1
|
Gong X, Fang Q, Gu R, Qiu S, Liu K, Lin D, Zhou C, Zhang G, Gong B, Liu Y, Li Y, Liu B, Wang Y, Wei H, Mi Y, Wang J. A pediatric-inspired regimen for adolescent and adult patients with Philadelphia chromosome-negative acute lymphoblastic leukemia: a prospective study from China. Haematologica 2024; 109:3146-3156. [PMID: 38235508 PMCID: PMC11443404 DOI: 10.3324/haematol.2023.284228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Indexed: 01/19/2024] Open
Abstract
Several international centers have used and reported on pediatric-inspired regimens to treat adolescent and adult patients with Philadelphia chromosome-negative acute lymphoblastic leukemia (Ph- ALL). However, there is a lack of prospective data from the Chinese population. We performed a prospective study with a pediatric-inspired regimen (IH-2014 regimen) to treat adolescent and adult Ph- ALL patients in our center. From 2014 to 2021, a total of 415 patients aged between 14 and 65 years (median age, 27 years) were included in this study. After a median follow-up of 40.8 months, the 5-year overall survival, disease-free survival, and event-free survival rates were 53.8%, 51.1% and 45.0%, respectively. The regimen was generally well tolerated and safe, and the overall chemotherapy-related mortality was 3.6%. Age ≥40 years and persistent detectable minimal residual disease (MRD) after induction were independent prognostic factors. Traditional risk factors for adult patients combined with post-induction MRD had predictive significance for survival and relapse, which is helpful in the selection of subsequent treatment. Patients with high-risk factors who can achieve a deep MRD response after induction do not derive benefit from allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Xiaoyuan Gong
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Qiuyun Fang
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Runxia Gu
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Shaowei Qiu
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Kaiqi Liu
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Dong Lin
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Chunlin Zhou
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Guangji Zhang
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Benfa Gong
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Yuntao Liu
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Yan Li
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Bingcheng Liu
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Ying Wang
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Hui Wei
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Yingchang Mi
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600
| | - Jianxiang Wang
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600.
| |
Collapse
|
2
|
Østergaard A, Fiocco M, de Groot-Kruseman H, Moorman AV, Vora A, Zimmermann M, Schrappe M, Biondi A, Escherich G, Stary J, Imai C, Imamura T, Heyman M, Schmiegelow K, Pieters R. ETV6::RUNX1 Acute Lymphoblastic Leukemia: how much therapy is needed for cure? Leukemia 2024; 38:1477-1487. [PMID: 38844578 PMCID: PMC11216990 DOI: 10.1038/s41375-024-02287-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 07/03/2024]
Abstract
Recent trials show 5-year survival rates >95% for ETV6::RUNX1 Acute Lymphoblastic Leukemia (ALL). Since treatment has many side effects, an overview of cumulative drug doses and intensities between eight international trials is presented to characterize therapy needed for cure. A meta-analysis was performed as a comprehensive summary of survival outcomes at 5 and 10 years. For drug dose comparison in non-high risk trial arms, risk group distribution was applied to split the trials into two groups: trial group A with ~70% (range: 63.5-75%) of patients in low risk (LR) (CCLSG ALL2004, CoALL 07-03, NOPHO ALL2008, UKALL2003) and trial group B with ~45% (range: 38.7-52.7%) in LR (AIEOP-BFM ALL 2000, ALL-IC BFM ALL 2002, DCOG ALL10, JACLS ALL-02). Meta-analysis did not show evidence of heterogeneity between studies in trial group A LR and medium risk (MR) despite differences in treatment intensity. Statistical heterogeneity was present in trial group B LR and MR. Trials using higher cumulative dose and intensity of asparaginase and pulses of glucocorticoids and vincristine showed better 5-year event-free survival but similar overall survival. Based on similar outcomes between trials despite differences in therapy intensity, future trials should investigate, to what extent de-escalation is feasible for ETV6::RUNX1 ALL.
Collapse
Affiliation(s)
- Anna Østergaard
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Department of Biomedical Science, Section Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hester de Groot-Kruseman
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, The Netherlands
| | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- United Kingdom Acute Lymphoblastic Leukaemia (UKALL) study group, Liverpool, UK
| | - Ajay Vora
- United Kingdom Acute Lymphoblastic Leukaemia (UKALL) study group, Liverpool, UK
- Department of Haematology, Great Ormond Street Hospital, London, UK
| | - Martin Zimmermann
- Department of Paediatric Haematology and Oncology, Hannover Medical School, 30625, Hannover, Germany
- Berlin-Frankfurt-Münster Study Group (BFM), Frankfurt, Germany
| | - Martin Schrappe
- Berlin-Frankfurt-Münster Study Group (BFM), Frankfurt, Germany
- Department of Paediatrics, University Medical Centre Schleswig-Holstein, Kiel, Germany
| | - Andrea Biondi
- Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
- Associazione Italiana di Ematologia e Oncologia Pediatrica (AIEOP), Bologna, Italy
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Childhood Acute Lymphoblastic Leukemia study group (CoALL), Hamburg, Germany
| | - Jan Stary
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Chihaya Imai
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Children's Cancer and Leukemia Study Group (CCLSG), Nagoya, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
- Japan Childhood Leukemia Study Group (JACLS), Nagoya, Japan
| | - Mats Heyman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
- Nordic Society of Paediatric Haematology and Oncology (NOPHO), Nordic Countries, Uppsala, Sweden
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
- Nordic Society of Paediatric Haematology and Oncology (NOPHO), Nordic and Baltic Countries, Uppsala, Sweden
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Dutch Childhood Oncology Group (DCOG), Utrecht, The Netherlands.
| |
Collapse
|
3
|
Ronceray L, Dworzak M, Dieckmann K, Ebetsberger-Dachs G, Glogova E, Haas OA, Jones N, Nebral K, Moser R, Lion T, Meister B, Panzer-Grümayer R, Strehl S, Peters C, Pötschger U, Urban C, Mann G, Attarbaschi A. Prospective use of molecular minimal residual disease for risk stratification in children and adolescents with acute lymphoblastic leukemia : Long-term results of the AIEOP-BFM ALL 2000 trial in Austria. Wien Klin Wochenschr 2024; 136:405-418. [PMID: 37535134 DOI: 10.1007/s00508-023-02249-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023]
Abstract
Since 1979 Austrian children and adolescents with acute lymphoblastic leukemia (ALL) have been treated according to protocols of the Berlin-Frankfurt-Münster (BFM) study group. The Associazione Italiana di Ematologia e Oncologia Pediatrica and BFM (AIEOP-BFM) ALL 2000 study was designed to prospectively study patient stratification into three risk groups using minimal residual disease (MRD) on two time points during the patient's early disease course. The MRD levels were monitored by detection of clone-specific rearrangements of the immunoglobulin and T‑cell receptor genes applying a quantitative polymerase chain reaction-based technique. The 7‑year event-free survival (EFS) and overall survival rates for all 608 Austrian patients treated between June 1999 and December 2009 within the AIEOP-BFM 2000 study were 84 ± 2% and 91 ± 1%, respectively, with a median observation time of 6.58 years. Event-free survival for patients with precursor B‑cell and T‑cell ALL were 84 ± 2% (n = 521) and 84 ± 4% (n = 87; p = 0.460), respectively. The MRD assessment was feasible in 94% of the patients and allowed the definition of precursor B‑cell ALL patients with a low, intermediate or high risk of relapse even on top of clinically relevant subgroups. A similar finding with respect to MRD relevance in T‑ALL patients was not possible due to the small number of patients and events. Since this pivotal international AIEOP-BFM ALL 2000 trial, molecular response to treatment has been continuously used with additional refinements to stratify patients into different risk groups in all successive trials of the AIEOP-BFM ALL study group.
Collapse
Affiliation(s)
- Leila Ronceray
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Kinderspitalgasse 6, 1090, Vienna, Austria
| | - Michael Dworzak
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Kinderspitalgasse 6, 1090, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Karin Dieckmann
- Department of Radiotherapy, Medical University of Vienna, Vienna, Austria
| | - Georg Ebetsberger-Dachs
- Department of Pediatrics and Adolescent Medicine, Kepler University Hospital Linz, Linz, Austria
| | - Evgenia Glogova
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Oskar A Haas
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Neil Jones
- Department of Pediatrics and Adolescent Medicine, University Clinics Salzburg, Salzburg, Austria
| | - Karin Nebral
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Reinhard Moser
- Department of Pediatrics and Adolescent Medicine, State Hospital Leoben, Leoben, Austria
| | - Thomas Lion
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Bernhard Meister
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Sabine Strehl
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Christina Peters
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Kinderspitalgasse 6, 1090, Vienna, Austria
| | - Ulrike Pötschger
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Christian Urban
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Georg Mann
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Kinderspitalgasse 6, 1090, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Kinderspitalgasse 6, 1090, Vienna, Austria.
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
| |
Collapse
|
4
|
Hayashi H, Makimoto A, Yuza Y. Treatment of Pediatric Acute Lymphoblastic Leukemia: A Historical Perspective. Cancers (Basel) 2024; 16:723. [PMID: 38398113 PMCID: PMC10887299 DOI: 10.3390/cancers16040723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common disease in pediatric oncology. The history of developmental therapeutics for ALL began in the 1960s with the repetition of "unreliable" medical interventions against this lethal disease. By the 1990s, the development of multi-agent chemotherapy and various types of supportive care rendered ALL treatable. Highly sophisticated, molecular, diagnostic techniques have enabled highly accurate prediction of the relapse risk, and the application of risk-adapted treatments has increased the survival rate in the standard-risk group to nearly 100% in most European nations and North America. Incorporation of state-of-the-art, molecularly targeted agents and novel treatments, including cell and immunotherapy, is further improving outcomes even in the high-risk group. On the other hand, the financial burden of treating children with ALL has increased, imperiling the availability of these diagnostic and treatment strategies to patients in low- and middle-income countries (LMICs). The fundamental treatment strategy, consisting of corticosteroid and classical cytotoxic therapy, has achieved fairly good outcomes and should be feasible in LMICs as well. The present review will discuss the history of developmental therapeutics for childhood ALL in various countries through an extensive literature review with the aim of proposing a model for a treatment backbone for pediatric ALL. The discussion will hopefully benefit LMICs and be useful as a base for future clinical trials of novel treatments.
Collapse
Affiliation(s)
- Hiroshi Hayashi
- Department of Hematology/Oncology, Tokyo Metropolitan Children’s Medical Center, 2-8-29 Musashidai, Fuchu 183-8561, Tokyo, Japan; (A.M.); (Y.Y.)
| | - Atsushi Makimoto
- Department of Hematology/Oncology, Tokyo Metropolitan Children’s Medical Center, 2-8-29 Musashidai, Fuchu 183-8561, Tokyo, Japan; (A.M.); (Y.Y.)
- Department of Laboratory Medicine, Tokyo Metropolitan Children’s Medical Center, 2-8-29 Musashidai, Fuchu 183-8561, Tokyo, Japan
| | - Yuki Yuza
- Department of Hematology/Oncology, Tokyo Metropolitan Children’s Medical Center, 2-8-29 Musashidai, Fuchu 183-8561, Tokyo, Japan; (A.M.); (Y.Y.)
| |
Collapse
|
5
|
Mattano LA, Devidas M, Loh ML, Raetz EA, Chen Z, Winick NJ, Hunger SP, Carroll WL, Larsen EC. Development of osteonecrosis and improved survival in B-ALL: results of Children's Oncology Group Trial AALL0232. Leukemia 2024; 38:258-265. [PMID: 38062123 PMCID: PMC11235418 DOI: 10.1038/s41375-023-02099-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 01/14/2024]
Abstract
Osteonecrosis is a significant toxicity of acute lymphoblastic leukemia (ALL) therapy. In retrospective analyses, superior event-free survival was noted among affected adolescents in an earlier trial. We prospectively assessed osteonecrosis incidence, characteristics, and risk factors in patients 1-30 years with newly diagnosed high-risk B-ALL on COG AALL0232. Patients were randomized to induction dexamethasone vs prednisone, and interim maintenance high-dose methotrexate vs escalating-dose Capizzi methotrexate/pegaspargase. Event-free and overall survival were compared between patients with/without imaging-confirmed osteonecrosis. Osteonecrosis developed in 322/2730 eligible, evaluable patients. The 5-year cumulative incidence was 12.2%. Risk was greater in patients ≥10 years (hazard ratio [HR], 7.23; P < 0.0001), particularly females (HR, 1.37; P = 0.0057), but lower in those with asparaginase allergy (HR, 0.60; P = 0.0077). Among rapid early responders ≥10 years, risk was greater with dexamethasone (HR, 1.84; P = 0.0003) and with prednisone/Capizzi (HR, 1.45; P = 0.044), even though neither therapy was independently associated with improved survival. Patients with osteonecrosis had higher 5-year event-free (HR, 0.51; P < 0.0001) and overall survival (HR, 0.42; P < 0.0001), and this was directly attributable to reduced relapse rates (HR, 0.57; P = 0.0014). Osteonecrosis in high-risk B-ALL patients is associated with improved survival, suggesting an important role for host factors in mediating both toxicity and enhanced efficacy of specific therapies.
Collapse
Affiliation(s)
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mignon L Loh
- Division of Pediatric Hematology, Oncology, Bone Marrow Transplantation, and Cellular Therapy, Seattle Children's Hospital, Seattle, WA, USA
- The Ben Towne Center for Childhood Cancer Research, University of Washington, Seattle, WA, USA
| | - Elizabeth A Raetz
- Department of Pediatrics, New York University Langone Medical Center, New York, NY, USA
- Perlmutter Cancer Center at New York University Langone Health, New York, NY, USA
| | - Zhiguo Chen
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Naomi J Winick
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephen P Hunger
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA, USA
| | - William L Carroll
- Department of Pediatrics, New York University Langone Medical Center, New York, NY, USA
- Perlmutter Cancer Center at New York University Langone Health, New York, NY, USA
| | - Eric C Larsen
- Maine Children's Cancer Program, Scarborough, ME, USA
| |
Collapse
|
6
|
Tosta Pérez M, Herrera Belén L, Letelier P, Calle Y, Pessoa A, Farías JG. L-Asparaginase as the gold standard in the treatment of acute lymphoblastic leukemia: a comprehensive review. Med Oncol 2023; 40:150. [PMID: 37060469 DOI: 10.1007/s12032-023-02014-9] [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: 12/06/2022] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
L-Asparaginase is an antileukemic drug long approved for clinical use to treat childhood acute lymphoblastic leukemia, the most common cancer in this population worldwide. However, the efficacy and its use as a drug have been subject to debate due to the variety of adverse effects that patients treated with it present, as well as the prompt elimination in plasma, the need for multiple administrations, and high rates of allergic reactions. For this reason, the search for new, less immunogenic variants has long been the subject of study. This review presents the main aspects of the L-asparaginase enzyme from a structural, pharmacological, and clinical point of view, from the perspective of its use in chemotherapy protocols in conjunction with other drugs in the different treatment phases.
Collapse
Affiliation(s)
- María Tosta Pérez
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile
| | - Lisandra Herrera Belén
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Santiago de Chile, Chile
| | - Pablo Letelier
- Precision Health Research Laboratory, Departamento de Procesos Diagnósticos y Evaluación, Facultad de Ciencias de La Salud, Universidad Católica de Temuco, Temuco, Chile
| | - Yolanda Calle
- Department of Life Sciences, Whitelands College, University of Roehampton, London, UK
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jorge G Farías
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile.
| |
Collapse
|
7
|
Li XY, Han XW, Huang K, Zhang YT, Xu HG, Zhou DH, Xu LH, Fang JP. Chidamide as maintenance after chemotherapy or hematopoietic stem cell transplantation in 27 children with T-cell lymphoblastic leukemia: A real-world prospective study. Front Med (Lausanne) 2023; 10:1096529. [PMID: 36817761 PMCID: PMC9932021 DOI: 10.3389/fmed.2023.1096529] [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: 11/12/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Background The long-term overall survival of children with T-cell acute lymphoblastic leukemia (T-ALL) is limited to approximately 80-85% because of a high incidence of relapse after achieving remission with intensive chemotherapy and hematopoietic stem cell transplantation (HSCT). Novel treatment strategies inducing long-term remission are needed to improve the outcome. Histone deacetylase inhibitors (HDACis) have been reported to be effective in a series of T-ALL cases. Preclinical studies suggested that T-ALL cells are sensitive to Chidamide, which is a selective HDACi. Methods This preliminary clinical study evaluated the efficacy and safety of Chidamide in combination with chemotherapy or post-HSCT for children with T-ALL at a dose of 0.5 mg/kg weight of patient twice per week for at least 6 months. Results In total, 27 children with a mean age of 7.88 years were included. The high-risk proportion was 66.7%. After a median follow-up period of 37.8 months (9.5-67.9 months), the overall survival and event-free survival in the patients treated with Chidamide were 94.1 and 95.2%, respectively. All patients except two maintained persistent remission with <0.01% blast cells in minimal residual disease. Conclusion The combination therapy with Chidamide in a case series of T-ALL shows the promising clinical efficacy and good safety in children. Clinical trial registration https://www.chictr.org.cn/, identifier ChiCTR2000030357.
Collapse
Affiliation(s)
- Xin-Yu Li
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xia-Wei Han
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke Huang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ya-Ting Zhang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong-Gui Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dun-Hua Zhou
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lu-Hong Xu
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian-Pei Fang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,*Correspondence: Jian-Pei Fang,
| |
Collapse
|
8
|
Prevalence and risk factors of disseminated intravascular coagulation in childhood acute lymphoblastic leukemia. Pediatr Res 2023:10.1038/s41390-023-02475-8. [PMID: 36670158 DOI: 10.1038/s41390-023-02475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND Few studies have examined disseminated intravascular coagulation (DIC) in childhood acute lymphoblastic leukemia (ALL). Our aims were to evaluate the prevalence, risk factors and outcomes of DIC at ALL presentation and during induction chemotherapy. METHODS The medical records of ALL patients aged <15 years were retrospectively reviewed. Logistic regression analysis was used to identify risk factors. The Kaplan-Meier method was used to depict survival. RESULTS Of the 312 patients, 48 (15.4%) and 76 (24.4%) had DIC at presentation and during induction chemotherapy, respectively. Risk factors for DIC at presentation (OR and 95% CI) were antibiotics prior to admission 2.34 (1.17-4.89), white blood cell count ≥100 × 109/L 2.39 (1.04-5.72), platelets <100 × 109/L 5.44 (1.84-23.4) and high National Cancer Institute (NCI) risk 2.68 (1.08-6.62). Risk factors for DIC during induction chemotherapy were antibiotics prior to admission 1.86 (1.07-3.27), high peripheral blasts 1.01 (1.00-1.02) and transaminitis 2.02 (1.18-3.48). Five-year overall survival of patients who had DIC was significantly lower than those who did not (45.0% vs. 74.1%, p <0.001). CONCLUSION Antibiotics prior to admission, hyperleukocytosis, thrombocytopenia and high NCI risk were risk factors of DIC at presentation. Antibiotics prior to admission, high peripheral blasts and transaminitis were risk factors of DIC during induction chemotherapy. IMPACT There are only two studies, both published before 2000, evaluating risk factors of DIC in pediatric ALL patients without reporting outcomes. DIC was associated with lower remission and survival rates in pediatric ALL patients. We identified the risk factors of DIC at presentation as antibiotics prior to admission, hyperleukocytosis, thrombocytopenia and high NCI risk. The risk factors of DIC during induction chemotherapy were antibiotics prior to admission, high peripheral blasts and aspartate transaminitis. Pediatric ALL patients who have the aforementioned risk factors should be closely monitored for DIC secondary to infection, and early treatment with appropriate antimicrobial agents is recommended.
Collapse
|
9
|
A Pediatric Case of Treatment-related Myelodysplastic Syndrome While on Therapy for Pre-B Acute Lymphoblastic Leukemia. J Pediatr Hematol Oncol 2023; 45:e518-e521. [PMID: 36706304 DOI: 10.1097/mph.0000000000002613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 12/01/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Treatment-related myelodysplastic syndrome (t-MDS) is a rare late effect of cancer therapy. After alkylating agents, this typically occurs years after completion of therapy. Treatment of t-MDS in pediatrics is an allogeneic stem cell transplant, however, the prognosis remains poor. OBSERVATIONS This case demonstrates t-MDS developing in a patient receiving treatment for pre-B acute lymphoblastic leukemia. This patient was treated with a combination of hematopoietic stem cell transplant and hypomethylating agents. CONCLUSIONS These agents should be considered for use in patients with t-MDS, before transplant to limit additional chemotherapy and as maintenance therapy post-transplant to reduce the risk of relapse.
Collapse
|
10
|
Moorman AV, Antony G, Wade R, Butler ER, Enshaei A, Harrison CJ, Moppett J, Hough R, Rowntree C, Hancock J, Goulden N, Samarasinghe S, Vora A. Time to Cure for Childhood and Young Adult Acute Lymphoblastic Leukemia Is Independent of Early Risk Factors: Long-Term Follow-Up of the UKALL2003 Trial. J Clin Oncol 2022; 40:4228-4239. [PMID: 35714315 DOI: 10.1200/jco.22.00245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The aim of the randomized trial, UKALL2003, was to adjust treatment intensity on the basis of minimal residual disease (MRD) stratification for children and young adults with acute lymphoblastic leukemia. We analyzed the 10-year randomized outcomes and the time for patients to be considered cured (ClinicalTrials.gov identifier: NCT00222612). METHODS A total of 3,113 patients were analyzed including 1,054 patients who underwent random assignment (521 MRD low-risk and 533 MRD high-risk patients). Time to cure was defined as the point at which the chance of relapse was < 1%. The median follow-up time was 10.98 (interquartile range, 9.19-13.02) years, and survival rates are quoted at 10 years. RESULTS In the low-risk group, the event-free survival was 91.7% (95% CI, 87.4 to 94.6) with one course of delayed intensification versus 93.7% (95% CI, 89.9 to 96.1) with two delayed intensifications (adjusted hazard ratio, 0.73; 95% CI, 0.38 to 1.40; P = .3). In the high-risk group, the event-free survival was 82.1% (95% CI, 76.9 to 86.2) with standard therapy versus 87.1% (95% CI, 82.4 to 90.6) with augmented therapy (adjusted hazard ratio, 0.68; 95% CI, 0.44 to 1.06; P = .09). Cytogenetic high-risk patients treated on augmented therapy had a lower relapse risk (22.1%; 95% CI, 15.1 to 31.6) versus standard therapy (52.4%; 95% CI, 28.9 to 80.1; P = .016). The initial risk of relapse differed significantly by sex, age, MRD, and genetics, but the risk of relapse for all subgroups quickly coalesced at around 6 years after diagnosis. CONCLUSION Long-term outcomes of the UKALL2003 trial confirm that low-risk patients can safely de-escalate therapy, while intensified therapy benefits patients with high-risk cytogenetics. Regardless of prognosis, the time to cure is similar across risk groups. This will facilitate communication to patients and families who pose the question "When am I/is my child cured?"
Collapse
Affiliation(s)
- Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Grace Antony
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rachel Wade
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ellie R Butler
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Amir Enshaei
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christine J Harrison
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John Moppett
- Department of Paediatric Oncology, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Rachael Hough
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Clare Rowntree
- Cardiff and Vale University Health Board (UHB), Wales, United Kingdom
| | - Jeremy Hancock
- Bristol Genetics Laboratory, North Bristol NHS Trust, Bristol, United Kingdom
| | - Nicholas Goulden
- Department of Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Sujith Samarasinghe
- Department of Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Ajay Vora
- Department of Haematology, Great Ormond Street Hospital, London, United Kingdom
| |
Collapse
|
11
|
Essa MF, Abdellatif R, Elimam N, Ballourah W, Alsudairy R, Alkaiyat M, Alsultan A, Jastaniah W. Outcomes of blinatumomab based therapy in children with relapsed, persistent, or refractory acute lymphoblastic leukemia: a multicenter study focusing on predictors of response and post-treatment immunoglobulin production. Pediatr Hematol Oncol 2022; 39:613-628. [PMID: 36200934 DOI: 10.1080/08880018.2022.2049936] [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: 10/18/2022]
Abstract
The management of Refractory/Relapsed B-cell Acute Lymphoblastic Leukemia (R/R ALL) remains challenging. Incorporating blinatumomab in R/R ALL treatment has shown encouraging results. We describe the outcome and predictors of response in children receiving blinatumomab as a bridge to definitive therapy. Immunoglobulin (Ig) G and viral serology before and after therapy were evaluated. Thirty-three patients that failed standard first-line treatments due to relapsed ALL (n = 22), persistent minimal residual disease (MRD) (n = 8), or refractory disease (n = 3) received blinatumomab. Grade 2 toxicity occurred in 27.2% of patients. MRD remission (<0.01%) was achieved in 72.7% of patients. Pre-blinatumomab absolute lymphocyte count (ALC) and MRD/ALC ratio significantly associated with MRD-response. Patients with t(1;19) translocation had lower response rate, compared to all other cytogenetic categories (p = 0.013). One-year event-free survival (EFS) and overall survival (OS) were 69.2% and 79.7%, respectively. Analysis of OS and EFS showed pre-blinatumomab MRD level, ALC, MRD/ALC ratio, t(1;19), and post-blinatumomab MRD remission associated with survival. Following blinatumomab, 83% (15/18) of tested patients had low IgG levels. IgG seronegative status was observed in 83% (12/15) for varicella zoster, 35% (6/17) for herpes zoster, 18% (3/17) for cytomegalovirus, and 26% (5/17) for Epstein Barr virus. Blinatumomab produced encouraging results in children with R/R ALL and low disease burden bridging to definitive therapy. Incorporating baseline genetics and biomarkers may help identify subgroups likely to be responsive/resistant to therapy. Viral serological testing pre- and post-blinatumomab is recommended to optimize supportive and preemptive therapy.Supplemental data for this article is available online at https://doi.org/10.1080/08880018.2022.2049936 .
Collapse
Affiliation(s)
- Mohammed F Essa
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Reem Abdellatif
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Naglla Elimam
- King Abdullah International Medical Research Center, National Guard Health Affairs, Riyadh, Saudi Arabia.,Department of Oncology, Princess Noorah Oncology Center, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Waleed Ballourah
- Department of Pediatric Hematology/Oncology, Comprehensive Cancer Center, King Fahd Medical City, Riyadh, Saudi Arabia
| | - Reem Alsudairy
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohammad Alkaiyat
- Department of Oncology, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdulrahman Alsultan
- Department of Oncology, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Wasil Jastaniah
- Department of Oncology, King Faisal Specialist Hospital & Research Center (Gen.Org.), Jeddah, Saudi Arabia
| |
Collapse
|
12
|
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: 2.5] [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
|
13
|
Abstract
Hematopoietic stem cell (HSC) regeneration is the remarkable process by which extremely rare, normally inactive cells of the bone marrow can replace an entire organ if called to do so by injury or harnessed by transplantation. HSC research is arguably the first quantitative single-cell science and the foundation of adult stem cell biology. Bone marrow transplant is the oldest and most refined technique of regenerative medicine. Here we review the intertwined history of the discovery of HSCs and bone marrow transplant, the molecular and cellular mechanisms of HSC self-renewal, and the use of HSCs and their derivatives for cell therapy.
Collapse
Affiliation(s)
- Mitch Biermann
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Tannishtha Reya
- Department of Medicine, University of California San Diego, La Jolla, California 92093
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
14
|
Patel PG, Panseriya HZ, Vala AK, Dave BP, Gosai HB. Exploring current scenario and developments in the field of microbial L-asparaginase production and applications: A review. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
15
|
Jain H, Rajendra A, Sengar M, Goli VB, Thorat J, Muthuluri H, Tongaonkar AH, Kota KK, Gupta H, Sharma N, Eipe T, Mehta H. The current treatment approach to adolescents and young adults with acute lymphoblastic leukemia (AYA-ALL): challenges and considerations. Expert Rev Anticancer Ther 2022; 22:845-860. [PMID: 35734814 DOI: 10.1080/14737140.2022.2093718] [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: 11/04/2022]
Abstract
INTRODUCTION AYA-ALL differs from pediatric ALL in terms of clinical, biological, psychosocial factors and access to care and has an inferior outcome. It is now being recognized that pediatric-inspired protocols are superior to adult protocols for this cohort, but given the lack of randomized trials, several questions remain unanswered. AREAS COVERED In this review, we discuss how AYA-ALL is different from the pediatric ALL population, compare AYA ALL with ALL in middle and older age adults, review the studies that have enrolled the AYA cohort, summarize risk-stratified and response-adapted approaches, describe the biological subtypes, and review the novel agents/approaches under evaluation. EXPERT OPINION AYA-ALL is a complex and challenging disease that needs multidisciplinary and focused care. Well-designed clinical trials that focus on this cohort are needed to further improve the outcomes.
Collapse
Affiliation(s)
- Hasmukh Jain
- Tata Memorial Hospital, Parel, Mumbai, Maharashtra, India
| | - Akhil Rajendra
- Tata Memorial Hospital, Parel, Mumbai, Maharashtra, India
| | - Manju Sengar
- Tata Memorial Hospital, Parel, Mumbai, Maharashtra, India
| | - Vasu Babu Goli
- Tata Memorial Hospital, Parel, Mumbai, Maharashtra, India
| | | | | | | | | | - Himanshi Gupta
- Tata Memorial Hospital, Parel, Mumbai, Maharashtra, India
| | - Neha Sharma
- Tata Memorial Hospital, Parel, Mumbai, Maharashtra, India
| | - Thomas Eipe
- Gloria, PRRA-143, Pallissery road, Palarivattom, Ernakulam, Kerala, India
| | - Hiral Mehta
- A/31, 65-D, Bafna Courts, West Ponnurangam Road, RS Puram, Coimbatore, India
| |
Collapse
|
16
|
Mostafavi E, Zarepour A, Barabadi H, Zarrabi A, Truong LB, Medina-Cruz D. Antineoplastic activity of biogenic silver and gold nanoparticles to combat leukemia: Beginning a new era in cancer theragnostic. BIOTECHNOLOGY REPORTS 2022; 34:e00714. [PMID: 35686001 PMCID: PMC9171450 DOI: 10.1016/j.btre.2022.e00714] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 12/13/2022]
Abstract
The American Cancer Society estimated around 61,090 new cases of leukemia were diagnosed, and around 23,660 people died from this disease in the United States alone in 2021. Due to its burden on society, there is an unmet need to explore innovative approaches to overcome leukemia. Among different strategies that have been explored, nanotechnology appears to be a promising and effective approach for therapeutics. Specifically, biogenic silver and gold nanoparticles (NPs) have attracted significant attention for their antineoplastic activity toward leukemia cancer cells due to their unique physicochemical properties. Indeed, these nanostructures have emerged as useful approaches in anti-leukemic applications, either as carriers to enhance drug bioavailability and its targeted delivery to a specific organ or as a novel therapeutic agent. This review explores recent advances in green synthesized nanomaterials and their potential use against leukemia, especially focusing on silver (Ag) and gold (Au) nanostructures. In detail, we have reviewed various eco-friendly methods of bio-synthesized NPs, their analytical properties, and toxicity effects against leukemic models. This overview confirms the satisfactory potency of biogenic NPs toward leukemic cells and desirable safety profiles against human native cells, which opens a promising door toward commercializing these types of nontherapeutic agents if challenges involve clinical validations, reproducibility, and scalability could be resolved.
Collapse
|
17
|
Ahmad SM, Ahmed BS, Khidhir KG, Rahman HS. Prospective quantitative gene expression analysis of kallikrein-related peptidase KLK10 as a diagnostic biomarker for childhood acute lymphoblastic leukemia. PeerJ 2022; 10:e13489. [PMID: 35669967 PMCID: PMC9165590 DOI: 10.7717/peerj.13489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/03/2022] [Indexed: 01/17/2023] Open
Abstract
Background The most common malignancy in children is acute lymphoblastic leukemia (ALL). This study aimed to explore KLK10 mRNA expression as a potential diagnostic biomarker for ALL in children and to examine the effect of chemotherapy on KLK10 mRNA expression following the induction and after three months of receiving chemotherapy. Methods In this prospective study, total RNA was extracted from blood samples of 23 pediatric ALL patients on diagnosis, after one month and three months of receiving chemotherapy. Healthy pediatric volunteers (n = 12) were selected as control individuals. After cDNA synthesis, KLK10 mRNA gene expression levels were quantified using quantitative real-time PCR (qRT-PCR). Results KLK10 mRNA expression levels were significantly decreased in leukemic cells compared to their levels in cells of normal blood samples (p = 0.0001). KLK10 expression levels in ALL patients after one month and three months of receiving chemotherapy decreased compared to normal blood samples (p < 0.0001 and p = 0.0175 respectively). The expression level of KLK10 mRNA in ALL patients after one month of chemotherapy was decreased compared to their level on diagnosis (p = 0.4413). KLK10 mRNA expression levels in ALL patients after three months of chemotherapy were increased compared to their level on diagnosis (p = 0.0602). The ROC curve illustrated that KLK10 mRNA expression could very efficiently discriminate ALL patients from normal counterparts (AUC=0.886, 95% CI [0.7720-1.000], SE = 0.0582, p = 0.0004). Conclusion KLK10 mRNA expression could serve as a potential diagnostic molecular biomarker for ALL in children.
Collapse
Affiliation(s)
- Shwan Majid Ahmad
- Department of Biochemistry, College of Medicine, University of Sulaimani, Sulaimaniyah, Iraq
| | - Basima Sadq Ahmed
- Department of Biochemistry & Clinical Chemistry, College of Pharmacy, University of Sulaimani, Sulaimaniyah, Iraq
| | - Karzan Ghafur Khidhir
- Department of Biology, College of Science, University of Sulaimani, Sulaimaniyah, Iraq
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Sulaimani, Sulaimaniyah, Iraq
| |
Collapse
|
18
|
Valtis YK, Place AE, Silverman LB, Vrooman LM, DeAngelo DJ, Luskin MR. Orthopaedic adverse events among adolescents and adults treated with asparaginase for acute lymphoblastic leukaemia. Br J Haematol 2022; 198:421-430. [PMID: 35312041 DOI: 10.1111/bjh.18093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 01/19/2023]
Abstract
Osteonecrosis (ON) is a complication of acute lymphoblastic leukaemia (ALL) treatment with patient- (age, female sex, genetic polymorphisms, presence of metabolic syndrome) and treatment-specific (glucocorticoid type and schedule) risk factors described. The potential role of asparaginase in increasing risk of ON via effects on coagulation, lipid metabolism, and steroid clearance is now also recognised. Paediatric studies consistently identify age as a key risk factor for ON, with adolescents at higher risk than young children. Fewer studies comprehensively report on risk of ON in adults, but available evidence suggests that adolescents and young adults (AYAs) treated with corticosteroid and asparaginase-containing paediatric-inspired regimens are more at risk than older adults treated with paediatric-inspired or traditional adult regimens. There are few proven strategies to prevent or mitigate the severity of ON and other orthopaedic complications of ALL therapy. Future clinical trials should carefully ascertain orthopaedic adverse events in adults. Evidence-based guidelines should be developed for management of orthopaedic adverse events in adults being treated for ALL, especially high-risk AYAs being treated with paediatric-inspired regimens.
Collapse
Affiliation(s)
- Yannis K Valtis
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Andrew E Place
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Pediatric Hematology Oncology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lewis B Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Pediatric Hematology Oncology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lynda M Vrooman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Pediatric Hematology Oncology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Marlise R Luskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Burke MJ, Devidas M, Chen Z, Salzer WL, Raetz EA, Rabin KR, Heerema NA, Carroll AJ, Gastier-Foster JM, Borowitz MJ, Wood BL, Winick NJ, Carroll WL, Hunger SP, Loh ML, Larsen EC. Outcomes in adolescent and young adult patients (16 to 30 years) compared to younger patients treated for high-risk B-lymphoblastic leukemia: report from Children's Oncology Group Study AALL0232. Leukemia 2021; 36:648-655. [PMID: 34725453 DOI: 10.1038/s41375-021-01460-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 10/14/2021] [Accepted: 10/20/2021] [Indexed: 12/31/2022]
Abstract
Adolescent and young adult (AYA) patients 16-30 years old with high-risk acute lymphoblastic leukemia (HR-ALL) have inferior outcomes compared to younger HR-ALL patients. AALL0232 was a Phase 3 randomized Children's Oncology Group trial for newly diagnosed HR B-ALL (1-30 years). Between 2004 and 2011, 3154 patients enrolled with 3040 eligible and evaluable for induction. AYA patients comprised 20% of patients (16-21 years, n = 551; 22-30 years, n = 46). 5-year event-free survival and overall survival was 65.4 ± 2.2% and 77.4 ± 2.0% for AYA patients compared to 78.1 ± 0.9% and 87.3 ± 0.7% for younger patients (p < 0.0001). Five-year cumulative incidence of relapse was 18.5 ± 1.7% for AYA patients and 13.5 ± 0.7% for younger patients (p = 0.006), largely due to increased marrow relapses (14.0 ± 1.5% versus 9.1 ± 0.6%; p < 0.0001). Additionally, induction failure rate was higher in AYA (7.2 ± 1.1% versus 3.5 ± 0.4%; p < 0.001) and post-induction remission deaths were significantly higher in AYA (5.7 ± 1.0% versus 2.4 ± 0.3%; p < 0.0001). AALL0232 enrolled the largest number of AYA B-ALL patients to date, demonstrating significantly inferior survival and greater rates of treatment-related toxicities compared to younger patients. Although treatment intensification has improved outcomes in younger patients, they have not been associated with the same degree of improvement for older patients.
Collapse
Affiliation(s)
- Michael J Burke
- Department of Pediatrics, Children's Hospital of Wisconsin, Milwaukee, WI, USA.
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhiguo Chen
- Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, FL, USA
| | - Wanda L Salzer
- U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| | - Elizabeth A Raetz
- Department of Pediatrics, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Karen R Rabin
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Nyla A Heerema
- Department of Pathology, The Ohio State University School of Medicine, Columbus, OH, USA
| | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Michael J Borowitz
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Brent L Wood
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Naomi J Winick
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - William L Carroll
- Department of Pediatrics, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Stephen P Hunger
- Department of Pediatrics, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Eric C Larsen
- Department of Pediatrics, Maine Children's Cancer Program, Scarborough, ME, USA
| |
Collapse
|
20
|
Baek DW, Kim DY, Sohn SK, Koh Y, Jung SH, Yhim HY, Choi Y, Moon JH. Pediatric-inspired regimen with late intensification and increased dose of L-asparaginase for adult acute lymphoblastic leukemia: the KALLA 1406/1407 study. Korean J Intern Med 2021; 36:1471-1485. [PMID: 34530526 PMCID: PMC8588968 DOI: 10.3904/kjim.2021.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 08/05/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS The objective of this study was to evaluate the efficacy and feasibility of the pediatric-inspired regimen of the adult acute lymphoblastic leukemia (ALL) Working Party, the Korean Society of Hematology. METHODS Data of 99 patients with newly diagnosed ALL, who were treated with the KALLA 1406/1407 protocol, were retrospectively analyzed. All patients equally received age-adjusted daunorubicin, vincristine, and prednisolone. L-asparaginase was additionally administered to Philadelphia (Ph)-negative patients according to age, whereas Ph-positive patients received 600 mg/day of imatinib. RESULTS A total of 99 patients were enrolled in this study, of whom 62 (62.6%) were diagnosed with Ph-negative ALL and 37 (37.3%) were diagnosed with Ph-positive ALL. The median age of patients in the Ph-negative ALL group was 46 years, and that of patients in the Ph-positive ALL group was 49 years. In patients with Ph-negative ALL, 57 (92%) patients achieved complete remission (CR) and CR with incomplete hematologic recovery (CRi). Disease-free survival (DFS) and overall survival (OS) rates at 2 years were estimated to be 42% and 63%, respectively. In patients with Ph-positive ALL, 32 (86%) patients achieved CR/CRi, and 2-year DFS and OS were 31.2% and 49.1%, respectively. Patients who were able to proceed to the allogeneic hematopoietic cell transplantation and younger patients showed significantly superior survival in both Ph-negative ALL and Ph-positive ALL. Neutropenic fever and bacterial infection were the most common and severe adverse events. CONCLUSION The KALLA 1406/1407 protocol showed tolerable toxicities in adult ALL patients. Especially, younger patients had more survival benefits with KALLA 1406/1407 protocol.
Collapse
Affiliation(s)
- Dong Won Baek
- Department of Hematology/Oncology, Kyungpook National University Hospital, Daegu,
Korea
| | - Dae Young Kim
- Department of Hematology and Oncology, Ewha Womans University School of Medicine, Seoul,
Korea
| | - Sang Kyun Sohn
- Department of Hematology/Oncology, Kyungpook National University Hospital, Daegu,
Korea
| | - Youngil Koh
- Department of Hematology/Oncology, Seoul National University Hospital, Seoul,
Korea
| | - Sung-Hoon Jung
- Department of Hematology/Oncology, Chonnam National University Hwasun Hospital, Hwasun,
Korea
| | - Ho-Young Yhim
- Department of Hematology/Oncology, Jeonbuk National University Hospital, Jeonju,
Korea
| | - Yunsuk Choi
- Department of Hematology/Oncology, Ulsan University Hospital, Ulsan,
Korea
| | - Joon Ho Moon
- Department of Hematology/Oncology, Kyungpook National University Hospital, Daegu,
Korea
| |
Collapse
|
21
|
Treatment outcomes for childhood acute lymphoblastic leukemia in low-middle income country before minimal residual disease risk stratification. Cancer Epidemiol 2021; 75:102040. [PMID: 34649157 DOI: 10.1016/j.canep.2021.102040] [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: 04/16/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Outcome of childhood acute lymphoblastic leukemia (ALL) in low- and middle-income countries is lagging in many aspects including diagnosis, risk stratification, access to treatment and supportive care. OBJECTIVE to report the outcome of childhood ALL at Ain Shams University Children's Hospitals with the use of risk-based protocols before the implementation of minimal residual disease technology and to evaluate the use of double delayed intensification (DDI) in standard risk patients. METHODS Two hundred and twenty patients with ALL diagnosed between January 2005 and December 2014 were included in the study. Patients were treated according to a modified CCG 1991 and 1961 for standard and high risk respectively. Patients were stratified into three risk groups: standard risk (SR), high-risk standard arm (HR-SA), and high-risk augmented arm (HR-AA). RESULTS Among the whole cohort, the 10-year event-free survival (EFS) and overall survival (OS) were 78.1% and 84.3% respectively. Patients with Pre-B immunophenotype (IPT) had significantly better outcome than T-cell IPT (EFS 82.0% versus 58.6%, p < 0.001; OS 86.9% versus 69%, p = 0.003 for Pre-B and T-cell respectively). Among the SR group, patients treated with single delayed intensification (SDI) had comparable EFS and OS rates when compared to patients treated with DDI with EFS 82.4% versus 87.5%, p = 0.825 and OS 88.2% versus 93.5%, p = 0.638 for SDI and DDI groups, respectively. CONCLUSION The use of risk-based protocol with simple laboratory techniques resulted in acceptable survival outcome in resource limited settings. The use of double delayed intensification showed no survival advantage in patients with standard risk.
Collapse
|
22
|
Brown PA, Shah B, Advani A, Aoun P, Boyer MW, Burke PW, DeAngelo DJ, Dinner S, Fathi AT, Gauthier J, Jain N, Kirby S, Liedtke M, Litzow M, Logan A, Luger S, Maness LJ, Massaro S, Mattison RJ, May W, Oluwole O, Park J, Przespolewski A, Rangaraju S, Rubnitz JE, Uy GL, Vusirikala M, Wieduwilt M, Lynn B, Berardi RA, Freedman-Cass DA, Campbell M. Acute Lymphoblastic Leukemia, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 19:1079-1109. [PMID: 34551384 DOI: 10.6004/jnccn.2021.0042] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Guidelines for Acute Lymphoblastic Leukemia (ALL) focus on the classification of ALL subtypes based on immunophenotype and cytogenetic/molecular markers; risk assessment and stratification for risk-adapted therapy; treatment strategies for Philadelphia chromosome (Ph)-positive and Ph-negative ALL for both adolescent and young adult and adult patients; and supportive care considerations. Given the complexity of ALL treatment regimens and the required supportive care measures, the NCCN ALL Panel recommends that patients be treated at a specialized cancer center with expertise in the management of ALL This portion of the Guidelines focuses on the management of Ph-positive and Ph-negative ALL in adolescents and young adults, and management in relapsed settings.
Collapse
Affiliation(s)
- Patrick A Brown
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | - Anjali Advani
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | | | - Shira Dinner
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | - Jordan Gauthier
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Nitin Jain
- The University of Texas MD Anderson Cancer Center
| | | | | | | | - Aaron Logan
- UCSF Helen Diller Family Comprehensive Cancer Center
| | - Selina Luger
- Abramson Cancer Center at the University of Pennsylvania
| | | | | | | | | | | | - Jae Park
- Memorial Sloan Kettering Cancer Center
| | | | | | - Jeffrey E Rubnitz
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Geoffrey L Uy
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | - Beth Lynn
- National Comprehensive Cancer Network
| | | | | | | |
Collapse
|
23
|
Geyer MB, Ritchie EK, Rao AV, Vemuri S, Flynn J, Hsu M, Devlin SM, Roshal M, Gao Q, Shukla M, Salcedo JM, Maslak P, Tallman MS, Douer D, Park JH. Pediatric-inspired chemotherapy incorporating pegaspargase is safe and results in high rates of minimal residual disease negativity in adults up to age 60 with Philadelphia chromosome-negative acute lymphoblastic leukemia. Haematologica 2021; 106:2086-2094. [PMID: 33054114 PMCID: PMC8327717 DOI: 10.3324/haematol.2020.251686] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Indexed: 11/23/2022] Open
Abstract
Administration of pediatric-inspired chemotherapy to adults up to age 60 with acute lymphoblastic leukemia (ALL) is challenging in part due to toxicities of asparaginase as well as myelosuppression. We conducted a multi-center phase II clinical trial (clinicaltrials gov. Identifier: NCT01920737) investigating a pediatric-inspired regimen, based on the augmented arm of the Children’s Cancer Group 1882 protocol, incorporating six doses of pegaspargase 2,000 IU/m2, rationally synchronized to avoid overlapping toxicity with other agents. We treated 39 adults aged 20-60 years (median age 38 years) with newly-diagnosed ALL (n=31) or lymphoblastic lymphoma (n=8). Grade 3-4 hyperbilirubinemia occurred frequently and at higher rates in patients aged 40-60 years (n=18) versus 18-39 years (n=21) (44% vs. 10%, P=0.025). However, eight of nine patients rechallenged with pegaspargase did not experience recurrent grade 3-4 hyperbilirubinemia. Grade 3-4 hypertriglyceridemia and hypofibrinogenemia were common (each 59%). Asparaginase activity at 7 days post-infusion reflected levels associated with adequate asparagine depletion, even among those with antibodies to pegaspargase. Complete response (CR)/CR with incomplete hematologic recovery was observed post-induction in 38 of 39 (97%) patients. Among patients with ALL, rates of minimal residual disease negativity by multi-parameter flow cytometry were 33% and 83% following induction phase I and phase II, respectively. Event-free and overall survival at 3 years (67.8% and 76.4%) compare favorably to outcomes observed in other series. These results demonstrate pegaspargase can be administered in the context of intensive multi-agent chemotherapy to adults aged ≤60 years with manageable toxicity. This regimen may serve as an effective backbone into which novel agents may be incorporated in future frontline studies. Trial registration: https://clinicaltrials. gov/ct2/show/NCT01920737
Collapse
Affiliation(s)
- Mark B Geyer
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Center for Cell Engineering, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Ellen K Ritchie
- Weill Cornell Medical College, Hematology and Medical Oncology, Joan and Sanford I. Weill Department of Medicine, New York
| | | | | | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Meier Hsu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - Qi Gao
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - Madhulika Shukla
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Jose M Salcedo
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Peter Maslak
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Martin S Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Dan Douer
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Jae H Park
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Center for Cell Engineering, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| |
Collapse
|
24
|
Pulse therapy with vincristine and dexamethasone for childhood acute lymphoblastic leukaemia (CCCG-ALL-2015): an open-label, multicentre, randomised, phase 3, non-inferiority trial. Lancet Oncol 2021; 22:1322-1332. [PMID: 34329606 DOI: 10.1016/s1470-2045(21)00328-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Vincristine plus dexamethasone pulses are generally used throughout maintenance treatment for childhood acute lymphoblastic leukaemia. However, previous studies remain inconclusive about the benefit of this maintenance therapy and the absence of randomised, controlled trials in patients with low-risk or high-risk acute lymphoblastic leukaemia provides uncertainty. We therefore aimed to determine if this therapy could be safely omitted beyond 1 year of treatment without leading to an inferior outcome in any risk subgroup of childhood acute lymphoblastic leukaemia. METHODS This open-label, multicentre, randomised, phase 3, non-inferiority trial involved 20 major medical centres across China. We enrolled patients who were aged 0-18 years with newly diagnosed acute lymphoblastic leukaemia that was subsequently in continuous remission for 1 year after initial treatment. Patients with secondary malignancy or primary immunodeficiency were excluded. Eligible patients were classified as having low-risk, intermediate-risk, or high-risk acute lymphoblastic leukaemia based on minimal residual disease and immunophenotypic and genetic features of leukaemic cells. Randomisation and analyses were done separately for the low-risk and intermediate-to-high-risk cohorts. Randomisation was generated by the study biostatistician with a block size of six. Stratification factors included participating centre, sex, and age at diagnosis; the low-risk cohort was additionally stratified for ETV6-RUNX1 status, and the intermediate-to-high-risk cohort for cell lineage. Patients in each risk cohort were randomly assigned (1:1) to either receive (ie, the control group) or not receive (ie, the experimental group) seven pulses of intravenous vincristine (1·5 mg/m2) plus oral dexamethasone (6 mg/m2 per day for 7 days) during the second year of treatment. The primary endpoint was difference in 5-year event-free survival between the experimental group and the control group for both the low-risk and intermediate-to-high-risk cohorts, with a non-inferiority margin of 0·05 (5%). The analysis was by intention to treat. This trial is registered with the Chinese Clinical Trial Registry, ChiCTR-IPR-14005706. FINDINGS Between Jan 1, 2015, and Feb 20, 2020, 6141 paediatric patients with newly diagnosed acute lymphoblastic leukaemia were registered to this study. Approximately 1 year after diagnosis and treatment, 5054 patients in continuous remission were randomly assigned, including 2923 (1442 in the control group and 1481 in the experimental group) with low-risk acute lymphoblastic leukaemia and 2131 (1071 control, 1060 experimental) with intermediate-to-high risk acute lymphoblastic leukaemia. Median follow-up for patients who were alive at the time of analysis was 3·7 years (IQR 2·8-4·7). Among patients with low-risk acute lymphoblastic leukaemia, no difference was observed in 5-year event-free survival between the control group and the experimental group (90·3% [95% CI 88·4-92·2] vs 90·2% [88·2-92·2]; p=0·90). The one-sided 95% upper confidence bound for the difference in 5-year event-free survival probability was 0·024, establishing non-inferiority. Among patients with intermediate-to-high-risk acute lymphoblastic leukaemia, no difference was observed in 5-year event-free survival between the control group and the experimental group (82·8% [95% CI 80·0-85·7] vs 80·8% [77·7-84·0]; p=0·90), but the one-sided 95% upper confidence bound for the difference in 5-year event-free survival probability was 0·055, giving a borderline inferior result for those in the experimental group. In the low-risk cohort, we found no differences in the rates of infections, symptomatic osteonecrosis, or other complications during the second year of maintenance treatment between patients in the control and experimental groups. Patients with intermediate-to-high-risk acute lymphoblastic leukaemia in the control group were more likely to develop grade 3-4 pneumonia (26 [2·4%] of 1071 vs ten [0·9%] of 1060) and vincristine-related peripheral neuropathy (17 [1·6%] vs six [0·6%]) compared with the experimental group. Incidence of grade 5 fatal infection was similar between the control group and the experimental group in both the low-risk cohort (two [0·1%] of 1442 vs five [0·3%] of 1481) and intermediate-to-high risk cohort (six [0·6%] of 1071 vs five [0·5%] of 1060). INTERPRETATION Vincristine plus dexamethasone pulses might be omitted beyond 1 year of treatment for children with low-risk acute lymphoblastic leukaemia. Additional studies are needed for intermediate-to-high-risk acute lymphoblastic leukaemia. FUNDING VIVA China Children's Cancer Foundation, the National Natural Science Foundation of China, the China fourth round of Three-Year Public Health Action Plan (2015-2017), Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences, US National Cancer Institute, St Baldrick's Foundation, and the American Lebanese Syrian Associated Charities. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
|
25
|
Karol SE, Pei D, Smith CA, Liu Y, Yang W, Kornegay NM, Panetta JC, Crews KR, Cheng C, Finch ER, Inaba H, Metzger ML, Rubnitz JE, Ribeiro RC, Gruber TA, Yang JJ, Evans WE, Jeha S, Pui CH, Relling MV. Comprehensive analysis of dose intensity of acute lymphoblastic leukemia chemotherapy. Haematologica 2021; 107:371-380. [PMID: 34196166 PMCID: PMC8804576 DOI: 10.3324/haematol.2021.278411] [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: 01/21/2021] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy dosages are often compromised, but most reports lack data on dosages that are actually delivered. In two consecutive acute lymphoblastic leukemia trials that differed in their asparaginase formulation, native E. coli L-asparaginase in St. Jude Total 15 (T15, n=365) and pegaspargase in Total 16 (T16, n=524), we tallied the dose intensities for all drugs on the low-risk or standard-risk arms, analyzing 504,039 dosing records. The median dose intensity for each drug ranged from 61-100%. Dose intensities for several drugs were more than 10% higher on T15 than on T16: cyclophosphamide (P<0.0001 for the standard- risk arm), cytarabine (P<0.0001 for the standard-risk arm), and mercaptopurine (P<0.0001 for the low-risk arm and P<0.0001 for the standardrisk arm). We attributed the lower dosages on T16 to the higher asparaginase dosages on T16 than on T15 (P<0.0001 for both the low-risk and standard-risk arms), with higher dose-intensity for mercaptopurine in those with anti-asparaginase antibodies than in those without (P=5.62x10- 3 for T15 standard risk and P=1.43x10-4 for T16 standard risk). Neutrophil count did not differ between protocols for low-risk patients (P=0.18) and was actually lower for standard-risk patients on T16 than on T15 (P<0.0001) despite lower dosages of most drugs on T16. Patients with low asparaginase dose intensity had higher methotrexate dose intensity with no impact on prognosis. The only dose intensity measure predicting a higher risk of relapse on both studies was higher mercaptopurine dose intensity, but this did not reach statistical significance (P=0.03 T15; P=0.07 T16). In these intensive multiagent trials, higher dosages of asparaginase compromised the dosing of other drugs for acute lymphoblastic leukemia, particularly mercaptopurine, but lower chemotherapy dose intensity was not associated with relapse.
Collapse
Affiliation(s)
- Seth E Karol
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN; Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Deqing Pei
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | - Colton A Smith
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Yiwei Liu
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Wenjian Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Nancy M Kornegay
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - John C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Kristine R Crews
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | - Emily R Finch
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Monika L Metzger
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN; Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN
| | - Jeffrey E Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Raul C Ribeiro
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Tanja A Gruber
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - William E Evans
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Sima Jeha
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN; Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Mary V Relling
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN.
| |
Collapse
|
26
|
Outcomes and prognostic factors in adolescents and young adults with ALL treated with a modified BFM-90 protocol. Blood Adv 2021; 5:1178-1193. [PMID: 33635331 DOI: 10.1182/bloodadvances.2020003526] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
The use of pediatrics-inspired protocols in adolescent and young adult (AYA) acute lymphoblastic leukemia (ALL) results in superior survival compared with the adult protocols. Pediatrics-inspired protocols carry an increased risk of toxicity and treatment-related mortality in low resource settings, which can offset the potential benefits. We studied the outcomes and prognostic factors in the treatment of AYA ALL with a pediatrics-inspired regimen. We retrieved data regarding demographics, investigations, treatment details, and toxicities from the electronic medical records of patients diagnosed with ALL in the 15- to 25-year-old age group who were initiated on a modified Berlin-Frankfurt-Münster 90 (BFM-90) protocol between January 2013 and December 2016 at the Tata Memorial Centre. A total of 349 patients in the 15- to 25-year-old age group were treated with a modified BFM-90 protocol. The use of this pediatrics-inspired protocol resulted in a 3-year event-free survival (EFS) and overall survival (OS) of 59.4% and 61.8%, respectively. Only 15 patients underwent an allogeneic stem cell transplant. Minimal residual disease (MRD) persistence postinduction emerged as the only factor predictive of poor outcomes. A modified BFM-90 protocol is an effective and safe regimen for AYA ALL with an OS and EFS comparable to the published literature.
Collapse
|
27
|
Sherief LM, Beshir M, Raafat N, Abdelkhalek ER, Mokhtar WA, Elgerby KM, Soliman BK, Salah HE, Mokhtar GA, Kamal NM, ELsayed H, Zakaria M. Genetic polymorphism of vitamin D receptors and plasminogen activator inhibitor-1 and osteonecrosis risk in childhood acute lymphoblastic leukemia. Mol Genet Genomic Med 2021; 9:e1700. [PMID: 34042331 PMCID: PMC8372120 DOI: 10.1002/mgg3.1700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/11/2021] [Accepted: 04/13/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Osteonecrosis (ON) is one of the major therapy-related complications in childhood acute lymphoblastic leukemia (ALL). The purpose of the current study is to assess the frequency of ON in children with ALL and to detect whether polymorphisms in vitamin D receptor gene (VDR) and plasminogen activator inhibitor-1 (PAI-1) gene can affect the risk of ON. PATIENTS AND METHODS Nighty-six ALL children were enrolled. Serum 25-hydroxyvitamin D 25(OH)D levels were performed in addition to the detection of polymorphisms in PAI-1and VDR genes by polymerase chain reaction. RESULTS Ten out of 96 patients had ON (four males and six females aged above 10 years) and had an insufficient level of 25(OH)D. Fifty-two percent of patients had PAI-1 GG genotype while 48% had PAI-1 GA genotype. PAI-1 polymorphism was detected in 60% of all ON cases. The frequencies of VDR genotypes were CT (56.3%), CC (39.6%), and TT (4.2%). Osteonecrosis was found in eight patients with CC genotype and in two patients with CT genotype. CONCLUSION Osteonecrosis can develop early during the therapy of ALL. Older age and insufficient level of 25(OH)D were considered important risk factor for the development of osteonecrosis. PAT-1 and VDR gene polymorphism may be a genetic risk factor in its pathogenesis.
Collapse
Affiliation(s)
- Laila M Sherief
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed Beshir
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Nermin Raafat
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Elhamy R Abdelkhalek
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Wesam A Mokhtar
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Khaled M Elgerby
- Department of Radiodiagnosis, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Basma K Soliman
- Department of Radiodiagnosis, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hosam E Salah
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ghada A Mokhtar
- Department of Medical Microbiology & Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Naglaa M Kamal
- Pediatric Department, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Heba ELsayed
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Marwa Zakaria
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
28
|
Fayed D, Donia T, El-Shanshory M, Ali EMM, Mohamed TM. Evaluation of MicroRNA92, MicroRNA638 in Acute Lymphoblastic Leukemia of Egyptian Children. Asian Pac J Cancer Prev 2021; 22:1567-1572. [PMID: 34048187 PMCID: PMC8408408 DOI: 10.31557/apjcp.2021.22.5.1567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Indexed: 01/09/2023] Open
Abstract
Objective: miRNA considers a small non-coding RNA molecule that has tumor suppressor or oncogenic functions and regulates gene expression. miRNA may be involved in the pathogenesis of acute lymphoblastic leukemia (ALL). miRNA was evaluated in patients with ALL to correlate their importance in the clinical prediction and the response to chemotherapy. Subject and methods: The study population included 30 healthy control and 71 children with ALL is divided into 4 groups: healthy, newly diagnosed, remitted, and relapsed groups. We quantify miRNA 92a, miRNA 638 expression using real-time PCR in childhood ALL. Results: plasma miRNA 92a and miRNA 638 expressions were elevated in ALL cases at the time of diagnosis (2.51 and 2.19 folds), and relapsed (2.1 and 1.61 folds) than that of patients with remitted ALL. There was a positive correlation between miRNA 92a and miRNA 638 patients with ALL. Also, total leukocyte and blast correlated with miRNA 92a and miRNA 638 unlike hemoglobin, and platelets didn’t correlate with miRNA 92a and miRNA 638. The sensitivity of miRNA 92a and miRNA 638 were 41.5% and 54.7% respectively while the specificity was 100 % of miRNA 92a and miRNA 638. Conclusion: miRNA 92a and miRNA 638 are recommended to be used as potential predictive and follow-up markers in children with ALL remitted and relapsed cases.
Collapse
Affiliation(s)
- Dina Fayed
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Thoria Donia
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed El-Shanshory
- Department of Pediatric, Hematology Unit, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ehab M M Ali
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Tarek M Mohamed
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
29
|
In-Hospital Management Might Reduce Induction Deaths in Pediatric Patients With Acute Lymphoblastic Leukemia: Results From a Japanese Cohort. J Pediatr Hematol Oncol 2021; 43:39-46. [PMID: 32852400 DOI: 10.1097/mph.0000000000001926] [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: 03/05/2020] [Accepted: 06/15/2020] [Indexed: 11/25/2022]
Abstract
Induction deaths (ID) remain a critical issue in the treatment of pediatric patients with acute lymphoblastic leukemia (ALL). The reported rate of ID in this population is 1% or higher. We speculate that this proportion might be lower in Japan because of mandatory hospitalization during induction therapy to manage complications. We retrospectively analyzed the incidence of ID among children with ALL enrolled in 4 Japanese study groups between 1994 and 2013. Among 5620 children, 41 (0.73%) cases of ID were noted. The median age was 6.5 years; 24 children were female, and 7 had T-cell ALL. Infection was the most common cause of ID (n=22), but the incidence (0.39%) was lower than that reported in western countries. Mortality within 48 hours from the onset of infection was low, comprising 25% of infection-related deaths. The incidence of infections caused by Bacillus species was low. Only 1 patient died because of Aspergillus infection. Fatal infections mostly occurred during the third week of induction therapy. Our findings suggest that close monitoring, stringent infection control, and immediate administration of appropriate antibiotics through hospitalization might be important strategies in reducing the rate of infection-related ID in pediatric patients with ALL.
Collapse
|
30
|
Mateos MK, Marshall GM, Barbaro PM, Quinn MC, George C, Mayoh C, Sutton R, Revesz T, Giles JE, Barbaric D, Alvaro F, Mechinaud F, Catchpoole D, Lawson JA, Chenevix-Trench G, MacGregor S, Kotecha RS, Dalla-Pozza L, Trahair TN. Methotrexate-related central neurotoxicity: clinical characteristics, risk factors and genome-wide association study in children treated for acute lymphoblastic leukemia. Haematologica 2021; 107:635-643. [PMID: 33567813 PMCID: PMC8883571 DOI: 10.3324/haematol.2020.268565] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 11/09/2022] Open
Abstract
Symptomatic methotrexate-related central neurotoxicity, 'MTX neurotoxicity', is a severe toxicity experienced during acute lymphoblastic leukemia (ALL) therapy with potential long-term neurologic complications. Risk factors and long-term outcomes require further study. We conducted a systematic, retrospective review of 1251 consecutive Australian children enrolled on BFM or COG-based protocols between 1998-2013. Clinical risk predictors for MTX neurotoxicity were analyzed using regression. A genome-wide association study (GWAS) was performed on 48 cases and 537 controls. The incidence of MTX neurotoxicity was 7.6% (n=95/1251), at a median of 4 months from ALL diagnosis and 8 days after intravenous or intrathecal MTX. Grade 3 elevation of serum aspartate aminotransferase (P=0.005, OR 2.31 (1.28-4.16)) in induction/consolidation was associated with MTX neurotoxicity, after accounting for the only established risk factor, age a10 years. Cumulative incidence of CNS relapse was increased in children where intrathecal MTX was omitted following symptomatic MTX neurotoxicity (n=48) compared to where intrathecal MTX was continued throughout therapy (n=1174) (P=0.047). Five-year CNS relapsefree survival was 89.2%±4.6% when intrathecal MTX was ceased compared to 95.4%±0.6% when intrathecal MTX was continued. Recurrence of MTX neurotoxicity was low (12.9%) for patients whose intrathecal MTX was continued after their first episode. The GWAS identified SNPs associated with MTX neurotoxicity near genes regulating neuronal growth, neuronal differentiation and cytoskeletal organization (P>1E-06). In conclusion, increased serum aspartate aminotransferase and age a10 years at diagnosis were independent risk factors for MTX neurotoxicity. Our data do not support cessation of intrathecal MTX after a first MTX neurotoxicity event.
Collapse
Affiliation(s)
- Marion K Mateos
- Kids Cancer Centre, Sydney Children's Hospital Randwick, Sydney, Australia; School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia; Northern Institute for Cancer Research, Wolfson Childhood Cancer Research Centre, Newcastle-Upon-Tyne
| | - Glenn M Marshall
- Kids Cancer Centre, Sydney Children's Hospital Randwick, Sydney, Australia; School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney
| | - Pasquale M Barbaro
- Children's Medical Research Institute, University of Sydney, Sydney, Australia; Department of Haematology, Queensland Children's Hospital, Brisbane
| | | | - Carly George
- Perth Children's Hospital, Perth, Australia; Division of Paediatrics, School of Medicine, University of Western Australia, Perth
| | - Chelsea Mayoh
- School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney
| | - Rosemary Sutton
- School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney
| | | | - Jodie E Giles
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney
| | - Draga Barbaric
- Kids Cancer Centre, Sydney Children's Hospital Randwick, Sydney
| | - Frank Alvaro
- John Hunter Children's Hospital, Newcastle, Australia; University of Newcastle, Newcastle
| | - Françoise Mechinaud
- The Royal Children's Hospital, Melbourne, Australia; Service d'Immuno-hématologie pédiatrique Hôpital Robert-Debré, Paris
| | - Daniel Catchpoole
- The Tumour Bank, Children's Cancer Research Unit, The Children's Hospital at Westmead, Sydney
| | - John A Lawson
- School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Department of Neurology, Sydney Children's Hospital Randwick, Sydney
| | | | | | - Rishi S Kotecha
- Perth Children's Hospital, Perth, Australia; Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia; School of Pharmacy and Biomedical Sciences, Curtin University, Perth
| | - Luciano Dalla-Pozza
- Children's Medical Research Institute, University of Sydney, Sydney, Australia; Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, Australia; Children's Cancer Research Unit, The Children's Hospital at Westmead, Sydney
| | - Toby N Trahair
- Kids Cancer Centre, Sydney Children's Hospital Randwick, Sydney, Australia; School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney.
| |
Collapse
|
31
|
Brown P, Inaba H, Annesley C, Beck J, Colace S, Dallas M, DeSantes K, Kelly K, Kitko C, Lacayo N, Larrier N, Maese L, Mahadeo K, Nanda R, Nardi V, Rodriguez V, Rossoff J, Schuettpelz L, Silverman L, Sun J, Sun W, Teachey D, Wong V, Yanik G, Johnson-Chilla A, Ogba N. Pediatric Acute Lymphoblastic Leukemia, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 18:81-112. [PMID: 31910389 DOI: 10.6004/jnccn.2020.0001] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy. Advancements in technology that enhance our understanding of the biology of the disease, risk-adapted therapy, and enhanced supportive care have contributed to improved survival rates. However, additional clinical management is needed to improve outcomes for patients classified as high risk at presentation (eg, T-ALL, infant ALL) and who experience relapse. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for pediatric ALL provide recommendations on the workup, diagnostic evaluation, and treatment of the disease, including guidance on supportive care, hematopoietic stem cell transplantation, and pharmacogenomics. This portion of the NCCN Guidelines focuses on the frontline and relapsed/refractory management of pediatric ALL.
Collapse
Affiliation(s)
- Patrick Brown
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Hiroto Inaba
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Colleen Annesley
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Susan Colace
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Mari Dallas
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Kara Kelly
- Roswell Park Comprehensive Cancer Center
| | | | | | | | - Luke Maese
- Huntsman Cancer Institute at the University of Utah
| | - Kris Mahadeo
- The University of Texas MD Anderson Cancer Center
| | | | | | | | - Jenna Rossoff
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - Laura Schuettpelz
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | - Weili Sun
- City of Hope National Medical Center
| | - David Teachey
- Abramson Cancer Center at the University of Pennsylvania
| | | | | | | | | |
Collapse
|
32
|
Arshad U, Jabbar N, Mansoor N, Haider M, Butt Z, Nadeem K. Central Nervous System Involvement in Childhood Acute Lymphoblastic Leukemia: An Analysis of Day-One Versus Day-Eight Lumbar Punctures in Remission Induction Therapy. Cureus 2021; 13:e12464. [PMID: 33552781 PMCID: PMC7856329 DOI: 10.7759/cureus.12464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Introduction Traumatic lumbar puncture (TLP+) can lead to the iatrogenic infiltration of the central nervous system (CNS) by circulating leukemic blast cells in childhood acute lymphoblastic leukemia (ALL). The risk of TLP+ is increased by a number of factors at the time of presentation of the disease, such as a high white cell count (WCC), T-ALL phenotype, and unstable clinical condition of the patient. For this reason, the first lumbar puncture (LP) was deferred until Day Eight of prednisolone prophase during remission induction therapy in one set of patients. The objective was to compare the historical cohort of Day-One LP with Day-Eight LP with respect to the incidence of TLP+ and de novo CNS leukemia. Methods A retrospective comparative data analysis of 1,185 childhood ALL patients aged 1-16 years was conducted based on the electronic medical records of the pediatric hematology-oncology department of The Indus Hospital (TIH), Karachi, from January 2010 to August 2018. A total of 600 patients whose LP was done on Day One (January 2010-May 2015) were placed in cohort A, whereas 585 patients whose LP was performed on Day Eight (June 2015-August 2018) were placed in cohort B. After the examination of the cerebrospinal fluid (CSF), the status of CNS infiltration was classified as CNS-1, CNS-2, CNS-3, and TLP+. Results A total of 1,185 patients were included in the study, of whom 600 patients were in cohort A and 585 patients in cohort B. The incidence of TLP+ was found to be lower in cohort B (1.7%) as compared with the incidence in cohort A (4.3%) (p-value=0.009). However, there was an increase in the incidence of CNS-3 cases in cohort B (8%) as compared to cohort A (3%) (p-value: <0.001). When the CNS status of both the cohorts was compared with that of the internationally published data, a low incidence of TLP+ cases was noted in patients with LP on Day Eight. Conclusion The modified approach of performing the first LP on Day Eight significantly reduced the incidence of TLP+ cases. However, an unusual finding of a significant increase in the CNS-3 leukemia was noted. More prospective studies are needed to investigate this significant increase in CNS-3 cases.
Collapse
Affiliation(s)
- Uzma Arshad
- Pediatrics, Jinnah Medical and Dental College, Karachi, PAK
| | - Naeem Jabbar
- Department of Pediatric Oncology, The Indus Hospital, Karachi, PAK
| | - Neelum Mansoor
- Department of Hematology, The Indus Hospital, Karachi, PAK
| | - Maryam Haider
- Pediatrics, Jinnah Medical and Dental College, Karachi, PAK.,Pediatrics, Dr. Ruth K. M. Pfau, Civil Hospital, Karachi, PAK
| | - Zainab Butt
- Department of Pediatric Oncology, The Indus Hospital, Karachi, PAK
| | - Kishwer Nadeem
- Department of Pediatric Oncology, The Indus Hospital, Karachi, PAK
| |
Collapse
|
33
|
Farweez BAT, Kassim NA, Abdelfataah MF, Hassan NM, Hassnien DEA, El-Sakhawy YN. Clinical impact of early minimal residual disease detection at day 15 in precursor B-childhood acute lymphoblastic leukemia: an Egyptian experience. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2020. [DOI: 10.1186/s43042-020-00065-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Chromosomal abnormalities in childhood acute lymphoblastic leukemia (ALL) are well-established prognostic markers and useful tools for minimal residual disease (MRD) assessment. This study aimed to stratify high-risk precursor B-childhood ALL (pre-B-ALL) patients according to standard prognostic factors (age and total leucocytic count), fluorescence in situ hybridization (FISH) analysis for these cytogenetic abnormalities [t (9;22) BCR/ABL, t(1;19)TCF3/PBX1, and 11q23 MLL gene rearrangement], and MRD status at day 15. Besides, we aimed to demonstrate the relation of these prognostic factors (standard and cytogenetic risk groups) to patients’ outcome at day 15 of induction therapy as well as exploring the impact of early MRD assessment during remission induction compared to other prognostic factors together with the ability to tailor investigations as needed especially in places with limited health resources without compromising the outcome. Seventy-two newly-diagnosed Egyptian children with pre-B-ALL, aged 6 months to 15.5 years, registered from February 2016 to February 2018 were included. They were treated according to the modified Children’s Oncology Group (COG) protocol. Patients were classified into (a) standard and high-risk groups according to standard prognostic factors. (b) Patients with the studied cytogenetic abnormalities and patients without the studied cytogenetic abnormalities. (c) Good outcome (negative MRD) and bad outcome (positive MRD) groups according to day 15 MRD status.
Results
The studied cytogenetic abnormalities were identified in 22.2% of patients, all of them were in the high-risk group, and 75% of them had a bad outcome (positive MRD) at day 15 of induction therapy.
Conclusion
Patients with favorable presenting features (standard risk) and undetectable MRD after 2 weeks remission induction therapy would not be in need to advanced molecular studies, while these studies should be considered for patients with high-risk presenting features and high levels of MRD after 2 weeks remission induction therapy. Therefore, this could provide a cost-effective guideline in countries suffering from financial challenges without affecting the outcome
Collapse
|
34
|
Elgaz S, Bonig H, Bader P. Mesenchymal stromal cells for osteonecrosis. J Transl Med 2020; 18:399. [PMID: 33081809 PMCID: PMC7576732 DOI: 10.1186/s12967-020-02565-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/10/2020] [Indexed: 12/24/2022] Open
Abstract
Osteonecrosis (ON) is an acquired debilitating skeletal disorder, which is caused by a multitude of traumatic and non-traumatic etiological factors. Vascular damage, mechanical stress and increased intraosseous pressure have been discussed as contributors to ON. The optimal treatment of ON remains to be determined, since the current gold standard, core decompression, is insufficiently effective. Specific properties of mesenchymal stromal cells (MSCs) provide the rationale for their assessment in advanced stages of ON: Osteoinductive potential has been demonstrated and MSC preparations of suitable quality for use as medicinal products have been developed. Here we review the scant information on the use of allogeneic or autologous MSCs in advanced ON as well as potentially supportive data from pre-clinical studies with autologous bone marrow mononuclear cells (auto BM-MNCs), which have been studied quite extensively and the presumed therapeutic effect of which was attributed to the rare MSCs contained in these cell products. Outcomes in clinical trials with MSCs and auto-BM-MNCs remain preliminary and non-definitive, at best promising, with respect to their pharmacological effect. Clearly, though, the application of any of these cell therapies was technically feasible and safe in that it was associated with low complication rates. The heterogeneity of cell type and source, study protocols, cell manufacturing, cell properties, cell doses and surgical techniques might contribute to inconsistent results.
Collapse
Affiliation(s)
- S Elgaz
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - H Bonig
- Institute of Transfusion Medicine and Immunohematology, and German Red Cross Blood Center Baden-Württemberg-Hessen, Goethe University, Frankfurt am Main, Germany
| | - P Bader
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
35
|
Gupta S, Hunger SP. Recent trends in the results of studies conducted by the Children’s Oncology Group acute lymphoblastic leukemia committee and implications for emerging cooperative trial groups in low- and middle-income countries. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2020. [DOI: 10.1016/j.phoj.2020.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
36
|
Jasinski S, De Los Reyes FA, Yametti GC, Pierro J, Raetz E, Carroll WL. Immunotherapy in Pediatric B-Cell Acute Lymphoblastic Leukemia: Advances and Ongoing Challenges. Paediatr Drugs 2020; 22:485-499. [PMID: 32860590 PMCID: PMC7537790 DOI: 10.1007/s40272-020-00413-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leukemia, most commonly B-cell acute lymphoblastic leukemia (B-ALL), accounts for about 30% of childhood cancer diagnoses. While there have been dramatic improvements in childhood ALL outcomes, certain subgroups-particularly those who relapse-fare poorly. In addition, cure is associated with significant short- and long-term side effects. Given these challenges, there is great interest in novel, targeted approaches to therapy. A number of new immunotherapeutic agents have proven to be efficacious in relapsed or refractory disease and are now being investigated in frontline treatment regimens. Blinatumomab (a bispecific T-cell engager that targets cluster of differentiation [CD]-19) and inotuzumab ozogamicin (a humanized antibody-drug conjugate to CD22) have shown the most promise. Chimeric antigen receptor T (CAR-T) cells, a form of adoptive immunotherapy, rely on the transfer of genetically modified effector T cells that have the potential to persist in vivo for years, providing ongoing long-term disease control. In this article, we discuss the clinical biology and treatment of B-ALL with an emphasis on the role of immunotherapy in overcoming the challenges of conventional cytotoxic therapy. As immunotherapy continues to move into the frontline of pediatric B-ALL therapy, we also discuss strategies to address unique side effects associated with these agents and efforts to overcome mechanisms of resistance to immunotherapy.
Collapse
Affiliation(s)
- Sylwia Jasinski
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | | | - Gloria Contreras Yametti
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Joanna Pierro
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Elizabeth Raetz
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - William L Carroll
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
37
|
Gupta S, Maude SL, O'Brien MM, Rau RE, McNeer JL. How the COG is Approaching the High-Risk Patient with ALL: Incorporation of Immunotherapy into Frontline Treatment. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2020; 20 Suppl 1:S8-S11. [PMID: 32862880 DOI: 10.1016/s2152-2650(20)30443-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Affiliation(s)
- Sumit Gupta
- Hospital for Sick Children and University of Toronto, 555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Shannon L Maude
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, 3501 Civic Center Blvd, Philadelphia, PA, 19104, United States
| | - Maureen M O'Brien
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, United States
| | - Rachel E Rau
- Baylor College of Medicine, Texas Children's Hospital, 1102 Bates Street, Houston, TX, 77030, United States
| | - Jennifer L McNeer
- University of Chicago Comer Children's Hospital, 5841 S Maryland Ave, Chicago, IL, 60637, United States.
| |
Collapse
|
38
|
Dunsmore KP, Winter SS, Devidas M, Wood BL, Esiashvili N, Chen Z, Eisenberg N, Briegel N, Hayashi RJ, Gastier-Foster JM, Carroll AJ, Heerema NA, Asselin BL, Rabin KR, Zweidler-Mckay PA, Raetz EA, Loh ML, Schultz KR, Winick NJ, Carroll WL, Hunger SP. Children's Oncology Group AALL0434: A Phase III Randomized Clinical Trial Testing Nelarabine in Newly Diagnosed T-Cell Acute Lymphoblastic Leukemia. J Clin Oncol 2020; 38:3282-3293. [PMID: 32813610 DOI: 10.1200/jco.20.00256] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Nelarabine is effective in inducing remission in patients with relapsed and refractory T-cell acute lymphoblastic leukemia (T-ALL) but has not been fully evaluated in those with newly diagnosed disease. PATIENTS AND METHODS From 2007 to 2014, Children's Oncology Group trial AALL0434 (ClinicalTrials.gov identifier: NCT00408005) enrolled 1,562 evaluable patients with T-ALL age 1-31 years who received the augmented Berlin-Frankfurt-Muenster (ABFM) regimen with a 2 × 2 pseudo-factorial randomization to receive escalating-dose methotrexate (MTX) without leucovorin rescue plus pegaspargase (C-MTX) or high-dose MTX (HDMTX) with leucovorin rescue. Intermediate- and high-risk patients were also randomly assigned after induction to receive or not receive six 5-day courses of nelarabine that was incorporated into ABFM. Patients who experienced induction failure were nonrandomly assigned to HDMTX plus nelarabine. Patients with overt CNS disease (CNS3; ≥ 5 WBCs/μL with blasts) received HDMTX and were randomly assigned to receive or not receive nelarabine. All patients, except those with low-risk disease, received cranial irradiation. RESULTS The 5-year event-free and overall survival rates were 83.7% ± 1.1% and 89.5% ± 0.9%, respectively. The 5-year disease-free survival (DFS) rates for patients with T-ALL randomly assigned to nelarabine (n = 323) and no nelarabine (n = 336) were 88.2% ± 2.4% and 82.1% ± 2.7%, respectively (P = .029). Differences between DFS in a four-arm comparison were significant (P = .01), with no interactions between the MTX and nelarabine randomizations (P = .41). Patients treated with the best-performing arm, C-MTX plus nelarabine, had a 5-year DFS of 91% (n = 147). Patients who received nelarabine had significantly fewer isolated and combined CNS relapses compared with patients who did not receive nelarabine (1.3% ± 0.63% v 6.9% ± 1.4%, respectively; P = .0001). Toxicities, including neurotoxicity, were acceptable and similar between all four arms. CONCLUSION The addition of nelarabine to ABFM therapy improved DFS for children and young adults with newly diagnosed T-ALL without increased toxicity.
Collapse
Affiliation(s)
| | - Stuart S Winter
- Children's Minnesota Cancer and Blood Disorders Program, Minneapolis, MN
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| | - Brent L Wood
- Laboratory Medicine, Seattle Children's Hospital, Seattle, WA
| | - Natia Esiashvili
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Zhiguo Chen
- Department of Biostatistics, Colleges of Medicine and Public Health and Health Professions, University of Florida, Gainesville, FL
| | - Nancy Eisenberg
- Pediatric Hematology/Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Nikki Briegel
- Pharmacy Department, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Robert J Hayashi
- Pediatric Hematology/Oncology, Washington School of Medicine, St Louis Children's Hospital, St Louis, MO
| | - Julie M Gastier-Foster
- Nationwide Children's Hospital, Columbus OH.,Department of Pathology, The Ohio State University, Columbus, OH
| | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL
| | - Nyla A Heerema
- Department of Pathology, The Ohio State University, Columbus, OH
| | - Barbara L Asselin
- Department of Pediatrics, University of Rochester Medical Center and Wilmot Cancer Institute, Rochester, NY
| | - Karen R Rabin
- Pediatric Hematology/Oncology, Baylor College of Medicine/Dan L. Duncan Comprehensive Cancer Center, Houston TX
| | | | - Elizabeth A Raetz
- Laura and Isaac Perlmutter Cancer Center at New York University Langone Health, New York, NY
| | - Mignon L Loh
- Department of Pediatrics, University of California, San Francisco Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Kirk R Schultz
- Pediatric Hematology-Oncology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Naomi J Winick
- Pediatric Hematology/Oncology, University of Texas Southwestern/Simmons Cancer Center, Dallas, TX
| | - William L Carroll
- Laura and Isaac Perlmutter Cancer Center at New York University Langone Health, New York, NY
| | - Stephen P Hunger
- Department of Pediatrics and The Center for Childhood Cancer Research, The Children's Hospital of Philadelphia and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
39
|
Salzer WL, Burke MJ, Devidas M, Dai Y, Hardy KK, Kairalla JA, Gore L, Hilden JM, Larsen E, Rabin KR, Zweidler-McKay PA, Borowitz MJ, Wood B, Heerema NA, Carroll AJ, Winick N, Carroll WL, Raetz EA, Loh ML, Hunger SP. Impact of Intrathecal Triple Therapy Versus Intrathecal Methotrexate on Disease-Free Survival for High-Risk B-Lymphoblastic Leukemia: Children's Oncology Group Study AALL1131. J Clin Oncol 2020; 38:2628-2638. [PMID: 32496902 DOI: 10.1200/jco.19.02892] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
PURPOSE The high-risk stratum of Children's Oncology Group Study AALL1131 was designed to test the hypothesis that postinduction CNS prophylaxis with intrathecal triple therapy (ITT) including methotrexate, hydrocortisone, and cytarabine would improve the postinduction 5-year disease-free survival (DFS) compared with intrathecal methotrexate (IT MTX), when given on a modified augmented Berlin-Frankfurt-Münster backbone. PATIENTS AND METHODS Children with newly diagnosed National Cancer Institute (NCI) high-risk B-cell acute lymphoblastic leukemia (HR B-ALL) or NCI standard-risk B-ALL with defined minimal residual disease thresholds during induction were randomly assigned to receive postinduction IT MTX or ITT. Patients with CNS3-status disease were not eligible. Postinduction IT therapy was given for a total of 21 to 26 doses. Neurocognitive assessments were performed during therapy and during 1 year off therapy. RESULTS Random assignment was closed to accrual in March 2018 after a futility boundary had been crossed, concluding that ITT could not be shown to be superior to IT MTX. The 5-year postinduction DFS and overall survival rates (± SE) of children randomly assigned to IT MTX versus ITT were 93.2% ± 2.1% v 90.6% ± 2.3% (P = .85), and 96.3% ± 1.5% v 96.7% ± 1.4% (P = .77), respectively. There were no differences in the cumulative incidence of isolated bone marrow relapse, isolated CNS relapse, or combined bone marrow and CNS relapse rates, or in toxicities observed for patients receiving IT MTX compared with ITT. There were no significant differences in neurocognitive outcomes for patients receiving IT MTX compared with ITT. CONCLUSION Postinduction CNS prophylaxis with ITT did not improve 5-year DFS for children with HR B-ALL. The standard of care for CNS prophylaxis for children with B-ALL and no overt CNS involvement remains IT MTX.
Collapse
Affiliation(s)
| | - Michael J Burke
- Department of Pediatrics, Children's Hospital of Wisconsin, Milwaukee, WI
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| | - Yunfeng Dai
- Department of Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, FL
| | | | - John A Kairalla
- Department of Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, FL
| | - Lia Gore
- Department of Pediatrics, Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO.,University of Colorado School of Medicine, Aurora, CO
| | - Joanne M Hilden
- Department of Pediatrics, Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO.,University of Colorado School of Medicine, Aurora, CO
| | - Eric Larsen
- Department of Pediatrics, Maine Children's Cancer Program, Scarborough, ME
| | - Karen R Rabin
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | | | - Michael J Borowitz
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Brent Wood
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | - Nyla A Heerema
- Department of Pathology, The Ohio State University School of Medicine, Columbus, OH
| | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL
| | - Naomi Winick
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - William L Carroll
- Department of Pediatrics, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY
| | - Elizabeth A Raetz
- Department of Pediatrics, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Stephen P Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA.,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
40
|
Prednisolone Prophase for a Week Versus Upfront Multiagent Chemotherapy in Childhood Acute Lymphoblastic Leukemia: An Analysis With Reference to Induction Mortality in a Developing Country. J Pediatr Hematol Oncol 2020; 42:181-184. [PMID: 31688627 DOI: 10.1097/mph.0000000000001636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In childhood acute lymphoblastic leukemia, high treatment-related mortality, especially in the induction phase of treatment, is a major challenge for developing countries. The reasons are multifactorial, including a late presentation with higher disease burden, malnourishment, and limited support services. These factors may aggravate the toxic effects of upfront multiagent chemotherapy in terms of severe neutropenic sepsis and tumor lysis. Therefore, instead of upfront chemotherapy, we offered prednisolone prophase for 1 week with the objective of balancing the antileukemic versus the toxic effect of treatment. The data of 538 patients who received induction with this approach (cohort B) are compared for induction mortality with previous records of 438 patients (cohort A) treated with upfront chemotherapy. In the presence of similar clinical characteristics including age, sex, risk group, and phenotype in both cohorts, a significant difference was found in overall induction mortality of 9% in cohort B versus 14% in cohort A (P<0.05). This difference was also significant in the high-risk and T-cell phenotype, which strengthens our hypothesis that patients with higher burden of disease may experience more fatal toxic effects with upfront intensive chemotherapy. Therefore, we suggest that the prednisolone prophase approach is beneficial to control the disease with less severe toxic effects in our settings.
Collapse
|
41
|
McNeer JL, Rau RE, Gupta S, Maude SL, O'Brien MM. Cutting to the Front of the Line: Immunotherapy for Childhood Acute Lymphoblastic Leukemia. Am Soc Clin Oncol Educ Book 2020; 40:1-12. [PMID: 32320280 DOI: 10.1200/edbk_278171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Although many children and young adults with B-cell acute lymphoblastic leukemia (B-ALL) are cured with modern, risk-adapted chemotherapy regimens, 10% to 15% of patients will experience relapse or have refractory disease. Recent efforts to further intensify cytotoxic chemotherapy regimens in the frontline setting have failed as a result of excessive toxicity or lack of improvement in efficacy. As a result, novel approaches will be required to achieve cures in more newly diagnosed patients. Multiple immune-based therapies have demonstrated considerable efficacy in the setting of relapsed or refractory (R/R) disease, including CD19 targeting with blinatumomab and tisagenlecleucel and CD22 targeting with inotuzumab ozogamicin. These agents are now under investigation by the Children's Oncology Group (COG) in clinical trials for newly diagnosed B-ALL, with integration into standard chemotherapy regimens based on clinically and biology-based risk stratification as well as disease response.
Collapse
Affiliation(s)
| | - Rachel E Rau
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Sumit Gupta
- The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Shannon L Maude
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Maureen M O'Brien
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
42
|
Maloney KW, Devidas M, Wang C, Mattano LA, Friedmann AM, Buckley P, Borowitz MJ, Carroll AJ, Gastier-Foster JM, Heerema NA, Kadan-Lottick N, Loh ML, Matloub YH, Marshall DT, Stork LC, Raetz EA, Wood B, Hunger SP, Carroll WL, Winick NJ. Outcome in Children With Standard-Risk B-Cell Acute Lymphoblastic Leukemia: Results of Children's Oncology Group Trial AALL0331. J Clin Oncol 2019; 38:602-612. [PMID: 31825704 DOI: 10.1200/jco.19.01086] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Children's Oncology Group (COG) AALL0331 tested whether intensified postinduction therapy that improves survival in children with high-risk B-cell acute lymphoblastic leukemia (ALL) would also improve outcomes for those with standard-risk (SR) ALL. PATIENTS AND METHODS AALL0331 enrolled 5,377 patients between 2005 and 2010. All patients received a 3-drug induction with dexamethasone, vincristine, and pegaspargase (PEG) and were then classified as SR low, SR average, or SR high. Patients with SR-average disease were randomly assigned to receive either standard 4-week consolidation (SC) or 8-week intensified augmented Berlin-Frankfurt-Münster (BFM) consolidation (IC). Those with SR-high disease were nonrandomly assigned to the full COG-augmented BFM regimen, including 2 interim maintenance and delayed intensification phases. RESULTS The 6-year event-free survival (EFS) rate for all patients enrolled in AALL0331 was 88.96% ± 0.46%, and overall survival (OS) was 95.54% ± 0.31%. For patients with SR-average disease, the 6-year continuous complete remission (CCR) and OS rates for SC versus IC were 87.8% ± 1.3% versus 89.1% ± 1.2% (P = .52) and 95.8% ± 0.8% versus 95.2% ± 0.8% (P = 1.0), respectively. Those with SR-average disease with end-induction minimal residual disease (MRD) of 0.01% to < 0.1% had an inferior outcome compared with those with lower MRD and no improvement with IC (6-year CCR: SC, 77.5% ± 4.8%; IC, 77.1% ± 4.8%; P = .71). At 6 years, the CCR and OS rates among 635 nonrandomly treated patients with SR-high disease were 85.55% ± 1.49% and 92.97% ± 1.08%, respectively. CONCLUSION The 6-year OS rate for > 5,000 children with SR ALL enrolled in AALL0331 exceeded 95%. The addition of IC to treatment for patients with SR-average disease did not improve CCR or OS, even in patients with higher MRD, in whom it might have been predicted to provide more value. The EFS and OS rates are excellent for this group of patients with SR ALL, with particularly good outcomes for those with SR-high disease.
Collapse
Affiliation(s)
- Kelly W Maloney
- Department of Pediatrics, University of Colorado, Aurora, CO.,Children's Hospital Colorado, Aurora, CO
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN
| | - Cindy Wang
- Department of Biostatistics, Colleges of Medicine, Public Health and Health Professions, University of Florida, Gainesville, FL
| | | | - Alison M Friedmann
- Department of Pediatrics, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Patrick Buckley
- Department of Pathology, Duke University Medical Center, Durham, NC
| | | | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL
| | - Julie M Gastier-Foster
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH.,Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH
| | - Nyla A Heerema
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH
| | | | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital, San Francisco, CA.,Helen Diller Family Comprehensive Cancer, University of California, San Francisco, CA
| | - Yousif H Matloub
- Department of Pediatrics, Rainbow Babies and Children's Hospital, Cleveland, OH
| | - David T Marshall
- Department of Radiation Oncology, Medical University of South Carolina, Charleston, SC
| | - Linda C Stork
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Elizabeth A Raetz
- Perlmutter Cancer Center, New York University (NYU) Langone Medical Center, New York, NY.,Department of Pediatrics, NYU Langone Medical Center, New York, NY
| | - Brent Wood
- Department of Pathology, University of Washington, Seattle, WA.,Department of Medicine, University of Washington, Seattle, WA
| | - Stephen P Hunger
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA.,Perelman School of Medicine at University of Philadelphia, Philadelphia, PA
| | - William L Carroll
- Perlmutter Cancer Center, New York University (NYU) Langone Medical Center, New York, NY.,Department of Pediatrics, NYU Langone Medical Center, New York, NY
| | - Naomi J Winick
- Department of Pediatrics, University of Texas (UT) Southwestern, Dallas, TX.,Simmons Cancer Center, UT Southwestern, Dallas, TX
| |
Collapse
|
43
|
Randomized post-induction and delayed intensification therapy in high-risk pediatric acute lymphoblastic leukemia: long-term results of the international AIEOP-BFM ALL 2000 trial. Leukemia 2019; 34:1694-1700. [DOI: 10.1038/s41375-019-0670-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/26/2019] [Accepted: 11/17/2019] [Indexed: 12/14/2022]
|
44
|
Frisch A, Ofran Y. How I diagnose and manage Philadelphia chromosome-like acute lymphoblastic leukemia. Haematologica 2019; 104:2135-2143. [PMID: 31582548 PMCID: PMC6821607 DOI: 10.3324/haematol.2018.207506] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/03/2019] [Indexed: 01/31/2023] Open
Abstract
Advances in our understanding of mechanisms of leukemogenesis and driver mutations in acute lymphoblastic leukemia (ALL) lead to a more precise and informative sub-classification, mainly of B-cell ALL. In parallel, in recent years, novel agents have been approved for the therapy of B-cell ALL, and many others are in active clinical research. Among the newly recognized disease subtypes, Philadelphia-chromosome-like ALL is the most heterogeneous and thus, diagnostically challenging. Given that this subtype of B-cell ALL is associated with a poorer prognosis, improvement of available therapeutic approaches and protocols is a burning issue. Herein, we summarize, in a clinically relevant manner, up-to-date information regarding diagnostic strategies developed for the identification of patients with Philadelphia-chromosome-like ALL. Common therapeutic dilemmas, presented as several case scenarios, are also discussed. It is currently acceptable that patients with B-cell ALL, treated with an aim of cure, irrespective of their age, be evaluated for a Philadelphia-chromosome-like signature as early as possible. Following Philadelphia-chromosome-like recognition, a higher risk of resistance or relapse must be realized and treatment should be modified based on the patient’s specific genetic driver and clinical features. However, while active targeted therapeutic options are limited, there is much more to do than just prescribe a matched inhibitor to the identified mutated driver genes. In this review, we present a comprehensive evidence-based approach to the diagnosis and management of Philadelphia-chromosome-like ALL at different time-points during the disease course.
Collapse
Affiliation(s)
- Avraham Frisch
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa
| | - Yishai Ofran
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa .,Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
45
|
Gaynon PS. Methotrexate and asparaginase: not so simple. Leuk Lymphoma 2019; 60:2849-2850. [PMID: 31558076 DOI: 10.1080/10428194.2019.1668941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Paul S Gaynon
- Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
46
|
Hunger SP. More Is Not Always Better: The Perils of Treatment Intensification in Pediatric Acute Lymphoblastic Leukemia. J Clin Oncol 2019; 37:1601-1603. [DOI: 10.1200/jco.19.00889] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Stephen P. Hunger
- Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
47
|
Strategies to Overcome Resistance Mechanisms in T-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2019; 20:ijms20123021. [PMID: 31226848 PMCID: PMC6627878 DOI: 10.3390/ijms20123021] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022] Open
Abstract
Chemoresistance is a major cause of recurrence and death from T-cell acute lymphoblastic leukemia (T-ALL), both in adult and pediatric patients. In the majority of cases, drug-resistant disease is treated by selecting a combination of other drugs, without understanding the molecular mechanisms by which malignant cells escape chemotherapeutic treatments, even though a more detailed genomic characterization and the identification of actionable disease targets may enable informed decision of new agents to improve patient outcomes. In this work, we describe pathways of resistance to common chemotherapeutic agents including glucocorticoids and review the resistance mechanisms to targeted therapy such as IL7R, PI3K-AKT-mTOR, NOTCH1, BRD4/MYC, Cyclin D3: CDK4/CDK6, BCL2 inhibitors, and selective inhibitors of nuclear export (SINE). Finally, to overcome the limitations of the current trial-and-error method, we summarize the experiences of anti-cancer drug sensitivity resistance profiling (DSRP) approaches as a rapid and relevant strategy to infer drug activity and provide functional information to assist clinical decision one patient at a time.
Collapse
|
48
|
Marini BL, Brown J, Benitez L, Walling E, Hutchinson RJ, Mody R, Jasty Rao R, Slagle L, Bishop L, Pettit K, Bixby DL, Burke PW, Perissinotti AJ. A single-center multidisciplinary approach to managing the global Erwinia asparaginase shortage. Leuk Lymphoma 2019; 60:2854-2868. [PMID: 31099289 DOI: 10.1080/10428194.2019.1608530] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The availability of Erwinia Asparaginase has been limited across the world due to manufacturing shortages or for some countries due to the high acquisition cost, putting patients at risk for inferior outcomes. This manuscript provides guidance on how to manage hypersensitivity reactions and utilize therapeutic drug monitoring (TDM) to conserve and limit Erwinia use. The clinical and financial impact of a multidisciplinary committee are also discussed. Faced with a global Erwinia shortage, a multidisciplinary asparaginase allergy committee was created to review all hypersensitivity reactions to asparaginase therapy, staff education was performed on the management of asparaginase hypersensitivity reactions, an institution-wide premedication policy was mandated, and standardized guidelines were created for TDM. This multidisciplinary approach reduced the PEG-asparaginase to Erwinia switch rate from 21% (35 of 163) to 7% (10 of 134) (p = .0035). A multifaceted approach can safely maintain patients on PEG-asparaginase and conserve Erwinia for patients who need it most.
Collapse
Affiliation(s)
- Bernard L Marini
- Department of Pharmacy Services and Clinical Sciences Michigan Medicine, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Julia Brown
- Department of Pharmacy Services and Clinical Sciences Michigan Medicine, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Lydia Benitez
- Department of Pharmacy Services and Clinical Sciences Michigan Medicine, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Emily Walling
- Department of Pediatrics and Communicable Diseases Division of Pediatric Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Raymond J Hutchinson
- Department of Pediatrics and Communicable Diseases Division of Pediatric Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rajen Mody
- Department of Pediatrics and Communicable Diseases Division of Pediatric Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rama Jasty Rao
- Department of Pediatrics and Communicable Diseases Division of Pediatric Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lynn Slagle
- Department of Pediatrics and Communicable Diseases Division of Pediatric Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lauren Bishop
- Department of Pediatrics and Communicable Diseases Division of Pediatric Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kristen Pettit
- Department of Internal Medicine Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Dale L Bixby
- Department of Internal Medicine Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Patrick W Burke
- Department of Internal Medicine Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anthony J Perissinotti
- Department of Pharmacy Services and Clinical Sciences Michigan Medicine, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
49
|
Kim H. Advances in the Treatment of Childhood Acute Lymphoblastic Leukemia. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2019. [DOI: 10.15264/cpho.2019.26.1.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Hyery Kim
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
50
|
Jastaniah W, Elimam N, Abdalla K, AlAzmi AA, Algamal A, Felimban S. Intrathecal dose intensification by CNS status at diagnosis in the treatment of children with acute lymphoblastic leukemia. ACTA ACUST UNITED AC 2019; 24:369-377. [PMID: 30885098 DOI: 10.1080/16078454.2019.1590962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Acute lymphoblastic leukemia (ALL) with CNS2 status predicts inferior outcome and a high rate of CNS relapse, similar to overt CNS leukemia (CNS3). The purpose of this study was to determine if intrathecal (IT) dose intensification during induction would improve outcomes and reduce CNS relapse for CNS2 disease. METHODS From January 2001 to December 2014, children (1-14 years) with newly diagnosed ALL were treated at the Princess Noorah Oncology Centre (PNOC) following modifications of the Children's Oncology Group (COG) protocols. We intensified IT methotrexate (ITM) during induction for patients with CNS2 disease. Patients were evaluated for overall survival (OS), disease-free survival (DFS), and cumulative incidence of relapse (CIR). RESULTS 449 children with T-cell (14.3%) or B-cell (85.7%) ALL were treated using PNOC-SR or PNOC-HR regimens (Jan 2001- Dec 2007) or CALL08 regimens (Arm A [SR], Arm B [IR], and Arm C [HR]) (Jan 2008 - Dec 2014). The 5-year OS, DFS, and CIR were 87.2 ± 1.6%, 81.7 ± 1.9%, and 13.0 ± 1.7%, respectively. The OS and DFS of patients with CNS2 were significantly superior to that of patients with CNS3 (P = 0.025 and P = 0.019, respectively). Patients with CNS2 had similar OS and DFS to those with CNS1. None of the patients with CNS2 at initial diagnosis experienced CNS relapse. CONCLUSIONS ITM intensification during induction was associated with elimination of CNS recurrence in patients with CNS2 disease and childhood ALL. Controlled studies are needed to confirm this observation.
Collapse
Affiliation(s)
- Wasil Jastaniah
- a Department of Pediatrics, Faculty of Medicine , Umm AlQura University , Makkah , Saudi Arabia.,b Princess Noorah Oncology Center , King Saud Bin Abdulaziz University for Health Sceinces and King Abdulaziz Medical City, Ministry of National Guard Health Affairs , Jeddah , Saudi Arabia
| | - Naglla Elimam
- b Princess Noorah Oncology Center , King Saud Bin Abdulaziz University for Health Sceinces and King Abdulaziz Medical City, Ministry of National Guard Health Affairs , Jeddah , Saudi Arabia
| | - Khalid Abdalla
- b Princess Noorah Oncology Center , King Saud Bin Abdulaziz University for Health Sceinces and King Abdulaziz Medical City, Ministry of National Guard Health Affairs , Jeddah , Saudi Arabia
| | - Aeshah A AlAzmi
- c Department of Pharmaceutical Care, Clinical Pharmacy, Pediatric Hematology/Oncology , King Saud Bin Abdulaziz University for Health Sciences and King Abdulaziz Medical City, Ministry of National Guard Health Affairs , Jeddah , Saudi Arabia
| | - Amal Algamal
- b Princess Noorah Oncology Center , King Saud Bin Abdulaziz University for Health Sceinces and King Abdulaziz Medical City, Ministry of National Guard Health Affairs , Jeddah , Saudi Arabia
| | - Sami Felimban
- b Princess Noorah Oncology Center , King Saud Bin Abdulaziz University for Health Sceinces and King Abdulaziz Medical City, Ministry of National Guard Health Affairs , Jeddah , Saudi Arabia
| |
Collapse
|