1
|
Cao Z, Wang W, Yang Z, Liu Y, Sun L, Zhang L, Li Z. Discovery of the FXR/CES2 dual modulator LE-77 for the treatment of irinotecan-induced delayed diarrhea. Bioorg Chem 2024; 153:107852. [PMID: 39362081 DOI: 10.1016/j.bioorg.2024.107852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/11/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Irinotecan (CPT-11) is a widely utilized topoisomerase I inhibitor in the treatment of colorectal cancer and other malignant tumors. However, severe and even life-threatening dose-limiting toxicity-delayed diarrhea affects the clinical application of CPT-11. The standard treatment for CPT-11-induced delayed diarrhea is prompt use of loperamide (LPA), however LPA can also cause constipation, diarrhea and even intestinal obstruction and has a high failure rate. Carboxylesterase 2 (CES2) is the main enzyme in the intestinal transformation of CPT-11, which can convert CPT-11 into toxic metabolite SN-38 and produce intestinal toxicity. Inhibiting CES2 activity can block the hydrolysis process of CPT-11 in the intestine and reduce SN-38 accumulation. Additionally, Farnesoid X receptor (FXR) agonists have anti-inflammatory, anti-secretory, and protective functions on intestinal barrier integrity that could potentially alleviate diarrhea. In this study, we investigated for the first time the anti-delayed diarrhea effect of FXR agonists, and the first time identified LE-77 as a potent dual modulator that activates FXR and inhibits CES2 through high-throughput screening. In the CPT-11-induced delayed diarrhea model, LE-77 demonstrated a dual modulator mechanism by activating FXR and inhibiting CES2, thereby reducing the accumulation of SN-38 in the intestine, alleviating intestinal inflammation, preserving intestinal mucosal integrity, and ultimately alleviating delayed diarrhea.
Collapse
Affiliation(s)
- Zhijun Cao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Wenxin Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Zhongcheng Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yuxia Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lidan Sun
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, PR China.
| | - Luyong Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, PR China.
| |
Collapse
|
2
|
Korver SK, Bowen JM, Gibson RJ, Ball IA, Secombe KR, Wain TJ, Logan RM, Tuke J, Mead KR, Richards AM, Karapetis CS, Keefe DM, Coller JK. Advanced statistics identification of participant and treatment predictors associated with severe adverse effects induced by fluoropyrimidine-based chemotherapy. Cancer Chemother Pharmacol 2023; 91:507-521. [PMID: 37162533 DOI: 10.1007/s00280-023-04538-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/23/2023] [Indexed: 05/11/2023]
Abstract
PURPOSE Adverse effects following fluoropyrimidine-based chemotherapy regimens are common. However, there are no current accepted diagnostic markers for prediction prior to treatment, and the underlying mechanisms remain unclear. This study aimed to determine genetic and non-genetic predictors of adverse effects. METHODS Genomic DNA was analyzed for 25 single nucleotide polymorphisms (SNPs). Demographics, comorbidities, cancer and fluoropyrimidine-based chemotherapy regimen types, and adverse effect data were obtained from clinical records for 155 Australian White participants. Associations were determined by bivariate analysis, logistic regression modeling and Bayesian network analysis. RESULTS Twelve different adverse effects were observed in the participants, the most common severe adverse effect was diarrhea (12.9%). Bivariate analysis revealed associations between all adverse effects except neutropenia, between genetic and non-genetic predictors, and between 8 genetic and 12 non-genetic predictors with more than 1 adverse effect. Logistic regression modeling of adverse effects revealed a greater/sole role for six genetic predictors in overall gastrointestinal toxicity, nausea and/or vomiting, constipation, and neutropenia, and for nine non-genetic predictors in diarrhea, mucositis, neuropathy, generalized pain, hand-foot syndrome, skin toxicity, cardiotoxicity and fatigue. The Bayesian network analysis revealed less directly associated predictors (one genetic and six non-genetic) with adverse effects and confirmed associations between six adverse effects, eight genetic predictors and nine non-genetic predictors. CONCLUSION This study is the first to link both genetic and non-genetic predictors with adverse effects following fluoropyrimidine-based chemotherapy. Collectively, we report a wealth of information that warrants further investigation to elucidate the clinical significance, especially associations with genetic predictors and adverse effects.
Collapse
Affiliation(s)
- Samantha K Korver
- Discipline of Pharmacology, School of Biomedicine, The University of Adelaide, L2 Helen Mayo South, Adelaide, SA, 5000, Australia
| | - Joanne M Bowen
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Rachel J Gibson
- School of Allied Health Science and Practice, The University of Adelaide, Adelaide, Australia
| | - Imogen A Ball
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Kate R Secombe
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Taylor J Wain
- Discipline of Pharmacology, School of Biomedicine, The University of Adelaide, L2 Helen Mayo South, Adelaide, SA, 5000, Australia
| | - Richard M Logan
- Adelaide Dental School, The University of Adelaide, Adelaide, Australia
| | - Jonathan Tuke
- School of Mathematical Sciences, The University of Adelaide, Adelaide, Australia
| | - Kelly R Mead
- Flinders Medical Centre, Bedford Park, Australia
| | | | - Christos S Karapetis
- Flinders Medical Centre, Bedford Park, Australia
- Flinders University, Bedford Park, Australia
| | - Dorothy M Keefe
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
| | - Janet K Coller
- Discipline of Pharmacology, School of Biomedicine, The University of Adelaide, L2 Helen Mayo South, Adelaide, SA, 5000, Australia.
| |
Collapse
|
3
|
Coller JK, Tuke J, Wain TJ, Quinn E, Steele L, Abreu M, Aenlle K, Klimas N, Sullivan K. Associations of Immune Genetic Variability with Gulf War Illness in 1990-1991 Gulf War Veterans from the Gulf War Illness Consortium (GWIC) Multisite Case-Control Study. Brain Sci 2021; 11:brainsci11111410. [PMID: 34827409 PMCID: PMC8615505 DOI: 10.3390/brainsci11111410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 11/16/2022] Open
Abstract
Gulf War illness (GWI) encompasses a constellation of persistent debilitating symptoms associated with significant changes in central nervous system (CNS) and immune functioning. Currently, there is no validated biomarker for GWI risk susceptibility. Given the impact of immune responses linked to GWI symptomology, genetic variability that causes persistent inflammatory/immune alterations may be key. This Boston University-based Gulf War Illness Consortium (GWIC) study investigated the impact of single nucleotide polymorphisms (SNPs) in variants of immune and pain genetic markers IL1B, IL2, IL6, IL6R, IL10, TNF, TGF, TLR2, TLR4, MD2, MYD88, BDNF, CRP, ICE, COMT and OPRM1 on GWI occurrence in a Caucasian subset of Gulf War (GW) veterans with (cases, n = 170) and without (controls, n = 34) GWI. Logistic regression modeling created a prediction model of GWI risk that associated genetic variability in TGF (rs1800469, p = 0.009), IL6R (rs8192284, p = 0.004) and TLR4 (rs4986791, p = 0.013) with GWI occurrence. This prediction model was specific and sensitive, with a receiver operator characteristic area under the curve of 71.4%. This is the first report of immune genetic variability being predictive of GWI and warrants validation in larger independent cohorts. Future reports will present interactions of these genetic risk factors with other characteristics of GW service.
Collapse
Affiliation(s)
- Janet K. Coller
- Discipline of Pharmacology, School of Biomedicine, University of Adelaide, Adelaide 5005, South Australia, Australia;
- Correspondence:
| | - Jonathan Tuke
- School of Mathematical Sciences, University of Adelaide, Adelaide 5005, South Australia, Australia;
| | - Taylor J. Wain
- Discipline of Pharmacology, School of Biomedicine, University of Adelaide, Adelaide 5005, South Australia, Australia;
| | - Emily Quinn
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA 02118, USA;
| | - Lea Steele
- Veterans Health Research Program, Beth K. and Stuart C. Yudofsky Division of Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Maria Abreu
- Institute for Neuroimmune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (M.A.); (K.A.); (N.K.)
- Department of Veterans Affairs, Research Service, Miami VA Healthcare System, Miami, FL 33125, USA
| | - Kristina Aenlle
- Institute for Neuroimmune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (M.A.); (K.A.); (N.K.)
- Department of Veterans Affairs, Research Service, Miami VA Healthcare System, Miami, FL 33125, USA
| | - Nancy Klimas
- Institute for Neuroimmune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (M.A.); (K.A.); (N.K.)
- Department of Veterans Affairs, Miami VA Healthcare System Geriatric Research Education and Clinical Center Healthcare System, Miami, FL 33125, USA
| | - Kimberly Sullivan
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA;
| |
Collapse
|
4
|
Giommoni E, Giorgione R, Paderi A, Pellegrini E, Gambale E, Marini A, Antonuzzo A, Marconcini R, Roviello G, Matucci-Cerinic M, Capaccioli D, Pillozzi S, Antonuzzo L. Eosinophil Count as Predictive Biomarker of Immune-Related Adverse Events (irAEs) in Immune Checkpoint Inhibitors (ICIs) Therapies in Oncological Patients. IMMUNO 2021; 1:253-263. [DOI: 10.3390/immuno1030017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Background: To date, no biomarkers are effective in predicting the risk of developing immune-related adverse events (irAEs) in patients treated with immune checkpoint inhibitors (ICIs). This study aims to evaluate the association between basal absolute eosinophil count (AEC) and irAEs during treatment with ICIs for solid tumors. Methods: We retrospectively evaluated 168 patients with metastatic melanoma (mM), renal cell carcinoma (mRCC), and non-small cell lung cancer (mNSCLC) receiving ICIs at our medical oncology unit. By combining baseline AEC with other clinical factors, we developed a mathematical model for predicting the risk of irAEs, which we validated in an external cohort of patients. Results: Median baseline AEC was 135/µL and patients were stratified into two groups accordingly; patients with high baseline AEC (>135/µL) were more likely to experience toxicity (p = 0.043) and have a better objective response rate (ORR) (p = 0.003). By constructing a covariance analysis model, it emerged that basal AEC correlated with the risk of irAEs (p < 0.01). Finally, we validated the proposed model in an independent cohort of 43 patients. Conclusions: Baseline AEC could be a predictive biomarker of ICI-related toxicity, as well as of response to treatment. The use of a mathematical model able to predict the risk of developing irAEs could be useful for clinicians for monitoring patients receiving ICIs.
Collapse
Affiliation(s)
- Elisa Giommoni
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Roberta Giorgione
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Agnese Paderi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Elisa Pellegrini
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Elisabetta Gambale
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Andrea Marini
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Andrea Antonuzzo
- Medical Oncology 1 and 2, Azienda Ospedaliero-Universitaria Pisana, 56127 Pisa, Italy
| | - Riccardo Marconcini
- Medical Oncology 1 and 2, Azienda Ospedaliero-Universitaria Pisana, 56127 Pisa, Italy
| | - Giandomenico Roviello
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - David Capaccioli
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Serena Pillozzi
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Lorenzo Antonuzzo
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
5
|
Prediction of mucositis risk secondary to cancer therapy: a systematic review of current evidence and call to action. Support Care Cancer 2020; 28:5059-5073. [PMID: 32592033 DOI: 10.1007/s00520-020-05579-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/12/2020] [Indexed: 01/25/2023]
Abstract
PURPOSE Despite advances in personalizing the efficacy of cancer therapy, our ability to identify patients at risk of severe treatment side effects and provide individualized supportive care is limited. This is particularly the case for mucositis (oral and gastrointestinal), with no comprehensive risk evaluation strategies to identify high-risk patients. We, the Multinational Association for Supportive Care in Cancer/International Society for Oral Oncology (MASCC/ISOO) Mucositis Study Group, therefore aimed to systematically review current evidence on that factors that influence mucositis risk to provide a foundation upon which future risk prediction studies can be based. METHODS We identified 11,018 papers from PubMed and Web of Science, with 197 records extracted for full review and 113 meeting final eligibility criteria. Data were then synthesized into tables to highlight the level of evidence for each risk predictor. RESULTS The strongest level of evidence supported dosimetric parameters as key predictors of mucositis risk. Genetic variants in drug-metabolizing pathways, immune signaling, and cell injury/repair mechanisms were also identified to impact mucositis risk. Factors relating to the individual were variably linked to mucositis outcomes, although female sex and smoking status showed some association with mucositis risk. CONCLUSION Mucositis risk reflects the complex interplay between the host, tumor microenvironment, and treatment specifications, yet the large majority of studies rely on hypothesis-driven, single-candidate approaches. For significant advances in the provision of personalized supportive care, coordinated research efforts with robust multiplexed approaches are strongly advised.
Collapse
|
6
|
Hu R, Barratt DT, Coller JK, Sallustio BC, Somogyi AA. No Major Effect of Innate Immune Genetics on Acute Kidney Rejection in the First 2 Weeks Post-Transplantation. Front Pharmacol 2020; 10:1686. [PMID: 32153387 PMCID: PMC7045476 DOI: 10.3389/fphar.2019.01686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/24/2019] [Indexed: 12/03/2022] Open
Abstract
Background Innate immunity contributes to acute rejection after kidney transplantation. Genetic polymorphisms affecting innate immunity may therefore influence patients’ risk of rejection. IL2 -330T > G, IL10 -1082G > A, -819C > T, and -592C > A, and TNF -308G > A are not associated with acute rejection incidence in Caucasian kidney transplant recipients receiving a calcineurin inhibitor, ciclosporin or tacrolimus (TAC). However, other important innate immune genetic polymorphisms have not yet been extensively studied in recipients and donors. In addition, innate immunogenetics have not been investigated in kidney transplant cohorts receiving only TAC as the calcineurin inhibitor. Objective To investigate the effect of recipient and donor CASP1, CRP, IL1B, IL2, IL6, IL6R, IL10, MYD88, TGFB, TLR2, TLR4, and TNF genetics on acute kidney rejection in the first 2 weeks post-transplant in TAC-treated kidney transplant recipients. Methods This study included 154 kidney transplant recipients and 81 donors successfully genotyped for 17 polymorphisms in these genes. All recipients were under triple immunosuppressant therapy of TAC, mycophenolate mofetil, and prednisolone. Recipient and donor genotype differences in acute rejection incidence within the first 2 weeks post-transplantation were assessed by logistic regression, adjusting for induction therapy, human leukocyte antigen mismatches, kidney transplant number, living donor, and peak panel-reactive antibody scores. Results A trend (Cochran-Armitage P = 0.031) of increasing acute rejection incidence was observed from recipient IL6 -6331 T/T (18%) to T/C (25%) to C/C (46%) genotype [C/C versus T/T odds ratio (95% confidence interval) = 6.6 (1.7 to 25.8) (point-wise P = 0.017)]. However, no genotype differences were significant after Bonferroni correction for multiple comparisons. Conclusions This study did not detect any statistically significant effects of recipient or donor innate immune genetics on acute rejection incidence in the first 2 weeks post-transplantation. However, the sample size was small, and future larger studies or meta-analyses are required to demonstrate conclusively if innate immune genetics such as IL6 influence the risk of acute rejection after kidney transplantation.
Collapse
Affiliation(s)
- Rong Hu
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel T. Barratt
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Janet K. Coller
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Benedetta C. Sallustio
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Department of Clinical Pharmacology, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Andrew A. Somogyi
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, SA, Australia
- *Correspondence: Andrew A. Somogyi,
| |
Collapse
|
7
|
Zhao D, Wu YH, Zhao TC, Jia ZF, Cao DH, Yang N, Wang YQ, Cao XY, Jiang J. Single-nucleotide polymorphisms in Toll-like receptor genes are associated with the prognosis of gastric cancer and are not associated with Helicobacter pylori infection. INFECTION, GENETICS AND EVOLUTION 2019; 73:384-389. [DOI: 10.1016/j.meegid.2019.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/22/2019] [Accepted: 06/03/2019] [Indexed: 01/17/2023]
|
8
|
Coller JK, Ramachandran J, John L, Tuke J, Wigg A, Doogue M. The impact of liver transplant recipient and donor genetic variability on tacrolimus exposure and transplant outcome. Br J Clin Pharmacol 2019; 85:2170-2175. [PMID: 31219197 DOI: 10.1111/bcp.14034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/15/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022] Open
Abstract
This study investigated the effect of recipient and donor genetic variability on dose-adjusted steady-state tacrolimus concentrations (Css ) and clinical outcomes 3 and 6 months after liver transplant. Twenty-nine recipients and matched donor blood samples were genotyped for 27 single nucleotide polymorphisms including CYP3A5*3 (rs776746), ABCB1 haplotype and immune genes. Associations between genetic variability and clinical parameters and Css and the occurrence of rejection and nephrotoxicity were analysed by multivariate and multinomial logistic regression modelling and Jonckheere-Terpstra tests examined the impact of combined donor/recipient CYP3A5 expression on Css . At 3 months post-transplant modelling revealed an association between tacrolimus Css and recipient CASP1 rs580523 genotype (P = 0.005), accounting for 52% Css variance. Jonckheere-Terpstra tests revealed that as combined donor/recipient CYP3A5 expression increased, Css decreased (P = 0.010 [3 months], 0.018 [6 months]). As this is the first report of CASP1 genetic variability influencing tacrolimus Css , further validation in larger cohorts is required.
Collapse
Affiliation(s)
- Janet K Coller
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Jeyamani Ramachandran
- Hepatology and Liver Transplantation Medicine Unit, Flinders Medical Centre, Bedford Park, Australia.,South Australian Liver Transplant Unit, Flinders Medical Centre, Bedford Park, Australia
| | - Libby John
- South Australian Liver Transplant Unit, Flinders Medical Centre, Bedford Park, Australia
| | - Jonathan Tuke
- School of Mathematical Sciences, University of Adelaide, Adelaide, Australia.,ARC Centre of Excellence for Mathematical & Statistical Frontiers, School of Mathematical Sciences, University of Adelaide, Adelaide, Australia
| | - Alan Wigg
- Hepatology and Liver Transplantation Medicine Unit, Flinders Medical Centre, Bedford Park, Australia.,South Australian Liver Transplant Unit, Flinders Medical Centre, Bedford Park, Australia
| | - Matthew Doogue
- Department of Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
9
|
Korver SK, Gibson RJ, Bowen JM, Coller JK. Toll-like receptor/interleukin-1 domain innate immune signalling pathway genetic variants are candidate predictors for severe gastrointestinal toxicity risk following 5-fluorouracil-based chemotherapy. Cancer Chemother Pharmacol 2018; 83:217-236. [PMID: 30474704 DOI: 10.1007/s00280-018-3729-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/08/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE Severe gastrointestinal (GI) toxicity is a common adverse effect following 5-fluorouracil (5-FU)-based chemotherapy treatment. The presence of severe GI toxicity leads to treatment revisions, sub-optimal therapy outcomes, and decreases to patients' quality of life. There are no adequate predictors for 5-FU-induced severe GI toxicity risk. The Toll-like receptor/interleukin-1 (TIR) domain innate immune signalling pathway is known to be a mediating pathway in the development of GI toxicity. Hence, genetic variability in this signalling pathway may alter the pathophysiology of GI toxicity and, therefore, be predictive of risk. However, little research has investigated the effects of TIR domain innate immune signalling pathway single nucleotide polymorphism (SNPs) on the risk and development of severe GI toxicity. METHODS This critical review surveyed the literature and reported on the in vitro, ex vivo and in vivo effects, as well as the genetic association, of selected TIR domain innate immune signalling pathway SNPs on disease susceptibility and gene functioning. RESULTS Of the TIR domain innate immune signalling pathway SNPs reviewed, evidence suggests interleukin-1 beta (IL1B) and tumour necrosis factor alpha (TNF) SNPs have the greatest potential as predictors for severe GI toxicity risk. These results warrant further research into the effect of IL1B and TNF SNPs on the risk and development of severe GI toxicity. CONCLUSIONS SNPs of the TIR domain innate immune signalling pathway have profound effects on disease susceptibility and gene functioning, making them candidate predictors for severe GI toxicity risk. The identification of a predictor for 5-FU-induced severe GI toxicity will allow the personalization of supportive care measures.
Collapse
Affiliation(s)
- Samantha K Korver
- Cancer Treatment Toxicities Group, Adelaide Medical School, Disciplines of Pharmacology and Physiology, University of Adelaide, Level 2, Helen Mayo South Building, Adelaide, SA, 5005, Australia.
| | - Rachel J Gibson
- Cancer Treatment Toxicities Group, Adelaide Medical School, Disciplines of Pharmacology and Physiology, University of Adelaide, Level 2, Helen Mayo South Building, Adelaide, SA, 5005, Australia.,Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Joanne M Bowen
- Cancer Treatment Toxicities Group, Adelaide Medical School, Disciplines of Pharmacology and Physiology, University of Adelaide, Level 2, Helen Mayo South Building, Adelaide, SA, 5005, Australia
| | - Janet K Coller
- Cancer Treatment Toxicities Group, Adelaide Medical School, Disciplines of Pharmacology and Physiology, University of Adelaide, Level 2, Helen Mayo South Building, Adelaide, SA, 5005, Australia
| |
Collapse
|
10
|
Secombe KR, Coller JK, Gibson RJ, Wardill HR, Bowen JM. The bidirectional interaction of the gut microbiome and the innate immune system: Implications for chemotherapy‐induced gastrointestinal toxicity. Int J Cancer 2018; 144:2365-2376. [DOI: 10.1002/ijc.31836] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/14/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Kate R. Secombe
- Cancer Treatment Toxicities Group, Discipline of Physiology, Adelaide Medical SchoolUniversity of Adelaide Adelaide South Australia Australia
| | - Janet K. Coller
- Cancer Treatment Toxicities Group, Discipline of PharmacologyAdelaide Medical School, University of Adelaide Adelaide South Australia Australia
| | - Rachel J. Gibson
- Cancer Treatment Toxicities Group, Discipline of Physiology, Adelaide Medical SchoolUniversity of Adelaide Adelaide South Australia Australia
- Division of Health SciencesUniversity of South Australia Adelaide South Australia Australia
| | - Hannah R. Wardill
- Cancer Treatment Toxicities Group, Discipline of Physiology, Adelaide Medical SchoolUniversity of Adelaide Adelaide South Australia Australia
- Department of Pediatric Oncology/Hematology, University of Groningen, Beatrix Children's HospitalUniversity Medical Center Groningen Groningen The Netherlands
| | - Joanne M. Bowen
- Cancer Treatment Toxicities Group, Discipline of Physiology, Adelaide Medical SchoolUniversity of Adelaide Adelaide South Australia Australia
| |
Collapse
|
11
|
Bai J, Behera M, Bruner DW. The gut microbiome, symptoms, and targeted interventions in children with cancer: a systematic review. Support Care Cancer 2017; 26:427-439. [PMID: 29168036 DOI: 10.1007/s00520-017-3982-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE The gut microbiome plays a critical role in maintaining children's health and in preventing and treating children's disease. Current application of the gut microbiome in childhood cancer is still lacking. This study aimed to systematically review the following: (1) alternations in the gut microbiome throughout cancer treatment trajectories in children, (2) the associations between the gut microbiome and gastrointestinal (GI) symptoms and psychoneurological symptoms (PNS), and (3) the efficacy of therapeutic interventions in the gut microbiome in children with cancer. METHODS PubMed, EMBASE, the Cochrane Library, and the American Society of Clinical Oncology abstract were searched. Eligible studies included all study types in which the gut microbiome was primarily reported in children with cancer. The Mixed Methods Assessment Tool was used to evaluate the methodology quality of included studies. Seven studies met our eligibility criteria, including two cohort studies, two case-control studies, and three randomized controlled trails. RESULTS The findings showed that the diversity estimates of the gut microbiome in children with cancer were lower than those of healthy controls both pre- and post-treatment. Children with cancer showed a significantly lower relative abundance of healthy gut microbiome (e.g., Clostridium XIVa and Bifidobacterium) during and after cancer treatment. No adequate literature was identified to support the associations between dysbiosis of the gut microbiome and GI symptoms/PNS. The use of prebiotics (fructooligosaccharides) and probiotics (Bifidobacterium or Lactobacilli) appears to improve the microenvironment of the gut around 1 month (4-5 weeks) during chemotherapy rather than at the beginning of treatment. Data also suggest that both prebiotic and probiotic interventions decrease clinical side effects (e.g., infection and morbidity risk) in children with cancer. CONCLUSIONS This study adds to the evidence that dysbiosis of the gut microbiome can be improved using prebiotic and probiotic supplementations in children with cancer. More well-designed experimental studies are needed to confirm this conclusion. Further studies are needed to examine the associations between the gut microbiome and GI symptoms/PNS in childhood cancer.
Collapse
Affiliation(s)
- Jinbing Bai
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA.
| | - Madhusmita Behera
- Department of Hematology and Oncology, School of Medicine, Emory University, Atlanta, GA, USA.,Winship Research Informatics, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Deborah Watkins Bruner
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA.,Education and Training, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
12
|
Toll-like receptors in the pathogenesis of chemotherapy-induced gastrointestinal toxicity. Curr Opin Support Palliat Care 2016; 10:157-64. [DOI: 10.1097/spc.0000000000000202] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
13
|
Dupuis LL, Lu X, Mitchell HR, Sung L, Devidas M, Mattano LA, Carroll WL, Winick N, Hunger SP, Maloney KW, Kadan-Lottick NS. Anxiety, pain, and nausea during the treatment of standard-risk childhood acute lymphoblastic leukemia: A prospective, longitudinal study from the Children's Oncology Group. Cancer 2016; 122:1116-25. [PMID: 26773735 DOI: 10.1002/cncr.29876] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/17/2015] [Accepted: 12/02/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND This prospective study describes the procedure-related anxiety, treatment-related anxiety, pain, and nausea experienced by children with standard-risk acute lymphoblastic leukemia (ALL) during the first year of treatment. METHODS This study was undertaken at 31 Children's Oncology Group (COG) sites. Eligible children who were 2 to 9.99 years old were enrolled in a COG trial for patients with newly diagnosed standard-risk ALL from 2005 to 2009. Parents completed a demographic survey at the baseline and the Pediatric Quality of Life Inventory 3.0 Cancer Module (proxy version) and the General Functioning Scale of the Family Assessment Device 1, 6, and 12 months after the diagnosis. The association between patient-related (age, sex, ethnicity, and treatment), parent-related (marital status and education), and family-related factors (functioning, income, and size) and symptom scores was evaluated. RESULTS The mean scores for procedure-related anxiety, treatment-related anxiety, and pain improved during the first year of treatment (P < .0389). The mean nausea score was poorer 6 months after the diagnosis in comparison with the other assessments (P = .0085). A younger age at diagnosis was associated with significantly worse procedure-related anxiety (P = .004). An older age (P = .0002) and assignment to the intensified consolidation study arm (P = .02) were associated with significantly worse nausea. CONCLUSIONS Children with ALL experienced decreasing treatment-related anxiety, procedure-related anxiety, and pain during the first year of treatment. In comparison with scores at 1 and 12 months, nausea was worse 6 months after the diagnosis. Minimization of procedure-related anxiety in younger children and improved nausea control in older children and those receiving more intensified treatment should be prioritized.
Collapse
Affiliation(s)
- L Lee Dupuis
- Department of Pharmacy, Research Institute, Hospital for Sick Children, Toronto, Canada.,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Xiaomin Lu
- Department of Biostatistics, Public Health and Health Professions, College of Medicine, University of Florida, Gainesville, Florida
| | | | - Lillian Sung
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Canada
| | - Meenakshi Devidas
- Department of Biostatistics, Public Health and Health Professions, College of Medicine, University of Florida, Gainesville, Florida
| | | | - William L Carroll
- Perlmutter Cancer Center, Langone Medical Center, New York University, New York, New York
| | - Naomi Winick
- Pediatric Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stephen P Hunger
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kelly W Maloney
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado
| | - Nina S Kadan-Lottick
- Section of Pediatric Hematology/Oncology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
14
|
Aprile G, Rihawi K, De Carlo E, Sonis ST. Treatment-related gastrointestinal toxicities and advanced colorectal or pancreatic cancer: A critical update. World J Gastroenterol 2015; 21:11793-11803. [PMID: 26557003 PMCID: PMC4631977 DOI: 10.3748/wjg.v21.i41.11793] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/06/2015] [Accepted: 09/15/2015] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal toxicities (GIT), including oral mucositis, nausea and vomiting, and diarrhea, are common side effects of chemotherapy and targeted agents in patients with advanced colorectal cancer and pancreatic cancer. Being often underreported, it is still difficult to precisely establish their burden in terms of both patient’s quality of life and cancer care costs. Moreover, with the use of more intensive upfront combination regimens, the frequency of these toxicities is rapidly growing with a potential negative effect also on patient’s outcome, as a result of dose reductions, delays or even discontinuation of active treatments. Thus, identifying patients at higher risk of developing GIT as well as an optimal management are paramount in order to improve patient’s compliance and outcome. After the description of the main treatment-induced GIT, we discuss the current knowledge on the pathophysiology of these side effects and comment the scales commonly used to assess and grade them. We then provide a critical update on GIT incidence based on the results of key randomized trials conducted in patients with metastatic colorectal cancer and advanced pancreatic cancer.
Collapse
|
15
|
Zhang H, Wang M, Shi T, Shen L, Liang L, Deng Y, Li G, Zhu J, Wu Y, Fan M, Deng W, Wei Q, Zhang Z. TNF rs1799964 as a Predictive Factor of Acute Toxicities in Chinese Rectal Cancer Patients Treated With Chemoradiotherapy. Medicine (Baltimore) 2015; 94:e1955. [PMID: 26559268 PMCID: PMC4912262 DOI: 10.1097/md.0000000000001955] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Acute toxicity is the main dose-limiting factor in the chemoradiotherapy of rectal cancer patients and depends on several pro-inflammatory factors, including interleukin-1 (IL-1), IL-6, and tumor necrosis factor-alpha (TNF-α). It is unknown whether genetic factors, such as single-nucleotide polymorphisms (SNPs) in the IL-1, IL-6, and TNF genes, are also associated with acute toxicity in the process.We genotyped 5 potentially functional SNPs in these 3 genes (TNF rs1799964, TNF rs1800629, IL-6 rs1800796, and IL-1 rs1143623, IL-1 rs1143627) and estimated their associations with severe acute radiation injury (grade ≥2) in 356 rectal cancer patients.We found a predictive role of the TNF rs1799964 T variant allele in the development of acute injury (for CT vs CC: adjusted odds ratio [OR] = 4.718, 95% confidence interval [CI] = 1.152-19.328, P = 0.031; for TT vs CC: adjusted OR = 4.443, 95% CI = 1.123-17.581, P = 0.034). In the dominant model, for CT/TT vs CC, the adjusted OR = 4.132, 95% CI = 1.069-15.966, and P = 0.04.Our results suggested that genetic variants in the TNF gene may influence acute injury in rectal cancer patients treated with chemoradiotherapy and may be a predictor for personalized treatment. Additional larger and independent studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Hui Zhang
- From the Department of Radiation Oncology (HZ, LS, LL, GL, JZ, YW, MF, WD, ZZ); Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China (MW, YD, QW); Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina (QW); Department of Obstetrics and Gynocology, Zhongshan Hospital (TS); and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (HZ, MW, LS, LL, YD, GL, JZ, YW, MF, WD, ZZ)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Gibson RJ, Bowen JM, Coller JK. What are the predictive factors in the risk and severity of chemotherapy-induced gastrointestinal toxicity? Future Oncol 2015; 11:2367-70. [PMID: 26270862 DOI: 10.2217/fon.15.138] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Rachel J Gibson
- Discipline of Anatomy & Pathology, School of Medicine, University of Adelaide, Adelaide SA 5005, Australia
| | - Joanne M Bowen
- Discipline of Physiology, School of Medicine, University of Adelaide, Adelaide SA 5005, Australia
| | - Janet K Coller
- Discipline of Pharmacology, School of Medicine, University of Adelaide, Adelaide SA 5005, Australia
| |
Collapse
|