1
|
Podpeskar A, Crazzolara R, Kropshofer G, Obexer P, Rabensteiner E, Michel M, Salvador C. Supportive methods for childhood acute lymphoblastic leukemia then and now: A compilation for clinical practice. Front Pediatr 2022; 10:980234. [PMID: 36172391 PMCID: PMC9510731 DOI: 10.3389/fped.2022.980234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
Survival of childhood acute lymphoblastic leukemia has significantly improved over the past decades. In the early years of chemotherapeutic development, improvement in survival rates could be attained only by increasing the cytostatic dose, also by modulation of the frequency and combination of chemotherapeutic agents associated with severe short- and long-time side-effects and toxicity in a developing child's organism. Years later, new treatment options have yielded promising results through targeted immune and molecular drugs, especially in relapsed and refractory leukemia, and are continuously added to conventional therapy or even replace first-line treatment. Compared to conventional strategies, these new therapies have different side-effects, requiring special supportive measures. Supportive treatment includes the prevention of serious acute and sometimes life-threatening events as well as managing therapy-related long-term side-effects and preemptive treatment of complications and is thus mandatory for successful oncological therapy. Inadequate supportive therapy is still one of the main causes of treatment failure, mortality, poor quality of life, and unsatisfactory long-term outcome in children with acute lymphoblastic leukemia. But nowadays it is a challenge to find a way through the flood of supportive recommendations and guidelines that are available in the literature. Furthermore, the development of new therapies for childhood leukemia has changed the range of supportive methods and must be observed in addition to conventional recommendations. This review aims to provide a clear and recent compilation of the most important supportive methods in the field of childhood leukemia, based on conventional regimes as well as the most promising new therapeutic approaches to date.
Collapse
Affiliation(s)
- Alexandra Podpeskar
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Roman Crazzolara
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Kropshofer
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Petra Obexer
- Department of Pediatrics II, Medical University of Innsbruck, Innsbruck, Austria
| | - Evelyn Rabensteiner
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Miriam Michel
- Division of Cardiology, Department of Pediatrics III, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Salvador
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Murphy L, Maloney K, Gore L, Blanchette E. Hypertension in Pediatric Acute Lymphoblastic Leukemia Patients: Prevalence, Impact, and Management Strategies. Integr Blood Press Control 2022; 15:1-10. [PMID: 35082528 PMCID: PMC8784271 DOI: 10.2147/ibpc.s242244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/17/2021] [Indexed: 01/27/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer diagnosed in children under the age of 18. While modern diagnostic technologies, risk-stratification, and therapy intensification have led to outstanding outcomes for many children with ALL, the side effects and consequences of therapy are not to be underestimated. Hypertension is a well-known acute and chronic side effect of treatment for childhood ALL, although limited data are available regarding the prevalence of hypertension in children undergoing treatment for ALL. In this review of hypertension in pediatric ALL patients, we examine the existing data on incidence and prevalence during treatment and in pediatric ALL survivors. We describe independent risk factors for development of hypertension along with treatment-related causes. Long-term consequences and the risk to survivors of pediatric ALL are further defined. While many ALL patients require antihypertensive medications during some portion of their treatment, there are no clear guidelines on treating inpatient hypertension given challenges that exist in recognizing and managing hypertension in this setting and in this population. Here, we propose an algorithmic approach to diagnose and treat pediatric ALL patients with HTN, along with monitoring and continuation versus cessation of antihypertensive therapy as an outpatient.
Collapse
Affiliation(s)
- Lindsey Murphy
- Department of Pediatrics, Sections of Hematology/Oncology/Bone Marrow Transplant-Cellular Therapeutics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| | - Kelly Maloney
- Department of Pediatrics, Sections of Hematology/Oncology/Bone Marrow Transplant-Cellular Therapeutics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| | - Lia Gore
- Department of Pediatrics, Sections of Hematology/Oncology/Bone Marrow Transplant-Cellular Therapeutics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
- Correspondence: Lia Gore Tel +17207776458Fax +17207777339 Email
| | - Eliza Blanchette
- Department of Pediatrics, Section of Nephrology, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
3
|
Determination of NUDT15 variants by targeted sequencing can identify compound heterozygosity in pediatric acute lymphoblastic leukemia patients. Sci Rep 2020; 10:14400. [PMID: 32873882 PMCID: PMC7463237 DOI: 10.1038/s41598-020-71468-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
Mercaptopurine intolerance is an adverse effect of mercaptopurine administration in pediatric acute lymphoblastic leukemia. Recently, NUDT15 variants were identified as a major determinant of mercaptopurine intolerance. Two NUDT15 variants, c.36_37insGGAGTC and c.415C > T, are located on exons 1 and 3, respectively. Patients with heterozygous c.36_37insGGAGTC and c.415C > T can be either compound heterozygous with two variants on different alleles or heterozygous with both variants on the same allele. Because patients with biallelic NUDT15 variants are extremely sensitive to mercaptopurine, clinical identification of NUDT15 diplotype would be advantageous. A cohort of 37 patients with c.36_37insGGAGTC and c.415C > T NUDT15 variants were selected for haplotyping by targeted sequencing. NUDT15 complementary DNA was amplified and sequenced by 300-bp paired-end sequencing on Illumina MiSeq. Of the 37 patients carrying NUDT15 variants, 35 had heterozygous NUDT15*1/*2 variants and two had compound heterozygous NUDT15*3/*6 and NUDT15*2/*7 variants. These two patients with compound heterozygous variants could only tolerate low doses of mercaptopurine, similar to patients with homozygous NUDT15 variants. Targeted sequencing of NUDT15 cDNA can be used to determine NUDT15 diplotype and identify patients with compound heterozygous NUDT15 variants.
Collapse
|
4
|
Hypertension is a modifiable risk factor for osteonecrosis in acute lymphoblastic leukemia. Blood 2019; 134:983-986. [PMID: 31409674 DOI: 10.1182/blood.2019000006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
5
|
Abstract
Advances in genomic research and risk-directed therapy have led to improvements in the long-term survival and quality of life outcomes of patients with childhood acute lymphoblastic leukaemia (ALL). The application of next-generation sequencing technologies, especially transcriptome sequencing, has resulted in the identification of novel molecular subtypes of ALL with prognostic and therapeutic implications, as well as cooperative mutations that account for much of the heterogeneity in clinical responses observed among patients with specific ALL subtypes. In addition, germline genetic variants have been shown to influence the risk of developing ALL and/or the responses of non-malignant and leukaemia cells to therapy; shared pathways for drug activation and metabolism are implicated in treatment-related toxicity and drug sensitivity or resistance, depending on whether the genetic changes are germline, somatic or both. Indeed, although once considered a non-hereditary disease, genomic investigations of familial and sporadic ALL have revealed a growing number of genetic alterations or conditions that predispose individuals to the development of ALL and treatment-related second cancers. The identification of these genetic alterations holds the potential to direct genetic counselling, testing and possibly monitoring for the early detection of ALL and other cancers. Herein, we review these advances in our understanding of the genomic landscape of childhood ALL and their clinical implications.
Collapse
|
6
|
Pavlovic S, Kotur N, Stankovic B, Zukic B, Gasic V, Dokmanovic L. Pharmacogenomic and Pharmacotranscriptomic Profiling of Childhood Acute Lymphoblastic Leukemia: Paving the Way to Personalized Treatment. Genes (Basel) 2019; 10:E191. [PMID: 30832275 PMCID: PMC6471971 DOI: 10.3390/genes10030191] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
Personalized medicine is focused on research disciplines which contribute to the individualization of therapy, like pharmacogenomics and pharmacotranscriptomics. Acute lymphoblastic leukemia (ALL) is the most common malignancy of childhood. It is one of the pediatric malignancies with the highest cure rate, but still a lethal outcome due to therapy accounts for 1%⁻3% of deaths. Further improvement of treatment protocols is needed through the implementation of pharmacogenomics and pharmacotranscriptomics. Emerging high-throughput technologies, including microarrays and next-generation sequencing, have provided an enormous amount of molecular data with the potential to be implemented in childhood ALL treatment protocols. In the current review, we summarized the contribution of these novel technologies to the pharmacogenomics and pharmacotranscriptomics of childhood ALL. We have presented data on molecular markers responsible for the efficacy, side effects, and toxicity of the drugs commonly used for childhood ALL treatment, i.e., glucocorticoids, vincristine, asparaginase, anthracyclines, thiopurines, and methotrexate. Big data was generated using high-throughput technologies, but their implementation in clinical practice is poor. Research efforts should be focused on data analysis and designing prediction models using machine learning algorithms. Bioinformatics tools and the implementation of artificial i Lack of association of the CEP72 rs924607 TT genotype with intelligence are expected to open the door wide for personalized medicine in the clinical practice of childhood ALL.
Collapse
Affiliation(s)
- Sonja Pavlovic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Nikola Kotur
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Biljana Stankovic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Branka Zukic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Vladimir Gasic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Lidija Dokmanovic
- University Children's Hospital, 11000 Belgrade, Serbia.
- University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia.
| |
Collapse
|
7
|
Incidence and Risk Factors for Hypertension During Childhood Acute Lymphoblastic Leukemia Therapy. Indian Pediatr 2018. [DOI: 10.1007/s13312-018-1399-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Warris LT, van den Akker ELT, Bierings MB, van den Bos C, Aarsen FK, Zwaan MC, Tissing WJE, Veening MA, Pieters R, van den Heuvel-Eibrink MM. Eating behavior during dexamethasone treatment in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2017; 64. [PMID: 28598548 DOI: 10.1002/pbc.26679] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 04/28/2017] [Accepted: 04/29/2017] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIM Large prospective studies on dexamethasone-induced changes in eating behavior, energy, and nutrient intake are lacking in pediatric acute lymphoblastic leukemia (ALL). We prospectively studied eating behavior, energy, nutrient intake, and the effect on leptin and adiponectin levels during dexamethasone administration in children with ALL. PATIENTS Parents of patients with ALL (3-16 years) completed a dietary diary for their child during 4 days of dexamethasone (6 mg/m2 ) administration. Energy intake and nutrient intake (energy percentage = E%) were assessed and compared with the recommended intake. The Dutch Eating Behavior Questionnaire for Children was completed before start and after 4 days of dexamethasone administration by patients of 7-12 years of age. Fasting leptin and adiponectin levels were also measured before start and after 4 days of dexamethasone administration. RESULTS Energy intake per day(kcal) (N = 44) increased significantly during dexamethasone (median day 1: 1,103 (717-1,572) versus day 4: 1,482 (1,176-1,822), P < 0.01), including an increase in total protein, fat, saturated fat, carbohydrate, and sodium intake. Intake of saturated fat (median day 4: 12 E%) and salt (median day 4: 1.9 g/day) exceeded the healthy range for age and gender. With respect to eating behavior, dexamethasone significantly decreased restrained eating (P = 0.04). Leptin levels as well as adiponectin levels increased significantly during the dexamethasone course. CONCLUSIONS Four days of dexamethasone treatment significantly increased energy intake, including excessive saturated fat and salt intake, and changed eating behavior in children with ALL. Nutritional and behavioral interventions during dexamethasone treatment are recommended to stimulate a healthy lifestyle.
Collapse
Affiliation(s)
- Lidewij T Warris
- Department of Pediatric Oncology, Erasmus MC Cancer Institute, Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pediatric Endocrinology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Erica L T van den Akker
- Department of Pediatric Endocrinology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Marc B Bierings
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pediatric Hematology and Oncology, University Medical Center Utrecht-Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Cor van den Bos
- Department of Pediatric Oncology, Academic Medical Center-Emma Children's Hospital, Amsterdam, The Netherlands
| | - Femke K Aarsen
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Michel C Zwaan
- Department of Pediatric Oncology, Erasmus MC Cancer Institute, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Wim J E Tissing
- Department of Pediatric Hematology and Oncology, Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Margreet A Veening
- Department of Pediatric Hematology and Oncology, VU Medical Center, Amsterdam, The Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | |
Collapse
|
9
|
England J, Drouin S, Beaulieu P, St-Onge P, Krajinovic M, Laverdière C, Levy E, Marcil V, Sinnett D. Genomic determinants of long-term cardiometabolic complications in childhood acute lymphoblastic leukemia survivors. BMC Cancer 2017; 17:751. [PMID: 29126409 PMCID: PMC5681795 DOI: 10.1186/s12885-017-3722-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 10/30/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND While cure rates for childhood acute lymphoblastic leukemia (cALL) now exceed 80%, over 60% of survivors will face treatment-related long-term sequelae, including cardiometabolic complications such as obesity, insulin resistance, dyslipidemia and hypertension. Although genetic susceptibility contributes to the development of these problems, there are very few studies that have so far addressed this issue in a cALL survivorship context. METHODS In this study, we aimed at evaluating the associations between common and rare genetic variants and long-term cardiometabolic complications in survivors of cALL. We examined the cardiometabolic profile and performed whole-exome sequencing in 209 cALL survivors from the PETALE cohort. Variants associated with cardiometabolic outcomes were identified using PLINK (common) or SKAT (common and rare) and a logistic regression was used to evaluate their impact in multivariate models. RESULTS Our results showed that rare and common variants in the BAD and FCRL3 genes were associated (p<0.05) with an extreme cardiometabolic phenotype (3 or more cardiometabolic risk factors). Common variants in OGFOD3 and APOB as well as rare and common BAD variants were significantly (p<0.05) associated with dyslipidemia. Common BAD and SERPINA6 variants were associated (p<0.05) with obesity and insulin resistance, respectively. CONCLUSIONS In summary, we identified genetic susceptibility loci as contributing factors to the development of late treatment-related cardiometabolic complications in cALL survivors. These biomarkers could be used as early detection strategies to identify susceptible individuals and implement appropriate measures and follow-up to prevent the development of risk factors in this high-risk population.
Collapse
Affiliation(s)
- Jade England
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
| | - Simon Drouin
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
| | - Patrick Beaulieu
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
| | - Pascal St-Onge
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
| | - Maja Krajinovic
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
| | - Caroline Laverdière
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
- Departments of Pediatrics, Université de Montréal, Montreal, Quebec, H3T 1C5 Canada
| | - Emile Levy
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
- Departments of Nutrition, Université de Montréal, Montreal, Quebec, H3T 1C5 Canada
| | - Valérie Marcil
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
- Departments of Nutrition, Université de Montréal, Montreal, Quebec, H3T 1C5 Canada
| | - Daniel Sinnett
- Research Centre, Sainte-Justine University Health Center, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec, H3T 1C5 Canada
- Departments of Pediatrics, Université de Montréal, Montreal, Quebec, H3T 1C5 Canada
| |
Collapse
|
10
|
Schmiegelow K, Attarbaschi A, Barzilai S, Escherich G, Frandsen TL, Halsey C, Hough R, Jeha S, Kato M, Liang DC, Mikkelsen TS, Möricke A, Niinimäki R, Piette C, Putti MC, Raetz E, Silverman LB, Skinner R, Tuckuviene R, van der Sluis I, Zapotocka E. Consensus definitions of 14 severe acute toxic effects for childhood lymphoblastic leukaemia treatment: a Delphi consensus. Lancet Oncol 2017; 17:e231-e239. [PMID: 27299279 DOI: 10.1016/s1470-2045(16)30035-3] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 01/19/2023]
Abstract
Although there are high survival rates for children with acute lymphoblastic leukaemia, their outcome is often counterbalanced by the burden of toxic effects. This is because reported frequencies vary widely across studies, partly because of diverse definitions of toxic effects. Using the Delphi method, 15 international childhood acute lymphoblastic leukaemia study groups assessed acute lymphoblastic leukaemia protocols to address toxic effects that were to be considered by the Ponte di Legno working group. 14 acute toxic effects (hypersensitivity to asparaginase, hyperlipidaemia, osteonecrosis, asparaginase-associated pancreatitis, arterial hypertension, posterior reversible encephalopathy syndrome, seizures, depressed level of consciousness, methotrexate-related stroke-like syndrome, peripheral neuropathy, high-dose methotrexate-related nephrotoxicity, sinusoidal obstructive syndrome, thromboembolism, and Pneumocystis jirovecii pneumonia) that are serious but too rare to be addressed comprehensively within any single group, or are deemed to need consensus definitions for reliable incidence comparisons, were selected for assessment. Our results showed that none of the protocols addressed all 14 toxic effects, that no two protocols shared identical definitions of all toxic effects, and that no toxic effect definition was shared by all protocols. Using the Delphi method over three face-to-face plenary meetings, consensus definitions were obtained for all 14 toxic effects. In the overall assessment of outcome of acute lymphoblastic leukaemia treatment, these expert opinion-based definitions will allow reliable comparisons of frequencies and severities of acute toxic effects across treatment protocols, and facilitate international research on cause, guidelines for treatment adaptation, preventive strategies, and development of consensus algorithms for reporting on acute lymphoblastic leukaemia treatment.
Collapse
Affiliation(s)
- Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark; Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Pediatric Oncology, Langone Medical Center, New York University, New York, NY, USA.
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St Anna Children's Hospital, Vienna, Austria; Department of Pediatric and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Shlomit Barzilai
- Schneider Children's Medical Center of Israel, Department of Pediatric Hematology Oncology, Petah-Tikva, Israel
| | - Gabriele Escherich
- University Medical Center Eppendorf, Clinic of Pediatric Hematology and Oncology, Hamburg, Germany
| | - Thomas Leth Frandsen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Christina Halsey
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Rachael Hough
- University College London's NHS Foundation Trust, London, UK
| | - Sima Jeha
- St Jude Children's Research Hospital, Memphis, TN, USA
| | - Motohiro Kato
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Der-Cherng Liang
- Division of Pediatric Hematology-Oncology, Mackay Memorial Hospital, Taipei, Taiwan
| | | | - Anja Möricke
- Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Department of Pediatrics, Kiel, Germany
| | - Riitta Niinimäki
- Department of Pediatrics, Oulu University Hospital, Oulu, Finland
| | - Caroline Piette
- EORTC Children's Leukemia Group and University Department of Pediatric Oncology CHR Citadelle, Liège, Belgium
| | - Maria Caterina Putti
- Clinic of Pediatric Hematology Oncology, Department of Women's and Children's Health, Padova, Italy
| | - Elizabeth Raetz
- University of Utah, Department of Pediatrics and Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Lewis B Silverman
- Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
| | - Roderick Skinner
- Department of Paediatric and Adolescent Haematology/Oncology, and Children's Haemopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Ruta Tuckuviene
- Department of Pediatrics, Aalborg University Hospital, Aalborg, Denmark
| | - Inge van der Sluis
- Dutch Childhood Oncology Group, The Hague, Netherlands; Erasmus Medical Center, Sophia Children's Hospital, Department of Pediatric Hematology-Oncology, Rotterdam, Netherlands
| | - Ester Zapotocka
- University Hospital Motol, Department of Pediatric Hematology/Oncology, Prague, Czech Republic
| | | |
Collapse
|
11
|
Song QQ, Xie WY, Tang YJ, Zhang J, Liu J. Genetic variation in the glucocorticoid pathway involved in interindividual differences in the glucocorticoid treatment. Pharmacogenomics 2017; 18:293-316. [PMID: 28112586 DOI: 10.2217/pgs-2016-0151] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoids (GCs) are widely used for treating asthma, rheumatoid arthritis, nephrotic syndrome, acute lymphoblastic leukemia and other autoimmune diseases. However, in a subgroup of patients, failure to respond to GCs is known as GC resistance or GC insensitivity. This represents an important barrier to effective treatment and a clinical problem requiring an urgent solution. Genetic variation in the GC pathway is a significant factor in interindividual differences in GC treatment. This article reviews the pharmacogenetics of GCs in diverse diseases based on the GC pathway.
Collapse
Affiliation(s)
- Qian-Qian Song
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, Hunan, P.R. China
| | - Wan-Ying Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, Hunan, P.R. China
| | - Yong-Jun Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, Hunan, P.R. China
| | - Jun Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Jie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, Hunan, P.R. China
| |
Collapse
|
12
|
Turolo S, Edefonti A, Lepore M, Ghio L, Cuzzoni E, Decorti G, Pasini A, Materassi M, Malaventura C, Pugliese F, Montini G. SXR rs3842689: a prognostic factor for steroid sensitivity or resistance in pediatric idiopathic nephrotic syndrome. Pharmacogenomics 2016; 17:1227-1233. [PMID: 27377607 DOI: 10.2217/pgs-2016-0029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/14/2016] [Indexed: 12/17/2022] Open
Abstract
AIM of the study was to analyse the impact of SXR rs3842689 polymorphism on the response to corticosteroids in pediatric idiopathic nephrotic syndrome. PATIENTS & METHODS 66 children (56 steroid-sensitive, ten steroid-resistant) were studied for SXR gene polymorphism distribution. RESULTS Steroid sensitive patients accounted for 96% of cases with In/In polymorphism, but only for 53% of cases with Del/Del polymorphism At odds ratio analysis, Del/Del represented a clear risk factor of steroid resistance (OR: 20.57; p = 0.009), while In/In was a favourable prognostic factor of steroid sensitivity. CONCLUSION The analysis of SXR polymorphism is a promising tool to predict both the favourable response to corticosteroids and the risk of developing steroid resistance.
Collapse
Affiliation(s)
- Stefano Turolo
- Pediatric Nephrology & Dialysis, Milano. Fondazione IRCCS Cà Grande Ospedale Maggiroe Policlinico, Italy
| | - Alberto Edefonti
- Pediatric Nephrology & Dialysis, Milano. Fondazione IRCCS Cà Grande Ospedale Maggiroe Policlinico, Italy
| | - Marta Lepore
- Pediatric Nephrology & Dialysis, Milano. Fondazione IRCCS Cà Grande Ospedale Maggiroe Policlinico, Italy
| | - Luciana Ghio
- Pediatric Nephrology & Dialysis, Milano. Fondazione IRCCS Cà Grande Ospedale Maggiroe Policlinico, Italy
| | - Eva Cuzzoni
- Università degli Studi di Trieste Dipartimento di Scienze della Vita, Trieste, Friuli-Venezia Giulia, Italy
| | - Giuliana Decorti
- Università degli Studi di Trieste Dipartimento di Scienze della Vita, Trieste, Friuli-Venezia Giulia, Italy
| | - Andrea Pasini
- Università degli Studi di Bologna Azienda Ospedaliera Sant/'Orsola-Malpighi, Bologna, Emilia-Romagna, Italy
| | | | - Cristina Malaventura
- Università degli Studi di Ferrara, Medical Science Ferrara, Emilia-Romagna, Italy
| | - Fabrizio Pugliese
- Agenzia sanitaria e sociale regionale Regione Emilia-Romagna, Ravenna, Emilia-Romagna, Italy
| | - Giovanni Montini
- Pediatric Nephrology & Dialysis, Milano. Fondazione IRCCS Cà Grande Ospedale Maggiroe Policlinico, Italy
- Università degli Studi di Milano, Department of Clinical Sciences and Community Health, Milan, Italy
| |
Collapse
|
13
|
Warris LT, van den Akker ELT, Bierings MB, van den Bos C, Zwaan CM, Sassen SDT, Tissing WJE, Veening MA, Pieters R, van den Heuvel-Eibrink MM. Acute Activation of Metabolic Syndrome Components in Pediatric Acute Lymphoblastic Leukemia Patients Treated with Dexamethasone. PLoS One 2016; 11:e0158225. [PMID: 27362350 PMCID: PMC4928792 DOI: 10.1371/journal.pone.0158225] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/13/2016] [Indexed: 11/18/2022] Open
Abstract
Although dexamethasone is highly effective in the treatment of pediatric acute lymphoblastic leukemia (ALL), it can cause serious metabolic side effects. Because studies regarding the effects of dexamethasone are limited by their small scale, we prospectively studied the direct effects of treating pediatric ALL with dexamethasone administration with respect to activation of components of metabolic syndrome (MetS); in addition, we investigated whether these side effects were correlated with the level of dexamethasone. Fifty pediatric patients (3–16 years of age) with ALL were studied during a 5-day dexamethasone course during the maintenance phase of the Dutch Childhood Oncology Group ALL-10 and ALL-11 protocols. Fasting insulin, glucose, total cholesterol, HDL, LDL, and triglycerides levels were measured at baseline (before the start of dexamethasone; T1) and on the fifth day of treatment (T2). Dexamethasone trough levels were measured at T2. We found that dexamethasone treatment significantly increased the following fasting serum levels (P<0.05): HDL, LDL, total cholesterol, triglycerides, glucose, and insulin. In addition, dexamethasone increased insulin resistance (HOMA-IR>3.4) from 8% to 85% (P<0.01). Dexamethasone treatment also significantly increased the diastolic and systolic blood pressure. Lastly, dexamethasone trough levels (N = 24) were directly correlated with high glucose levels at T2, but not with other parameters. These results indicate that dexamethasone treatment acutely induces three components of the MetS. Together with the weight gain typically associated with dexamethasone treatment, these factors may contribute to the higher prevalence of MetS and cardiovascular risk among survivors of childhood leukemia who received dexamethasone treatment.
Collapse
Affiliation(s)
- Lidewij T. Warris
- Department of Pediatric Oncology, Erasmus MC- Sophia Children’s Hospital, Rotterdam, The Netherlands
- Department of Pediatric Endocrinology, Erasmus MC- Sophia Children’s Hospital, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- * E-mail:
| | - Erica L. T. van den Akker
- Department of Pediatric Endocrinology, Erasmus MC- Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Marc B. Bierings
- Department of Pediatric Hematology and Oncology, University Medical Center Utrecht – Wilhelmina Children’s Hospital, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Cor van den Bos
- Department of Pediatric Oncology, Academic Medical Center – Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Christian M. Zwaan
- Department of Pediatric Oncology, Erasmus MC- Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Sebastiaan D. T. Sassen
- Department of Pediatric Oncology, Erasmus MC- Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Wim J. E. Tissing
- Department of Pediatric Hematology and Oncology, University of Groningen Medical Center, Groningen, The Netherlands
| | - Margreet A. Veening
- Department of Pediatric Hematology and Oncology, VU Medical Center, Amsterdam, The Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marry M. van den Heuvel-Eibrink
- Department of Pediatric Oncology, Erasmus MC- Sophia Children’s Hospital, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
14
|
A common polymorphism rs1800247 in osteocalcin gene is associated with hypertension and diastolic blood pressure levels: the Shanghai Changfeng study. J Hum Hypertens 2016; 30:679-684. [DOI: 10.1038/jhh.2016.16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 02/14/2016] [Accepted: 02/25/2016] [Indexed: 12/15/2022]
|
15
|
Mei L, Ontiveros EP, Griffiths EA, Thompson JE, Wang ES, Wetzler M. Pharmacogenetics predictive of response and toxicity in acute lymphoblastic leukemia therapy. Blood Rev 2015; 29:243-9. [PMID: 25614322 DOI: 10.1016/j.blre.2015.01.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 12/26/2014] [Accepted: 01/06/2015] [Indexed: 12/19/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is a relatively rare disease in adults accounting for no more than 20% of all cases of acute leukemia. By contrast with the pediatric population, in whom significant improvements in long term survival and even cure have been achieved over the last 30years, adult ALL remains a significant challenge. Overall survival in this group remains a relatively poor 20-40%. Modern research has focused on improved pharmacokinetics, novel pharmacogenetics and personalized principles to optimize the efficacy of the treatment while reducing toxicity. Here we review the pharmacogenetics of medications used in the management of patients with ALL, including l-asparaginase, glucocorticoids, 6-mercaptopurine, methotrexate, vincristine and tyrosine kinase inhibitors. Incorporating recent pharmacogenetic data, mainly from pediatric ALL, will provide novel perspective of predicting response and toxicity in both pediatric and adult ALL therapies.
Collapse
Affiliation(s)
- Lin Mei
- Leukemia Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Evelena P Ontiveros
- Leukemia Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Elizabeth A Griffiths
- Leukemia Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - James E Thompson
- Leukemia Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Eunice S Wang
- Leukemia Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Meir Wetzler
- Leukemia Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| |
Collapse
|
16
|
Cancer and bone: A complex complex. Arch Biochem Biophys 2014; 561:159-66. [DOI: 10.1016/j.abb.2014.07.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 07/03/2014] [Accepted: 07/08/2014] [Indexed: 12/13/2022]
|
17
|
Lopez-Lopez E, Gutierrez-Camino A, Bilbao-Aldaiturriaga N, Pombar-Gomez M, Martin-Guerrero I, Garcia-Orad A. Pharmacogenetics of childhood acute lymphoblastic leukemia. Pharmacogenomics 2014; 15:1383-98. [DOI: 10.2217/pgs.14.106] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the major pediatric cancer in developed countries. Although treatment outcome has improved owing to advances in chemotherapy, there is still a group of patients for which therapy fails while some patients experience severe toxicity. In the last few years, several pharmacogenetic studies have been performed to search for markers of outcome and toxicity in pediatric ALL. However, to date, TPMT is the only pharmacogenetic marker in ALL with clinical guidelines for drug dosing. In this article, we will provide an overview of the most important findings carried out in pharmacogenetics for pediatric ALL, such as the interest drawn by methotrexate transporters in the context of methotrexate treatment. Even if most of the studies are centered on coding genes, we will also point to new approaches focusing on noncoding regions and epigenetic variation that could be interesting for consideration in the near future.
Collapse
Affiliation(s)
- Elixabet Lopez-Lopez
- Department of Genetics, Physical Anthropology & Animal Physiology, Faculty of Medicine & Odontology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Angela Gutierrez-Camino
- Department of Genetics, Physical Anthropology & Animal Physiology, Faculty of Medicine & Odontology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Nerea Bilbao-Aldaiturriaga
- Department of Genetics, Physical Anthropology & Animal Physiology, Faculty of Medicine & Odontology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Maria Pombar-Gomez
- Department of Genetics, Physical Anthropology & Animal Physiology, Faculty of Medicine & Odontology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Idoia Martin-Guerrero
- Department of Genetics, Physical Anthropology & Animal Physiology, Faculty of Medicine & Odontology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Africa Garcia-Orad
- Department of Genetics, Physical Anthropology & Animal Physiology, Faculty of Medicine & Odontology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
- BioCruces Health Research Institute, Leioa, Spain
| |
Collapse
|
18
|
Gragnoli C. Hypothesis of the neuroendocrine cortisol pathway gene role in the comorbidity of depression, type 2 diabetes, and metabolic syndrome. APPLICATION OF CLINICAL GENETICS 2014; 7:43-53. [PMID: 24817815 PMCID: PMC4012344 DOI: 10.2147/tacg.s39993] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Depression, type 2 diabetes (T2D), and metabolic syndrome (MetS) are often comorbid. Depression per se increases the risk for T2D by 60%. This risk is not accounted for by the use of antidepressant therapy. Stress causes hyperactivation of the hypothalamic–pituitary–adrenal (HPA) axis, by triggering the hypothalamic corticotropin-releasing hormone (CRH) secretion, which stimulates the anterior pituitary to release the adrenocorticotropin hormone (ACTH), which causes the adrenal secretion of cortisol. Depression is associated with an increased level of cortisol, and CRH and ACTH at inappropriately “normal” levels, that is too high compared to their expected lower levels due to cortisol negative feedback. T2D and MetS are also associated with hypercortisolism. High levels of cortisol can impair mood as well as cause hyperglycemia and insulin resistance and other traits typical of T2D and MetS. We hypothesize that HPA axis hyperactivation may be due to variants in the genes of the CRH receptors (CRHR1, CRHR2), corticotropin receptors (or melanocortin receptors, MC1R-MC5R), glucocorticoid receptor (NR3C1), mineralocorticoid receptor (NR3C2), and of the FK506 binding protein 51 (FKBP5), and that these variants may be partially responsible for the clinical association of depression, T2D and MetS. In this review, we will focus on the correlation of stress, HPA axis hyperactivation, and the possible genetic role of the CRHR1, CRHR2, MCR1–5, NR3C1, and NR3C2 receptors and FKBP5 in the susceptibility to the comorbidity of depression, T2D, and MetS. New studies are needed to confirm the hypothesized role of these genes in the clinical association of depression, T2D, and MetS.
Collapse
Affiliation(s)
- Claudia Gragnoli
- Center for Biotechnology and Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA ; Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy
| |
Collapse
|
19
|
Dulucq S, Laverdière C, Sinnett D, Krajinovic M. Pharmacogenetic considerations for acute lymphoblastic leukemia therapies. Expert Opin Drug Metab Toxicol 2014; 10:699-719. [PMID: 24673379 DOI: 10.1517/17425255.2014.893294] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Advances in our understanding of the pathobiology of childhood acute lymphoblastic leukemia (ALL) have led to risk-targeted treatment regimens and remarkable improvement in survival rates. Still, up to 20% of patients experience treatment failure due to drug resistance. Treatment-related toxicities are often life-threatening and are the primary cause of treatment interruption, while ALL survivors may develop complications due to exposure to chemotherapy and/or irradiation during a vulnerable period of development. Different factors may contribute to variable treatment outcomes including patient genetics that has been shown to play important role. AREAS COVERED This review summarizes candidate gene and genome-wide association studies that identified common polymorphisms underlying variability in treatment responses including a few studies addressing late effects of the treatment. Genetic variants influencing antileukemic drug effects or leukemic cell biology have been identified, including for example variants in folate-dependent enzymes, influx and efflux transporters, metabolizing enzymes, drug receptor or apoptotic proteins. EXPERT OPINION Many pharmacogenetic studies have been conducted in ALL and a variety of potential markers have been identified. Yet more comprehensive insight into genome variations influencing drug responses is needed. Whole exome/genome sequencing, careful study design, mechanistic explanation of association found and collaborative studies will ultimately lead to personalized treatment and improved therapeutic and health outcomes.
Collapse
Affiliation(s)
- Stéphanie Dulucq
- University Health Center Bordeaux, Heamatology Laboratory , Bordeaux , France
| | | | | | | |
Collapse
|
20
|
Slominski AT, Zmijewski MA, Zbytek B, Tobin DJ, Theoharides TC, Rivier J. Key role of CRF in the skin stress response system. Endocr Rev 2013; 34:827-84. [PMID: 23939821 PMCID: PMC3857130 DOI: 10.1210/er.2012-1092] [Citation(s) in RCA: 303] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 08/02/2013] [Indexed: 02/08/2023]
Abstract
The discovery of corticotropin-releasing factor (CRF) or CRH defining the upper regulatory arm of the hypothalamic-pituitary-adrenal (HPA) axis, along with the identification of the corresponding receptors (CRFRs 1 and 2), represents a milestone in our understanding of central mechanisms regulating body and local homeostasis. We focused on the CRF-led signaling systems in the skin and offer a model for regulation of peripheral homeostasis based on the interaction of CRF and the structurally related urocortins with corresponding receptors and the resulting direct or indirect phenotypic effects that include regulation of epidermal barrier function, skin immune, pigmentary, adnexal, and dermal functions necessary to maintain local and systemic homeostasis. The regulatory modes of action include the classical CRF-led cutaneous equivalent of the central HPA axis, the expression and function of CRF and related peptides, and the stimulation of pro-opiomelanocortin peptides or cytokines. The key regulatory role is assigned to the CRFR-1α receptor, with other isoforms having modulatory effects. CRF can be released from sensory nerves and immune cells in response to emotional and environmental stressors. The expression sequence of peptides includes urocortin/CRF→pro-opiomelanocortin→ACTH, MSH, and β-endorphin. Expression of these peptides and of CRFR-1α is environmentally regulated, and their dysfunction can lead to skin and systemic diseases. Environmentally stressed skin can activate both the central and local HPA axis through either sensory nerves or humoral factors to turn on homeostatic responses counteracting cutaneous and systemic environmental damage. CRF and CRFR-1 may constitute novel targets through the use of specific agonists or antagonists, especially for therapy of skin diseases that worsen with stress, such as atopic dermatitis and psoriasis.
Collapse
Affiliation(s)
- Andrzej T Slominski
- MD, PhD, Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center; 930 Madison Avenue, Suite 500, Memphis, Tennessee 38163.
| | | | | | | | | | | |
Collapse
|
21
|
Fardet L. Effets indésirables métaboliques et cardiovasculaires des corticothérapies systémiques. Rev Med Interne 2013; 34:303-9. [DOI: 10.1016/j.revmed.2012.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 12/10/2012] [Indexed: 02/07/2023]
|
22
|
Liu X, Wang Y, Qu H, Hou M, Cao W, Ma Z, Wang H. Associations of polymorphisms of rs693 and rs1042031 in apolipoprotein B gene with risk of breast cancer in Chinese. Jpn J Clin Oncol 2013; 43:362-8. [PMID: 23444115 DOI: 10.1093/jjco/hyt018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Lipid synthesis is an integrated result of genetic, epigenetic and environmental factors, and also can promote growth and survival of cancer cells. Apolipoprotein B plays a central role in lipid metabolism as the major protein component of very-low-density lipoprotein and low-density lipoprotein. METHODS We investigated the associations of polymorphisms of rs693 (-7673C>T) and rs1042031 (-12669 G>A) in the APOB gene with risk of breast cancer in 675 blood-unrelated Chinese patients with breast cancer and 712 healthy controls. RESULTS Polymorphisms of -12669 G>A and -7673C>T in the APOB gene were significantly associated with an increased risk of breast cancer (P = 0.000), especially for postmenopausal women (P = 0.000, 0.023). The positive associations still remained after further analysis of the two polymorphisms' distribution according to body mass index. However, no statistical associations were found between -12669 G>A and -7673C>T polymorphisms and other clinical characteristics, including tumor size, lymph node metastasis, histological grade, estrogen and progesterone receptor status and Her-2 status. CONCLUSIONS rs693 and rs1042031 polymorphisms in the APOB gene increased the risk of breast cancer in Chinese, and this role of the two polymorphisms in connection with breast cancer was not dependent on body mass index.
Collapse
Affiliation(s)
- Xiaoyi Liu
- Department of Galactophore, Qingdao University, Qingdao, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Dubovsky AN, Arvikar S, Stern TA, Axelrod L. The Neuropsychiatric Complications of Glucocorticoid Use: Steroid Psychosis Revisited. PSYCHOSOMATICS 2012; 53:103-15. [DOI: 10.1016/j.psym.2011.12.007] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 12/08/2011] [Accepted: 12/12/2011] [Indexed: 12/21/2022]
|
24
|
Yang JJ, Mehta PA, Relling MV, Davies SM. Pharmacogenetic and Pharmacogenomic Considerations in the Biology and Treatment of Childhood Leukemia. CHILDHOOD LEUKEMIA 2011. [DOI: 10.1007/978-3-642-13781-5_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
25
|
De Iudicibus S, Stocco G, Martelossi S, Londero M, Ebner E, Pontillo A, Lionetti P, Barabino A, Bartoli F, Ventura A, Decorti G. Genetic predictors of glucocorticoid response in pediatric patients with inflammatory bowel diseases. J Clin Gastroenterol 2011; 45:e1-e7. [PMID: 20697295 DOI: 10.1097/mcg.0b013e3181e8ae93] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Glucocorticoids (GCs) are used in moderate-to-severe inflammatory bowel diseases (IBD) but their effect is often unpredictable. AIM To determine the influence of 4 polymorphisms in the GC receptor [nuclear receptor subfamily 3, group C, member 1 (NR3C1)], interleukin-1β (IL-1β), and NACHT leucine-rich-repeat protein 1 (NALP1) genes, on the clinical response to steroids in pediatric patients with IBD. METHODS One hundred fifty-four young IBD patients treated with GCs for at least 30 days and with a minimum follow-up of 1 year were genotyped. The polymorphisms considered are the BclI in the NR3C1 gene, C-511T in IL-1β gene, and Leu155His and rs2670660/C in NALP1 gene. Patients were grouped as responder, dependant, and resistant to GCs. The relation between GC response and the genetic polymorphisms considered was examined using univariate, multivariate, and Classification and Regression Tree (CART) analysis. RESULTS Univariate analysis showed that BclI polymorphism was more frequent in responders compared with dependant patients (P=0.03) and with the combined dependant and resistant groups (P=0.02). Moreover, the NALP1 Leu155His polymorphism was less frequent in the GC responsive group compared with resistant (P=0.0059) and nonresponder (P=0.02) groups. Multivariate analysis comparing responders and nonresponders confirmed an association between BclI mutated genotype and steroid response (P=0.030), and between NALP1 Leu155His mutant variant and nonresponders (P=0.033). An association between steroid response and male sex was also observed (P=0.034). In addition, Leu155His mutated genotype was associated with steroid resistance (P=0.034). Two CART analyses supported these findings by showing that BclI and Leu155His polymorphisms had the greatest effect on steroid response (permutation P value=0.046). The second CART analysis also identified age of disease onset and male sex as important variables affecting response. CONCLUSIONS These results confirm that genetic and demographic factors may affect the response to GCs in young patients with IBD and strengthen the importance of studying high-order interactions for predicting response.
Collapse
|
26
|
Staatz CE, Goodman LK, Tett SE. Effect of CYP3A and ABCB1 single nucleotide polymorphisms on the pharmacokinetics and pharmacodynamics of calcineurin inhibitors: Part II. Clin Pharmacokinet 2010; 49:207-21. [PMID: 20214406 DOI: 10.2165/11317550-000000000-00000] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The calcineurin inhibitors ciclosporin (cyclosporine) and tacrolimus are immunosuppressant drugs used for the prevention of organ rejection following transplantation. Both agents are metabolic substrates for cytochrome P450 (CYP) 3A enzymes - in particular, CYP3A4 and CYP3A5 - and are transported out of cells via P-glycoprotein (ABCB1). Several single nucleotide polymorphisms (SNPs) have been identified in the genes encoding for CYP3A4, CYP3A5 and P-glycoprotein, including CYP3A4 -392A>G (rs2740574), CYP3A5 6986A>G (rs776746), ABCB1 3435C>T (rs1045642), ABCB1 1236C>T (rs1128503) and ABCB1 2677G>T/A (rs2032582). The aim of this review is to provide the clinician with an extensive overview of the recent literature on the known effects of these SNPs on the pharmacodynamics of ciclosporin and tacrolimus in solid-organ transplant recipients. Literature searches were performed and all relevant primary research articles were critiqued and summarized. There is no evidence that the CYP3A4 -392A>G SNP has an effect on the pharmacodynamics of either ciclosporin or tacrolimus; however, studies have been limited. For patients prescribed ciclosporin, the CYP3A5 6986A>G SNP may influence long-term survival, possibly because of a different metabolite pattern over time. This SNP has no clear association with acute rejection during ciclosporin therapy. Despite a strong association between the CYP3A5 6986A>G SNP and tacrolimus pharmacokinetics, there is no consistent evidence of organ rejection as a result of genotype-related under-immunosuppression. This is likely to be explained by the practice of performing tacrolimus dose adjustments in the early phase after transplantation. The effect of the CYP3A5 6986A>G SNP on ciclosporin- and tacrolimus-related nephrotoxicity and development of hypertension is unclear. Similarly, the ABCB1 SNPs exert no clear influence on either ciclosporin or tacrolimus pharmacodynamics, with studies showing conflicting results in regard to the main parameters of acute rejection and nephrotoxicity. In kidney transplant patients, consideration of the donor kidney genotype rather than the recipient genotype may be more important when assessing development of nephrotoxicity. Studies with low patient numbers may account for many inconsistent results to date. The majority of studies have only evaluated the effects of individual SNPs; however, multiple polymorphisms may interact to produce a combined effect. Further haplotype analyses are likely to be useful, particularly ones that consider both donor and recipient genotype. The effects of polymorphisms associated with the pregnane X receptor, organic anion transporting polypeptides, calcineurin inhibitor target sites and immune response pathways need to be further investigated. A large standardized clinical trial is now required to evaluate the relationship between the pharmacokinetics and pharmacodynamics of CYP3A5-mediated tacrolimus metabolism, particularly in regard to the outcomes of acute rejection and nephrotoxicity. It is not yet clear whether pharmacogenetic profiling of calcineurin inhibitors will be a useful clinical tool for personalizing immunosuppressant therapy.
Collapse
|
27
|
Meeker ND, Yang JJ, Schiffman JD. Pharmacogenomics of pediatric acute lymphoblastic leukemia. Expert Opin Pharmacother 2010; 11:1621-32. [DOI: 10.1517/14656566.2010.484019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
28
|
Roche SL, O'Sullivan JJ, Kantor PF. Hypertension after pediatric cardiac transplantation: detection, etiology, implications and management. Pediatr Transplant 2010; 14:159-68. [PMID: 19624603 DOI: 10.1111/j.1399-3046.2009.01205.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
While it may rescue children with end-stage heart failure from impending catastrophe, cardiac transplantation leaves 50-70% of pediatric recipients with new-onset hypertension. Given the unique vulnerability of the heart and kidneys in these children, we can expect long-term uncontrolled hypertension to shorten both graft and patient survival. In this review we discuss the multi-factorial etiology of post-transplant hypertension, highlighting current uncertainties and emphasizing mechanisms specific to cardiac recipients. We consider the optimal means of monitoring BP and in particular, the advantages of 24 h-ABP over intermittent clinic measurements. We also review BP treatment after cardiac transplantation, drawing attention to specific cautions appropriate when prescribing antihypertensive agents in these circumstances.
Collapse
Affiliation(s)
- S Lucy Roche
- Department of Pediatric Cardiology, The Hospital for Sick Children, Toronto, ON, Canada
| | | | | |
Collapse
|
29
|
Stocco G, Crews KR, Evans WE. Genetic polymorphism of inosine-triphosphate-pyrophosphatase influences mercaptopurine metabolism and toxicity during treatment of acute lymphoblastic leukemia individualized for thiopurine-S-methyl-transferase status. Expert Opin Drug Saf 2010; 9:23-37. [PMID: 20021291 DOI: 10.1517/14740330903426151] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IMPORTANCE OF THE FIELD Although genetic polymorphisms in the gene encoding human thiopurine methyltransferase (TPMT) are known to have a marked effect on mercaptopurine metabolism and toxicity, there are many patients with wild-type TPMT who develop toxicity. Furthermore, when mercaptopurine dosages are adjusted in patients who are heterozygous at the TPMT locus, there are still some patients who develop toxicity for reasons that are not fully understood. Therefore, we recently studied the effects of a common polymorphism in another gene encoding an enzyme involved in mercaptopurine metabolism (SNP rs1127354 in inosine-triphospate-pyrophosphatase, ITPA), showing that genetic polymorphism of ITPA is a significant determinant of mercaptopurine metabolism and of febrile neutropenia following combination chemotherapy of acute lymphoblastic leukemia (ALL) in which mercaptopurine doses are individualized based on TPMT genotype. AREA COVERED IN THIS REVIEW In this review, we summarize the knowledge available about the effect and clinical relevance of TPMT and ITPA on mercaptopurine pharmacogenomics, with a particular focus on the use of this medication in pediatric patients with ALL. WHAT THE READER WILL GAIN Reader will gain insights into: i) the effects of pharmacogenomic traits on mercaptopurine toxicity and efficacy for the treatment of ALL and ii) individualization strategies that can be used to mitigate toxicity without compromising efficacy in pediatric patients with ALL. TAKE HOME MESSAGE Mercaptopurine dose can be adjusted on the basis of TPMT genotype to mitigate toxicity in pediatric patients with ALL. As treatment is individualized in this way for the most relevant genetic determinant of drug response (i.e., for mercaptopurine, TPMT), the importance of other genetic polymorphisms emerges (e.g., ITPA).
Collapse
Affiliation(s)
- Gabriele Stocco
- St. Jude Children's Research Hospital, Department of Pharmaceutical Sciences, 262 Danny Thomas Place MS 272, Memphis, TN 38105, USA
| | | | | |
Collapse
|
30
|
Abstract
Hypertension is the first single modifiable cause of disease burden worldwide. Genes encoding proteins that are involved in the metabolism (CYP3A5) and transport (ABCB1) of drugs and hormones might contribute to blood pressure control in humans. Indeed, recent data have suggested that CYP3A5 and ABCB1 gene polymorphisms are associated with blood pressure in the rat as well as in humans. Interestingly, the effects of these genes on blood pressure appear to be modified by dietary salt intake. This review summarizes what is known regarding the relationships of the ABCB1 and CYP3A5 genes with blood pressure, and discusses the potential underlying mechanisms of the association. If the role of these genes in blood pressure control is confirmed in other populations and other ethnic groups, these findings would point toward a new pathway for blood pressure control in humans.
Collapse
Affiliation(s)
- Murielle Bochud
- Institute of Social and Preventive Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) et Université de Lausanne, Rue du Bugnon 17, CH-1005 Lausanne, Switzerland.
| | | | | | | |
Collapse
|
31
|
Abstract
Progress in the treatment of acute lymphoblastic leukemia (ALL) in children has been remarkable, from a disease being lethal four decades ago to current cure rates exceeding 80%. This exemplary progress is largely due to the optimization of existing treatment modalities rather than the discovery of new antileukemic agents. However, despite these high cure rates, the annual number of children whose leukemia relapses after their initial therapy remains greater than that of new cases of most types of childhood cancers. The aim of pharmacogenetics is to develop strategies to personalize treatment and tailor therapy to individual patients, with the goal of optimizing efficacy and safety through better understanding of human genome variability and its influence on drug response. In this review, we summarize recent pharmacogenomic studies related to the treatment of pediatric ALL. These studies illustrate the promise of pharmacogenomics to further advance the treatment of human cancers, with childhood leukemia serving as a paradigm.
Collapse
Affiliation(s)
- Meyling H Cheok
- Jean-Pierre Aubert Research Center, INSERM U837, Genomics Core IRCL-IMPRT, Lille, France.
| | | | | | | |
Collapse
|