablative conditioning regimen. Posttransplant cyclophosphamide, tacrolimus, and mycophenolate mofetil were administered for the prophylaxis of graft-versus-host disease. At 23 months, she was in complete remission. Although the administration of inotuzumab ozogamicin followed by haploidentical peripheral blood stem cell transplantation with posttransplant cyclophosphamide has been limited in children, this strategy may be an effective treatment for pediatric refractory acute lymphoblastic leukemia.
Collapse
|
100
|
Bartram J, Wright G, Adams S, Archer P, Brooks T, Edwards D, Hancock J, Knecht H, Inglott S, Mountjoy E, Roynane M, Wakeman S, Moppett J, Hubank M, Goulden N. High-throughput sequencing of peripheral blood for minimal residual disease monitoring in childhood precursor B-cell acute lymphoblastic leukemia: A prospective feasibility study. Pediatr Blood Cancer 2022; 69:e29513. [PMID: 34971078 DOI: 10.1002/pbc.29513] [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: 06/28/2021] [Revised: 10/28/2021] [Accepted: 11/20/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Minimal residual disease (MRD) measured on end-of-induction bone marrow (BM) is the most important biomarker for guiding therapy in pediatric acute lymphoblastic leukemia (ALL). Due to limited sensitivity of current approaches, peripheral blood (PB) is not a reliable source for identifying patients needing treatment changes. We sought to determine if high-throughput sequencing (HTS) (next-generation sequencing) of rearranged immunoglobulin and T-cell receptor genes can overcome this and be used to measure MRD in PB. PROCEDURE We employed a quantitative HTS approach to accurately measure MRD from one million cell equivalents of DNA from 17 PB samples collected at day 29 after induction therapy in patients with precursor B-cell ALL. We compared these results to the gold-standard real-time PCR result obtained from their paired BM samples, median follow-up 49 months. RESULTS With the increased sensitivity, detecting up to one abnormal cell in a million normal cells, we were able to detect MRD in the PB by HTS in all those patients requiring treatment intensification (MRD ≥ 0.005% in BM). CONCLUSION This is proof of principle that using the increased sensitivity of HTS, PB can be used to measure MRD and stratify children with ALL. The method is cost effective, rapid, accurate, and reproducible, with inherent advantages in children. Importantly, increasing the frequency testing by PB as opposed to intermittent BM sampling may allow extension of the dynamic range of MRD, giving a more complete picture of the kinetics of disease remission while improving relapse prediction and speed of detection.
Collapse
Affiliation(s)
- Jack Bartram
- Depatment of Haematology, Great Ormond Street Hospital for Children, London, UK.,Cancer Section, Institute of Child Health, University College London, UK
| | - Gary Wright
- Depatment of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Stuart Adams
- Depatment of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Paul Archer
- Bristol Genetics Laboratory, Southmead Hospital, North Bristol NHS Trust, UK
| | - Tony Brooks
- UCL Genomics, Institute of Child Health, University College London, UK
| | - Darren Edwards
- Cancer Section, Institute of Child Health, University College London, UK
| | - Jerry Hancock
- Bristol Genetics Laboratory, Southmead Hospital, North Bristol NHS Trust, UK
| | - Henrik Knecht
- Department of Hematology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sarah Inglott
- Depatment of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Edward Mountjoy
- School of Social and Community Medicine, University of Bristol, UK
| | - Marie Roynane
- Depatment of Haematology, Great Ormond Street Hospital for Children, London, UK
| | - Stephanie Wakeman
- Bristol Genetics Laboratory, Southmead Hospital, North Bristol NHS Trust, UK
| | - John Moppett
- Department of Paediatric Haematology/Oncology, Royal Hospital for Children, Bristol, UK
| | - Mike Hubank
- Centre for Molecular Pathology, The Royal Marsden, Sutton, UK
| | - Nick Goulden
- Depatment of Haematology, Great Ormond Street Hospital for Children, London, UK.,Trapehade, Monferran-Plavès, France
| |
Collapse
|