1
|
Sayer M, Hamano H, Nagasaka M, Lee BJ, Doh J, Patel PM, Zamami Y, Ozaki AF. Time dependent predictors of cardiac inflammatory adverse events in cancer patients receiving immune checkpoint inhibitors. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:40. [PMID: 40296103 PMCID: PMC12036232 DOI: 10.1186/s40959-025-00331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Cardio-inflammatory immune related adverse events (irAEs) while receiving immune checkpoint inhibitor (ICI) therapy are particularly consequential due to their associations with poorer treatment outcomes. Evaluation of predictive factors of these serious irAEs with a time dependent approach allows better understanding of patients most at risk. OBJECTIVE To identify different elements of patient data that are significant predictors of early and late-onset or delayed cardio-inflammatory irAEs through various predictive modeling strategies. METHODS A cohort of patients receiving ICI therapy from January 1, 2010 to May 1, 2022 was identified from TriNetX meeting inclusion/exclusion criteria. Patient data collected included occurrence of early and later cardio-inflammatory irAEs, patient survival time, patient demographic information, ICI therapies, comorbidities, and medication histories. Predictive and statistical modeling approaches identified unique risk factors for early and later developing cardio-inflammatory irAEs. RESULTS A cohort of 66,068 patients on ICI therapy were identified in the TriNetX platform; 193 (0.30%) experienced early cardio-inflammatory irAEs and 175 (0.26%) experienced later cardio-inflammatory irAEs. Significant predictors for early irAEs included: anti-PD-1 therapy at index, combination ICI therapy at index, and history of peripheral vascular disease. Significant predictors for later irAEs included: a history of myocarditis and/or pericarditis, cerebrovascular disease, and history of non-steroidal anti-inflammatory medication use. CONCLUSIONS Cardio-inflammatory irAEs can be divided into clinically meaningful categories of early and late based on time since initiation of ICI therapy. Considering distinct risk factors for early-onset and late-onset events may allow for more effective patient monitoring and risk assessment.
Collapse
Affiliation(s)
- Michael Sayer
- School of Pharmacy & Pharmaceutical Sciences, University of California, 802 W Peltason Dr, Room 106A, Irvine, CA, 92617, USA
| | - Hirofumi Hamano
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Misako Nagasaka
- Division of Hematology and Oncology, University of California, Irvine, CA, USA
| | - Benjamin J Lee
- Department of Pharmacy, University of California Irvine Health, Orange, CA, USA
| | - Jean Doh
- Department of Pharmacy, University of California Irvine Health, Orange, CA, USA
| | - Pranav M Patel
- Division of Cardiology, Department of Medicine, University of California, Irvine, CA, USA
| | - Yoshito Zamami
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Aya F Ozaki
- School of Pharmacy & Pharmaceutical Sciences, University of California, 802 W Peltason Dr, Room 106A, Irvine, CA, 92617, USA.
| |
Collapse
|
2
|
Alwaili MA, Abu-Almakarem AS, El-Said KS, Eid TM, Mobasher MA, Alsabban AH, Alburae NA, Banjabi AA, Soliman MM. Shikimic acid protects against doxorubicin-induced cardiotoxicity in rats. Sci Rep 2025; 15:8126. [PMID: 40057537 PMCID: PMC11890735 DOI: 10.1038/s41598-025-90549-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 02/13/2025] [Indexed: 05/13/2025] Open
Abstract
Doxorubicin (DOX) is used to treat a variety of malignancies; however, its cardiotoxicity limits its effectiveness. Shikimic acid (SA) showed several promising biomedical applications. This study investigated the protective effect of SA on DOX-induced cardiotoxicity in male rats. The ADMETlab 2.0 web server was used to predict the pharmacokinetic properties of SA. Molecular docking studies were conducted using AutoDock Vina. Fifty male rats were divided into 4 groups (n = 10); G1 was a negative control; G2 was injected with 4 mg/kg of DOX intraperitoneally (i.p.) once a week for a month; G3 was gavaged by 1/10 of SA LD50 (280 mg/kg) daily for a month, and G4 was injected with DOX as in G2 and with SA as in G3. After a month, hematological, biochemical, molecular, and histopathological investigations were assessed. The results showed that SA treatment led to significant amelioration of the DOX-induced cardiotoxicity in rats by restoring hematological, biochemical, inflammatory biomarkers, antioxidant gene expression, and cardiac histopathological alterations. Importantly, the impact of SA treatment against DOX-promoted cardiac deterioration is by targeting the Nrf-2/Keap-1/HO-1/NQO-1 signaling pathway, which in turn induces the antioxidant agents. These findings suggest that SA treatment could potentially mitigate cardiac toxicity during DOX-based chemotherapy.
Collapse
Affiliation(s)
- Maha Abdullah Alwaili
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 11671, Riyadh, Saudi Arabia
| | - Amal S Abu-Almakarem
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Karim Samy El-Said
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
- Biochemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Thamir M Eid
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Maysa A Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, 72388, Sakaka, Saudi Arabia
| | - Ashwaq Hassan Alsabban
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Unit of Neurological Disorders, Department of Genetic Medicine, Faculty of Medicine, Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER.HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Najla Ali Alburae
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abeer A Banjabi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Mohamed Mostafa Soliman
- Department of Biology, College of Science, Jazan University, Kingdom of Saudi Arabia, P.O. Box. 114, 45142, Jazan, Saudi Arabia
- Department of Zoology, Faculty of Science, Ain Shams University, PO Box 11566, Cairo, Egypt
| |
Collapse
|
3
|
Huang Y, Chen X, Chen M, Lin Y, Chen B, Gao H, Chen M. Drug-induced heart failure: a real-world pharmacovigilance study using the FDA adverse event reporting system database. Front Pharmacol 2025; 15:1523136. [PMID: 39881876 PMCID: PMC11775474 DOI: 10.3389/fphar.2024.1523136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/24/2024] [Indexed: 01/31/2025] Open
Abstract
Objective Although there are certain drug categories associated with heart failure (HF), most of the associated risks are unclear. We investigated the top drugs associated with HF and acute HF (AHF) reported in the FDA Adverse Event Reporting System (FAERS). Methods We reviewed publicly available FAERS databases from 2004 to 2023. Using the search terms "cardiac failure" or "cardiac failure acute" and classifying cases by drug name, we processed and analyzed drug reports related to HF or AHF. Results From 2004 to 2023, 17,379,609 adverse drug events were reported by FAERS, of which 240,050 (1.38%) were reported as HF. Among those with HF, the male-to-female ratio was 0.94% and 52.37% were >65 years old; 46.2% were from the United States. There were 5,971 patients with AHF. We identified 38 drugs and 13 drug classes with a potential high risk of causing HF, and 41 drugs and 19 drug classes were associated with AHF. The median onset times of HF and AHF were 83 days (IQR: 11-416) and 49 days (IQR: 8-259), respectively. The Weibull shape parameter (WSP) test showed early failure-type profile characteristics. Conclusion This study highlights key drugs associated with drug-induced HF and AHF, emphasizing the importance of early risk assessment and close monitoring, particularly during the initial stages of treatment. These findings contribute to a better understanding of drug-induced HF and provide a basis for future research on its underlying mechanisms.
Collapse
Affiliation(s)
- Youqi Huang
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xiaowen Chen
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Mingyu Chen
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Yuze Lin
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bingqi Chen
- Department of pharmacy, Xiamen Medical College, Xiamen, China
| | - Hongjin Gao
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Min Chen
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| |
Collapse
|
4
|
Igawa Y, Hamano H, Esumi S, Takeda T, Kajizono M, Kikuoka R, Kimura I, Zamami Y. Cardiovascular toxicity risk assessment of tyrosine kinase inhibitors: a pharmacovigilance study using the VigiBase database. Front Pharmacol 2024; 15:1472008. [PMID: 39687301 PMCID: PMC11646760 DOI: 10.3389/fphar.2024.1472008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Advances in the early detection and treatment of cancer have significantly improved the prognosis of patients with cancer. Tyrosine kinase inhibitors (TKIs) are effective targeted treatments for various malignancies that act by inhibiting kinase activity. Although these drugs share a common mechanism of action, they differ in their targeted kinases, pharmacokinetics, and side effects. TKIs can cause cardiovascular side effects, which adversely affect the prognosis of cancer survivors. This study aimed to assess the risk of cardiac toxicity associated with TKIs using the World Health Organization Global Database, VigiBase. Methods We conducted a cross-sectional analysis of data from VigiBase, a comprehensive global database of suspected drug reactions. The dataset included reports up to December 2022. We identified patients treated with Food and Drug Administration-approved TKIs and analyzed their age and sex data. The primary outcome was cardiovascular impairment, defined by 21 preferred terms in the Medical Dictionary for Regulatory Activities Terminology version 25.1. Disproportionality analysis using the reported odds ratio was performed to detect adverse cardiovascular signals. Statistical analyses were conducted using R 3.3.2, with a P-value <0.05 considered significant. Results Of the 32, 520, 983 reports in VigiBase, 23, 181, 539 were eligible for the analysis. Significant cardiovascular signals were identified for 17 TKIs, including erlotinib, gefitinib, and imatinib. Stratified analyses revealed potential sex- and age-related differences in the risk of adverse events. Heatmaps indicated significant signals for drugs such as lapatinib in males and gefitinib in younger patients. Discussion Our findings indicate that some TKIs, particularly those classified as VEGFR, BCR-ABL, and BTK, pose similar risks of cardiotoxicity, while others, including EGFR, HER2, and ALK TKIs, exhibit varied risk profiles. These results underscore the importance of individualized risk assessment and management of TKI-treated patients. In conclusion, this study provides valuable insights into the cardiotoxic risk of TKIs, which is essential for developing tailored treatment plans.
Collapse
Affiliation(s)
- Yusuke Igawa
- Department of Clinical Pharmacology and Pharmacy, Okayama University, Okayama, Japan
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Hirofumi Hamano
- Department of Clinical Pharmacology and Pharmacy, Okayama University, Okayama, Japan
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Satoru Esumi
- Department of Clinical Drug Evaluation, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Tatsuaki Takeda
- Department of Education and Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Makoto Kajizono
- Department of Pharmacy, Okayama City Hospital, Okayama, Japan
| | - Ryo Kikuoka
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Ikuya Kimura
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Yoshito Zamami
- Department of Clinical Pharmacology and Pharmacy, Okayama University, Okayama, Japan
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
5
|
Neumann J, Hesse C, Hofmann B, Gergs U. Mosapride stimulates human 5-HT 4-serotonin receptors in the heart. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6705-6720. [PMID: 38498060 PMCID: PMC11422274 DOI: 10.1007/s00210-024-03047-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024]
Abstract
Mosapride (4-amino-5-chloro-2-ethoxy-N-[[4-[(4-fluorophenyl) methyl]-2-morpholinyl]-methyl] benzamide) is a potent agonist at gastrointestinal 5-HT4 receptors. Mosapride is an approved drug to treat several gastric diseases. We tested the hypothesis that mosapride also stimulates 5-HT4 receptors in the heart. Mosapride increased the force of contraction and beating rate in isolated atrial preparations from mice with cardiac overexpression of human 5-HT4-serotonin receptors (5-HT4-TG). However, it is inactive in wild-type mouse hearts (WT). Mosapride was less effective and potent than serotonin in raising the force of contraction or the beating rate in 5-HT4-TG. Only in the presence of cilostamide (1 μM), a phosphodiesterase III inhibitor, mosapride, and its primary metabolite time dependently raised the force of contraction under isometric conditions in isolated paced human right atrial preparations (HAP, obtained during open heart surgery). In HAP, mosapride (10 μM) reduced serotonin-induced increases in the force of contraction. Mosapride (10 µM) shifted the concentration-response curves to serotonin in HAP to the right. These data suggest that mosapride is a partial agonist at 5-HT4-serotonin receptors in HAP.
Collapse
Affiliation(s)
- Joachim Neumann
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany.
| | - Christin Hesse
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany
| | - Britt Hofmann
- Department of Cardiac Surgery, Mid-German Heart Center, University Hospital Halle, Ernst-Grube-Straße 40, 06097, Halle (Saale), Germany
| | - Ulrich Gergs
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Straße 4, 06112, Halle (Saale), Germany
| |
Collapse
|
6
|
Cocco M, Carnovale C, Clementi E, Barbieri MA, Battini V, Sessa M. Exploring the impact of co-exposure timing on drug-drug interactions in signal detection through spontaneous reporting system databases: a scoping review. Expert Rev Clin Pharmacol 2024; 17:441-453. [PMID: 38619027 DOI: 10.1080/17512433.2024.2343875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/12/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Drug-drug interactions (DDIs) are defined as the pharmacological effects produced by the concomitant administration of two or more drugs. To minimize false positive signals and ensure their validity when analyzing Spontaneous Reporting System (SRS) databases, it has been suggested to incorporate key pharmacological principles, such as temporal plausibility. AREAS COVERED The scoping review of the literature was completed using MEDLINE from inception to March 2023. Included studies had to provide detailed methods for identifying DDIs in SRS databases. Any methodological approach and adverse event were accepted. Descriptive analyzes were excluded as we focused on automatic signal detection methods. The result is an overview of all the available methods for DDI signal detection in SRS databases, with a specific focus on the evaluation of the co-exposure time of the interacting drugs. It is worth noting that only a limited number of studies (n = 3) have attempted to address the issue of overlapping drug administration times. EXPERT OPINION Current guidelines for signal validation focus on factors like the number of reports and temporal association, but they lack guidance on addressing overlapping drug administration times, highlighting a need for further research and method development.
Collapse
Affiliation(s)
- Marianna Cocco
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Carla Carnovale
- Pharmacovigilance & Clinical Research, International Centre for Pesticides and Health Risk Prevention, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università Degli Studi di Milano, Milan, Italy
| | - Emilio Clementi
- Pharmacovigilance & Clinical Research, International Centre for Pesticides and Health Risk Prevention, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università Degli Studi di Milano, Milan, Italy
- Scientific Institute, IRCCS E. Medea, Bosisio Parini, LC, Italy
| | - Maria Antonietta Barbieri
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Vera Battini
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Maurizio Sessa
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Yanagida S, Kawagishi H, Kanda Y. [Cardiotoxicity risk assessment of anti-cancer drugs and future perspectives]. Nihon Yakurigaku Zasshi 2024; 159:83-89. [PMID: 38432924 DOI: 10.1254/fpj.23094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Cardiotoxicity is a serious adverse effect of anti-cancer drugs. Anti-cancer drug-induced cardiotoxicity are arrhythmia, cardiac contractile dysfunction, coronary artery disease, and hypertension, which affect to the quality of life in patients with cancer. In particular, cardiac contractile dysfunction is a life-threatening symptom leading to heart failure, suggesting that it is very important to predict the risk of developing the contractile dysfunction by anti-cancer drugs. Recently, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to assess the risk of drug-induced arrhythmias. This prompts us to evaluate other cardiotoxic effects such as contractility dysfunction and structural toxicity with hiPSC-CMs. Since anti-cancer drug-induced contractility dysfunction are considered to be induced by chronic exposure, we have developed a method to assess chronic contractility dysfunction by imaging analysis of hiPSC-CMs. BMS-986094, which failed in clinical trials due to the occurrence of heart failure, was used as a positive compound. We found that chronic exposure to BMS-986094 decreased the contraction and relaxation velocity in hiPSC-CMs. Doxorubicin was observed to decrease cytotoxicity and both contraction and relaxation velocities in hiPSC-CMs. We are currently further evaluating other anti-cancer drugs with different mode-of-actions using hiPSC-CMs and assess the predictivity and utility of contractile assessment using hiPSC-CMs by comparing with real-world data. Here, we introduce our novel method to assess the chronic contractility of hiPSC-CMs by imaging analysis and discuss the future perspectives for assessing the anti-cancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences
| | | | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences
| |
Collapse
|
8
|
Hamano H, Zamami Y, Ushio S, Niimura T, Goda M, Izawa-Ishizawa Y, Ishizawa K. [Development of Preventive Methods for Drug-induced Cardiotoxicity Using a Large-scale Medical Information Database]. YAKUGAKU ZASSHI 2024; 144:257-264. [PMID: 38432934 DOI: 10.1248/yakushi.23-00164-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Cancer therapies have evolved considerably thereby substantially improving the survival of patients with cancer. However, cardiotoxicity, such as myocarditis and heart failure, induced by anticancer drugs, including immune checkpoint inhibitor(ICI)s and doxorubicin, present serious challenges. Numerous observations have indicated increased risks of cardiotoxicity- and cancer-related mortality in patients with drug-induced cardiotoxicity. Therefore, the prevention and management of drug-induced cardiotoxicity should be prioritized to enable sustainable long-term treatment while preserving patients' quality of life. Recently, medical research has been primarily focused on elucidation of therapeutic benefits and adverse events using medical big data, including worldwide databases of adverse events. The aim of the present study was to establish prevention strategies for drug-induced cardiotoxicity and advance data analytics. A data-driven approach was adopted to comprehensively analyze patient data and drug-induced cardiotoxicity. These data analytics revealed numerous risk factors, leading to the development of drugs that mitigate these factors. Furthermore, many unknown adverse events with molecularly targeted drugs were brought to light. Consequently, the importance of managing adverse events, guided by insights from data science, is predicted to increase. In this symposium review, we introduce our research exemplifying pharmaceutical studies utilizing medical big data. In particular, we discuss in detail the risk factors associated with myocarditis induced by immune checkpoint inhibitors along with prophylactic agents to mitigate doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | - Takahiro Niimura
- Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital
| | - Mitsuhiro Goda
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences
| | - Yuki Izawa-Ishizawa
- Department of Pharmacology, Tokushima University Graduate School of Biomedical Sciences
| | - Keisuke Ishizawa
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences
| |
Collapse
|
9
|
Zhou L, Yang J, Xiao M, Shan H, Liu M, Lu Y, Zou Y, Wu B. Severe cutaneous adverse reactions due to antibiotics therapy: a pharmacovigilance analysis of FDA adverse event reporting system events. Expert Opin Drug Saf 2023:1-8. [PMID: 37937960 DOI: 10.1080/14740338.2023.2278685] [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: 06/06/2023] [Accepted: 09/15/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND The aim of this study was to monitor, identify and evaluate severe cutaneous adverse reactions (SCAR) induced by antibiotics in patients. METHODS Disproportionality algorithms were performed in data mining to screen suspected SCAR after using nine categories of antibiotics based on the FDA's Adverse Event Reporting System (FAERS) from January 2004 to December 2022. The drug information and demographic characteristics of antibiotic-associated SCAR were also investigated. RESULTS The FAERS database received 12,212 antibiotic-associated SCAR cases. Approximately half of the SCAR patients were females, the majority of them were adults aged 18-65 years (48.35%), and 47.68% of SCAR patients required hospitalization. The highest SCAR signals RORs (95% CI) for antibiotics were: sulfonamides 23.30 (22.05-24.62), glycopeptides 21.27 (20.26-22.33), penicillins 16.00 (15.44-16.59), carbapenems 10.46 (9.57-11.44), and cephalosporins 13.27 (12.57-14.00). Cefotaxime, sulfamethoxazole/trimethoprim, cefixime, vancomycin, piperacillin, ceftriaxone, amoxicillin, and meropenem had stronger associations with the SCAR than the other antibiotics. However, sulfonamides-associated SCAR cases had the lowest fatality rate (6.23%), penicillin-associated SCAR cases had the highest hospitalization rate (54.16%), and carbapenem-associated SCAR cases seemingly resulted in the highest risk of death (19.03%). CONCLUSION Data mining of FAERS identified 30 antibiotic-associated SCAR signals, and provided a referable evidence of the occurrence, characteristics and prognosis of antibiotic-related SCARs.
Collapse
Affiliation(s)
- Lu Zhou
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Jing Yang
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Min Xiao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - HuiFang Shan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - MaoZhu Liu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yun Lu
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Ya Zou
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Bin Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Clayton ZS, Rossman MJ, Mahoney SA, Venkatasubramanian R, Maurer GS, Hutton DA, VanDongen NS, Greenberg NT, Longtine AG, Ludwig KR, Brunt VE, LaRocca TJ, Campisi J, Melov S, Seals DR. Cellular Senescence Contributes to Large Elastic Artery Stiffening and Endothelial Dysfunction With Aging: Amelioration With Senolytic Treatment. Hypertension 2023; 80:2072-2087. [PMID: 37593877 PMCID: PMC10530538 DOI: 10.1161/hypertensionaha.123.21392] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Here, we assessed the role of cellular senescence and the senescence associated secretory phenotype (SASP) in age-related aortic stiffening and endothelial dysfunction. METHODS We studied young (6-8 mo) and old (27-29 mo) p16-3MR mice, which allows for genetic-based clearance of senescent cells with ganciclovir (GCV). We also treated old C57BL/6N mice with the senolytic ABT-263. RESULTS In old mice, GCV reduced aortic stiffness assessed by aortic pulse wave velocity (PWV; 477±10 vs. 382±7 cm/s, P<0.05) to young levels (old-GCV vs. young-vehicle, P=0.35); ABT-263 also reduced aortic PWV in old mice (446±9 to 356±11 cm/s, P<0.05). Aortic adventitial collagen was reduced by GCV (P<0.05) and ABT-263 (P=0.12) in old mice. To show an effect of the circulating SASP, we demonstrated that plasma exposure from Old-vehicle p16-3MR mice, but not from Old-GCV mice, induced aortic stiffening assessed ex vivo (elastic modulus; P<0.05). Plasma proteomics implicated glycolysis in circulating SASP-mediated aortic stiffening. In old p16-3MR mice, GCV increased endothelial function assessed via peak carotid artery endothelium-dependent dilation (EDD; Old-GCV, 94±1% vs. Old-vehicle, 84±2%, P<0.05) to young levels (Old-GCV vs. young-vehicle, P=0.98), and EDD was higher in old C57BL/6N mice treated with ABT-263 vs. vehicle (96±1% vs. 82±3%, P<0.05). Improvements in endothelial function were mediated by increased nitric oxide (NO) bioavailability (P<0.05) and reduced oxidative stress (P<0.05). Circulating SASP factors related to NO signaling were associated with greater NO-mediated EDD following senescent cell clearance. CONCLUSIONS Cellular senescence and the SASP contribute to vascular aging and senolytics hold promise for improving age-related vascular function.
Collapse
Affiliation(s)
- Zachary S. Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Matthew J. Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Sophia A. Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | | | - Grace S. Maurer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - David A. Hutton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | | | - Nathan T. Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Abigail G. Longtine
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Katelyn R. Ludwig
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Vienna E. Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Thomas J. LaRocca
- Department of Health & Exercise Science, Colorado State University, Fort Collins, CO
- Center for Healthy Aging, Colorado State University, Fort Collins, CO
| | - Judith Campisi
- The Buck Institute for Research on Aging, Novato, CA
- Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Simon Melov
- The Buck Institute for Research on Aging, Novato, CA
| | - Douglas R. Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| |
Collapse
|
11
|
Liu P, Wu J, Yu X, Guo L, Zhao L, Ban T, Huang Y. Metabolomics and Network Analyses Reveal Phenylalanine and Tyrosine as Signatures of Anthracycline-Induced Hepatotoxicity. Pharmaceuticals (Basel) 2023; 16:797. [PMID: 37375744 DOI: 10.3390/ph16060797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/09/2023] [Accepted: 05/20/2023] [Indexed: 06/29/2023] Open
Abstract
The chemotherapy drug doxorubicin (DOX) is an anthracycline with over 30% incidence of liver injury in breast cancer patients, yet the mechanism of its hepatotoxicity remains unclear. To identify potential biomarkers for anthracycline-induced hepatotoxicity (AIH), we generated clinically-relevant mouse and rat models administered low-dose, long-term DOX. These models exhibited significant liver damage but no decline in cardiac function. Through untargeted metabolic profiling of the liver, we identified 27 differential metabolites in a mouse model and 28 in a rat model. We then constructed a metabolite-metabolite network for each animal model and computationally identified several potential metabolic markers, with particular emphasis on aromatic amino acids, including phenylalanine, tyrosine, and tryptophan. We further performed targeted metabolomics analysis on DOX-treated 4T1 breast cancer mice for external validation. We found significant (p < 0.001) reductions in hepatic levels of phenylalanine and tyrosine (but not tryptophan) following DOX treatment, which were strongly correlated with serum aminotransferases (ALT and AST) levels. In summary, the results of our study present compelling evidence supporting the use of phenylalanine and tyrosine as metabolic signatures of AIH.
Collapse
Affiliation(s)
- Peipei Liu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Jing Wu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xinyue Yu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China
| | - Linling Guo
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ling Zhao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Tao Ban
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| | - Yin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|