1
|
Perrone G, Rigacci L, Roviello G, Landini I, Fabbri A, Iovino L, Puccini B, Cencini E, Orciuolo E, Bocchia M, Bosi A, Mini E, Nobili S. Validation of single nucleotide polymorphisms potentially related to R-CHOP resistance in diffuse large B-cell lymphoma patients. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:21. [PMID: 38835350 PMCID: PMC11149109 DOI: 10.20517/cdr.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024]
Abstract
Aim: Diffuse large B-cell lymphoma (DLBCL) is the most common B-cell non-Hodgkin lymphoma (NHL). Despite the availability of clinical and molecular algorithms applied for the prediction of prognosis, in up to 30%-40% of patients, intrinsic or acquired drug resistance occurs. Constitutional genetics may help to predict R-CHOP resistance. This study aimed to validate previously identified single nucleotide polymorphisms (SNPs) in the literature as potential predictors of R-CHOP resistance in DLBCL patients, SNPs. Methods: Twenty SNPs, involved in R-CHOP pharmacokinetics/pharmacodynamics or other pathobiological processes, were investigated in 185 stage I-IV DLBCL patients included in a multi-institution pharmacogenetic study to validate their previously identified correlations with resistance to R-CHOP. Results: Correlations between rs2010963 (VEGFA gene) and sex (P = 0.046), and rs1625895 (TP53 gene) and stage (P = 0.003) were shown. After multivariate analyses, a concordant effect (i.e., increased risk of disease progression and death) was observed for rs1883112 (NCF4 gene) and rs1800871 (IL10 gene). When patients were grouped according to the revised International Prognostic Index (R-IPI), both these SNPs further discriminated progression-free survival (PFS) and overall survival (OS) of the R-IPI-1-2 subgroup. Overall, patients harboring the rare allele showed shorter PFS and OS compared with wild-type patients. Conclusions: Two out of the 20 study SNPs were validated. Thus, these results support the role of previously identified rs1883112 and rs1800871 in predicting DLBCL resistance to R-CHOP and highlight their ability to further discriminate the prognosis of R-IPI-1-2 patients. These data point to the need to also focus on host genetics for a more comprehensive assessment of DLBCL patient outcomes in future prospective trials.
Collapse
Affiliation(s)
- Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence 50139, Italy
| | - Luigi Rigacci
- Research Unit of Hematology, Department of Medicine and Surgery, Campus Biomedico University, Rome 00128, Italy
| | | | - Ida Landini
- Department of Health Sciences, University of Florence, Florence 50139, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Lorenzo Iovino
- Unit of Hematology, Santa Chiara University Hospital, University of Pisa, Pisa 56126, Italy
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109-4433, USA
| | - Benedetta Puccini
- Unit of Hematology, Careggi University-Hospital, Florence 50134, Italy
| | - Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Enrico Orciuolo
- Unit of Hematology, Santa Chiara University Hospital, University of Pisa, Pisa 56126, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Alberto Bosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence 50139, Italy
- Authors contributed equally
| | - Stefania Nobili
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy
- Authors contributed equally
| |
Collapse
|
2
|
Lingaratnam S, Shah M, Nicolazzo J, Michael M, Seymour JF, James P, Lazarakis S, Loi S, Kirkpatrick CMJ. A systematic review and meta-analysis of the impacts of germline pharmacogenomics on severe toxicity and symptom burden in adult patients with cancer. Clin Transl Sci 2024; 17:e13781. [PMID: 38700261 PMCID: PMC11067509 DOI: 10.1111/cts.13781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/12/2024] [Accepted: 03/14/2024] [Indexed: 05/05/2024] Open
Abstract
The clinical application of Pharmacogenomics (PGx) has improved patient safety. However, comprehensive PGx testing has not been widely adopted in clinical practice, and significant opportunities exist to further optimize PGx in cancer care. This systematic review and meta-analysis aim to evaluate the safety outcomes of reported PGx-guided strategies (Analysis 1) and identify well-studied emerging pharmacogenomic variants that predict severe toxicity and symptom burden (Analysis 2) in patients with cancer. We searched MEDLINE, EMBASE, CENTRAL, clinicaltrials.gov, and International Clinical Trials Registry Platform from inception to January 2023 for clinical trials or comparative studies evaluating PGx strategies or unconfirmed pharmacogenomic variants. The primary outcomes were severe adverse events (SAE; ≥ grade 3) or symptom burden with pain and vomiting as defined by trial protocols and assessed by trial investigators. We calculated pooled overall relative risk (RR) and 95% confidence interval (95%CI) using random effects models. PROSPERO, registration number CRD42023421277. Of 6811 records screened, six studies were included for Analysis 1, 55 studies for Analysis 2. Meta-analysis 1 (five trials, 1892 participants) showed a lower absolute incidence of SAEs with PGx-guided strategies compared to usual therapy, 16.1% versus 34.0% (RR = 0.72, 95%CI 0.57-0.91, p = 0.006, I2 = 34%). Meta-analyses 2 identified nine medicine(class)-variant pairs of interest across the TYMS, ABCB1, UGT1A1, HLA-DRB1, and OPRM1 genes. Application of PGx significantly reduced rates of SAEs in patients with cancer. Emergent medicine-variant pairs herald further research into the expansion and optimization of PGx to improve systemic anti-cancer and supportive care medicine safety and efficacy.
Collapse
Affiliation(s)
- Senthil Lingaratnam
- Pharmacy DepartmentPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Monash Institute of Pharmaceutical Sciences, Monash UniversityMelbourneVictoriaAustralia
| | - Mahek Shah
- Faculty of Pharmacy and Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Joseph Nicolazzo
- Monash Institute of Pharmaceutical Sciences, Monash UniversityMelbourneVictoriaAustralia
| | - Michael Michael
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Department of Medical OncologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - John F. Seymour
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Department of Clinical HaematologyPeter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVictoriaAustralia
| | - Paul James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVictoriaAustralia
| | - Smaro Lazarakis
- Health Sciences LibraryRoyal Melbourne HospitalMelbourneVictoriaAustralia
| | - Sherene Loi
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Division of Cancer ResearchPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Carl M. J. Kirkpatrick
- Monash Institute of Pharmaceutical Sciences, Monash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
3
|
Franczyk B, Rysz J, Miłoński J, Konecki T, Rysz-Górzyńska M, Gluba-Brzózka A. Will the Use of Pharmacogenetics Improve Treatment Efficiency in COVID-19? Pharmaceuticals (Basel) 2022; 15:739. [PMID: 35745658 PMCID: PMC9230944 DOI: 10.3390/ph15060739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
The COVID-19 pandemic is associated with a global health crisis and the greatest challenge for scientists and doctors. The virus causes severe acute respiratory syndrome with an outcome that is fatal in more vulnerable populations. Due to the need to find an efficient treatment in a short time, there were several drugs that were repurposed or repositioned for COVID-19. There are many types of available COVID-19 therapies, including antiviral agents (remdesivir, lopinavir/ritonavir, oseltamivir), antibiotics (azithromycin), antiparasitics (chloroquine, hydroxychloroquine, ivermectin), and corticosteroids (dexamethasone). A combination of antivirals with various mechanisms of action may be more efficient. However, the use of some of these medicines can be related to the occurrence of adverse effects. Some promising drug candidates have been found to be ineffective in clinical trials. The knowledge of pharmacogenetic issues, which translate into variability in drug conversion from prodrug into drug, metabolism as well as transport, could help to predict treatment efficiency and the occurrence of adverse effects in patients. However, many drugs used for the treatment of COVID-19 have not undergone pharmacogenetic studies, perhaps as a result of the lack of time.
Collapse
Affiliation(s)
- Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| | - Jarosław Miłoński
- Department of Otolaryngology, Laryngological Oncology, Audiology and Phoniatrics, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Tomasz Konecki
- Department of Urology, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Magdalena Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| |
Collapse
|
4
|
Lavacchi D, Landini I, Perrone G, Roviello G, Mini E, Nobili S. Pharmacogenetics in diffuse large B-cell lymphoma treated with R-CHOP: Still an unmet challenge. Pharmacol Ther 2021; 229:107924. [PMID: 34175369 DOI: 10.1016/j.pharmthera.2021.107924] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023]
Abstract
DLBCL is the most common lymphoma representing approximately one third of all non-Hodgkin lymphomas and about 40% of patients do not benefit of the standard first-line immune-chemotherapeutic treatment (i.e., R-CHOP - rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) that is administered as upfront therapy to substantially all patients independently from the stage of disease and other prognostic parameters. The administration of other pharmacological treatments is in fact limited to selected patients, unfitting for R-CHOP. Although clinical prognostic scores, i.e. International Prognostic Index (IPI), and molecular classifiers based on the cell of origin are available, at present no biomarkers predictive of R-CHOP response has been identified and validated. Constitutional polymorphisms of genes involved in the mechanism of action of drugs included in R-CHOP have been suggested by many authors to play a role in the efficacy and in some case in the toxicity of this treatment. Thus, it is conceivable that in the future, after proper validation, some polymorphisms can be used as pharmacogenetic biomarkers of therapeutic outcome in this disease setting. This review discusses the status of the art on molecular biomarkers predictive of DLBCL prognosis and deals with the relevant issue of the variability in response to DLBCL drug treatment. Overall, this review focuses on single nucleotide polymorphisms (SNPs) that, based on a candidate gene approach or on a GWAS analysis, have been suggested to play a role in response to R-CHOP. In particular, SNPs discovered by a candidate gene approach are related to gene involved in drug transport (i.e. ATP-binding cassette transporters), drug metabolism, drug detoxification enzymes, oxidative stress, apoptosis, DNA repair, immunity and angiogenesis. Data from a GWAS analysis performed in DLBCL patients treated with R-CHOP, identified two SNPs associated with clinical outcomes related to genes involved in pivotal cellular processes and in transcriptional regulation and cell cycle progression, respectively. Ongoing prospective pharmacogenetic clinical trials, including a GWAS study we performed, have also been discussed.
Collapse
Affiliation(s)
- Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, Florence, Italy; DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence, Italy; DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, University of Florence, Florence, Italy; DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy; DENOTHE Excellence Center, University of Florence, Florence, Italy; Cancer Pharmacology Working Group of the Italian Society of Pharmacology, Milan, Italy.
| | - Stefania Nobili
- Cancer Pharmacology Working Group of the Italian Society of Pharmacology, Milan, Italy; Department of Neurosciences, Imaging and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
5
|
Bell RMB, Villalobos E, Nixon M, Miguelez-Crespo A, Murphy L, Fawkes A, Coutts A, Sharp MGF, Koerner MV, Allan E, Meijer OC, Houtman R, Odermatt A, Beck KR, Denham SG, Lee P, Homer NZM, Walker BR, Morgan RA. Carbonyl reductase 1 amplifies glucocorticoid action in adipose tissue and impairs glucose tolerance in lean mice. Mol Metab 2021; 48:101225. [PMID: 33785425 PMCID: PMC8095185 DOI: 10.1016/j.molmet.2021.101225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/09/2021] [Accepted: 03/24/2021] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Carbonyl reductase 1 (Cbr1), a recently discovered contributor to tissue glucocorticoid metabolism converting corticosterone to 20β-dihydrocorticosterone (20β-DHB), is upregulated in adipose tissue of obese humans and mice and may contribute to cardiometabolic complications of obesity. This study tested the hypothesis that Cbr1-mediated glucocorticoid metabolism influences glucocorticoid and mineralocorticoid receptor activation in adipose tissue and impacts glucose homeostasis in lean and obese states. METHODS The actions of 20β-DHB on corticosteroid receptors in adipose tissue were investigated first using a combination of in silico, in vitro, and transcriptomic techniques and then in vivo administration in combination with receptor antagonists. Mice lacking one Cbr1 allele and mice overexpressing Cbr1 in their adipose tissue underwent metabolic phenotyping before and after induction of obesity with high-fat feeding. RESULTS 20β-DHB activated both the glucocorticoid and mineralocorticoid receptor in adipose tissue and systemic administration to wild-type mice induced glucose intolerance, an effect that was ameliorated by both glucocorticoid and mineralocorticoid receptor antagonism. Cbr1 haploinsufficient lean male mice had lower fasting glucose and improved glucose tolerance compared with littermate controls, a difference that was abolished by administration of 20β-DHB and absent in female mice with higher baseline adipose 20β-DHB concentrations than male mice. Conversely, overexpression of Cbr1 in adipose tissue resulted in worsened glucose tolerance and higher fasting glucose in lean male and female mice. However, neither Cbr1 haploinsfficiency nor adipose overexpression affected glucose dyshomeostasis induced by high-fat feeding. CONCLUSIONS Carbonyl reductase 1 is a novel regulator of glucocorticoid and mineralocorticoid receptor activation in adipose tissue that influences glucose homeostasis in lean mice.
Collapse
Affiliation(s)
- Rachel M B Bell
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Elisa Villalobos
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Mark Nixon
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Allende Miguelez-Crespo
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Lee Murphy
- Genetics Core, Edinburgh Clinical Research Facility, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.
| | - Angie Fawkes
- Genetics Core, Edinburgh Clinical Research Facility, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.
| | - Audrey Coutts
- Genetics Core, Edinburgh Clinical Research Facility, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.
| | - Matthew G F Sharp
- Transgenics Core, Bioresearch & Veterinary Services, University of Edinburgh, Edinburgh, United Kingdom.
| | - Martha V Koerner
- Transgenics Core, Bioresearch & Veterinary Services, University of Edinburgh, Edinburgh, United Kingdom.
| | - Emma Allan
- Transgenics Core, Bioresearch & Veterinary Services, University of Edinburgh, Edinburgh, United Kingdom.
| | - Onno C Meijer
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Renè Houtman
- Pamgene International, Den Bosch, the Netherlands.
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| | - Katharina R Beck
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| | - Scott G Denham
- Mass Spectrometry Core Laboratory, Wellcome Trust Clinical Research Facility, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Patricia Lee
- Mass Spectrometry Core Laboratory, Wellcome Trust Clinical Research Facility, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Natalie Z M Homer
- Mass Spectrometry Core Laboratory, Wellcome Trust Clinical Research Facility, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Brian R Walker
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Clinical and Translational Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Ruth A Morgan
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom.
| |
Collapse
|
6
|
Magdy T, Burridge PW. Use of hiPSC to explicate genomic predisposition to anthracycline-induced cardiotoxicity. Pharmacogenomics 2021; 22:41-54. [PMID: 33448871 PMCID: PMC7923254 DOI: 10.2217/pgs-2020-0104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
The anticancer agents of the anthracycline family are commonly associated with the potential to cause severe toxicity to the heart. To solve the question of why particular a patient is predisposed to anthracycline-induced cardiotoxicity (AIC), researchers have conducted numerous pharmacogenomic studies and identified more than 60 loci associated with AIC. To date, none of these identified loci have been developed into US FDA-approved biomarkers for use in routine clinical practice. With advances in the application of human-induced pluripotent stem cell-derived cardiomyocytes, sequencing technologies and genomic editing techniques, variants associated with AIC can now be validated in a human model. Here, we provide a comprehensive overview of known genetic variants associated with AIC from the perspective of how human-induced pluripotent stem cell-derived cardiomyocytes can be used to help better explain the genomic predilection to AIC.
Collapse
Affiliation(s)
- Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
7
|
Zubiaur P, Koller D, Saiz‐Rodríguez M, Navares‐Gómez M, Abad‐Santos F. Important Pharmacogenetic Information for Drugs Prescribed During the SARS-CoV-2 Infection (COVID-19). Clin Transl Sci 2020; 13:1023-1033. [PMID: 32936528 PMCID: PMC7719396 DOI: 10.1111/cts.12866] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
In December 2019, the severe acute respiratory syndrome virus-2 pandemic began, causing the coronavirus disease 2019. A vast variety of drugs is being used off-label as potential therapies. Many of the repurposed drugs have clinical pharmacogenetic guidelines available with therapeutic recommendations when prescribed as indicated on the drug label. The aim of this review is to provide a comprehensive summary of pharmacogenetic biomarkers available for these drugs, which may help to prescribe them more safely.
Collapse
Affiliation(s)
- Pablo Zubiaur
- Clinical Pharmacology DepartmentHospital Universitario de La PrincesaInstituto Teófilo HernandoUniversidad Autónoma de Madrid (UAM)Instituto de Investigación Sanitaria La Princesa (IP)MadridSpain
- UICEC Hospital Universitario de La PrincesaPlataforma SCReN (Spanish Clinical Research Network)Instituto de Investigación Sanitaria La Princesa (IP)MadridSpain
| | - Dora Koller
- Clinical Pharmacology DepartmentHospital Universitario de La PrincesaInstituto Teófilo HernandoUniversidad Autónoma de Madrid (UAM)Instituto de Investigación Sanitaria La Princesa (IP)MadridSpain
| | - Miriam Saiz‐Rodríguez
- Clinical Pharmacology DepartmentHospital Universitario de La PrincesaInstituto Teófilo HernandoUniversidad Autónoma de Madrid (UAM)Instituto de Investigación Sanitaria La Princesa (IP)MadridSpain
- Research UnitFundación Burgos por la Investigación de la SaludHospital Universitario de BurgosBurgosSpain
| | - Marcos Navares‐Gómez
- Clinical Pharmacology DepartmentHospital Universitario de La PrincesaInstituto Teófilo HernandoUniversidad Autónoma de Madrid (UAM)Instituto de Investigación Sanitaria La Princesa (IP)MadridSpain
| | - Francisco Abad‐Santos
- Clinical Pharmacology DepartmentHospital Universitario de La PrincesaInstituto Teófilo HernandoUniversidad Autónoma de Madrid (UAM)Instituto de Investigación Sanitaria La Princesa (IP)MadridSpain
- UICEC Hospital Universitario de La PrincesaPlataforma SCReN (Spanish Clinical Research Network)Instituto de Investigación Sanitaria La Princesa (IP)MadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
8
|
ABCB1 and ABCC2 genetic polymorphism as risk factors for neutropenia in esophageal cancer patients treated with docetaxel, cisplatin, and 5-fluorouracil chemotherapy. Cancer Chemother Pharmacol 2020; 86:315-324. [PMID: 32748110 DOI: 10.1007/s00280-020-04118-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 07/19/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE The combination of docetaxel, cisplatin and 5-fluorouracil (DCF) is a newly developed chemotherapy regimen for esophageal cancer. Severe neutropenia is dose-limiting toxicity of docetaxel and it is well known to be frequently occurred during DCF chemotherapy. This study aimed to investigate the relationship between severe neutropenia and genetic polymorphisms in patients treated with preoperative DCF chemotherapy. METHODS A total of 158 patients were investigated for their absolute neutrophil count (ANC) within the first cycle of DCF chemotherapy at the National Cancer Center (NCC) Hospital East. DNA samples obtained from the NCC Biobank Registry were used for the analysis of nine genetic polymorphisms related to docetaxel pharmacokinetics. These genotypes were evaluated for their association with severe neutropenia, and further their risk factors were examined using a multivariate logistic regression. RESULTS A total 81 (51.3%) patients developed severe neutropenia. Multivariate analysis revealed that age (OR 1.054; CI 1.008-1.102, P = 0.022), baseline ANC (OR 1.019; CI 1.002-1.037, P = 0.030), ABCB1 3435C>T (OR 2.191; CI 1.087-4.417, P = 0.028) and ABCC2 *+9383C>G (OR 2.342; CI 1.108-4.948, P = 0.026) were significant risk factors for severe neutropenia development. The results from this study showed that age, ANC, ABCB1 3435C>T, and ABCC2 *+9383 G>C increased the incidence of severe neutropenia with the number of identified risk factors. CONCLUSIONS In addition to age and baseline ANC, ABCB1 3435C>T and ABCC2 *+9383C>G were identified as independent predictors for severe neutropenia in esophageal cancer patients treated with DCF chemotherapy.
Collapse
|
9
|
Cencini E, Sicuranza A, Fabbri A, Ferrigno I, Rigacci L, Cox MC, Raspadori D, Bocchia M. Study of gene polymorphisms as predictors of treatment efficacy and toxicity in patients with indolent non-hodgkin lymphomas and mantle cell lymphoma receiving bendamustine and rituximab. Br J Haematol 2018; 184:223-231. [PMID: 30203425 DOI: 10.1111/bjh.15582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022]
Abstract
Bendamustine is used in combination with rituximab (BR) to treat indolent non-Hodgkin lymphomas (iNHL) and mantle cell lymphoma (MCL). The variability in treatment efficacy and toxicity could be related to single nucleotide polymorphisms (SNPs) in immune response genes. We would like to show a correlation between SNPs and treatment outcome in iNHL and MCL patients receiving BR. We investigated some SNPs that had already been associated with NHL outcome. Samples were genotyped for the IL2 (rs2069762), IL10 (rs1800890, rs10494879), VEGFA (rs3025039), IL8 (rs4073), CFH (rs1065489) and MTHFR (rs1801131) SNPs by allelic discrimination assays. We enrolled 70 patients that received rituximab 375 mg/m2 and bendamustine 90 mg/m2 every 28 days, both as first-line treatment and ≥ second-line regimens. Overall response rate was 97·1% (complete response [CR] rate 73·9%). Treatment toxicity included grade 3-4 neutropenia (24/70 patients), infections (21/70 patients; 1/70 grade 3), skin rash (26/70 patients; 2/70 grade 3). After a median follow-up of 24 months we did find any correlation between the analysed SNPs, CR rate and PFS. However, we demonstrated an association between the SNP in IL2 (rs2069762) and the onset of skin rash (P = 0·0001). Our study suggests a role for cytokine SNPs in bendamustine-related toxicity, which could represent a promising research field.
Collapse
Affiliation(s)
- Emanuele Cencini
- Unit of Haematology, Azienda Ospedaliera Universitaria Senese, Siena, Italy.,University of Siena, Siena, Italy
| | - Anna Sicuranza
- Unit of Haematology, Azienda Ospedaliera Universitaria Senese, Siena, Italy.,University of Siena, Siena, Italy
| | - Alberto Fabbri
- Unit of Haematology, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Ilaria Ferrigno
- Unit of Haematology, Azienda Ospedaliera Universitaria Senese, Siena, Italy.,University of Siena, Siena, Italy
| | - Luigi Rigacci
- Haematology Department, University of Florence, Florence, Italy
| | - Maria C Cox
- Haematology Department, Ospedale S. Andrea, Roma, Italy
| | | | - Monica Bocchia
- Unit of Haematology, Azienda Ospedaliera Universitaria Senese, Siena, Italy.,University of Siena, Siena, Italy
| |
Collapse
|
10
|
Tecza K, Pamula-Pilat J, Lanuszewska J, Butkiewicz D, Grzybowska E. Pharmacogenetics of toxicity of 5-fluorouracil, doxorubicin and cyclophosphamide chemotherapy in breast cancer patients. Oncotarget 2018; 9:9114-9136. [PMID: 29507678 PMCID: PMC5823653 DOI: 10.18632/oncotarget.24148] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/02/2018] [Indexed: 01/28/2023] Open
Abstract
The differences in patients' response to the same medication, toxicity included, are one of the major problems in breast cancer treatment. Chemotherapy toxicity makes a significant clinical problem due to decreased quality of life, prolongation of treatment and reinforcement of negative emotions associated with therapy. In this study we evaluated the genetic and clinical risk factors of FAC chemotherapy-related toxicities in the group of 324 breast cancer patients. Selected genes and their polymorphisms were involved in FAC drugs transport (ABCB1, ABCC2, ABCG2,SLC22A16), metabolism (ALDH3A1, CBR1, CYP1B1, CYP2C19, DPYD, GSTM1, GSTP1, GSTT1, MTHFR,TYMS), DNA damage recognition, repair and cell cycle control (ATM, ERCC1, ERCC2, TP53, XRCC1). The multifactorial risk models that combine genetic risk modifiers and clinical characteristics were constructed for 12 toxic symptoms. The majority of toxicities was dependent on the modifications in components of more than one pathway of FAC drugs, while the impact level of clinical factors was comparable to the genetic ones. For the carriers of multiple high risk factors the chance of developing given symptom was significantly elevated which proved the factor-dosage effect. We found the strongest associations between concurrent presence of clinical factors - overall and recurrent anemia, nephrotoxicity and early nausea and genetic polymorphisms in genes responsible for DNA repair, drugs metabolism and transport pathways. These results indicate the possibility of selection of the patients with expected high tolerance to FAC treatment and consequently with high chance of chemotherapy completion without the dose reduction, treatment delays and decline in the quality of life.
Collapse
Affiliation(s)
- Karolina Tecza
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Jolanta Pamula-Pilat
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Joanna Lanuszewska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Dorota Butkiewicz
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Ewa Grzybowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| |
Collapse
|
11
|
Liu D, Wu N, Sun H, Dong M, Guo T, Chi P, Li G, Sun D, Jin Y. ABCG2 and NCF4 polymorphisms are associated with clinical outcomes in diffuse large B-cell lymphoma patients treated with R-CHOP. Oncotarget 2017; 8:58292-58303. [PMID: 28938556 PMCID: PMC5601652 DOI: 10.18632/oncotarget.16869] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/09/2017] [Indexed: 02/06/2023] Open
Abstract
The impact of pharmacogenetics on predicting survival in diffuse large B-cell lymphoma (DLBCL) remains unclear. We tested 337 DLBCL patients treated with rituximab-cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) for 9 single nucleotide polymorphisms from 6 genes (CD20, FCGR2A, NAD(P)H, ABCC2, ABCG2 and CYP3A5). Patients who carried the NCF4 rs1883112 GG genotype showed significantly shorter progression-free survival (PFS) (P = 0.023) and event-free survival (EFS) (P < 0.001) comparing with A allele. A significantly shortened PFS (P = 0.013) and EFS (P = 0.002) was also observed in the patients with ABCG2 rs2231137 GG genotype. Furthermore, the elder (> 60 years old) or male patients with ABCG2 rs2231137 GG genotype had poorer PFS and EFS than A allele. Moreover, CD20 rs2070770 CC and RAC2 rs13058338 AT genotypes were independent predictors of chemotherapy-induced toxicity. Cox proportional hazards analyses demonstrated that the GG genotype of ABCG2 rs2231137 and NCF4 rs1883112 were risk factors in DLBCL patients. In conclusion, the identified polymorphisms provide guide for the identification of DLBCL patients who are likely to benefit from chemotherapy.
Collapse
Affiliation(s)
- Duo Liu
- Department of Medical Genetics, Harbin Medical University, Harbin, Heilongjiang, China.,Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Nan Wu
- Department of Medical Genetics, Harbin Medical University, Harbin, Heilongjiang, China
| | - Haiming Sun
- Department of Medical Genetics, Harbin Medical University, Harbin, Heilongjiang, China
| | - Mei Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Tianzhu Guo
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Peng Chi
- Department of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Guofu Li
- Department of Neurosurvery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Donglin Sun
- Department of Medical Genetics, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yan Jin
- Department of Medical Genetics, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
12
|
Magdy T, Burmeister BT, Burridge PW. Validating the pharmacogenomics of chemotherapy-induced cardiotoxicity: What is missing? Pharmacol Ther 2016; 168:113-125. [PMID: 27609196 DOI: 10.1016/j.pharmthera.2016.09.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The cardiotoxicity of certain chemotherapeutic agents is now well-established, and has led to the development of the field of cardio-oncology, increased cardiac screening of cancer patients, and limitation of patients' maximum cumulative chemotherapeutic dose. The effect of chemotherapeutic regimes on the heart largely involves cardiomyocyte death, leading to cardiomyopathy and heart failure, or the induction of arrhythmias. Of these cardiotoxic drugs, those resulting in clinical cardiotoxicity can range from 8 to 26% for doxorubicin, 7-28% for trastuzumab, or 5-30% for paclitaxel. For tyrosine kinase inhibitors, QT prolongation and arrhythmia, ischemia and hypertension have been reported in 2-35% of patients. Furthermore, newly introduced chemotherapeutic agents are commonly used as part of changed combinational regimens with significantly increased incidence of cardiotoxicity. It is widely believed that the mechanism of action of these drugs is often independent of their cardiotoxicity, and the basis for why these drugs specifically affect the heart has yet to be established. The genetic rationale for why certain patients experience cardiotoxicity whilst other patients can tolerate high chemotherapy doses has proven highly illusive. This has led to significant genomic efforts using targeted and genome-wide association studies (GWAS) to divine the pharmacogenomic cause of this predilection. With the advent of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), the putative risk and protective role of single nucleotide polymorphisms (SNPs) can now be validated in a human model. Here we review the state of the art knowledge of the genetic predilection to chemotherapy-induced cardiotoxicity and discuss the future for establishing and validating the role of the genome in this disease.
Collapse
Affiliation(s)
- Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Brian T Burmeister
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, USA.
| |
Collapse
|
13
|
Boušová I, Skálová L, Souček P, Matoušková P. The modulation of carbonyl reductase 1 by polyphenols. Drug Metab Rev 2015; 47:520-33. [DOI: 10.3109/03602532.2015.1089885] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|