1
|
Tian L, Mi N, Wang L, Huang C, Fu W, Bai M, Gao L, Ma H, Zhang C, Lu Y, Zhao J, Zhang X, Jiang N, Lin Y, Yue P, Xia B, He Q, Yuan J, Meng W. Regular use of paracetamol and risk of liver cancer: a prospective cohort study. BMC Cancer 2024; 24:33. [PMID: 38178090 PMCID: PMC10765829 DOI: 10.1186/s12885-023-11767-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Paracetamol induces hepatotoxicity and subsequent liver injury, which may increase the risk of liver cancer, but epidemiological evidence remains unclear. We conducted this study to evaluate the association between paracetamol use and the risk of liver cancer. METHODS This prospective study included 464,244 participants free of cancer diagnosis from the UK Biobank. Incident liver cancer was identified through linkage to cancer and death registries and the National Health Service Central Register using the International Classification of Diseases (ICD)-10 codes (C22). An overlap-weighted Cox proportional hazards model was utilized to calculate the hazard ratio (HR) and 95% confidence interval (CI) for the risk of liver cancer associated with paracetamol use. The number needed to harm (NNH) was calculated at 10 years of follow-up. RESULTS During a median of 12.6 years of follow-up, 627 cases of liver cancer were identified. Paracetamol users had a 28% higher risk of liver cancer than nonusers (HR 1.28, 95% CI 1.06-1.54). This association was robust in several sensitivity analyses and subgroup analyses, and the quantitative bias analysis indicated that the result remains sturdy to unmeasured confounding factors (E-value 1.88, lower 95% CI 1.31). The NNH was 1106.4 at the 10 years of follow-up. CONCLUSION The regular use of paracetamol was associated with a higher risk of liver cancer. Physicians should be cautious when prescribing paracetamol, and it is recommended to assess the potential risk of liver cancer to personalize the use of paracetamol.
Collapse
Affiliation(s)
- Liang Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Ningning Mi
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Leiqing Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Chongfei Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Wenkang Fu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Mingzhen Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Long Gao
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Haidong Ma
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Chao Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Yawen Lu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jinyu Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Xianzhuo Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Ningzu Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Yanyan Lin
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Ping Yue
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Bin Xia
- Clinical Research Center, Big Data Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Qiangsheng He
- Clinical Research Center, Big Data Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China.
| | - Jinqiu Yuan
- Clinical Research Center, Big Data Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China.
| | - Wenbo Meng
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
2
|
Battista C, Shoda LKM, Watkins PB, Groettrup-Wolfers E, Rottmann A, Raschke M, Generaux GT. Quantitative Systems Toxicology Identifies Independent Mechanisms for Hepatotoxicity and Bilirubin Elevations Due to AKR1C3 Inhibitor BAY1128688. Clin Pharmacol Ther 2023; 114:1023-1032. [PMID: 37501650 DOI: 10.1002/cpt.3010] [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: 05/17/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
BAY1128688 is a selective inhibitor of AKR1C3, investigated recently in a trial that was prematurely terminated due to drug-induced liver injury. These unexpected observations prompted use of the quantitative systems toxicology model, DILIsym, to determine possible mechanisms of hepatotoxicity. Using mechanistic in vitro toxicity data as well as clinical exposure data, DILIsym predicted the potential for BAY1128688 to cause liver toxicity (elevations in serum alanine aminotransferase (ALT)) and elevations in serum bilirubin. Initial simulations overpredicted hepatotoxicity and bilirubin elevations, so the BAY1128688 representation within DILIsym underwent optimization. The liver partition coefficient Kp was altered to align simulated bilirubin elevations with those observed clinically. Altering the mode of bile acid canalicular and basolateral efflux inhibition was necessary to accurately predict ALT elevations. Optimization results support that bilirubin elevations observed early during treatment are due to altered bilirubin metabolism and transporter inhibition, which is independent of liver injury. The modeling further supports that on-treatment ALT elevations result from inhibition of bile acid transporters, particularly the bile salt excretory pump, leading to accumulation of toxic bile acids. The predicted dose-dependent intrinsic hepatotoxicity may increase patient susceptibility to an adaptive immune response, accounting for ALT elevations observed after completion of treatment. These BAY1128688 simulations provide insight into the mechanisms behind hepatotoxicity and bilirubin elevations and may inform the potential risk posed by future compounds.
Collapse
Affiliation(s)
- Christina Battista
- DILIsym Services division, Simulations Plus, Inc., Durham, North Carolina, USA
| | - Lisl K M Shoda
- DILIsym Services division, Simulations Plus, Inc., Durham, North Carolina, USA
| | - Paul B Watkins
- Eshelman School of Pharmacy, Institute for Drug Safety Sciences, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Antje Rottmann
- Pharmaceuticals Division, Research & Early Development, Bayer AG, Berlin, Germany
| | - Marian Raschke
- Pharmaceuticals Division, Research & Early Development, Bayer AG, Berlin, Germany
| | | |
Collapse
|
3
|
Zhou J, De Jonghe S, Codd EE, Weiner S, Gallacher D, Stahle P, Kelley MF, Kuffner EK, Flores CM, Eichenbaum GE. Preclinical safety assessment of JNJ-10450232 (NTM-006), a structural analog of acetaminophen, that does not cause hepatotoxicity at supratherapeutic doses. Regul Toxicol Pharmacol 2023:105334. [PMID: 36608923 DOI: 10.1016/j.yrtph.2023.105334] [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: 08/01/2022] [Revised: 12/05/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
JNJ-10450232 (NTM-006) is a new molecular entity that is structurally related to acetaminophen. A comprehensive non-clinical safety program was conducted to support first-in-human and clinical efficacy studies based on preclinical data suggesting that the compound has comparable or enhanced antinociceptive and antipyretic efficacy without causing hepatotoxicity at supratherapeutic doses. No hepatic toxicity was noted in a mouse model sensitive to acetaminophen hepatotoxicity or in rats, dogs, and non-human primates in 28-day repeat dose toxicity studies at and above doses/exposures at which acetaminophen is known to cause hepatotoxicity. In the 28-day toxicity studies, all treatment-related findings were monitorable and reversible. Methemoglobinemia, which was observed in dogs and to a lesser extent in rats, is also observed with acetaminophen. This finding is considered not relevant to humans due to species differences in metabolism. Thyroid hypertrophy and hyperplasia were also observed in dogs and were shown to be a consequence of a species-specific UGT induction also demonstrated with increased thyroid hormone metabolism. Indirect bilirubin elevation was observed in rats as a result of UGT1A1 Inhibition. JNJ-10450232 (NTM-006) had no toxicologically relevant findings in safety pharmacology or genotoxicity studies. Together, these data supported progressing into safety and efficacy studies in humans.
Collapse
Affiliation(s)
- Junguo Zhou
- Janssen Research & Development, LLC, Raritan, NJ, USA
| | - Sandra De Jonghe
- Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Ellen E Codd
- Janssen Research & Development, LLC, Raritan, NJ, USA
| | - Sandy Weiner
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - David Gallacher
- Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Paul Stahle
- Janssen Research & Development, LLC, Spring House, PA, USA
| | | | - Edwin K Kuffner
- Johnson & Johnson Consumer Companies, Fort Washington, PA, USA.
| | | | - Gary E Eichenbaum
- Office of the Chief Medical Officer, Johnson & Johnson, New Brunswick, NJ, USA
| |
Collapse
|
4
|
Unraveling the effect of intra- and intercellular processes on acetaminophen-induced liver injury. NPJ Syst Biol Appl 2022; 8:27. [PMID: 35933513 PMCID: PMC9357019 DOI: 10.1038/s41540-022-00238-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/20/2022] [Indexed: 11/09/2022] Open
Abstract
In high dosages, acetaminophen (APAP) can cause severe liver damage, but susceptibility to liver failure varies across individuals and is influenced by factors such as health status. Because APAP-induced liver injury and recovery is regulated by an intricate system of intra- and extracellular molecular signaling, we here aim to quantify the importance of specific modules in determining the outcome after an APAP insult and of potential targets for therapies that mitigate adversity. For this purpose, we integrated hepatocellular acetaminophen metabolism, DNA damage response induction and cell fate into a multiscale mechanistic liver lobule model which involves various cell types, such as hepatocytes, residential Kupffer cells and macrophages. Our model simulations show that zonal differences in metabolism and detoxification efficiency are essential determinants of necrotic damage. Moreover, the extent of senescence, which is regulated by intracellular processes and triggered by extracellular signaling, influences the potential to recover. In silico therapies at early and late time points after APAP insult indicated that prevention of necrotic damage is most beneficial for recovery, whereas interference with regulation of senescence promotes regeneration in a less pronounced way.
Collapse
|
5
|
Quantitative Systems Toxicology and Drug Development: The DILIsym Experience. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2486:181-196. [PMID: 35437723 DOI: 10.1007/978-1-0716-2265-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
DILIsym® is a Quantitative Systems Toxicology (QST) model that has been developed over the last decade by a public-private partnership to predict the liver safety liability in new drug candidates. DILIsym integrates the quantitative abilities of parent and relevant metabolites to cause oxidative stress, mitochondrial dysfunction, and alter bile acid homeostasis. Like the prediction of drug-drug interactions, the data entered into DILIsym are assessed in the laboratory in human experimental systems, and combined with estimates of liver exposure to predict the outcome. DILIsym is now frequently used in decision-making within the pharmaceutical industry and its modeling results are increasingly included in regulatory communications and NDA submissions. DILIsym can be used to identify dominant mechanisms underlying liver toxicity and this information is increasingly being used to identify patient-specific risk factors, including certain disease states. DILIsym is also increasingly used to optimize the interpretation of liver injury biomarkers. DILIsym provides an example of how QST modeling can help speed the delivery of safer new drugs to the patients who need them.
Collapse
|
6
|
Jaeschke H. Comments on "DNA-binding activities of compounds acting as enzyme inhibitors, ion channel blockers and receptor binders.". Chem Biol Interact 2022; 351:109761. [PMID: 34843691 PMCID: PMC8989075 DOI: 10.1016/j.cbi.2021.109761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 01/07/2023]
Abstract
I read with interest the article "DNA-binding activities of compounds acting as enzyme inhibitors, ion channel blockers and receptor binders" recently published in Chemico-Biological Interactions. The authors suggested that acetaminophen, one of the most used drugs worldwide, alkylates DNA at therapeutic doses and is genotoxic. Given the implications of this statements for public health, it is important for the reader to hear a different perspective that is based on the entire literature on this subject. Everything considered, there is no credible evidence that acetaminophen is a genotoxic hazard or a carcinogen at therapeutic doses.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- University of Kansas Medical Center, Department of Pharmacology, Toxicology & Therapeutics, Kansas City, KS, USA.
| |
Collapse
|
7
|
Jaeschke H, Adelusi OB, Akakpo JY, Nguyen NT, Sanchez-Guerrero G, Umbaugh DS, Ding WX, Ramachandran A. Recommendations for the use of the acetaminophen hepatotoxicity model for mechanistic studies and how to avoid common pitfalls. Acta Pharm Sin B 2021; 11:3740-3755. [PMID: 35024303 PMCID: PMC8727921 DOI: 10.1016/j.apsb.2021.09.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/22/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen (APAP) is a widely used analgesic and antipyretic drug, which is safe at therapeutic doses but can cause severe liver injury and even liver failure after overdoses. The mouse model of APAP hepatotoxicity recapitulates closely the human pathophysiology. As a result, this clinically relevant model is frequently used to study mechanisms of drug-induced liver injury and even more so to test potential therapeutic interventions. However, the complexity of the model requires a thorough understanding of the pathophysiology to obtain valid results and mechanistic information that is translatable to the clinic. However, many studies using this model are flawed, which jeopardizes the scientific and clinical relevance. The purpose of this review is to provide a framework of the model where mechanistically sound and clinically relevant data can be obtained. The discussion provides insight into the injury mechanisms and how to study it including the critical roles of drug metabolism, mitochondrial dysfunction, necrotic cell death, autophagy and the sterile inflammatory response. In addition, the most frequently made mistakes when using this model are discussed. Thus, considering these recommendations when studying APAP hepatotoxicity will facilitate the discovery of more clinically relevant interventions.
Collapse
Key Words
- AIF, apoptosis-inducing factor
- AMPK, AMP-activated protein kinase
- APAP, acetaminophen
- ARE, antioxidant response element
- ATG, autophagy-related genes
- Acetaminophen hepatotoxicity
- Apoptosis
- Autophagy
- BSO, buthionine sulfoximine
- CAD, caspase-activated DNase
- CYP, cytochrome P450 enzymes
- DAMPs, damage-associated molecular patterns
- DMSO, dimethylsulfoxide
- Drug metabolism
- EndoG, endonuclease G
- FSP1, ferroptosis suppressing protein 1
- Ferroptosis
- GPX4, glutathione peroxidase 4
- GSH, glutathione
- GSSG, glutathione disulfide
- Gclc, glutamate–cysteine ligase catalytic subunit
- Gclm, glutamate–cysteine ligase modifier subunit
- HMGB1, high mobility group box protein 1
- HNE, 4-hydroxynonenal
- Innate immunity
- JNK, c-jun N-terminal kinase
- KEAP1, Kelch-like ECH-associated protein 1
- LAMP, lysosomal-associated membrane protein
- LC3, light chain 3
- LOOH, lipid hydroperoxides
- LPO, lipid peroxidation
- MAP kinase, mitogen activated protein kinase
- MCP-1, monocyte chemoattractant protein-1
- MDA, malondialdehyde
- MPT, mitochondrial permeability transition
- Mitochondria
- MnSOD, manganese superoxide dismutase
- NAC, N-acetylcysteine
- NAPQI, N-acetyl-p-benzoquinone imine
- NF-κB, nuclear factor κB
- NQO1, NAD(P)H:quinone oxidoreductase 1
- NRF2
- NRF2, nuclear factor erythroid 2-related factor 2
- PUFAs, polyunsaturated fatty acids
- ROS, reactive oxygen species
- SMAC/DIABLO, second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI
- TLR, toll like receptor
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- UGT, UDP-glucuronosyltransferases
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Olamide B. Adelusi
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jephte Y. Akakpo
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Nga T. Nguyen
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Giselle Sanchez-Guerrero
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - David S. Umbaugh
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Kirkland D, Kovochich M, More SL, Murray FJ, Monnot AD, Miller JV, Jaeschke H, Jacobson-Kram D, Deore M, Pitchaiyan SK, Unice K, Eichenbaum G. A comprehensive weight of evidence assessment of published acetaminophen genotoxicity data: Implications for its carcinogenic hazard potential. Regul Toxicol Pharmacol 2021; 122:104892. [PMID: 33592196 DOI: 10.1016/j.yrtph.2021.104892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 11/17/2022]
Abstract
In 2019, the California Office of Environmental Health Hazard Assessment initiated a review of the carcinogenic hazard potential of acetaminophen, including an assessment of its genotoxicity. The objective of this analysis was to inform this review process with a weight-of-evidence assessment of more than 65 acetaminophen genetic toxicology studies that are of widely varying quality and conformance to accepted standards and relevance to humans. In these studies, acetaminophen showed no evidence of induction of point or gene mutations in bacterial and mammalian cell systems or in in vivo studies. In reliable, well-controlled test systems, clastogenic effects were only observed in unstable, p53-deficient cell systems or at toxic and/or excessively high concentrations that adversely affect cellular processes (e.g., mitochondrial respiration) and cause cytotoxicity. Across the studies, there was no clear evidence that acetaminophen causes DNA damage in the absence of toxicity. In well-controlled clinical studies, there was no meaningful evidence of chromosomal damage. Based on this weight-of-evidence assessment, acetaminophen overwhelmingly produces negative results (i.e., is not a genotoxic hazard) in reliable, robust high-weight studies. Its mode of action produces cytotoxic effects before it can induce the stable, genetic damage that would be indicative of a genotoxic or carcinogenic hazard.
Collapse
|
9
|
Jaeschke H, Murray FJ, Monnot AD, Jacobson-Kram D, Cohen SM, Hardisty JF, Atillasoy E, Hermanowski-Vosatka A, Kuffner E, Wikoff D, Chappell GA, Bandara SB, Deore M, Pitchaiyan SK, Eichenbaum G. Assessment of the biochemical pathways for acetaminophen toxicity: Implications for its carcinogenic hazard potential. Regul Toxicol Pharmacol 2021; 120:104859. [PMID: 33388367 DOI: 10.1016/j.yrtph.2020.104859] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
In 2019 California's Office of Environmental Health Hazard Assessment (OEHHA) initiated a review of the carcinogenic hazard potential of acetaminophen. In parallel with this review, herein we evaluated the mechanistic data related to the steps and timing of cellular events following therapeutic recommended (≤4 g/day) and higher doses of acetaminophen that may cause hepatotoxicity to evaluate whether these changes indicate that acetaminophen is a carcinogenic hazard. At therapeutic recommended doses, acetaminophen forms limited amounts of N-acetyl-p-benzoquinone-imine (NAPQI) without adverse cellular effects. Following overdoses of acetaminophen, there is potential for more extensive formation of NAPQI and depletion of glutathione, which may result in mitochondrial dysfunction and DNA damage, but only at doses that result in cell death - thus making it implausible for acetaminophen to induce the kind of stable, genetic damage in the nucleus indicative of a genotoxic or carcinogenic hazard in humans. The collective data demonstrate a lack of a plausible mechanism related to carcinogenicity and are consistent with rodent cancer bioassays, epidemiological results reviewed in companion manuscripts in this issue, as well as conclusions of multiple international health authorities.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- University of Kansas Medical Center, Department of Pharmacology, Toxicology & Therapeutics, Kansas City, KS, USA
| | | | | | | | - Samuel M Cohen
- University of Nebraska Medical Center, Havlik-Wall Professor of Oncology, Department of Pathology and Microbiology, Omaha, NE, USA
| | - Jerry F Hardisty
- Experimental Pathology Laboratories, Inc., Research Triangle Park, NC, USA
| | | | | | - Edwin Kuffner
- Johnson & Johnson Consumer Health, Fort Washington, PA, USA
| | | | | | | | | | | | | |
Collapse
|