1
|
Dong N, Ma HX, Liu XQ, Li D, Liu LH, Shi Q, Ju XL. Histidine re-sensitizes pediatric acute lymphoblastic leukemia to 6-mercaptopurine through tetrahydrofolate consumption and SIRT5-mediated desuccinylation. Cell Death Dis 2024; 15:216. [PMID: 38485947 PMCID: PMC10940622 DOI: 10.1038/s41419-024-06599-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
Despite progressive improvements in the survival rate of pediatric B-cell lineage acute lymphoblastic leukemia (B-ALL), chemoresistance-induced disease progression and recurrence still occur with poor prognosis, thus highlighting the urgent need to eradicate drug resistance in B-ALL. The 6-mercaptopurine (6-MP) is the backbone of ALL combination chemotherapy, and resistance to it is crucially related to relapse. The present study couples chemoresistance in pediatric B-ALL with histidine metabolism deficiency. Evidence was provided that histidine supplementation significantly shifts the 6-MP dose-response in 6-MP-resistant B-ALL. It is revealed that increased tetrahydrofolate consumption via histidine catabolism partially explains the re-sensitization ability of histidine. More importantly, this work provides fresh insights into that desuccinylation mediated by SIRT5 is an indispensable and synergistic requirement for histidine combination therapy against 6-MP resistance, which is undisclosed previously and demonstrates a rational strategy to ameliorate chemoresistance and protect pediatric patients with B-ALL from disease progression or relapse.
Collapse
Affiliation(s)
- Na Dong
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Hui-Xian Ma
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Xue-Qin Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Dong Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Ling-Hong Liu
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Qing Shi
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Xiu-Li Ju
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
2
|
Chisholm KM, Bohling SD. Childhood Myelodysplastic Syndrome. Clin Lab Med 2023; 43:639-655. [PMID: 37865508 DOI: 10.1016/j.cll.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
Myelodysplastic syndrome (MDS) in children is rare, accounting for < 5% of all childhood hematologic malignancies. With the advent of next-generation sequencing, the etiology of many childhood MDS (cMDS) cases has been elucidated with the finding of predisposing germline mutations in one-quarter to one-third of cases; somatic mutations have also been identified, indicating that cMDS is different than adult MDS. Herein, cMDS classification schema, clinical presentation, laboratory values, bone marrow histology, differential diagnostic considerations, and the recent molecular findings of cMDS are described.
Collapse
Affiliation(s)
- Karen M Chisholm
- Hematopathology, Department of Laboratories, Seattle Children's Hospital, 4800 Sand Point Way Northeast, FB.4.510, Seattle, WA 98105, USA; Department of Laboratory Medicine and Pathology, University of Washington Medical Center, 4800 Sand Point Way Northeast, FB.4.510, Seattle, WA 98105, USA.
| | - Sandra D Bohling
- Hematopathology, Department of Laboratories, Seattle Children's Hospital, 4800 Sand Point Way Northeast, FB.4.510, Seattle, WA 98105, USA; Department of Laboratory Medicine and Pathology, University of Washington Medical Center, 4800 Sand Point Way Northeast, FB.4.510, Seattle, WA 98105, USA
| |
Collapse
|
3
|
Anand U, Dey A, Chandel AKS, Sanyal R, Mishra A, Pandey DK, De Falco V, Upadhyay A, Kandimalla R, Chaudhary A, Dhanjal JK, Dewanjee S, Vallamkondu J, Pérez de la Lastra JM. Cancer chemotherapy and beyond: Current status, drug candidates, associated risks and progress in targeted therapeutics. Genes Dis 2023; 10:1367-1401. [PMID: 37397557 PMCID: PMC10310991 DOI: 10.1016/j.gendis.2022.02.007] [Citation(s) in RCA: 269] [Impact Index Per Article: 269.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 11/28/2022] Open
Abstract
Cancer is an abnormal state of cells where they undergo uncontrolled proliferation and produce aggressive malignancies that causes millions of deaths every year. With the new understanding of the molecular mechanism(s) of disease progression, our knowledge about the disease is snowballing, leading to the evolution of many new therapeutic regimes and their successive trials. In the past few decades, various combinations of therapies have been proposed and are presently employed in the treatment of diverse cancers. Targeted drug therapy, immunotherapy, and personalized medicines are now largely being employed, which were not common a few years back. The field of cancer discoveries and therapeutics are evolving fast as cancer type-specific biomarkers are progressively being identified and several types of cancers are nowadays undergoing systematic therapies, extending patients' disease-free survival thereafter. Although growing evidence shows that a systematic and targeted approach could be the future of cancer medicine, chemotherapy remains a largely opted therapeutic option despite its known side effects on the patient's physical and psychological health. Chemotherapeutic agents/pharmaceuticals served a great purpose over the past few decades and have remained the frontline choice for advanced-stage malignancies where surgery and/or radiation therapy cannot be prescribed due to specific reasons. The present report succinctly reviews the existing and contemporary advancements in chemotherapy and assesses the status of the enrolled drugs/pharmaceuticals; it also comprehensively discusses the emerging role of specific/targeted therapeutic strategies that are presently being employed to achieve better clinical success/survival rate in cancer patients.
Collapse
Affiliation(s)
- Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Arvind K. Singh Chandel
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Rupa Sanyal
- Department of Botany, Bhairab Ganguly College (affiliated to West Bengal State University), Kolkata, West Bengal 700056, India
| | - Amarnath Mishra
- Faculty of Science and Technology, Amity Institute of Forensic Sciences, Amity University Uttar Pradesh, Noida 201313, India
| | - Devendra Kumar Pandey
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Valentina De Falco
- Institute of Endocrinology and Experimental Oncology (IEOS), National Research Council (CNR), Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, Naples 80131, Italy
| | - Arun Upadhyay
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandar Sindari, Kishangarh Ajmer, Rajasthan 305817, India
| | - Ramesh Kandimalla
- CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007, India
- Department of Biochemistry, Kakatiya Medical College, Warangal, Telangana 506007, India
| | - Anupama Chaudhary
- Orinin-BioSystems, LE-52, Lotus Road 4, CHD City, Karnal, Haryana 132001, India
| | - Jaspreet Kaur Dhanjal
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-D), Okhla Industrial Estate, Phase III, New Delhi 110020, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Jayalakshmi Vallamkondu
- Department of Physics, National Institute of Technology-Warangal, Warangal, Telangana 506004, India
| | - José M. Pérez de la Lastra
- Biotechnology of Macromolecules Research Group, Instituto de Productos Naturales y Agrobiología, IPNA-CSIC, San Cristóbal de La Laguna 38206, Tenerife, Spain
| |
Collapse
|
4
|
Elitzur S, Vora A, Burkhardt B, Inaba H, Attarbaschi A, Baruchel A, Escherich G, Gibson B, Liu HC, Loh M, Moorman AV, Möricke A, Pieters R, Uyttebroeck A, Baird S, Bartram J, Barzilai-Birenboim S, Batra S, Ben-Harosh M, Bertrand Y, Buitenkamp T, Caldwell K, Drut R, Geerlinks AV, Gilad G, Grainger J, Haouy S, Heaney N, Huang M, Ingham D, Krenova Z, Kuhlen M, Lehrnbecher T, Manabe A, Niggli F, Paris C, Revel-Vilk S, Rohrlich P, Sinno MG, Szczepanski T, Tamesberger M, Warrier R, Wolfl M, Nirel R, Izraeli S, Borkhardt A, Schmiegelow K. EBV-driven lymphoid neoplasms associated with pediatric ALL maintenance therapy. Blood 2023; 141:743-755. [PMID: 36332176 DOI: 10.1182/blood.2022016975] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/19/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The development of a second malignancy after the diagnosis of childhood acute lymphoblastic leukemia (ALL) is a rare event. Certain second malignancies have been linked with specific elements of leukemia therapy, yet the etiology of most second neoplasms remains obscure and their optimal management strategies are unclear. This is a first comprehensive report of non-Hodgkin lymphomas (NHLs) following pediatric ALL therapy, excluding stem-cell transplantation. We analyzed data of patients who developed NHL following ALL diagnosis and were enrolled in 12 collaborative pediatric ALL trials between 1980-2018. Eighty-five patients developed NHL, with mature B-cell lymphoproliferations as the dominant subtype (56 of 85 cases). Forty-six of these 56 cases (82%) occurred during or within 6 months of maintenance therapy. The majority exhibited histopathological characteristics associated with immunodeficiency (65%), predominantly evidence of Epstein-Barr virus-driven lymphoproliferation. We investigated 66 cases of post-ALL immunodeficiency-associated lymphoid neoplasms, 52 from our study and 14 additional cases from a literature search. With a median follow-up of 4.9 years, the 5-year overall survival for the 66 patients with immunodeficiency-associated lymphoid neoplasms was 67.4% (95% confidence interval [CI], 56-81). Five-year cumulative risks of lymphoid neoplasm- and leukemia-related mortality were 20% (95% CI, 10.2-30) and 12.4% (95% CI, 2.7-22), respectively. Concurrent hemophagocytic lymphohistiocytosis was associated with increased mortality (hazard ratio, 7.32; 95% CI, 1.62-32.98; P = .01). A large proportion of post-ALL lymphoid neoplasms are associated with an immunodeficient state, likely precipitated by ALL maintenance therapy. Awareness of this underrecognized entity and pertinent diagnostic tests are crucial for early diagnosis and optimal therapy.
Collapse
Affiliation(s)
- Sarah Elitzur
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ajay Vora
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Birgit Burkhardt
- Pediatric Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Andre Baruchel
- Department of Pediatric Hematology, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncoogy, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Brenda Gibson
- Department of Paediatric Haematology, Royal Hospital for Children, Glasgow, United Kingdom
| | - Hsi-Che Liu
- Division of Pediatric Hematology/Oncology, Mackay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Mignon Loh
- Division of Pediatric Hematology, Oncology, Bone Marrow Transplant and Cellular Therapy, Seattle Children's Hospital and the Ben Towne Center for Childhood Cancer Research, University of Washington, Seattle, WA
| | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Wolfson Childhood Cancer Centre, Clinical and Translational Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anja Möricke
- Department of Pediatrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Anne Uyttebroeck
- Department of Paediatric Haematology and Oncology, University Hospital Leuven, Leuven, Leuven, Belgium
| | - Susan Baird
- Department of Haematology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| | - Jack Bartram
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Shlomit Barzilai-Birenboim
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Sandeep Batra
- Pediatric Hematology/Oncology, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN
| | - Miriam Ben-Harosh
- Department of Pediatric Hemato-Oncology, Soroka Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yves Bertrand
- Institut d'Hematologie et d'Oncologie Pediatrique, Hospices Civils de Lyon, Lyon, France
| | - Trudy Buitenkamp
- Amsterdam Academic Medical Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Kenneth Caldwell
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, FL
| | - Ricardo Drut
- Department of Pathology, School of Medicine, La Plata National University, La Plata, Argentina
| | | | - Gil Gilad
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - John Grainger
- Faculty of Medical & Human Sciences, University of Manchester and Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stephanie Haouy
- Department of Pediatric Oncology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Nicholas Heaney
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Mary Huang
- Department of Pediatric Hematology Oncology, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA
| | - Danielle Ingham
- Paediatric Oncology, Leeds Children's Hospital, Leeds, United Kingdom
| | - Zdenka Krenova
- Department of Pediatric Oncology and Department of Pediatrics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michaela Kuhlen
- Pediatrics and Adolescent Medicine, University of Augsburg, Augsburg, Germany
| | - Thomas Lehrnbecher
- Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Graduate School of Medicine, Sapporo, Japan
| | - Felix Niggli
- Department of Pediatric Oncology, University Children's Hospital, Zurich, Switzerland
| | - Claudia Paris
- Department of Pediatric Oncology and Hematology, Hospital Luis Calvo Mackenna, Santiago, Chile
| | - Shoshana Revel-Vilk
- Shaare Zedek Medical Centre and The Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | | | - Mohamad G Sinno
- Phoenix Children's Hospital, Center for Cancer and Blood Disorders, Phoenix, AZ
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Zabrze and Medical University of Silesia, Katowice, Poland
| | - Melanie Tamesberger
- Department of Pediatrics and Adolescent Medicine, Kepler University Clinic, Linz, Austria
| | | | - Matthias Wolfl
- Pediatric Oncology, Hematology and Stem Cell Transplantation Program, University Children's Hospital Würzburg, Würzburg, Germany
| | - Ronit Nirel
- Department of Statistics and Data Science, Hebrew University, Jerusalem, Israel
| | - Shai Izraeli
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Arndt Borkhardt
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, The University Hospital, Rigshospitalet, and Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Saultier P, Simonin M, Beaumais TAD, Rialland F, Alby-Laurent F, Lubnau M, Desplantes C, Jacqz-Aigrain E, Rohrlich P, Reguerre Y, Rabian F, Sirvent N, Plat GW, Petit A. [Practical management during maintenance therapy of pediatric acute lymphoblastic leukemia: Recommendations of the French Society for Childhood and Adolescent Cancer and Leukemia (SFCE)]. Bull Cancer 2022; 109:1132-1143. [PMID: 35863954 DOI: 10.1016/j.bulcan.2022.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022]
Abstract
Maintenance therapy is the last phase of treatment for acute lymphoblastic leukemia in children and adolescents. Although maintenance therapy is associated with toxicities and specific management issues, it is an essential phase of treatment that reduces the risk of relapse. The objective of this work is to propose a guide for the initiation, administration, and monitoring of maintenance therapy, and for the management of food, schooling, leisure, community life, risk of infection and links with family medicine.
Collapse
Affiliation(s)
- Paul Saultier
- Hôpital de la Timone Enfants, APHM, service d'hématologie, immunologie et oncologie pédiatrique, Marseille, France.
| | - Mathieu Simonin
- AP-HP, hôpital Armand-Trousseau, Sorbonne université, service d'hématologie et oncologie pédiatrique, Paris, France
| | | | - Fanny Rialland
- CHU de Nantes, service d'onco-hématologie pédiatrique, Nantes, France
| | - Fanny Alby-Laurent
- AP-HP, hôpital Armand-Trousseau, Sorbonne université, service d'hématologie et oncologie pédiatrique, Paris, France
| | - Marion Lubnau
- CHU de Nancy, service d'onco-hématologie pédiatrique, Nancy, France
| | | | - Evelyne Jacqz-Aigrain
- AP-HP, hôpital Saint-Louis, département de pharmacologie et pharmacogénétique, Paris, France
| | - Pierre Rohrlich
- CHU de Nice, service d'hématologie pédiatrique, Nice, France
| | - Yves Reguerre
- CHU de la Réunion, service d'hémato-oncologie pédiatrique, Réunion, France
| | - Florence Rabian
- AP-HP, hôpital Saint-Louis, service d'hématologie adolescents et jeunes adultes, Paris, France
| | - Nicolas Sirvent
- CHU de Montpellier, service d'hématologie et oncologie pédiatrique, Montpellier, France
| | - Geneviève Willson Plat
- CHU de Toulouse, service d'hématologie oncologie et immunologie pédiatrique, Toulouse, France
| | - Arnaud Petit
- AP-HP, hôpital Armand-Trousseau, Sorbonne université, service d'hématologie et oncologie pédiatrique, Paris, France
| |
Collapse
|
6
|
Maintenance therapy for acute lymphoblastic leukemia: basic science and clinical translations. Leukemia 2022; 36:1749-1758. [PMID: 35654820 PMCID: PMC9252897 DOI: 10.1038/s41375-022-01591-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 01/21/2023]
Abstract
Maintenance therapy (MT) with oral methotrexate (MTX) and 6-mercaptopurine (6-MP) is essential for the cure of acute lymphoblastic leukemia (ALL). MTX and 6-MP interfere with nucleotide synthesis and salvage pathways. The primary cytotoxic mechanism involves the incorporation of thioguanine nucleotides (TGNs) into DNA (as DNA-TG), which may be enhanced by the inhibition of de novo purine synthesis by other MTX/6-MP metabolites. Co-medication during MT is common. Although Pneumocystis jirovecii prophylaxis appears safe, the benefit of glucocorticosteroid/vincristine pulses in improving survival and of allopurinol to moderate 6-MP pharmacokinetics remains uncertain. Numerous genetic polymorphisms influence the pharmacology, efficacy, and toxicity (mainly myelosuppression and hepatotoxicity) of MTX and thiopurines. Thiopurine S-methyltransferase (encoded by TPMT) decreases TGNs but increases methylated 6-MP metabolites (MeMPs); similarly, nudix hydrolase 15 (encoded by NUDT15) also decreases TGNs available for DNA incorporation. Loss-of-function variants in both genes are currently used to guide MT, but do not fully explain the inter-patient variability in thiopurine toxicity. Because of the large inter-individual variations in MTX/6-MP bioavailability and metabolism, dose adjustments are traditionally guided by the degree of myelosuppression, but this does not accurately reflect treatment intensity. DNA-TG is a common downstream metabolite of MTX/6-MP combination chemotherapy, and a higher level of DNA-TG has been associated with a lower relapse hazard, leading to the development of the Thiopurine Enhanced ALL Maintenance (TEAM) strategy-the addition of low-dose (2.5-12.5 mg/m2/day) 6-thioguanine to the 6-MP/MTX backbone-that is currently being tested in a randomized ALLTogether1 trial (EudraCT: 2018-001795-38). Mutations in the thiopurine and MTX metabolism pathways, and in the mismatch repair genes have been identified in early ALL relapses, providing valuable insights to assist the development of strategies to detect imminent relapse, to facilitate relapse salvage therapy, and even to bring about changes in frontline ALL therapy to mitigate this relapse risk.
Collapse
|
7
|
Toksvang LN, Als-Nielsen B, Bacon C, Bertasiute R, Duarte X, Escherich G, Helgadottir EA, Johannsdottir IR, Jónsson ÓG, Kozlowski P, Langenskjöld C, Lepik K, Niinimäki R, Overgaard UM, Punab M, Räty R, Segers H, van der Sluis I, Smith OP, Strullu M, Vaitkevičienė G, Wik HS, Heyman M, Schmiegelow K. Thiopurine Enhanced ALL Maintenance (TEAM): study protocol for a randomized study to evaluate the improvement in disease-free survival by adding very low dose 6-thioguanine to 6-mercaptopurine/methotrexate-based maintenance therapy in pediatric and adult patients (0-45 years) with newly diagnosed B-cell precursor or T-cell acute lymphoblastic leukemia treated according to the intermediate risk-high group of the ALLTogether1 protocol. BMC Cancer 2022; 22:483. [PMID: 35501736 PMCID: PMC9063225 DOI: 10.1186/s12885-022-09522-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/10/2022] [Indexed: 12/05/2022] Open
Abstract
Background A critical challenge in current acute lymphoblastic leukemia (ALL) therapy is treatment intensification in order to reduce the relapse rate in the subset of patients at the highest risk of relapse. The year-long maintenance phase is essential in relapse prevention. The Thiopurine Enhanced ALL Maintenance (TEAM) trial investigates a novel strategy for ALL maintenance. Methods TEAM is a randomized phase 3 sub-protocol to the ALLTogether1 trial, which includes patients 0–45 years of age with newly diagnosed B-cell precursor or T-cell ALL, and stratified to the intermediate risk-high (IR-high) group, in 13 European countries. In the TEAM trial, the traditional methotrexate (MTX)/6-mercaptopurine (6MP) maintenance backbone (control arm) is supplemented with low dose (2.5–12.5 mg/m2/day) oral 6-thioguanine (6TG) (experimental arm), while the starting dose of 6MP is reduced from 75 to 50 mg/m2/day. A total of 778 patients will be included in TEAM during ~ 5 years. The study will close when the last included patient has been followed for 5 years from the end of induction therapy. The primary objective of the study is to significantly improve the disease-free survival (DFS) of IR-high ALL patients by adding 6TG to 6MP/MTX-based maintenance therapy. TEAM has 80% power to detect a 7% increase in 5-year DFS through a 50% reduction in relapse rate. DFS will be evaluated by intention-to-treat analysis. In addition to reducing relapse, TEAM may also reduce hepatotoxicity and hypoglycemia caused by high levels of methylated 6MP metabolites. Methotrexate/6MP metabolites will be monitored and low levels will be reported back to clinicians to identify potentially non-adherent patients. Discussion TEAM provides a novel strategy for maintenance therapy in ALL with the potential of improving DFS through reducing relapse rate. Potential risk factors that have been considered include hepatic sinusoidal obstruction syndrome/nodular regenerative hyperplasia, second cancer, infection, and osteonecrosis. Metabolite monitoring can potentially increase treatment adherence in both treatment arms. Trial registration EudraCT, 2018–001795-38. Registered 2020-05-15, Clinicaltrials.gov, NCT04307576. Registered 2020-03-13, https://clinicaltrials.gov/ct2/show/NCT04307576 Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09522-3.
Collapse
Affiliation(s)
- Linea Natalie Toksvang
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Bodil Als-Nielsen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | | | - Ruta Bertasiute
- Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
| | - Ximo Duarte
- Instituto Português de Oncologia Lisboa Francisco Gentil Departamento de Pediatria, Lisbon, Portugal
| | | | | | | | | | | | | | | | - Riitta Niinimäki
- Oulu University Hospital and PEDEGRO Research Unit, University of Oulu, Oulu, Finland
| | | | - Mari Punab
- Tartu University Hospital, Tartu, Estonia
| | - Riikka Räty
- Helsinki University Central Hospital, Helsinki, Finland
| | - Heidi Segers
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | | | | | - Marion Strullu
- Université de Paris, hôpital universitaire Robert-Debré (APHP), Paris, France
| | - Goda Vaitkevičienė
- Center for Pediatric Oncology and Hematology, Vilnius University, Vilnius, Lithuania
| | | | - Mats Heyman
- Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.,University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Brady SW, Gout AM, Zhang J. Therapeutic and prognostic insights from the analysis of cancer mutational signatures. Trends Genet 2022; 38:194-208. [PMID: 34483003 PMCID: PMC8752466 DOI: 10.1016/j.tig.2021.08.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 02/08/2023]
Abstract
The somatic mutations in each cancer genome are caused by multiple mutational processes, each of which leaves a characteristic imprint (or 'signature'), potentially caused by specific etiologies or exposures. Deconvolution of these signatures offers a glimpse into the evolutionary history of individual tumors. Recent work has shown that mutational signatures may also yield therapeutic and prognostic insights, including the identification of cell-intrinsic signatures as biomarkers of drug response and prognosis. For example, mutational signatures indicating homologous recombination deficiency are associated with poly(ADP)-ribose polymerase (PARP) inhibitor sensitivity, whereas APOBEC-associated signatures are associated with ataxia telangiectasia and Rad3-related kinase (ATR) inhibitor sensitivity. Furthermore, therapy-induced mutational signatures implicated in cancer progression have also been uncovered, including the identification of thiopurine-induced TP53 mutations in leukemia. In this review, we explore the various ways mutational signatures can reveal new therapeutic and prognostic insights, thus extending their traditional role in identifying disease etiology.
Collapse
Affiliation(s)
- Samuel W Brady
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Alexander M Gout
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
9
|
Yoshino H, Gemma Y, Miyazawa N, Bessho F, Yan K. Therapy-related acute megakaryoblastic leukemia with severe myelofibrosis. Pediatr Int 2022; 64:e14842. [PMID: 35103363 DOI: 10.1111/ped.14842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 04/24/2021] [Accepted: 05/12/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Hiroshi Yoshino
- Department of Pediatrics, Kyorin University School of Medicine, Tokyo, Japan
| | - Yuki Gemma
- Department of Pediatrics, Kyorin University School of Medicine, Tokyo, Japan
| | - Noritaka Miyazawa
- Department of Pediatrics, Kyorin University School of Medicine, Tokyo, Japan
| | - Fumio Bessho
- Department of Pediatrics, Kyorin University School of Medicine, Tokyo, Japan.,Department of Health Science, Nihon Institute of Medical Science, Moroyama, Japan
| | - Kunimasa Yan
- Department of Pediatrics, Kyorin University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Kudo K, Sato T, Takahashi Y, Yuzawa K, Kobayashi A, Kamio T, Sasaki S, Shimada J, Otani K, Tusjimoto S, Kato M, Toki T, Terui K, Ito E. Association of Multiple Gene Polymorphisms Including Homozygous NUDT15 R139C With Thiopurine Intolerance During the Treatment of Acute Lymphoblastic Leukemia. J Pediatr Hematol Oncol 2021; 43:e1173-e1176. [PMID: 33625081 DOI: 10.1097/mph.0000000000002085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/11/2020] [Indexed: 11/26/2022]
Abstract
Although thiopurine is a crucial drug for treating acute lymphoblastic leukemia, individual variations in intolerance are observed due to gene polymorphisms. A 3-year-old boy with B-cell precursor acute lymphoblastic leukemia who was administered thiopurine developed mucositis, sepsis, and hemophagocytic lymphohistiocytosis due to prolonged hematologic toxicity, chronic disseminated candidiasis, and infective endocarditis that triggered multiple brain infarctions. The patient was found to harbor 3 gene polymorphisms associated with thiopurine intolerance including homozygous NUDT15 R139C, heterozygous ITPA C94A, and homozygous MTHFR C677T and heterozygous RFC1 G80A. Thus, the combined effect of intolerance via multiple gene polymorphisms should be considered in case of unexpected adverse reactions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Shinichi Tusjimoto
- Department of Pediatric Hematology and Oncology Research, National Center for Child Health and Development
| | - Motohiro Kato
- Department of Pediatric Hematology and Oncology Research, National Center for Child Health and Development
- Department of Transplantation and Cell Therapy, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | | | | | - Etsuro Ito
- Community Medicine, Hirosaki University Graduate School of Medicine, Hirosaki
| |
Collapse
|
11
|
NUDT15 variants confer high incidence of second malignancies in children with acute lymphoblastic leukemia. Blood Adv 2021; 5:5420-5428. [PMID: 34662904 PMCID: PMC9153020 DOI: 10.1182/bloodadvances.2021005507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/19/2021] [Indexed: 11/20/2022] Open
Abstract
NUDT15 variants may confer a risk of second cancers after treatment with 6-MP in patients with ALL.
The effect of genetic variation on second malignant neoplasms (SMNs) remains unclear. First, we identified the pathogenic germline variants in cancer-predisposing genes among 15 children with SMNs after childhood leukemia/lymphoma using whole-exome sequencing. Because the prevalence was low, we focused on the association between SMNs and NUDT15 in primary acute lymphoblastic leukemia (ALL) cases. NUDT15 is one of the 6-mercaptopurine (6-MP) metabolic genes, and its variants are common in East Asian individuals. The prevalence of NUDT15 hypomorphic variants was higher in patients with SMNs (n = 14; 42.9%) than in the general population in the gnomAD database (19.7%; P = .042). In the validation study with a cohort of 438 unselected patients with ALL, the cumulative incidence of SMNs was significantly higher among those with (3.0%; 95% confidence interval [CI], 0.6% to 9.4%) than among those without NUDT15 variants (0.3%; 95% CI, 0.0% to 1.5%; P = .045). The 6-MP dose administered to patients with ALL with a NUDT15 variant was higher than that given to those without SMNs (P = .045). The 6-MP–related mutational signature was observed in SMN specimens after 6-MP exposure. In cells exposed to 6-MP, a higher level of 6-MP induced DNA damage in NUDT15-knockdown induced pluripotent stem cells. Our study indicates that NUDT15 variants may confer a risk of SMNs after treatment with 6-MP in patients with ALL.
Collapse
|
12
|
Baba SM, Pandith AA, Shah ZA, Geelani SA, Bhat J, Gul A, Guru SA, El-Serehy HA, Koul AM, Mansoor S. GSTT1 null and rs156697 Polymorphism in GSTO2 Influence the Risk and Therapeutic Outcome of B-Acute Lymphoblastic Leukemia Patients. Front Oncol 2021; 11:714421. [PMID: 34722260 PMCID: PMC8552530 DOI: 10.3389/fonc.2021.714421] [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: 05/25/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Glutathione S-transferase (GST) gene deletion or polymorphic sequence variations lead to decreased enzyme activity that influences susceptibility and response to chemotherapy in acute lymphoblastic leukemia (ALL). This case-control study investigated the association of GST gene polymorphisms with the etiology and therapeutic outcome of B-ALL among Kashmiri population. METHODS A total of 300 individuals including 150 newly diagnosed B-ALL patients and an equal number of age and gender matched controls were genotyped for five GST gene polymorphisms by polymerase chain reaction-restriction fragment length polymorphism technique (PCR-RFLP) and multiplex PCR techniques. RESULTS Higher frequency of GSTT1 null, GSTO2-AG, and GSTO2-GG genotypes was observed in ALL cases compared to controls that associated significantly with ALL risk (GSTT1 null: OR = 2.93, p = 0.0001; GSTO2-AG: OR = 2.58, p = 0.01; GSTO2-GG: OR = 3.13, p = 0.01). GSTM1, GSTP1, and GSTO1 SNPs showed no significant association (p > 0.05). Combined genotype analysis revealed significant association of GSTT1 null/GSTM1 null (OR = 4.11, p = 0.011) and GSTT1 null/GSTP1-AG (OR = 4.93, p = 0.0003) with B-ALL susceptibility. Haplotype analysis of rs4925 and rs156697 revealed that carriers of CG haplotype had increased risk of B-ALL (p = 0.04). Kaplan-Meier plots revealed significantly inferior 3-year disease-free survival for GSTO2-GG carriers (p = 0.002). Multivariate analysis confirmed GSTO2-GG as an independent poor prognostic factor for DFS (HR = 4.5, p = 0.034). Among combined genotypes, only GSTT1 null/GSTP1-AG associated significantly with poorer DFS rates (p = 0.032). CONCLUSION This study demonstrated that GSTT1 null individually or in combination with GSTM1null and GSTP1-AG genotypes associated with increased B-ALL risk. Also, rs156697 variant genotypes (AG and GG) associated with B-ALL, whereas the GG genotype of rs156697 influenced the treatment outcome.
Collapse
Affiliation(s)
- Shahid M. Baba
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | | | - Zafar A. Shah
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | | | - Javid R. Bhat
- Advanced Centre for Human Genetics, SKIMS, Srinagar, India
| | - Ayaz Gul
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | - Sameer A. Guru
- Department of Developmental and System Biology, Lurie Children’s Hospital Northwest University, Chicago, IL, United States
| | - Hamed A. El-Serehy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abid M. Koul
- Advanced Centre for Human Genetics, SKIMS, Srinagar, India
| | - Sheikh Mansoor
- Advanced Centre for Human Genetics, SKIMS, Srinagar, India
| |
Collapse
|
13
|
Liang J, Xuan M, Wu W, Li J. GSH-responsive nanofibrous prodrug formed by a short naphthylacetic acid-terminated peptide for 6-mercaptopurine delivery. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Abstract
Pediatric myelodysplastic syndromes (MDS) comprise less than 5% of childhood malignancies. Approximately 30% to 45% of pediatric MDS cases are associated with an underlying genetic predisposition syndrome. A subset of patients present with MDS/acute myeloid leukemia (AML) following intensive chemotherapy for an unrelated malignancy. A definitive diagnosis of MDS can often only be rendered pending a comprehensive clinical and laboratory-based evaluation, which frequently includes ancillary testing in a reference laboratory. Clinical subtypes, the current diagnostic schema, and the results of more recently performed next-generation sequencing studies in pediatric MDS are discussed here.
Collapse
Affiliation(s)
- Sanjay S Patel
- Division of Hematopathology, Weill Cornell Medical College/NewYork-Presbyterian Hospital, 525 East 68th Street, Starr 711A, New York, NY 10065, USA.
| |
Collapse
|
15
|
No association between relapse hazard and thiopurine methyltransferase geno- or phenotypes in non-high risk acute lymphoblastic leukemia: a NOPHO ALL2008 sub-study. Cancer Chemother Pharmacol 2021; 88:271-279. [PMID: 33928426 DOI: 10.1007/s00280-021-04281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/14/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE 6-mercaptopurine(6MP)/methotrexate maintenance therapy is essential to reduce relapse of childhood acute lymphoblastic leukemia (ALL). Common germline variants in TPMT cause low activity of thiopurine methyltransferase (TPMT) and higher 6MP metabolite (TGN) levels. Higher levels of TGNs incorporated into DNA (DNA-TG) and low TPMT activity have previously been associated with a lower relapse risk. We explored if TPMT geno- or phenotype was associated with DNA-TG levels and relapse rate in NOPHO ALL2008. METHODS TPMT genotype, repeated phenotyping, and DNA-TG measurements were collected in 918 children with non-high risk ALL (NOPHO ALL2008 maintenance therapy study). Maintenance therapy started with 6MP at 50 and 75 mg/m2 for TPMT heterozygous and wildtype patients and was adjusted to a target WBC of 1.5 - 3.0 × 109/L. RESULTS Of 918 patients, 78 (8.5%) were TPMT heterozygous and 903 had at least one TPMT measurement (total 3063). Mean TPMT activities were higher with wildtype than heterozygous TPMT (N = 752, 16.6 versus 9.6 U/mL ery., p < 0.001). The 5-year cumulative incidence of relapse was 6.4% and 6.0% for TPMT heterozygous and wildtype patients, and there was no association between genotype and relapse rate (N = 918, hazard ratio = 1.01, 95% confidence interval [CI] 0.40 - 2.54, p = 0.98). Although TPMT heterozygous patients had higher DNA-TG (N = 548, median 760.9 [interquartile range (IQR) 568.7 - 890.3] versus 492.7 [IQR 382.1 - 634.6] fmol/µg, p < 0.001), TPMT activity was not associated with relapse rate (N = 813; hazard ratio = 0.98 per one U/mL ery. increase in TPMT activity, 95% CI 0.91 - 1.06, p = 0.67). CONCLUSION TPMT geno- and phenotype were not associated with relapse in non-high risk NOPHO ALL2008.
Collapse
|
16
|
Dreisig K, Brünner ED, Marquart HV, Helt LR, Nersting J, Frandsen TL, Jonsson OG, Taskinen M, Vaitkeviciene G, Lund B, Abrahamsson J, Lepik K, Schmiegelow K. TPMT polymorphisms and minimal residual disease after 6-mercaptopurine post-remission consolidation therapy of childhood acute lymphoblastic leukaemia. Pediatr Hematol Oncol 2021; 38:227-238. [PMID: 33205673 DOI: 10.1080/08880018.2020.1842570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bone marrow minimal residual disease (MRD) is the strongest predictor of relapse in children with acute lymphoblastic leukemia (ALL). 6-mercaptopurine (6MP) in ALL therapy has wide inter-individual variation in disposition and is strongly influenced by polymorphisms in the thiopurine methyltransferase (TPMT) gene. In 952 patients treated according to the NOPHO ALL2008 protocol, we explored the association between thiopurine disposition, TPMT genotypes and MRD levels after consolidation therapy with 6MP, high-dose methotrexate (HD-MTX), asparaginase, and vincristine. The levels of the cytotoxic DNA-incorporated thioguanine were significantly higher on day 70-79 in G460A/A719G TPMT heterozygous (TPMTHZ) compared to TPMT wild type (TPMTWT) patients (mean: 230.7 vs. 149.7 fmol/µg DNA, p = 0.002). In contrast, TPMT genotype did not associate with the end of consolidation MRD levels irrespective of randomization of the patients to fixed dose (25 mg/m2/day) or 6MP escalation (up to 50 or 75 mg/m2/day) during consolidation therapy.
Collapse
Affiliation(s)
- Karin Dreisig
- Pediatric Oncology research laboratory, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Emilie Damgaard Brünner
- Pediatric Oncology research laboratory, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hanne V Marquart
- The Tissue Typing Laboratory, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Louise Rold Helt
- Pediatric Oncology research laboratory, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jacob Nersting
- Pediatric Oncology research laboratory, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Thomas Leth Frandsen
- Department of Pediatrics and adolescent medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Mervi Taskinen
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Children and Adolescents, Helsinki University Hospital, Helsinki, Finland
| | - Goda Vaitkeviciene
- Children's Hospital, Vilnius University Hospital Santaros Klinikos and Vilnius University, Vilnius, Lithuania
| | - Bendik Lund
- Department of Pediatrics, St. Olavs Hospital, Trondheim; Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jonas Abrahamsson
- Department of Pediatrics, Institution for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Kjeld Schmiegelow
- Pediatric Oncology research laboratory, The University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Pediatrics and adolescent medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Denmark
| |
Collapse
|
17
|
Sous D, Armstrong AE, Huang JT, Shah S, Carlberg VM, Coughlin CC. Cutaneous reactions to pediatric cancer treatment: Part I. Conventional chemotherapy. Pediatr Dermatol 2021; 38:8-17. [PMID: 33170534 DOI: 10.1111/pde.14418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chemotherapies often cause side effects of the skin, nails, and mucosal surfaces. These mucocutaneous toxicities contribute to morbidity and affect quality of life. Identification and management of these drug-induced eruptions is vital to allow for continuation of essential therapies. This review demonstrates the wide range of chemotherapy-induced cutaneous toxicities in children and includes clues for diagnosis as well as tips for counseling and management.
Collapse
Affiliation(s)
- Dana Sous
- Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Amy E Armstrong
- Division of Pediatric Oncology, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.,St. Louis Children's Hospital, St. Louis, MO, USA
| | - Jennifer T Huang
- Dermatology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sonal Shah
- Department of Dermatology, University of California, San Francisco, CA, USA
| | - Valerie M Carlberg
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carrie C Coughlin
- St. Louis Children's Hospital, St. Louis, MO, USA.,Division of Dermatology, Departments of Medicine and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| |
Collapse
|
18
|
Mohanty P, Sahoo N, Das D. Malignant Melanoma: A Double Malignancy or Second Malignant Neoplasm in a Patient of Acute Lymphoblastic Leukemia Following Therapy with a Composite Karyotype. Int J Appl Basic Med Res 2020; 10:218-221. [PMID: 33088748 PMCID: PMC7534717 DOI: 10.4103/ijabmr.ijabmr_143_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 03/26/2020] [Accepted: 05/25/2020] [Indexed: 11/04/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood malignancy of lymphoid origin seen with a peak incidence between 2 and 5 years. New drug regimen has increased the cure rate, although the risk of developing a second malignancy still persists. The common second malignant neoplasms in survivors of childhood ALL are hematolymphoid malignancies, central nervous system tumors, carcinomas, sarcomas, and bone tumors with a median latency of at least 10 years. There are also examples of nonmelanotic skin tumors such as basal cell carcinoma following ALL chemotherapy, but malignant melanoma is an extremely uncommon malignancy encountered. Melanoma is associated with genetic mutations such as CDKN2A, and CDK4 with an increased prevalence of second malignancy such as the lung, pancreas, and breast. However, double malignancy of melanoma with ALL is rare. Here, we report a rare case of malignant melanoma following ALL therapy associated with composite karyotype and early relapse.
Collapse
Affiliation(s)
- Pranita Mohanty
- Department of Pathology, IMS and SUM Hospital, Bhubaneswar, Odisha, India
| | - Nibedita Sahoo
- Department of Pathology, IMS and SUM Hospital, Bhubaneswar, Odisha, India
| | - Debasmita Das
- Department of Pathology, IMS and SUM Hospital, Bhubaneswar, Odisha, India
| |
Collapse
|
19
|
Belitskiy GA, Kirsanov KI, Lesovaya EA, Yakubovskaya MG. Drug-Related Carcinogenesis: Risk Factors and Approaches for Its Prevention. BIOCHEMISTRY (MOSCOW) 2020; 85:S79-S107. [PMID: 32087055 DOI: 10.1134/s0006297920140059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The review summarizes the data on the role of metabolic and repair systems in the mechanisms of therapy-related carcinogenesis and the effect of their polymorphism on the cancer development risk. The carcinogenic activity of different types of drugs, from the anticancer agents to analgesics, antipyretics, immunomodulators, hormones, natural remedies, and non-cancer drugs, is described. Possible approaches for the prevention of drug-related cancer induction at the initiation and promotion stages are discussed.
Collapse
Affiliation(s)
- G A Belitskiy
- Blokhin Russian Cancer Research Center, Ministry of Health of Russian Federation, Moscow, 115478, Russia
| | - K I Kirsanov
- Blokhin Russian Cancer Research Center, Ministry of Health of Russian Federation, Moscow, 115478, Russia. .,Peoples' Friendship University of Russia, Moscow, 117198, Russia
| | - E A Lesovaya
- Blokhin Russian Cancer Research Center, Ministry of Health of Russian Federation, Moscow, 115478, Russia.,Pavlov Ryazan State Medical University, Ryazan, 390026, Russia
| | - M G Yakubovskaya
- Blokhin Russian Cancer Research Center, Ministry of Health of Russian Federation, Moscow, 115478, Russia
| |
Collapse
|
20
|
After 95 years, it's time to eRASe JMML. Blood Rev 2020; 43:100652. [PMID: 31980238 DOI: 10.1016/j.blre.2020.100652] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 12/07/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022]
Abstract
Juvenile myelomonocytic leukaemia (JMML) is a rare clonal disorder of early childhood. Constitutive activation of the RAS pathway is the initial event in JMML. Around 90% of patients diagnosed with JMML carry a mutation in the PTPN11, NRAS, KRAS, NF1 or CBL genes. It has been demonstrated that after this first genetic event, an additional somatic mutation or epigenetic modification is involved in disease progression. The available genetic and clinical data have enabled researchers to establish relationships between JMML and several clinical conditions, including Noonan syndrome, Ras-associated lymphoproliferative disease, and Moyamoya disease. Despite scientific progress and the development of more effective treatments, JMML is still a deadly disease: the 5-year survival rate is ~50%. Here, we report on recent research having led to a better understanding of the genetic and molecular mechanisms involved in JMML.
Collapse
|
21
|
Karppinen S, Lohi O, Vihola M. Prediction of leukocyte counts during paediatric acute lymphoblastic leukaemia maintenance therapy. Sci Rep 2019; 9:18076. [PMID: 31792398 PMCID: PMC6889389 DOI: 10.1038/s41598-019-54492-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 11/12/2019] [Indexed: 12/29/2022] Open
Abstract
Maintenance chemotherapy with oral 6-mercaptopurine and methotrexate remains a cornerstone of modern therapy for acute lymphoblastic leukaemia. The dosage and intensity of therapy are based on surrogate markers such as peripheral blood leukocyte and neutrophil counts. Dosage based leukocyte count predictions could provide support for dosage decisions clinicians face trying to find and maintain an appropriate dosage for the individual patient. We present two Bayesian nonlinear state space models for predicting patient leukocyte counts during the maintenance therapy. The models simplify some aspects of previously proposed models but allow for some extra flexibility. Our second model is an extension which accounts for extra variation in the leukocyte count due to a treatment adversity, infections, using C-reactive protein as a surrogate. The predictive performances of our models are compared against a model from the literature using time series cross-validation with patient data. In our experiments, our simplified models appear more robust and deliver competitive results with the model from the literature.
Collapse
Affiliation(s)
- Santeri Karppinen
- University of Jyväskylä, Department of Mathematics and Statistics, Jyväskylä, FI-40014, Finland.
| | - Olli Lohi
- Tampere Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, FI-33521, Finland
| | - Matti Vihola
- University of Jyväskylä, Department of Mathematics and Statistics, Jyväskylä, FI-40014, Finland
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Myelodysplastic syndromes (MDSs) are rare disorders in children, showing peculiar clinical manifestations and biological features. This review will summarize biological, genetic and clinical features of childhood MDS and will provide an update of the algorithm of treatment of the different disease variants. RECENT FINDINGS The most recent classification of MDS includes refractory cytopenia of childhood (RCC), advanced and therapy-related MDS. Importantly, in children, these clonal hematopoietic disorders may be often associated with inherited bone marrow failure syndromes, this representing a challenge for diagnostic work-up and treatment. Moreover, germline syndromes predisposing to develop MDS/acute myeloid leukemia have been recently identified, such as those caused by mutations in GATA2, ETV6, SRP72 and SAMD9/SAMD9-L. SUMMARY Treatment of childhood MDS varies according to specific disease features; allogeneic hematopoietic stem cell transplantation (HSCT) using a Human Leukocyte antigen (HLA)-identical donor, whenever available, represents the treatment of choice for most of these children. HSCT is indicated in MDS with excess of blasts, or in therapy-related MDS. For RCC patients, HSCT is recommended for RCC associated with monosomy 7, or complex karyotype and for patients showing severe neutropenia or transfusion dependence. Novel approaches of HSCT from an HLA-haploidentical relative after selective graft manipulation allow reducing transplant-related complications.
Collapse
|
23
|
Quantification of Thiopurine Nucleotides in Erythrocytes and Clinical Application to Pediatric Acute Lymphoblastic Leukemia. Ther Drug Monit 2019; 41:75-85. [PMID: 30507626 PMCID: PMC6358190 DOI: 10.1097/ftd.0000000000000575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background: Concentrations of 6-thioguanine (6TG) nucleotides and 6-methylmercaptopurine (6MMP) nucleotides in RBCs were measured using liquid chromatography-tandem mass spectrometry (LC-MS/MS). This assay was validated for clinical use and was applied to blood samples from patients taking mercaptopurine (6MP). Methods: RBCs were hemolyzed and deproteinized using perchloric acid, followed by heating for the hydrolysis of nucleotides, and the resultant base was measured using LC-MS/MS. Precision, recovery, linearity, matrix effect, and limit of quantification was validated for clinical application. Our results were compared with another institution's established LC-MS/MS assay. We measured the concentrations of 6TG and 6MMP in RBCs of pediatric patients with acute lymphoblastic leukemia (ALL), and the clinical impact of those metabolites was investigated. Results: The imprecision coefficient of variations of 6TG and 6MMP were 5.7%–8.1%, and the bias was within 5%. Lower limits of quantification were set at 54 ng/mL for 6TG and 1036 ng/mL for 6MMP. Correlation coefficients for 6TG and 6MMP were 0.997 and 1.0 in a comparison study. For clinical proof-of-concept, 74 blood samples were collected from 37 pediatric ALL patients receiving maintenance therapy. Concentration of 6TG ranged from 16.1 to 880 pmol/8 × 108 RBCs and that of 6MMP from 55 to 20,937 pmol/8 × 108 RBCs. The 6MP metabolites were not correlated with WBC or absolute neutrophil count. On the other hand, the higher 6MMP level was associated with elevated alanine aminotransferase and aspartate aminotransferase. Conclusions: In this study, an assay for the quantification of 6TG and 6MMP in RBCs was established and applied to pediatric ALL patients. Interindividual variability in 6MP metabolite concentrations was considerable and associated with elevation of liver enzymes, which may be useful in the clinical monitoring of 6MP maintenance therapy in pediatric ALL patients.
Collapse
|
24
|
Abstract
Over the last 50 years, the survival rates in children with acute lymphoblastic leukemia (ALL) have increased remarkably. The optimal use of antileukemic agents in cooperative group protocols, central nervous system-directed treatment, improvements in supportive care, and recognition of biological, clinical, and treatment response characteristics that predict patients with a higher or a lower risk of treatment failure have improved 5-year event-free survival rates, reaching more than 85%, and 5-year overall survival rates, reaching more than 90%. Consequently, it has become increasingly important to characterize the occurrence of long-term late effects. ALL treatments have been associated with increased risks for adverse outcomes such as late mortality, secondary malignancies, and neurological, cardiac, endocrine, and social/psychological disorders. In recent decades, cooperative groups in Europe and in the United States have provided essential information about the long-term effects of ALL therapy, giving recommendations for screening as well as facilitating new approaches for reducing late-term morbidity and mortality. Current frontline protocols continue to examine ways to lower the intensity and amount of therapy to reduce late effects, whereas survivorship studies attempt to predict such adverse effects precisely and develop targeted prevention and treatment strategies.
Collapse
Affiliation(s)
- Hande Kızılocak
- Istanbul University-Cerrahpaşa Faculty of Medicine, Department of Pediatric Hematology and Oncology, İstanbul, Turkey
| | - Fatih Okcu
- Texas Children’s Hematology and Oncology Centers, Baylor College of Medicine, Department of Pediatrics, Division of Hematology and Oncology, Houston, TX, USA
| |
Collapse
|
25
|
Ishida Y, Maeda M, Adachi S, Inada H, Kawaguchi H, Hori H, Ogawa A, Kudo K, Kiyotani C, Shichino H, Rikiishi T, Kobayashi R, Sato M, Okamura J, Goto H, Manabe A, Yoshinaga S, Qiu D, Fujimoto J, Kuroda T. Secondary cancer after a childhood cancer diagnosis: viewpoints considering primary cancer. Int J Clin Oncol 2018; 23:1178-1188. [DOI: 10.1007/s10147-018-1303-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/02/2018] [Indexed: 11/24/2022]
|
26
|
How I treat myelodysplastic syndromes of childhood. Blood 2018; 131:1406-1414. [PMID: 29438960 DOI: 10.1182/blood-2017-09-765214] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/27/2018] [Indexed: 02/06/2023] Open
Abstract
Pediatric myelodysplastic syndromes (MDSs) are a heterogeneous group of clonal disorders with an annual incidence of 1 to 4 cases per million, accounting for less than 5% of childhood hematologic malignancies. MDSs in children often occur in the context of inherited bone marrow failure syndromes, which represent a peculiarity of myelodysplasia diagnosed in pediatric patients. Moreover, germ line syndromes predisposing individuals to develop MDS or acute myeloid leukemia have recently been identified, such as those caused by mutations in GATA2, ETV6, SRP72, and SAMD9/SAMD9-L Refractory cytopenia of childhood (RCC) is the most frequent pediatric MDS variant, and it has specific histopathologic features. Allogeneic hematopoietic stem cell transplantation (HSCT) is the treatment of choice for many children with MDSs and is routinely offered to all patients with MDS with excess of blasts, to those with MDS secondary to previously administered chemoradiotherapy, and to those with RCC associated with monosomy 7, complex karyotype, severe neutropenia, or transfusion dependence. Immune-suppressive therapy may be a treatment option for RCC patients with hypocellular bone marrow and the absence of monosomy 7 or a complex karyotype, although the response rate is lower than that observed in severe aplastic anemia, and a relevant proportion of these patients will subsequently need HSCT for either nonresponse or relapse.
Collapse
|
27
|
Piette C, Suciu S, Clappier E, Bertrand Y, Drunat S, Girard S, Yakouben K, Plat G, Dastugue N, Mazingue F, Grardel N, van Roy N, Uyttebroeck A, Costa V, Minckes O, Sirvent N, Simon P, Lutz P, Ferster A, Pluchart C, Poirée M, Freycon C, Dresse MF, Millot F, Chantrain C, van der Werff Ten Bosch J, Norga K, Gilotay C, Rohrlich PS, Benoit Y, Cavé H. Differential impact of drugs on the outcome of ETV6-RUNX1 positive childhood B-cell precursor acute lymphoblastic leukaemia: results of the EORTC CLG 58881 and 58951 trials. Leukemia 2018; 32:244-248. [PMID: 29064485 DOI: 10.1038/leu.2017.289] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- C Piette
- University Department of Pediatrics, Pediatric Onco-Hematology, CHR Citadelle, Liège, Belgium
| | - S Suciu
- EORTC Headquarters, Brussels, Belgium
| | - E Clappier
- Département de Génétique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Institut Universitaire d'Hématologie, Université Paris Diderot, Paris-Sorbonne-Cité, Paris, France
| | - Y Bertrand
- Department of Pediatric Hematology, IHOP, Hospices Civils de Lyon and Claude Bernard Lyon University, Lyon, France
| | - S Drunat
- Département de Génétique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
| | - S Girard
- Laboratory of Hematology, East Lyon University Hospital, Hospices Civils de Lyon, Lyon, France
| | - K Yakouben
- Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Robert Debré, Service d'Immuno-Hématologie pédiatrique, Paris, France
| | - G Plat
- Department of Pediatric Onco-Hematology, Purpan University Hospital, Toulouse, France
| | - N Dastugue
- Department of Pediatric Onco-Hematology, Purpan University Hospital, Toulouse, France
| | - F Mazingue
- Department of Pediatric Hematology-Oncology, Lille University Hospital, Lille, France
| | - N Grardel
- Department of Pediatric Hematology-Oncology, Lille University Hospital, Lille, France
| | - N van Roy
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - A Uyttebroeck
- Department of Pediatrics, University Hospital Gasthuisberg, Leuven, Belgium
| | - V Costa
- Department of Pediatrics, Portuguese Oncology Institute, Porto, Portugal
| | - O Minckes
- Department of Pediatric Onco-Hematology, Caen University Hospital, Caen, France
| | - N Sirvent
- Department of Pediatric Onco-Hematology, Montpellier University Hospital, Montpellier, France
| | - P Simon
- Department of Pediatric Onco-Hematology, Besançon University Hospital, Besançon, France
| | - P Lutz
- Department of Hematology, Hautepierre University Hospital, Strasbourg, France
| | - A Ferster
- Department of Pediatric Onco-Hematology, Hôpital Universitaire des Enfants Reine Fabiola (ULB), Brussels, Belgium
| | - C Pluchart
- Department of Pediatric Onco-Hematology, Reims University Hospital, Reims, France
| | - M Poirée
- Department of Pediatric Onco-Hematology, Nice University Hospital, Nice, France
| | - C Freycon
- Department of Pediatric Onco-Hematology, Grenoble University Hospital, Grenoble, France
| | - M-F Dresse
- University Department of Pediatrics, Pediatric Onco-Hematology, CHR Citadelle, Liège, Belgium
| | - F Millot
- Department of Pediatric Onco-Hematology, Poitiers University Hospital, Poitiers, France
| | - C Chantrain
- Department of Pediatrics, Clinique de l'Espérance, CHC, Liège, Belgium
| | | | - K Norga
- Pediatric Onco-Hematology Unit, Antwerp University Hospital, Antwerp, Belgium
| | - C Gilotay
- EORTC Headquarters, Brussels, Belgium
| | - P-S Rohrlich
- Department of Pediatric Onco-Hematology, Nice University Hospital, Nice, France
| | - Y Benoit
- Department of Pediatric Hematology-Oncology, Ghent University Hospital, Ghent, Belgium
| | - H Cavé
- Département de Génétique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Institut Universitaire d'Hématologie, Université Paris Diderot, Paris-Sorbonne-Cité, Paris, France
| |
Collapse
|
28
|
Roblin X, Paul S, Nancey S, Flourie B. Letter: thiopurines - is less really more? Authors' reply. Aliment Pharmacol Ther 2018; 47:150. [PMID: 29226415 DOI: 10.1111/apt.14385] [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: 12/08/2022]
Affiliation(s)
- X Roblin
- Department of Gastroenterology, University Hospital Saint Etienne, France
| | - S Paul
- Department of Gastroenterology, University Hospital Saint Etienne, France
| | - S Nancey
- Department of Gastroenterology, University Hospital Lyon, Lyon, France
| | - B Flourie
- Department of Gastroenterology, University Hospital Lyon, Lyon, France
| |
Collapse
|
29
|
Nielsen SN, Eriksson F, Rosthoej S, Andersen MK, Forestier E, Hasle H, Hjalgrim LL, Aasberg A, Abrahamsson J, Heyman M, Jónsson ÓG, Pruunsild K, Vaitkeviciené GE, Vettenranta K, Schmiegelow K. Children with low-risk acute lymphoblastic leukemia are at highest risk of second cancers. Pediatr Blood Cancer 2017; 64. [PMID: 28500740 DOI: 10.1002/pbc.26518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 02/02/2017] [Accepted: 02/09/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The improved survival rates for childhood acute lymphoblastic leukemia (ALL) may be jeopardized by the development of a second cancer, which has been associated with thiopurine therapy. PROCEDURE We retrospectively analyzed three sequential Nordic Society of Paediatric Haematology and Oncology's protocols characterized by increasing intensity of thiopurine-based maintenance therapy. We explored the risk of second cancer in relation to protocols, risk group, thiopurine methyltransferase (TPMT) activity, ALL high hyperdiploidy (HeH), and t(12;21)[ETV6/RUNX1]. RESULTS After median 9.5 years (interquartile range, 5.4-15.3 yrs) of follow-up, 40 of 3,591 patients had developed a second cancer, of whom 38 had non-high-risk B-cell precursor ALL. Patients with standard-risk ALL, who received the longest maintenance therapy, had the highest adjusted hazard of second cancer (hazard ratio [HR], intermediate vs. standard risk: 0.16, 95% CI: 0.06-0.43, P < 0.001; HR, high vs. standard risk: 0.09, 95% CI: 0.02-0.49, P = 0.006); no significant effects of protocol, age, or white blood cell count at diagnosis, ALL HeH, or t(12;21)[ETV6/RUNX1] were observed. A subset analysis on the patients with standard-risk ALL did not show an increased hazard of second cancer from either HeH or t(12;21) (adjusted HR 2.02, 95% CI: 0.69-5.96, P = 0.20). The effect of low TPMT low activity was explored in patients reaching maintenance therapy in clinical remission (n = 3,368); no association with second cancer was observed (adjusted HR 1.43, 95% CI: 0.54-3.76, P = 0.47). CONCLUSIONS The rate of second cancer was generally highest in patients with low-risk ALL, but we could not identify a subset at higher risk than others.
Collapse
Affiliation(s)
- Stine N Nielsen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Frank Eriksson
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Rosthoej
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Mette K Andersen
- Department of Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
| | - Erik Forestier
- Department of Medical Biosciences, Clinical Genetics, Umeå University, Umeå, Sweden
| | - Henrik Hasle
- Department of Paediatrics, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Lisa L Hjalgrim
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ann Aasberg
- Department of Paediatrics, University Hospital of Trondheim, Trondheim, Norway
| | - Jonas Abrahamsson
- Department of Pediatrics, Institution for Clinical Sciences Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mats Heyman
- Department of Paediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Ólafur G Jónsson
- Pediatric Hematology-Oncology, Children's Hospital, Barnaspitali Hringsins, Landspitali University Hospital, Reykjavik, Iceland
| | - Kaie Pruunsild
- Department of Oncology and Hematology, Tallinn Children's Hospital, Tallinn, Estonia
| | - Goda E Vaitkeviciené
- Centre for Paediatric Oncology and Haematology, University Children's Hospital, Vilnius, Lithuania
| | - Kim Vettenranta
- Department of Paediatrics, University of Tampere, Tampere, Finland
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Roblin X, Boschetti G, Williet N, Nancey S, Marotte H, Berger A, Phelip JM, Peyrin-Biroulet L, Colombel JF, Del Tedesco E, Paul S, Flourie B. Azathioprine dose reduction in inflammatory bowel disease patients on combination therapy: an open-label, prospective and randomised clinical trial. Aliment Pharmacol Ther 2017; 46:142-149. [PMID: 28449228 DOI: 10.1111/apt.14106] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/09/2017] [Accepted: 03/26/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND Infliximab (IFX) combined with azathioprine (AZA) is more effective than IFX monotherapy in inflammatory bowel disease (IBD). AIM To identify the AZA optimal dose that is required for efficacy when receiving combination therapy. METHODS Patients with IBD in durable remission on combination therapy were enrolled in a 1-year, open-label, prospective trial after randomisation into three groups: AZA steady (2-2.5 mg/kg/day, n=28) vs AZA down (dose was halved 1-1.25 mg/kg/day, n=27) vs AZA stopped (n=26). Primary endpoint was failure defined as occurrence of a clinical relapse and/or any change in IBD therapy. RESULTS Eighty-one patients were included. Five (17.9%), 3 (11.1%), and 8 (30.8%) patients experienced failure at 1 year in groups AZA steady, AZA down and AZA stopped, respectively (P=.1 across the groups). The median trough levels of IFX at inclusion were close to those measured at the end of follow-up in group AZA steady (3.65 vs 3.45 μg/mL, P=.9) and in group AZA down (3.95 vs 3.60 μg/mL, P=.5), whereas these levels dropped from 4.25 to 2.15 μg/mL (P=.02) in group AZA stopped. Four (14.3%), four (14.8%) and 11 (42.3%) patients experienced an unfavourable evolution of IFX pharmacokinetics in groups AZA steady, AZA down and AZA stopped, respectively. A threshold of 6-TGN <105 pmoles/8.108 RBC was associated with an unfavourable evolution of IFX pharmacokinetics. CONCLUSIONS Under combination therapy, AZA dose reduction, but not withdrawal, appears to be as effective as continuation of AZA at full dose.
Collapse
Affiliation(s)
- X Roblin
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - G Boschetti
- Department of Gastroenterology, Hospices Civils de Lyon, INSERM U1111, Lyon, France
| | - N Williet
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - S Nancey
- Department of Gastroenterology, Hospices Civils de Lyon, INSERM U1111, Lyon, France
| | - H Marotte
- Department of Rheumatology, University Hospital of Saint Etienne, Saint Etienne, France
| | - A Berger
- Department of Immunology, CIC1408, GIMAP EA3064, University Hospital of Saint Etienne, Saint Etienne, France
| | - J M Phelip
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - L Peyrin-Biroulet
- Department of Gastroenterology, University Hospital of Nancy, Nancy, France
| | - J F Colombel
- Division of Gastroenterology, Inflammatory Bowel Disease Center, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - E Del Tedesco
- Department of Gastroenterology, University Hospital of Saint Etienne, Saint Etienne, France
| | - S Paul
- Department of Immunology, CIC1408, GIMAP EA3064, University Hospital of Saint Etienne, Saint Etienne, France
| | - B Flourie
- Department of Gastroenterology, Hospices Civils de Lyon, INSERM U1111, Lyon, France
| |
Collapse
|
31
|
Shah RM. Do cytogenetics of acute lymphoblastic leukaemia blasts affect required duration and intensity of maintenance therapy? Lancet Oncol 2017; 18:e291. [PMID: 28593846 DOI: 10.1016/s1470-2045(17)30326-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/02/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Ravi M Shah
- Department of Haematology, Great Ormond Street Hospital, London WC1N 3JH, UK.
| |
Collapse
|
32
|
Abaji R, Krajinovic M. Thiopurine S-methyltransferase polymorphisms in acute lymphoblastic leukemia, inflammatory bowel disease and autoimmune disorders: influence on treatment response. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2017; 10:143-156. [PMID: 28507448 PMCID: PMC5428801 DOI: 10.2147/pgpm.s108123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The thiopurine S-methyltransferase (TPMT) gene encodes for the TPMT enzyme that plays a crucial role in the metabolism of thiopurine drugs. Genetic polymorphisms in this gene can affect the activity of the TPMT enzyme and have been correlated with variability in response to treatment with thiopurines. Advances in the pharmacogenetics of TPMT allowed the development of dosing recommendations and treatment strategies to optimize and individualize prescribing thiopurine in an attempt to enhance treatment efficacy while minimizing toxicity. The influence of genetic polymorphisms in the TPMT gene on clinical outcome has been well-documented and replicated in many studies. In this review, we provide an overview of the evolution, results, conclusions and recommendations of selected studies that investigated the influence of TPMT pharmacogenetics on thiopurine treatment in acute lymphoblastic leukemia, inflammatory bowel disease and autoimmune disorders. We focus mainly on prospective studies that explored the impact of individualized TPMT-based dosing of thiopurines on clinical response. Together, these studies demonstrate the importance of preemptive TPMT genetic screening and subsequent dose adjustment in mitigating the toxicity associated with thiopurine treatment while maintaining treatment efficacy and favorable long-term outcomes. In addition, we briefly address the cost-effectiveness of this pharmacogenetics approach and its impact on clinical practice as well as the importance of recent breakthrough advances in sequencing and genotyping techniques in refining the TPMT genetic screening process.
Collapse
Affiliation(s)
| | - Maja Krajinovic
- Departments of Pediatrics and Pharmacology, CHU Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
33
|
Cîrstea M, Coliță A, Ionescu B, Ghiaur A, Vasilescu D, Dobrea C, Jardan C, Dragomir M, Gheorghe A, Várady Z, Lupu AR. Therapy-related myelodysplastic syndrome after successful treatment of acute promyelocytic leukemia: case report and literature review. REV ROMANA MED LAB 2017. [DOI: 10.1515/rrlm-2017-0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Abstract
In the 2016 revision of the World Health Organization classification the term therapy-related myeloid neoplasia (t-MN) defines a subgroup of acute myeloid leukemia (AML) comprising patients who develop myelodysplastic syndrome (MDS-t) or acute myeloid leukemia (AML-t) after treatment with cytotoxic and/or radiation therapy for various malignancies or autoimmune disorders. We report the case of a 36 year old patient with t-MN (t-MDS) after achieving complete remission (CR) of a PML-RARA positive acute promyelocytic leukemia (APL) at 32 months after diagnosis. Initially classified as low risk APL and treated according to the AIDA protocol - induction and 3 consolidation cycles - the patient achieved a complete molecular response in September 2013 and started maintenance therapy. On follow-up PML-RARA transcript remained negative. In January 2016 leukopenia and thrombocytopenia developed and a peripheral blood smear revealed hypogranular and agranular neutrophils. Immunophenotyping in the bone marrow aspirate identified undifferentiated blast cells that did not express cytoplasmic myeloperoxidase. The cytogenetic study showed normal karyotype. The molecular biology tests not identified PMLRARA transcript. A diagnosis of t-MDS (AREB-2 - WHO 2008) was established. Treatment of AML was started with 2 “3+7” regimens and 1 MEC cycle. Two months from diagnosis, while in CR, an allogeneic HSCT from an unrelated HLA compatible donor was performed after myeloablative regimen. An unfavorable clinical evolution was followed by death on day 9 after transplantation. The occurrence of t-MNs during CR of APL represents a particular problem in terms of follow-up and differential diagnosis of relapse and constitutes a dramatic complication for a disease with a favorable prognosis.
This work was supported by the grants PN 41-087 /PN2-099 from the Romanian Ministry of Research and Technology
Collapse
Affiliation(s)
- Mihaela Cîrstea
- Department of Onco-hematology, Fundeni Clinical Institute , Bucharest , Romania
| | - Adriana Coliță
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | - Bogdan Ionescu
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | | | | | - Camelia Dobrea
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | | | | | - Anca Gheorghe
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | - Zsofia Várady
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | | |
Collapse
|
34
|
DNA-thioguanine nucleotide concentration and relapse-free survival during maintenance therapy of childhood acute lymphoblastic leukaemia (NOPHO ALL2008): a prospective substudy of a phase 3 trial. Lancet Oncol 2017; 18:515-524. [PMID: 28258828 DOI: 10.1016/s1470-2045(17)30154-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/09/2017] [Accepted: 01/17/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Adjustment of mercaptopurine and methotrexate maintenance therapy of acute lymphoblastic leukaemia by leucocyte count is confounded by natural variations. Cytotoxicity is primarily mediated by DNA-incorporated thioguanine nucleotides (DNA-TGN). The aim of this study was to establish whether DNA-TGN concentrations in blood leucocytes during maintenance therapy are associated with relapse-free survival. METHODS In this substudy of the NOPHO ALL2008 phase 3 trial done in 23 hospitals in seven European countries (Denmark, Estonia, Finland, Iceland, Lithuania, Norway, and Sweden), we analysed data from centralised and blinded analyses of 6-mercaptopurine and methotrexate metabolites in blood samples from patients with non-high-risk childhood acute lymphoblastic leukaemia. Eligible patients were aged 1·0-17·9 years; had been diagnosed with non-high-risk precursor B-cell or T-cell leukaemia; had been treated according to the Nordic Society of Pediatric Hematology and Oncology ALL2008 protocol; and had reached maintenance therapy in first remission. Maintenance therapy was (mercaptopurine 75 mg/m2 once per day and methotrexate 20 mg/m2 once per week, targeted to a leucocyte count of 1·5-3·0 × 109 cells per L). We measured DNA-TGN and erythrocyte concentrations of TGN nucleotides, methylated mercaptopurine metabolites, and methotrexate polyglutamates. The primary objective was the association of DNA-TGN concentrations and 6-mercaptopurine and methotrexate metabolites with relapse-free survival. The secondary endpoint was the assessment of DNA-TGN concentration and 6-mercaptopurine and methotrexate metabolites during maintenance therapy phase 2. FINDINGS Between Nov 26, 2008 and June 14, 2016, 1509 patients from the NOPHO ALL2008 study were assessed for eligibility in the DNA-TGN substudy, of which 918 (89%) of 1026 eligible patients had at least one DNA-TGN measurement and were included in the analyses. Median follow-up was 4·6 years (IQR 3·1-6·1). Relapse-free survival was significantly associated with DNA-TGN concentration (adjusted hazard ratio 0·81 per 100 fmol/μg DNA increase, 95% CI 0·67-0·98; p=0·029). In patients with at least five blood samples, erythrocyte concentrations of TGN, methylated mercaptopurine metabolites, and methotrexate polyglutamates were associated with DNA-TGN concentration (all p<0·0001). INTERPRETATION Our results suggest the need for intervention trials to identify clinically applicable strategies for individualised drug dosing to increase DNA-TGN concentration, and randomised studies to investigate whether such strategies improve cure rates compared with current dose adjustments based on white blood cell counts. FUNDING Danish Cancer Society, Childhood Cancer Foundation (Denmark), Childhood Cancer Foundation (Sweden), Nordic Cancer Union, Otto Christensen Foundation, University Hospital Rigshospitalet, and Novo Nordic Foundation.
Collapse
|
35
|
Williet N, Roblin X. Trend towards dose reduction of azathioprine as monotherapy in inflammatory bowel disease patients: what about for combination therapy? Therap Adv Gastroenterol 2017; 10:5-10. [PMID: 28286554 PMCID: PMC5330604 DOI: 10.1177/1756283x16670074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Nicolas Williet
- Department of Gastroenterology, University of Saint-Etienne, France
| | - Xavier Roblin
- Department of Hepatogastroenterology, University Hospital of Saint-Etienne, Allee Albert Raimond, 42270 Saint-Priest en Jarez, France
| |
Collapse
|
36
|
Schmiegelow K, Nersting J, Nielsen SN, Heyman M, Wesenberg F, Kristinsson J, Vettenranta K, Schrøeder H, Weinshilboum R, Jensen KL, Grell K, Rosthoej S. Maintenance therapy of childhood acute lymphoblastic leukemia revisited-Should drug doses be adjusted by white blood cell, neutrophil, or lymphocyte counts? Pediatr Blood Cancer 2016; 63:2104-2111. [PMID: 27447547 DOI: 10.1002/pbc.26139] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/16/2016] [Accepted: 06/22/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND 6-Mercaptopurine (6MP) and methotrexate (MTX) based maintenance therapy is a critical phase of childhood acute lymphoblastic leukemia treatment. Wide interindividual variations in drug disposition warrant frequent doses adjustments, but there is a lack of international consensus on dose adjustment guidelines. PROCEDURE To identify relapse predictors, we collected 28,255 data sets on drug doses and blood counts (median: 47/patient) and analyzed erythrocyte (Ery) levels of cytotoxic 6MP/MTX metabolites in 9,182 blood samples (median: 14 samples/patient) from 532 children on MTX/6MP maintenance therapy targeted to a white blood cell count (WBC) of 1.5-3.5 × 109 /l. RESULTS After a median follow-up of 13.8 years for patients in remission, stepwise Cox regression analysis did not find age, average doses of 6MP and MTX, hemoglobin, absolute lymphocyte counts, thrombocyte counts, or Ery levels of 6-thioguanine nucleotides or MTX (including its polyglutamates) to be significant relapse predictors. The parameters significantly associated with risk of relapse (N = 83) were male sex (hazard ratio [HR] 2.0 [1.3-3.1], P = 0.003), WBC at diagnosis (HR = 1.04 per 10 × 109 /l rise [1.00-1.09], P = 0.048), the absolute neutrophil count (ANC; HR = 1.7 per 109 /l rise [1.3-2.4], P = 0.0007), and Ery thiopurine methyltransferase activity (HR = 2.7 per IU/ml rise [1.1-6.7], P = 0.03). WBC was significantly related to ANC (Spearman correlation coefficient, rs = 0.77; P < 0.001), and only a borderline significant risk factor for relapse (HR = 1.28 [95% CI: 1.00-1.64], P = 0.046) when ANC was excluded from the Cox model. CONCLUSIONS This study indicates that a low neutrophil count is likely to be the best hematological target for dose adjustments of maintenance therapy.
Collapse
Affiliation(s)
- Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark. .,Faculty of Medicine, Institute of Clinical Medicine, University of Copenhagen, Denmark.
| | - Jacob Nersting
- Department of Pediatrics and Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Stine Nygaard Nielsen
- Department of Pediatrics and Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Mats Heyman
- Astrid Lindgrens Barnsjukhus, Stockholm, Sweden
| | - Finn Wesenberg
- Department of Pediatric Oncology, The University Hospital Rikshospitalet, Oslo, Norway
| | - Jon Kristinsson
- Department of Pediatric Oncology, The National Hospital, Reykjavik, Iceland
| | - Kim Vettenranta
- Department of Pediatric Oncology, The University Hospital, Helsinki, Finland
| | - Henrik Schrøeder
- Department of Pediatric Oncology, Århus University Hospital, Denmark
| | | | - Katrine Lykke Jensen
- Section of Biostatistics, Department of Public Health, The University of Copenhagen, Copenhagen, Denmark
| | - Kathrine Grell
- Section of Biostatistics, Department of Public Health, The University of Copenhagen, Copenhagen, Denmark
| | - Susanne Rosthoej
- Section of Biostatistics, Department of Public Health, The University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Levinsen M, Marquart HV, Groth-Pedersen L, Abrahamsson J, Albertsen BK, Andersen MK, Frandsen TL, Harila-Saari A, Pronk C, Ulvmoen A, Vaitkevičienė G, Lähteenmäki PM, Niinimäki R, Taskinen M, Jeppesen M, Schmiegelow K. Leukemic blasts are present at low levels in spinal fluid in one-third of childhood acute lymphoblastic leukemia cases. Pediatr Blood Cancer 2016; 63:1935-42. [PMID: 27447373 DOI: 10.1002/pbc.26128] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/30/2016] [Accepted: 06/08/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Central nervous system (CNS) involvement is associated with relapse in childhood acute lymphoblastic leukemia (ALL) and is a diagnostic challenge. PROCEDURE In a Nordic/Baltic prospective study, we assessed centralized flow cytometry (FCM) of locally fixed cerebrospinal fluid (CSF) samples versus local conventional cytospin-based cytology (CC) for detecting leukemic cells and evaluating kinetics of elimination of leukemic cells in CSF. RESULTS Among 300 patients with newly diagnosed ALL, 87 (29%) had CSF involvement by FCM, while CC was positive in 30 (10%) of 299 patients with available CC data (P < 0.001). Patients with FCM+/CC+ had higher CSF leukemic blast counts compared to patients positive by FCM only (medians: 0.10 vs. 0.017 leukemic blasts/μl, P = 0.006). Patients positive by FCM had higher white blood cell counts in peripheral blood than patients negative by FCM (medians: 45 × 10(9) /l vs. 10 × 10(9) /l, P < 0.001), were younger (medians: 3 years vs. 4 years, P = 0.03), and more frequently had T-cell ALL (18/87 vs. 16/213, P = 0.001). At treatment day 15, five of 52 patients (10%) who had CSF positive by FCM at diagnosis remained so despite at least two doses of weekly intrathecal chemotherapy. CONCLUSIONS Longer follow-up is needed to clarify whether FCM positivity has prognostic significance and is an indicator for intensified CNS-directed therapy.
Collapse
Affiliation(s)
- Mette Levinsen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hanne V Marquart
- Department of Clinical Immunology, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Line Groth-Pedersen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jonas Abrahamsson
- Department of Pediatrics, Institution of Clinical Sciences, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Mette K Andersen
- Department of Clinical Genetics, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Thomas L Frandsen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Arja Harila-Saari
- Department of Pediatrics, Astrid Lindgrens Hospital, Stockholm, Sweden
| | - Cornelis Pronk
- Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - Aina Ulvmoen
- Department of Pediatrics, Oslo University Hospital, Norway
| | - Goda Vaitkevičienė
- Centre for Pediatric Oncology and Hematology, University Children's Hospital, Vilnius, Lithuania
| | | | - Riitta Niinimäki
- Department of Pediatrics, Oulu University Hospital, Oulu, Finland
| | - Mervi Taskinen
- Children and Adolescents, Helsinki University Hospital, Helsinki, Finland
| | - Maria Jeppesen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark.
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
- Division of Pediatric Hematology/Oncology, Perlmutter Cancer Center, NYU Langone Medical Center, New York.
| | | |
Collapse
|
38
|
Long-term outcome of 6-month maintenance chemotherapy for acute lymphoblastic leukemia in children. Leukemia 2016; 31:580-584. [PMID: 27698447 DOI: 10.1038/leu.2016.274] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/29/2016] [Accepted: 09/06/2016] [Indexed: 12/27/2022]
Abstract
In the treatment of childhood acute lymphoblastic leukemia (ALL), excess shortening of maintenance therapy resulted in high relapse rate, as shown by our previous trial, TCCSG L92-13, in which maintenance therapy was terminated at 1 year from initiation of treatment. In this study, we aimed to confirm the long-term outcome of L92-13, and to identify who can or cannot be cured by shorter duration of maintenance therapy. To obtain sentinel cytogenetics information that had been missed before, we performed genetic analysis with genomic microarray and target intron-capture sequencing from diagnostic bone marrow smear. Disease-free survival (DFS) at 10 years from the end of therapy was 66.0±2.8%. Females (n=138) had better DFS (74.6±3.7%) than males (n=142, 57.5±4.2%, P=0.002). Patients with TCF3-PBX1 (n=11) and ETV6-RUNX1 (n=16) had excellent DFS (90.9±8.7% and 93.8±6.1%, respectively), whereas high hyperdiploidy (n=23) was the most unfavorable subgroup, with 56.6±10.3% of DFS. Short duration of therapy can cure more than half of pediatric ALL, especially females, TCF3-PBX1 and ETV6-RUNX1. Our retrospective observations suggest a gender/karyotype inhomogeneity on the impact of brief therapy.
Collapse
|
39
|
Abstract
6-thioguanine nucleotides levels are associated with clinical remission in patients with inflammatory bowel disease (IBD) on thiopurine monotherapy. Recently, few studies investigated the interaction between thiopurine metabolism and anti-tumor necrosis factor therapy among patients with IBD on combotherapy. Two studies demonstrated that infliximab therapy increases 6-thioguanine nucleotides level, while such effect could not be observed with adalimumab. Three studies showed that a Delta mean corpuscular volume >7 and high 6-thioguanine nucleotides levels are associated with favorable outcomes, i.e., greater mucosal healing rates, and have a positive impact on the pharmacokinetics of infliximab. These results suggest a synergistic effect between thiopurine metabolism and anti-tumor necrosis factor therapy, especially with infliximab. We propose here some algorithms for clinical practice integrating thiopurine metabolism in patients with IBD on combotherapy. Further randomized controlled trials are needed to further investigate the relationships between thiopurine metabolism and anti-tumor necrosis factor therapy and to establish the clinical utility of measuring thiopurines' metabolites in these patients in clinical practice. Whether measuring thiopurine metabolism can be used to guide decision-making in patients with IBD on combotherapy when considering drug de-escalation or discontinuation will require further investigation.
Collapse
|
40
|
Lamm N, Maoz K, Bester AC, Im MM, Shewach DS, Karni R, Kerem B. Folate levels modulate oncogene-induced replication stress and tumorigenicity. EMBO Mol Med 2016. [PMID: 26197802 PMCID: PMC4568948 DOI: 10.15252/emmm.201404824] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Chromosomal instability in early cancer stages is caused by replication stress. One mechanism by which oncogene expression induces replication stress is to drive cell proliferation with insufficient nucleotide levels. Cancer development is driven by alterations in both genetic and environmental factors. Here, we investigated whether replication stress can be modulated by both genetic and non-genetic factors and whether the extent of replication stress affects the probability of neoplastic transformation. To do so, we studied the effect of folate, a micronutrient that is essential for nucleotide biosynthesis, on oncogene-induced tumorigenicity. We show that folate deficiency by itself leads to replication stress in a concentration-dependent manner. Folate deficiency significantly enhances oncogene-induced replication stress, leading to increased DNA damage and tumorigenicity in vitro. Importantly, oncogene-expressing cells, when grown under folate deficiency, exhibit a significantly increased frequency of tumor development in mice. These findings suggest that replication stress is a quantitative trait affected by both genetic and non-genetic factors and that the extent of replication stress plays an important role in cancer development.
Collapse
Affiliation(s)
- Noa Lamm
- Department of Genetics, The Alexander Silberman Institute of Life Sciences Edmond J. Safra Campus The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Karin Maoz
- Department of Genetics, The Alexander Silberman Institute of Life Sciences Edmond J. Safra Campus The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Assaf C Bester
- Department of Genetics, The Alexander Silberman Institute of Life Sciences Edmond J. Safra Campus The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael M Im
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Donna S Shewach
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Rotem Karni
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Batsheva Kerem
- Department of Genetics, The Alexander Silberman Institute of Life Sciences Edmond J. Safra Campus The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
41
|
Gu S, Wu Y, Yang J. Screening of cytoprotectors against methotrexate-induced cytogenotoxicity from bioactive phytochemicals. PeerJ 2016; 4:e1983. [PMID: 27190706 PMCID: PMC4867711 DOI: 10.7717/peerj.1983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 04/07/2016] [Indexed: 01/14/2023] Open
Abstract
As a well known anti-neoplastic drug, the cytogenotoxicity of methotrexate (MTX) has received more attention in recent years. To develop a new cytoprotector to reduce the risk of second cancers caused by methotrexate, an umu test combined with a micronucleus assay was employed to estimate the cytoprotective effects of ten kinds of bioactive phytochemicals and their combinations. The results showed that allicin, proanthocyanidins, polyphenols, eleutherosides and isoflavones had higher antimutagenic activities than other phytochemicals. At the highest dose tested, the MTX genetoxicity was suppressed by 34.03%∼67.12%. Of all the bioactive phytochemical combinations, the combination of grape seed proanthocyanidins and eleutherosides from Siberian ginseng as well as green tea polyphenols and eleutherosides exhibited stronger antimutagenic effects; the inhibition rate of methotrexate-induced genotoxicity separately reached 74.7 ± 6.5% and 71.8 ± 4.7%. Pretreatment of Kunming mice with phytochemical combinations revealed an obvious reduction in micronucleus and sperm abnormality rates following exposure to MTX (p < 0.01). Moreover, significant increases in thymus and spleen indices were observed in cytoprotector candidates in treated groups. The results indicated that bioactive phytochemicals combinations had the potential to be used as new cytoprotectors.
Collapse
Affiliation(s)
- Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Luoyang Engineering and Technology Research Center of Microbial Fermentationon, Luoyang, China
| | - Ying Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Henan Engineering Research Center of Food Material, Luoyang, China
| | - Jianbo Yang
- Rice Research Institute, Anhui Academy of Agricultural Science, Hefei, China
| |
Collapse
|
42
|
Teng CJ, Huon LK, Hu YW, Yeh CM, Chien SH, Chen SC, Liu CJ. Secondary Solid Organ Neoplasm in Patients with Acute Lymphoblastic Leukemia: A Nationwide Population-Based Study in Taiwan. PLoS One 2016; 11:e0152909. [PMID: 27035574 PMCID: PMC4817987 DOI: 10.1371/journal.pone.0152909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 03/21/2016] [Indexed: 11/23/2022] Open
Abstract
Background Acute lymphoblastic leukemia (ALL) is more common in children than in adults. Secondary neoplasms (SNs) in childhood ALL have been widely reported. However, only one study has demonstrated SNs in adult ALL. Because of the poorer survival of adult ALL, the incidence might be underestimated. Objective To evaluate the incidence and risk factors of secondary solid organ neoplasms among adult and child ALL patients. Methods Newly diagnosed ALL patients between 1997 and 2011 were recruited from the Taiwan National Health Insurance database. Those who had antecedent or combined malignancies were excluded. Standardized incidence ratios (SIRs) were analyzed to compare the risk of our cohort to general population in the same age, sex and calendar year. Risk factors for SN development were analyzed by Cox proportional hazards models. Effects of treatments were treated as time-dependent variables. Results The 15-year cumulative incidence of SN was 1.9% and 8.4% in 1,381 child and 2,154 adult ALL patients, respectively. The SIR was significantly increased in child ALL (SIR 6.06), but not in adult ALL (SIR 1.16). The SIRs of follow-up periods were 5.14, 2.24, .87 and .71 at ≥ 10 years, 5–10 years, 1–5 years and 0–1, respectively. Overall, 15 SNs developed, and CNS tumors (SIR 11.56) were the most common type. Multivariate analysis showed that age ≥ 20 years (hazard ratio [HR] 5.04), end-stage renal disease (HR 18.98) and cranial irradiation (HR 8.12) were independent risk factors for cancer development. Conclusions When compared with the general population, child ALL shows a increased risk of developing SNs. CNS tumors are the most common type, and cranial irradiation is an independent risk factor. With longer follow-up, the risk of SNs increases. Hence, physicians need to pay more attention on the risk of developing SNs in long-term ALL survivors with risk factors.
Collapse
Affiliation(s)
- Chung-Jen Teng
- Division of Hematology and Oncology, Department of Medicine, Far Eastern Memorial Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Leh-Kiong Huon
- Department of Otolaryngology-Head & Neck Surgery, Cathay General Hospital, Taipei, Taiwan
- School of Medicine, Fu Jen Catholic University, Taipei, Taiwan
| | - Yu-Wen Hu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chiu-Mei Yeh
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sheng-Hsuan Chien
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - San-Chi Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (SC); (CL)
| | - Chia-Jen Liu
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (SC); (CL)
| |
Collapse
|
43
|
Roy LM, Zur RM, Uleryk E, Carew C, Ito S, Ungar WJ. Thiopurine S-methyltransferase testing for averting drug toxicity in patients receiving thiopurines: a systematic review. Pharmacogenomics 2016; 17:633-56. [PMID: 27020704 PMCID: PMC4931919 DOI: 10.2217/pgs.16.12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AIM Thiopurine S-methyltransferase (TPMT) testing is used in patients receiving thiopurines to identify enzyme deficiencies and risk for adverse drug reactions. It is uncertain whether genotyping is superior to phenotyping. The objectives were to conduct a systematic review of TPMT-test performance studies. MATERIALS & METHODS Electronic and grey literature sources were searched for studies reporting test performance compared with a reference standard. Sixty-six eligible studies were appraised for quality. RESULTS Thirty phenotype-genotype and six phenotype-phenotype comparisons were of high quality. The calculated sensitivity and specificity for genotyping to identify a homozygous mutation ranged from 0.0-100.0% and from 97.8-100.0%, respectively. CONCLUSION Clinical decision-makers require high-quality evidence of clinical validity and clinical utility of TPMT genotyping to ensure appropriate use in patients.
Collapse
Affiliation(s)
- Lilla M Roy
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Richard M Zur
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Elizabeth Uleryk
- Library Services, The Hospital for Sick Children, Toronto, Canada
| | - Chris Carew
- Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Shinya Ito
- Division of Clinical Pharmacology & Toxicology, The Hospital for Sick Children, Toronto, Canada
- Departments of Pharmacology & Pharmacy, Faculty of Medicine Department of Paediatrics, University of Toronto, Canada
| | - Wendy J Ungar
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute, Toronto, Canada
- Institute for Health Policy, Management & Evaluation, University of Toronto, Canada
| |
Collapse
|
44
|
Jayachandran D, Laínez-Aguirre J, Rundell A, Vik T, Hannemann R, Reklaitis G, Ramkrishna D. Model-Based Individualized Treatment of Chemotherapeutics: Bayesian Population Modeling and Dose Optimization. PLoS One 2015; 10:e0133244. [PMID: 26226448 PMCID: PMC4520687 DOI: 10.1371/journal.pone.0133244] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 06/25/2015] [Indexed: 11/18/2022] Open
Abstract
6-Mercaptopurine (6-MP) is one of the key drugs in the treatment of many pediatric cancers, auto immune diseases and inflammatory bowel disease. 6-MP is a prodrug, converted to an active metabolite 6-thioguanine nucleotide (6-TGN) through enzymatic reaction involving thiopurine methyltransferase (TPMT). Pharmacogenomic variation observed in the TPMT enzyme produces a significant variation in drug response among the patient population. Despite 6-MP's widespread use and observed variation in treatment response, efforts at quantitative optimization of dose regimens for individual patients are limited. In addition, research efforts devoted on pharmacogenomics to predict clinical responses are proving far from ideal. In this work, we present a Bayesian population modeling approach to develop a pharmacological model for 6-MP metabolism in humans. In the face of scarcity of data in clinical settings, a global sensitivity analysis based model reduction approach is used to minimize the parameter space. For accurate estimation of sensitive parameters, robust optimal experimental design based on D-optimality criteria was exploited. With the patient-specific model, a model predictive control algorithm is used to optimize the dose scheduling with the objective of maintaining the 6-TGN concentration within its therapeutic window. More importantly, for the first time, we show how the incorporation of information from different levels of biological chain-of response (i.e. gene expression-enzyme phenotype-drug phenotype) plays a critical role in determining the uncertainty in predicting therapeutic target. The model and the control approach can be utilized in the clinical setting to individualize 6-MP dosing based on the patient's ability to metabolize the drug instead of the traditional standard-dose-for-all approach.
Collapse
Affiliation(s)
- Devaraj Jayachandran
- School of Chemical Engineering, Purdue University, 480 Stadium Mall Way, West Lafayette, IN, 47907, United States of America
| | - José Laínez-Aguirre
- School of Chemical Engineering, Purdue University, 480 Stadium Mall Way, West Lafayette, IN, 47907, United States of America
| | - Ann Rundell
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN, 47907, United States of America
| | - Terry Vik
- Riley Hospital for Children, 702 Barnhill Drive, Indianapolis, IN, 46202, United States of America
| | - Robert Hannemann
- School of Chemical Engineering, Purdue University, 480 Stadium Mall Way, West Lafayette, IN, 47907, United States of America
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN, 47907, United States of America
| | - Gintaras Reklaitis
- School of Chemical Engineering, Purdue University, 480 Stadium Mall Way, West Lafayette, IN, 47907, United States of America
| | - Doraiswami Ramkrishna
- School of Chemical Engineering, Purdue University, 480 Stadium Mall Way, West Lafayette, IN, 47907, United States of America
| |
Collapse
|
45
|
Tanaka Y, Kato M, Hasegawa D, Urayama KY, Nakadate H, Kondoh K, Nakamura K, Koh K, Komiyama T, Manabe A. Susceptibility to 6-MP toxicity conferred by aNUDT15variant in Japanese children with acute lymphoblastic leukaemia. Br J Haematol 2015; 171:109-15. [DOI: 10.1111/bjh.13518] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/16/2015] [Indexed: 11/25/2022]
Affiliation(s)
- Yoichi Tanaka
- Department of Clinical Pharmacy; Centre for Clinical Pharmacy and Sciences; School of Pharmacy; Kitasato University; Tokyo Japan
| | - Motohiro Kato
- Department of Paediatrics; The University of Tokyo; Tokyo Japan
| | - Daisuke Hasegawa
- Department of Paediatrics; St. Luke's International Hospital; Tokyo Japan
| | - Kevin Y. Urayama
- Department of Human Genetics and Disease Diversity; Tokyo Medical and Dental University; Tokyo Japan
- Centre for Clinical Epidemiology; St. Luke's Life Science Institute; Tokyo Japan
| | - Hisaya Nakadate
- Department of General Paediatrics & Interdisciplinary Medicine; National Centre for Child Health and Development; Tokyo Japan
| | - Kensuke Kondoh
- Department of Paediatrics; St. Marianna University School of Medicine; Kanagawa Japan
| | - Kozue Nakamura
- Department of Paediatrics; Teikyo University Hospital; Tokyo Japan
| | - Katsuyoshi Koh
- Department of Haematology/Oncology; Saitama Children's Medical Centre; Saitama Japan
| | - Takako Komiyama
- Department of Clinical Pharmacy; Centre for Clinical Pharmacy and Sciences; School of Pharmacy; Kitasato University; Tokyo Japan
| | - Atsushi Manabe
- Department of Paediatrics; St. Luke's International Hospital; Tokyo Japan
| |
Collapse
|
46
|
Matimba A, Li F, Livshits A, Cartwright CS, Scully S, Fridley BL, Jenkins G, Batzler A, Wang L, Weinshilboum R, Lennard L. Thiopurine pharmacogenomics: association of SNPs with clinical response and functional validation of candidate genes. Pharmacogenomics 2015; 15:433-47. [PMID: 24624911 DOI: 10.2217/pgs.13.226] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM We investigated candidate genes associated with thiopurine metabolism and clinical response in childhood acute lymphoblastic leukemia. MATERIALS & METHODS We performed genome-wide SNP association studies of 6-thioguanine and 6-mercaptopurine cytotoxicity using lymphoblastoid cell lines. We then genotyped the top SNPs associated with lymphoblastoid cell line cytotoxicity, together with tagSNPs for genes in the 'thiopurine pathway' (686 total SNPs), in DNA from 589 Caucasian UK ALL97 patients. Functional validation studies were performed by siRNA knockdown in cancer cell lines. RESULTS SNPs in the thiopurine pathway genes ABCC4, ABCC5, IMPDH1, ITPA, SLC28A3 and XDH, and SNPs located within or near ATP6AP2, FRMD4B, GNG2, KCNMA1 and NME1, were associated with clinical response and measures of thiopurine metabolism. Functional validation showed shifts in cytotoxicity for these genes. CONCLUSION The clinical response to thiopurines may be regulated by variation in known thiopurine pathway genes and additional novel genes outside of the thiopurine pathway.
Collapse
Affiliation(s)
- Alice Matimba
- Division of Clinical Pharmacology, Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Treatment-related myelodysplastic syndrome in a child with acute myeloid leukemia and TPMT heterozygosity. J Pediatr Hematol Oncol 2015; 37:e242-4. [PMID: 25000470 DOI: 10.1097/mph.0000000000000211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION We describe a patient diagnosed with acute myeloid leukemia (AML) and low activity of thiopurine methyltransferase (TPMT) who developed secondary myelodysplastic syndrome after treatment. OBSERVATION A 10-year-old boy presented with AML-M2 with t(8;21)(q22;q22) and genotyping revealing 3*B TPMT heterozygosity. The patient was treated according to the NOPHO-AML 2004 protocol. Two years after the treatment, the patient presented with neutropenia and thrombocytopenia. Bone marrow, including fluorescent in situ hybridization and retrospective aCGH analysis, verified therapy-related myelodysplastic syndrome with ring chromosome 6. DISCUSSION The clinical course of this patient raises the possibility that low-activity TPMT genotypes may influence 6TG toxicity in patients with AML and lead to an increased risk of developing secondary malignant neoplasms.
Collapse
|
48
|
Niehues T. Optimizing treatment in paediatric rheumatology--lessons from oncology. Nat Rev Rheumatol 2015; 11:493-9. [PMID: 25900208 DOI: 10.1038/nrrheum.2015.50] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Treatment of children with cancer, in particular with acute lymphoblastic leukaemia (ALL), has been highly successful in the past two decades owing to the implementation of treatment optimization studies. Study centres appointed by scientific societies design treatment optimization study protocols (TOSPs) that address an investigator-initiated research question and detail treatment procedures according to these aims. Nearly all children with malignant diseases are treated within TOSPs, whereas children with juvenile idiopathic arthritis (JIA) and other common paediatric rheumatic diseases are mostly treated outside TOSPs and clinical trials. Despite the differences in natural course and prognosis between malignant and inflammatory diseases, aiming for the recruitment of all children with defined rheumatic diseases into TOSPs or similar protocols would enable the longitudinal collection of crucial clinical data and improve evidence-based approaches. Successful research networks already exist in paediatric rheumatology that could facilitate the implementation of this approach. Paediatric rheumatic diseases have a considerable impact on patients and their families; thus, I propose that research networks in paediatric rheumatology should recruit most--if not all--children with rheumatic diseases into study protocols with standardized treatment and outcome measures.
Collapse
Affiliation(s)
- Tim Niehues
- HELIOS Klinikum Krefeld, Lutherplatz 40, 47805 Krefeld, Germany
| |
Collapse
|
49
|
Gregers J, Gréen H, Christensen IJ, Dalhoff K, Schroeder H, Carlsen N, Rosthoej S, Lausen B, Schmiegelow K, Peterson C. Polymorphisms in the ABCB1 gene and effect on outcome and toxicity in childhood acute lymphoblastic leukemia. THE PHARMACOGENOMICS JOURNAL 2015; 15:372-9. [PMID: 25582575 PMCID: PMC4762905 DOI: 10.1038/tpj.2014.81] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/12/2014] [Accepted: 11/05/2014] [Indexed: 01/16/2023]
Abstract
The membrane transporter P-glycoprotein, encoded by the ABCB1 gene, influences the pharmacokinetics of anti-cancer drugs. We hypothesized that variants of ABCB1 affect outcome and toxicity in childhood acute lymphoblastic leukemia (ALL). We studied 522 Danish children with ALL, 93% of all those eligible. Risk of relapse was increased 2.9-fold for patients with the 1199GA variant versus 1199GG (P=0.001), and reduced 61% and 40%, respectively, for patients with the 3435CT or 3435TT variants versus 3435CC (overall P=0.02). The degree of bone marrow toxicity during doxorubicin, vincristine and prednisolone induction therapy was more prominent in patients with 3435TT variant versus 3435CT/3435CC (P=0.01/P<0.0001). We observed more liver toxicity after high-dose methotrexate in patients with 3435CC variant versus 3435CT/TT (P=0.03). In conclusion, there is a statistically significant association between ABCB1 polymorphisms, efficacy and toxicity in the treatment of ALL, and ABCB1 1199G>A may be a new possible predictive marker for outcome in childhood ALL.
Collapse
Affiliation(s)
- J Gregers
- 1] Clinical Pharmacology, Faculty of Health Sciences, Division of Drug Research, Department of Medical and Health Sciences, Linköpings Universitet, Linköping, Sweden [2] Laboratory of Molecular Medicine, Department of Clinical Immunology 7631, Rigshospitalet, University Hospital in Copenhagen, Copenhagen, Denmark
| | - H Gréen
- 1] Clinical Pharmacology, Faculty of Health Sciences, Division of Drug Research, Department of Medical and Health Sciences, Linköpings Universitet, Linköping, Sweden [2] Science for Life Laboratory, Division of Gene Technology, School of Biotechnology, KTH Royal Institute of Technology, Solna, Sweden [3] Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | | | - K Dalhoff
- Department of Clinical Pharmacology, Bispebjerg Hospital, Copenhagen, Denmark
| | - H Schroeder
- Department of Pediatric, University Hospital in Skejby, Aarhus, Denmark
| | - N Carlsen
- Department of Pediatric, University Hospital in Odense, Odense, Denmark
| | - S Rosthoej
- Department of Pediatric, University Hospital in Aalborg, Aalborg, Denmark
| | - B Lausen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - K Schmiegelow
- 1] Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark [2] The Medical Faculty, Institute of Gynecology, Obstetrics and Pediatrics, University of Copenhagen, Copenhagen, Denmark
| | - C Peterson
- Clinical Pharmacology, Faculty of Health Sciences, Division of Drug Research, Department of Medical and Health Sciences, Linköpings Universitet, Linköping, Sweden
| |
Collapse
|
50
|
Pui CH. Genomic and pharmacogenetic studies of childhood acute lymphoblastic leukemia. Front Med 2014; 9:1-9. [DOI: 10.1007/s11684-015-0381-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/15/2014] [Indexed: 11/30/2022]
|