1.1
The cancer mode of action for 1,4-DX plays a critical role in estimating cancer potency and setting threshold levels for this chemical. Please indicate your confidence rating on the weight of evidence supporting/refuting the potential mode of action for the carcinogenic effects of 1,4-DX in the rodent liver (-5=strong refuting evidence, 0=lack of or equivocal evidence, 5=strong supporting evidence)
Results
-5 | -4 | -3 | -2 | -1 | 0 | 1 | 2 | 3 | 4 | 5 | Total | |
---|---|---|---|---|---|---|---|---|---|---|---|---|
Direct genotoxicity | 66.67% 4 | 0.00% 0 | 16.67% 1 | 0.00% 0 | 0.00% 0 | 16.67% 1 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 6 |
Indirect genotoxicity | 16.67% 1 | 0.00% 0 | 16.67% 1 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 16.67% 1 | 33.33% 2 | 16.67% 1 | 6 |
Proliferative regeneration induced by cytotoxicity | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 16.67% 1 | 0.00% 0 | 0.00% 0 | 33.33% 2 | 16.67% 1 | 33.33% 2 | 6 |
Oxidative stress | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 16.67% 1 | 16.67% 1 | 0.00% 0 | 33.33% 2 | 0.00% 0 | 16.67% 1 | 16.67% 1 | 6 |
Activation of nuclear receptors and associated transcription factors (e.g., CAR/PXR) | 0.00% 0 | 33.33% 2 | 0.00% 0 | 0.00% 0 | 16.67% 1 | 0.00% 0 | 33.33% 2 | 0.00% 0 | 16.67% 1 | 0.00% 0 | 0.00% 0 | 6 |
Other (please explain) | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 0.00% 0 | 25.00% 1 | 25.00% 1 | 25.00% 1 | 0.00% 0 | 25.00% 1 | 4 |
Answer Explanations
- Expert 5
-5 -4 -3 -2 -1 0 1 2 3 4 5 Direct genotoxicity 1 0 0 0 0 0 0 0 0 0 0 Indirect genotoxicity 1 0 0 0 0 0 0 0 0 0 0 Proliferative regeneration induced by cytotoxicity 0 0 0 0 0 0 0 0 0 1 0 Oxidative stress 0 0 0 0 0 1 0 0 0 0 0 Activation of nuclear receptors and associated transcription factors (e.g., CAR/PXR) 0 0 0 0 0 0 1 0 0 0 0 Other (please explain) Oxidative stress
The issue of oxidative stress induction as a mode of action is compounded by the fact that toxicity (necrosis) and resulting inflammation results in oxdiative stress and damge. Oxidative damage and stress should be considered a part of the cytotoxicity pathway. Oxidative damage could also modify the response seen in the surviving (non necrotic) proliferating cells , thus enhancing the response. I am not convinced that oxidative stress /damage is a key event. Even in the case of metal overload (Fe and Cu) in the liver which produces oxidatve stress/damage, the liver tumor induction appears to be through cytotoxicity.
CAR/PXR
The metabolism of 1,4 DX appears to be through CYP2b and/or 2E. Therfore the participation of CAR in the CYP 2b pathway is expected. While many of the key events linked to a cytotoxic MOA overlap with the CAR (another nuclear receptor MOA), this is expected since the multistep liver cancer process is similar subsequent to the initial key event. While the data are not strong for a CAR MOA a further review of the availble results and possibly an additional experiemnt specifically address CAR activation might be warranted. - Expert 6
-5 -4 -3 -2 -1 0 1 2 3 4 5 Direct genotoxicity 1 0 0 0 0 0 0 0 0 0 0 Indirect genotoxicity 0 0 0 0 0 0 0 0 0 1 0 Proliferative regeneration induced by cytotoxicity 0 0 0 0 0 0 0 0 0 0 1 Oxidative stress 0 0 0 0 0 0 0 1 0 0 0 Activation of nuclear receptors and associated transcription factors (e.g., CAR/PXR) 0 0 0 0 0 0 1 0 0 0 0 Other (please explain) 0 0 0 0 0 0 0 1 0 0 0 Mo et al. (2022) show data suggesting an activation of TRPV1, vanilloid receptor by 1,4-DX. This may open a link to calcium influx, hypertrophy, necrosis/apoptosis, cell death. One alternative option may be 'oxidative stress'?? - Expert 4
-5 -4 -3 -2 -1 0 1 2 3 4 5 Direct genotoxicity 0 0 1 0 0 0 0 0 0 0 0 Indirect genotoxicity 0 0 0 0 0 0 0 0 0 1 0 Proliferative regeneration induced by cytotoxicity 0 0 0 0 0 0 0 0 1 0 0 Oxidative stress 0 0 0 0 0 0 0 1 0 0 0 Activation of nuclear receptors and associated transcription factors (e.g., CAR/PXR) 0 0 0 0 1 0 0 0 0 0 0 Other (please explain) 0 0 0 0 0 0 0 0 1 0 0 Based on current data and particularly, recent studies relevant to consideration of early key events (e.g., Chen et al., 2022; Lafranconi et al., 2023), the most likely mode of action for the carcinogenic effects of 1,4-DX in the rodent liver appears to be indirect genotoxicity, mediated through the induction of cyp2E1 metabolism, oxidative stress, DNA damage, hepatotoxicity, sustained proliferation and tumours. This MOA is consistent with key events described in two existing AOPs, namely OECD 220 (https://aopwiki.org/aops/2200) and Cho et al., 2022 (DOI: 10.1002/em.22479). This hypothesized MOA subsumes several of the options noted in this question (oxidative stress and proliferative regeneration) as key events; the confidence ratings should be considered in this context. (i.e., confidence in an indirect genotoxic mode of action involving early oxidative stress and later proliferative regeneration, the basis for the response to "other"). Responses for oxidative stress and proliferative regeneration are based on their likelihood of being key events in the hypothesized mode of action.
With respect to the distinction of "direct" and "indirect" genotoxicity, U.S. EPA (2007) indicates that: “Key data for a mutagenic mode of action may be evidence that the carcinogen or a metabolite is DNA reactive and/or has the ability to bind DNA. Also, such carcinogens usually produce positive effects in multiple test systems for different genetic endpoints, particularly gene mutations and structural chromosome aberrations, and in tests performed in vivo which generally are supported by positive tests in vitro.” (https://archive.epa.gov/osa/mmoaframework/web/html/index.html). There is no evidence that 1,4-DX is reactive with DNA nor that it is metabolized to reactive electrophiles. Based on a relatively robust dataset in both in vitro and in vivo assays, 1,2-DX is not mutagenic and only weakly genotoxic (US EPA, 2013). The pattern of positive results in genotoxicity assays (namely DNA damage and single strand breaks, cell transformation and sister chromatid exchange in vitro and DNA damage, micronucleus formation and DNA synthesis in vivo generally at high concentrations) doesn't necessarily imply direct interaction (reactivity) with DNA; for example, the positive results for replicative DNA synthesis provide support for a non-genotoxic mode of action. The nature of reported DNA adducts following 1,4-DX exposure (i.e., 8-oxo-dG, a well-known oxidative damage biomarker but no DNA alkylation) is also supportive of an indirect mode of action involving oxidative stress. The additional observation by US EPA (2013) that responses in genotoxicity assays were positive generally only “in the presence of toxicity” is also consistent with genetic toxicity being a secondary consequence of earlier key events such as oxidative stress and/or hepatic damage/persistent regenerative proliferation.
The extent of the evidence for hypothesized modes of action is normally considered through formal analysis based on the modified Bradford Hill considerations of biological plausibility, essentiality of key events, empirical support (dose-response and temporal concordance), consistency and analogy [US EPA, 2005 (https://www.epa.gov/risk/guidelines-carcinogen-risk-assessment), WHO IPCS,2014,(https://www.who.int/publications/i/item/9789241563499), and OECD, 2023, (https://aopwiki.org/handbooks/4)]. These considerations are rank ordered as listed above with biological plausibility being most influential, followed by the essentiality of key events and so on.
The biological plausibility of the MOA hypothesized above is well documented in existing AOPs (https://aopwiki.org/aops/2200 and Cho et al., 2022 (DOI: 10.1002/em.22479) though the availability of data on the early key events associated with 1,4-DX is limited. There is, however, only limited identified direct evidence of the essentiality of the key events in the hypothesized MOA addressed through knockout models (e.g., for cyp2E1 induction) or reversibility studies (e.g., for hepatotoxic effects), as noted also by Health Canada (2018). There is also limited evidence of consistency of the hypothesized mode of action in the core reference material identified for this review based on, for example, knowledge of species and strain variations in cyp2E1 induction. There is, however, some evidence of analogy with the results of QSAR analysis predicting that 1,4-DX would not induce mutagenicity in Salmonella, chromosomal aberrations in Chinese hamster ovary cells, or unscheduled DNA synthesis in rat hepatocytes but would induce sister chromatid exchange in Chinese hamster ovary cells, micronuclei in the bone marrow of rats and cancer in rats and mice (Rosenkranz and Klopman, 1992 in Health Canada, 2018).
For empirical support, the review materials identified for this poll focused principally on dose-response concordance across hypothesized late key events (often with separate consideration of temporal aspects), a function in part of the limited available data on the nature of and dose-response for early key events. None included integrated analyses of dose-response and temporal concordance for key events for hypothesized MOAs based on benchmark doses (BMDs), though many of the relevant BMD/Cs for late key events were developed and considered separately for cancer and non-cancer effects by US EPA (2013). The most extensive and recent analyses are those of Dourson et al. (2017) and Lancanfroni et al. (2023). Though challenging due to varied study designs, Dourson et al. (2017) provided helpful graphical illustration of the comparative incidence of the hepatic precursor and neoplastic lesions in the key carcinogenesis bioassays and related subchronic studies for the late key events in the hypothesized MOA (namely, hepatic toxicity, regenerative proliferation and tumours) based on translated versions of the full study reports for the key Japanese carcinogenesis bioassays in rats and mice and a reread of the slides for hepatic effects in rats and mice in NCI studies (JRBC, 1990a, 1990b, NCI, 1978, McConnell reread of NCI, 1978). Lacanfroni et al. (2023) extended analyses to include results from more recent mechanistic studies including transcriptomic analyses relevant to earlier key events in the hypothesized mode of action (Lacanfroni et al., 2021; Chappell et al., 2021; Charkoftaki et al., 2021; Chen et al., 2022).
Based on the conducted analyses, with few exceptions, the observed patterns of dose-response and temporal concordance broadly support the late key events in the hypothesized mode of action above (e.g., Dourson et al., 2017, Health Canada, 2018; Lafranconi et al., 2023), with associated dose ranges in rats and mice, as follows (data from Dourson et al., 2017 with variations of Lancanfroni et al., 2023, noted; inconsistent information italicized ):
rats: metabolic saturation at 30 to 100 mg/kg, cellular swelling, hypertrophy and liver weight increases at 42 to 55 mg/kg-day, necrosis and/or inflammation at 94 to 219 mg/kg-day, increased DNA synthesis at 330 mg/kg-day, hyperplasia development at 55 to 330 mg/kg-day, basophilic and mixed cell (when measured) foci development at 55 to 389 mg/kg-day and adenomas and carcinomas at doses of 66 to 1015 mg/kg-day (> 30 mg/kg bw-day in Lancanfroni et al., 2023).
Mice: metabolic saturation at ~200 mg/kg – day, cellular swelling, hypertrophy and liver weight increase, in the range of 190 to 200 mg/kg-day, necrosis and/or inflammation in the same range, hyperplasia at doses as low as 380 mg/kg-day in one study but not the other and foci development at doses as low as 380 mg/kg-day in one study but not the other and adenomas and carcinomas at doses of 66 to 1015 mg/kg-day (200 – 400 mg/kg-day in Lancanfroni et al., 2023)
One of the important outliers noted in the italicized text above is the lack of observation of later hepatic effects (i.e., proliferation) prior to tumour development in mice in the Japanese carcinogenesis bioassay, which was influential in US EPA’s (2013) considering the data inadequate to support the hypothesized cytotoxic mode of action of 1,4-DX in liver. Dourson et al. (2017) and Lacanfroni et al. (2023) note, as well, the seeming inconsistency with the observation of non-neoplastic liver pathology in the 13-week drinking water study in mice conducted by the same investigators (Kano et al., 2008) as well as the increases in liver enzymes in the cancer bioassay, expected to be associated with adverse hepatic effects.
Recent studies incorporating genomic analyses prompted in part by these seemingly discrepant observations have provided additional data on likely early key events in the mode of induction of tumours by 1,4-DX (Lacanfroni et al., 2021, Chappell et al., 2021; Charkoftaki et al., 2021; Chen et al., 2022). This evolving evidence is consistent with the pattern of results for 1,4-DX in genotoxicity studies in vitro and in vivo. However, data relevant to consideration of empirical support for hypothesized early key events such as oxidative stress and DNA damage in this dataset are limited, due to the paucity of dose-response information available from the studies (with effects having been observed generally only at the highest doses associated also with cyp 2E1 induction).
For example, Lafranconi et al. (2021) evaluated both the dose-response and time course of hepatic events in female B6D2F1 mice treated with 20, 40, 200, 600, 2000 or 6000 ppm 1,4-DX in drinking water for 7, 28 or 90 days. Liver weight increases after 90 days of exposure were accompanied by evidence of increased pan-lobular hepatocellular proliferation as determined by increased BrdU incorporation at the top dose. Other than limited evidence of single-cell necrosis typical of apoptosis, there was no histological or biochemical evidence of cytotoxicity at any of the exposures used in this study. There was evidence of changes in genomic signaling only at 2000 ppm (337–391 mg/kg/d) and 6000 ppm (895–1063 mg/kg/d) from whole transcriptome analyses consistent with mitotic events (Chappell et al., 2021).
Based on histopathological and transcriptomic analyses of mouse liver, Charkoftaki et al. (2021) reported alterations in oxidative stress response accompanying DNA damage following 4 weeks exposure to high doses of 1,4-DX (5,000 ppm) in drinking water based on the enrichment of Nrf2and GSH mediated pathways in transcriptomic analyses. In follow up investigation, this group also provided some evidence of the essentiality of oxidative stress as a key event. Following exposure for one week (1000 ppm) or 90 days (5000 ppm) in wild type and GSH-deficient glutamate-cysteine ligase modifier subunit (Gclm)-null mice, at 90 days, mild liver cytoxocity, progressive CYP2E1 induction, oxidative stress (persistent NRF2 induction), oxidation of the GSH pool and accumulation of the lipid peroxidation by-product 4-hydroxynonenal, and elevations in oxidative DNA damage and DNA repair response were observed. These DX-elicited changes were exaggerated in GSH-deficient mice (Chen et al., 2022).
Collectively, then, available data support an indirectly genotoxic mode of action with early key events, as indicated above - i.e., induction of cyp 2E1, oxidative stress, DNA damage, hepatoxicity, sustained proliferation and tumours. Uncertainty is greatest for the early key events between the induction of cyp 2E1 and hepatotoxicity, for which there is limited evidence for 1,2-DX of empirical support (temporal and dose-response concordance) but some experimental support for the essentiality of oxidative stress. - Expert 2
-5 -4 -3 -2 -1 0 1 2 3 4 5 Direct genotoxicity 0 0 0 0 0 1 0 0 0 0 0 Indirect genotoxicity 0 0 0 0 0 0 0 0 0 0 1 Proliferative regeneration induced by cytotoxicity 0 0 0 0 0 0 0 0 0 0 1 Oxidative stress 0 0 0 0 0 0 0 0 0 0 1 Activation of nuclear receptors and associated transcription factors (e.g., CAR/PXR) 0 0 0 0 0 0 0 0 1 0 0 Other (please explain) 0 0 0 0 0 0 0 0 0 0 1 The onset of hepatic DNA damage is evidenced by DSBs with gH2AX accumulation and unscheduled DNA synthesis as elicited by the BrdU or 3H-thymidine tracers. These culminate in hepatic growth-arrest as demonstrated by the accumulation of highly polyploid hepatocytes with meganuclei. This is noted in Chen et al. 2022 (Fig. 1B showing H&E images - note panels on the extreme right). The polyploid cells contain extensive DSBs with gH2AX expression as noted in Chen et al. 2022 (Fig. 5C shows H2AX-positive polyploid cells in panels on the extreme right).
The designation of "proliferative regeneration" is incorrect because the studies of DNA synthesis in situ through the above markers are insufficient for establishing cell division. The onset of mitochondrial DNA damage leading to their inability for fission followed by the transfer to daughter cells is another mechanism for interruptions in cytokinesis leading to polyploidy, but this aspect has not been studied.
The likelihood is that when hepatocytes are damaged by excessive oxidative stress, this will restrict these from cycling, leading to the origin and proliferation of alternative cell populations substantially at risk for genetic mutations contributing to oncogenesis and liver tumors. Such a mechanism will be similar to other major causes of liver cancers, including alcohol-associated steatosis, metabolic dysfunction-associated steatohepatitis (MASH), chronic hepatitis B and C, hemochromatosis, hereditary tyrosinemia, etc.
The role of cell interactions in this process has not been developed. The bulk transcriptomics and other omics assays of whole liver lysates are insufficiently informative because of the dilution of key events in DNA damage response, cell cycling, and cell survival events by various cell types. - Expert 3
-5 -4 -3 -2 -1 0 1 2 3 4 5 Direct genotoxicity 1 0 0 0 0 0 0 0 0 0 0 Indirect genotoxicity 0 0 1 0 0 0 0 0 0 0 0 Proliferative regeneration induced by cytotoxicity 0 0 0 0 0 1 0 0 0 0 0 Oxidative stress 0 0 0 0 1 0 0 0 0 0 0 Activation of nuclear receptors and associated transcription factors (e.g., CAR/PXR) 0 1 0 0 0 0 0 0 0 0 0 Other (please explain) 0 0 0 0 0 0 1 0 0 0 0 There is little/no evidence of direct genotoxicity certainly in vitro and only high dose related activity (above tumorigenic doses); however, there may be at best a very contribution of oxidative stress. However, caution must be exercised in attributing oxidative stress to a genotoxic carcinogenic MoA in that relatively potent oxidative stress inducers (e.g., paraquat and diquat, oxidative stress caused by redox cycling) are not rodent carcinogens. However, recent transcriptomic data provides supports for a potentially strong mitogenic liver cell proliferation, and CAR/PXR responses are not consistent with responses associated with known NR liver carcinogens, e.g., phenobarbital. Metabolism/saturation of cyp2E1 appears very critical to outcome in that DX, and suggests a contributing (but not necessarily a genotoxic) role of oxidative stress associated with cyp2e1 metabolism/induction (supported by attentuation of liver toxicity and oxidative stress in cyp2e1 knockout mice).
Expert 6
05/11/2024 09:36The role of induction of CYP2E1 in the chain of events 'cytotoxicity-regererative proliferation-tumour' reads well but there are open questions. Wang et al. (2022) state, e.g., that 'Unexpectedly, subchronic exposure to high dose DX induced a trend towards an increase in oxidative DNA damage (8-OHdG levels) and suppression of DNA damage repair (γH2AX/H2AX ratio) in the liver of Cyp2e1KO mice'.
Expert 2
05/13/2024 12:14Might the metabolic basis of oxidative stress for 1,4-DX reside in mitochondrial dysfunction leading to damage in both mitochondrial and nuclear genomes to constitute the initiating event of growth arrest in damaged cells followed by continued somatic mutations in other cell populations that eventually will result in oncogenic cell clone? Despite the absence of a "direct" genotoxic mechanism for 1,4-DX, the mutagenic selection pressure and clonal origin of cancers are highly probable. This should require focused studies. Similarly, the clonal carcinogenesis aspect is likely to be amplified in the setting of ongoing metabolic or other injuries in the liver, which too should require further studies.
The role of CYP2E1 for mutagenesis is neither unique since multiple other CYP isoforms and detoxification mechanisms are typically involved nor should this be an independent initiator of clonal carcinogenesis.
Expert 2
05/14/2024 09:29Expert 1
05/16/2024 08:24In my view irrespective of origin oxidative stress caused by high doses of dioxane this is a threshold based MOA and BMD should apply.
Secondly if more work is to be done I suggested to use the design put forth by US EPA in the EPA Transcriptomic Assessment Product (see: https://www.epa.gov/etap): a 5 Day transcriptomic study in liver will fulfill 2 criteria - insight in to MOA to identify tipping points for key events (e.g., NrF2 transcription regulator of liver oxidative stress response) and as importantly develop a traditional threshold based RfD for use in risk assessment - EPA has "endorsed" this approach based on several studies showing that a short term (5-28 day) transcriptomic based BMD is similar to a 2 yr cancer bioassay based BMD. Combined with PBPK modeling and reverse dosimetry a margin of exposure and can be calculated that will reflect the high dose induced biological effects not relevant to levels of exposure experienced in the environment.
Oxidative stress induced cytotoxicity by high dose dioxane causing cell liver regeneration and proliferation can "fix" mutations caused by either ROS-DNA adducts or lipid peroxides as liver cells undergo necrosis/apoptosis. This MOA is threshold based.
Expert 5
05/16/2024 09:10Expert 1
05/17/2024 07:31The studies measuring ROS only had these types of data (H2AX, Cyp2e1 induction) or high dose dioxane. My bias would be to use the EPA's design for a 5 day transcriptome study that includes liver histopathology - I think this approach will provide a RfD that EPA has endorsed (this approach is ongoing for 18 PFAS compounds). I served as an ad hoc reviewer of the EPA ETAP and the data appear to be very much in line with 2 yr bioassay data.
Secondly not sure it "matters" on source of ROS or specific ROS species since there could be many and could take yrs chasing high dose artefacts.
For number 2, I am uncertain if specific xenobiotic metabolic saturation can be considered a key event. Regulatory toxicology OECD testing requires testing up to 1000 mg/kg day, so in my biased view this is likely true for all regulatory toxicology studies; OECD 408 (90 day repeat dose study states "the highest dose level should be chosen with the aim to induce toxicity but not death or severe suffering"). This is why careful dose-response study capturing dose levels used in the cancer bioassay can be used to define that shape of the dose-response curve at low doses, determine an accurate NOEL for use in risk assessment.
Given the high dose needed to elicit the biological effects of dioxane in rats and an ROS based MOA as a key event a transcriptome based BMD will likely reveal a large margin of exposure to humans.
Expert 4
05/21/2024 13:57Expert 2
05/23/2024 10:17Expert 3
05/23/2024 15:47Consideration of initial and continuing repeated dosing sufficient to overwhelm metabolic capacity represents perhaps the most important molecular initiating event in that the overall data indicate that at dose levels insufficient to overwhelm metabolic DX clearance, many if not most of the following postulated KEs critical to tumor outcomes do not occur. Such is seemingly clearly apparent with DX, i.e., tumors are consistently restricted to doses that are greater than both initial and post-induction metabolic capacities of the animals. Picking up on this consideration, international guidance on the design of animal rodent toxicity bioassays strongly and consistently encourages that a priori consideration of dose-dependent toxicokinetics should play a pivotal role in top dose selection for animal toxicology studies (and logically conversely, facilitate interpretation of quantitative relevance of rodent tumor human risk relevance when such a DX-like toxicokinetic picture is developed post-hoc to bioassay conduct). This advice rests on long-established and extensive evidence that saturation of ADME can set in motion a spectrum of potentially interactive and complex MoAs that are unique to the resulting dose-disproportionately high plasma/tissue concentrations.
For DX, evidence of metabolic saturation was observed only shortly after the earliest DX chronic bioassays, e.g., the Young et al late 1970s-era studies. Given the above contention/perspective, however, the current status of dose-dependent toxicokinetic studies could and should be supplemented to better characterize at what doses and by what human-relevant routes and modes of administration (inhalation and drinking water) likely present metabolism-saturating conditions. At present there is a particular absence of appropriately designed single- and repeat-dose inhalation toxicokinetic studies that would more clearly define the range of doses at which DX metabolism transitions to clear evidence of parent DX dose disproportionality. Such studies would ideally characterize plasma and/or liver DX and HEAA, urine HEAA and exhaled DX, and include sufficient time-course data to identify Cmax and AUC determinations. Although similar studies could considered for drinking water, the Lafranconi et al. (2021, Fig.4) drinking water toxicokinetic study is likely adequate to demonstrate that metabolic saturation is present at 2000 ppm and greater. This study's conclusions might have been more robustly supported if a differing timepoint sampling regimen had been used for each of the selected doses and sampling on study days 7, 28 and 90. Instead of collecting just a single plasma sample from each animal (possibly reflecting a Cmax value depending on when the samples were collected), recent toxicokinetic studies reported by Saghir et al. have shown that for chemicals with short parent rodent plasma half-lives (as is case for DX), sampling of animals as few as 3 times during the course of a 24 hr period while remaining on diet or drinking water treatments can reasonably provide an estimate of 24 hr AUCs of parent and possibly key metabolite(s). Conduct of such toxicokinetic refinement studies, reflecting dose/exposure conditions reflecting dose-response conditions used for DX chronic bioassays, would be (in this expert's opinion) an extremely value-added dataset in demonstrating whether or not tumors findings are confidently restricted to doses only sufficient to saturate DX metabolism (both initially and after onset of metabolic induction). Given the dose-specific tumor profile of DX, such information would substantially support a conclusion that, absent doses sufficient to saturate metabolism, subsequent postulated key events would likely be sufficiently attenuated or even absent such that tumorigenicity does not result. The existing DX MoA data highlighted by other experts certainly support this, and particularly the state-of-art toxicogenomic studies indicating that DX-induced toxicological/biological signals are restricted largely and/or only to doses saturating DX metabolism (further informed by refined toxicokinetics). Also consistent with the metabolic saturation hypothesis is the commentary of Wang et al (2022; a member of the Charkoftaki and Chen research team) in which they describe an unpublished12 week 5000 ppm drinking water study in cyp2e1 knockout mice that reduced plasma HEAA by approximately 85% and attenuated both hepatic oxidative stress and the Nrf2 response. However, they also noted "unexpected" increased in liver 8OHdG and suppression of DNA repair and postulated that such events might be due to presumed DX-parent mediated events independent of cyp2e1 induction key event. Such speculation is not implausible in that, unless an unidentified supplemental clearance mechanism beyond cyp2e1 metabolism was available in these KO mice (e.g., triggering of CAR activation), the plasma/liver concentration of DX in would reasonably have otherwise been extremely high with possibility of initiating any number of unidentified MoAs.
The point of offering this additional perspective is that if it can be more confidently shown that DX tumorigenicity hinges entirely on doses saturating DX metabolism and under dosing regimens associated with cancers, the existing DX MoA database should be viewed as complementary sufficient to support a high-dose specific MoA that is quantitatively not relevant to human risk given the wide disparity between the animal tumor-producing and metabolically saturating DX doses and those realistically encountered by any human chronic exposure scenario.
Expert 4
05/24/2024 08:11