Next Article in Journal
Analysis of Genetic Diversity and Phylogenetic Relationships of Wheat (Triticum aestivum L.) Genotypes Using Phenological, Molecular and DNA Barcoding Markers
Previous Article in Journal
Comparative Gene Enrichment Analysis for Drought Tolerance in Contrasting Maize Genotype
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Novel Insights into Potential Cannabis-Related Cancerogenesis from Recent Key Whole Epigenome Screen of Cannabis Dependence and Withdrawal: Epidemiological Commentary and Explication of Schrott et al.

by
Albert Stuart Reece
1,2,* and
Gary Kenneth Hulse
1,2
1
Division of Psychiatry, University of Western Australia, Crawley, Perth, QLD 6009, Australia
2
School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
*
Author to whom correspondence should be addressed.
Genes 2023, 14(1), 32; https://doi.org/10.3390/genes14010032
Submission received: 15 November 2022 / Revised: 14 December 2022 / Accepted: 21 December 2022 / Published: 22 December 2022
(This article belongs to the Section Genes & Environments)

Abstract

:
Whilst the cannabis-cancer link has been traditionally described as controversial recent whole nation and whole continent studies have demonstrated that well documented laboratory-based multimodal cannabinoid genotoxicity is indeed reflected in numerous cancer types in larger epidemiological series. A recent longitudinal human sperm epigenome-wide DNA methylation screen in both cannabis dependence and cannabis withdrawal has revealed remarkable insights into the manner in which widespread perturbations of DNA methylation may lead to cancerogenic changes in both the exposed and subsequent generations as a result of both cannabis exposure and withdrawal. These results therefore powerfully strengthen and further robustify the causal nature of the relationship between cannabinoid exposure and cancerous outcomes well beyond the previously published extensive mechanistic literature on cannabinoid genotoxicity. The reported epigenomic results are strongly hypothesis generating and call powerfully for further work to investigate oncogenic mechanisms in many tissues, organs and preclinical models. These epigenomic results provide an extraordinarily close predictive account for the epidemiologically observed pattern of cannabis-related malignant disease and indicate that malignant and multigenerational cannabinoid epigenotoxicity is potentially a significant and major public health concern.

1. Introduction

The epidemiology of the relationship between cannabis and cancer is often seen as confusing and controversial with both positive [1,2,3,4,5,6,7,8,9,10,11,12] and negative [6,13,14] studies being available. Earlier studies linked cannabis exposure with cancers in adults affecting the brain, head and neck, larynx, lung, prostate, testis, brain, urothelium [1,2,3,4,5,6,7,8,9,10,11] and in several of these studies dose–response relationships were demonstrated [1,3,4,7]. Risk elevation in most studies was between two- and six- fold. Several childhood cancers have also been described following parental gestational exposure to cannabis including rhabdomyosarcoma, neuroblastoma and non-lymphoblastic leukaemia [12,15,16,17,18,19] and such childhood cancers are presumed to be related to inheritable carcinogenic teratogenesis consequent on parental genotoxicity [20,21]. The literature however is controversial with some studies failing to demonstrate a link [6,13]. These studies were reviewed in 2009 by the Californian Environmental Protection Agency who found that six of eleven studies in adults at that time were positive and five of six studies in children were able to confirm a link between parental cannabis exposure and childhood cancer [22].
Provocative new epidemiological studies of community cannabis exposure demonstrate that the cannabis—testicular cancer link [7,23,24,25] has driven the 100% rise in testicular cancer 1975–2018 in USA [26] and is also involved in several common or rapidly growing cancer incidences including breast, liver, thyroid and pancreatic cancer in adults [19] and pediatric acute myeloid leukaemia [16,17,18,19] and fourteen other adult and childhood cancers in USA [27,28,29,30] and Europe [30,31]. Of even greater concern is the recent demonstration that cannabis is driving both acute lymphoid leukaemia the commonest cancer of childhood [32] and also the 50% rise in USA pediatric cancer 1975–2018 [33]. Again these pediatric data raise major issues of transgenerationally heritable teratogenic carcinogenesis [20,21].
More recent epidemiological studies of community cannabis exposure in US have now presented epidemiologically causal linkages between 25 cancers and either cannabis, Δ9-tetrahydrocannabinol (THC) or cannabidiol exposure [27,28,29]. The list of cancers identified was all cancers, acute and chronic lymphoid and myeloid leukaemias, bladder, brain, breast, colorectal, Hodgkins, Kaposi, kidney, liver, melanoma, myeloma, Non-Hodgkins lymphoma, esophagus, ovary, pancreas, prostate, stomach, testis and thyroid [27,28,29].
It is important to note that the genotoxic moiety of cannabinoids has been shown to be the olivetol nucleus on the C-ring [34,35,36] which is shared by many cannabinoids so that cannabinoid genotoxicity/epigenotoxicity is likely to be a class effect shared by numerous cannabinoids.
Moreover, exponential effects for cannabinoid genotoxicity have been well demonstrated on many occasions in the laboratory [37,38,39,40,41,42,43,44] and this finding has been subsequently confirmed epidemiologically [27,28,29].
The group of Schrott and colleagues have recently published an enormously helpful whole epigenome screening study by both whole genome bisulphite sequencing and reduced representation bisulphite sequencing performed in both rats and humans both before and after a 77-day period of documented refraining from cannabis exposure which represents one human sperm cycle [45]. The paper was useful in many ways. It carefully documented functional annotations from Ingenuity Pathway Analysis (IPA) which highlighted cellular development, cell morphology, developmental disorders and nervous system functions during cannabis dependence. Following the period of abstinence cardiovascular system, cell death and survival, nervous system development, organ morphology and organismal death were notable pathways. In both cases the investigators removed cancer-associated annotations apparently because they felt that the IPA was biased towards cancer-related pathways.
The purpose of the present report was to unearth, examine and summarize the cancer-related findings of this noteworthy study and to consider how these remarkable results might fit within the extent published literature both on cellular pathophysiological mechanisms and recent major epidemiological studies.

2. Methodology

Data. The source data from the Schrott database [45] relating to DNA methylation changes in semen has been extracted and is provided as a Supplementary File (CaEpi.txt). The genes identified are those which have previously been related to cancer by the research literature.
Analysis. Each mention of the various tumours from the Schrott data appendix was extracted. The p-values extracted from the report of Schrott and colleagues was not further processed. Data were grouped and analyzed by the mean, median, minimum and maximum values within each tumour group. These data are presented in Tables and Figures and text. The computational and analytical code in R is also provided as a Supplementary File (translated into MS Word).
The experimental conditions were considered namely overall findings, and findings related to cannabis dependence and cannabis withdrawal considered separately.
Some technical comments are appropriate. Some cancers were not mentioned and were thus unassessable. Gastroesophageal cancers were assigned to both gastric and esophageal classes as their incidence is not dissimilar (about 8 and 5/100,000 according to the Centres for Control (CDC) Surveillance, Epidemiology and End Results (SEER) dataset [46]).
A formal literature review including search terms was not conducted. Rather the focus of this study was on unearthing and explicating the truly remarkable results of Schrott and colleagues and placing them within a conceptual and theoretical position within the mechanistic framework of the published cannabinoid pathophysiological literature.
Ethical Approval. Ethical permission for this study was granted through the University of Western Australia Human Research Ethics Committee on 24 September 2021 with HREC Number 2019/RA/4/20/4724.

3. Results

The 359 pages of Supplementary Material provided along with the paper mentioned cancer 487 times, carcinoma 84 times, neoplasm 28 times, leukemia 32 times and lymphoma 20 times which confirmed that tumourigenesis was indeed a major theme of this dataset. The 176 annotations relating to the 25 tumour types recently identified [27,28,29] may then be extracted and they are shown as Supplementary Table S1 listed by tumour type and by ascending p-value.
The data may be summarized by tumour type, the number of annotations, the median and cumulative number of genes referenced and the mean and cumulative p-value (Table 1). This table is listed in order of increasing median p-values.
The finding that all 20 identifiable cancers which had been linked with cannabis in recent nationwide epidemiological studies and were identifiable in this dataset were positively identified with highly significant differential DNA methylation signals is quite remarkable.
The data may also be divided into findings in cannabis dependence and eleven weeks later in cannabis withdrawal. These comparative data are presented in Table 2. This Table lists the median p-value in dependence and the median p-value in withdrawal along with their ratio and shows that in most cases the p-value in cannabis dependence is much greater, just as the authors note. On the right hand side of this Table appears the numbers of genes annotated in dependence and withdrawal and again notes that the ratio of dependence to withdrawal generally exceeds unity again confirming that the changes of dependence exceed those of withdrawal.
Tabular findings are displayed graphically in Figure 1, Figure 2 and Figure S1. Figure 1 shows the median number of genes implicated and the applicable median p-values by cancer type in the whole dataset. Supplementary Figure S1 shows similar metrics in cannabis dependence. Figure 2 shows the ratio of the p-values and the gene numbers in cannabis dependence to withdrawal.
As highlighted in Figure 2 the exception to the generality of these observations is the acute myeloid leukaemia [16,17,18,19] where the signal is much stronger in withdrawal than dependence (median 36.0 genes to 5.5 genes; median p = 6.26 × 10−4 to 0.0017). This is an important finding as some cases of acute myeloid leukaemia occur in early childhood indicating that intergenerational mutagenesis may be at play. This further suggests that in these cases the activation of leukaemogenic gene cassettes by the cannabinoid withdrawal syndrome following birth may actually be activating development of this tumour.

4. Discussion

From the perspective of offering a detailed explanation of the diverse pattern of tumourigenesis noted in recent epidemiological studies these results are astounding. They indicate that in both cannabis dependence and cannabis withdrawal DNA methylation changes occur which may in part explain the diverse pattern of tumourigenesis observed both in USA and in Europe.
Cannabinoid genotoxicity however is a aetiopathologically complex involving multiple chromosomal toxicities [39,47,48,49,50,51,52], mitochondrial toxicities [53,54,55,56,57,58,59,60] (which underpin and support the epigenome with substrates and energy amongst other actions) [61], induction of single- and double- stranded DNA breaks [34,36,42,62], oxidation of the bases of DNA [42] and micronucleus induction [62,63,64] and has been reviewed elsewhere [19,27,28,29,45,64,65,66,67,68].
Attributing molecular causal mechanisms may therefore involve parsing out the relative importance of this complex interplay of chromosomal, metabolic, genomic and epigenomic disruptions to properly apportion the importance of the different toxicities which may vary across tissues.
It has been noted that cannabis is often co-administered with other drugs particularly tobacco [69]. Whilst this might perhaps introduce a measure of complexity in epidemiological studies it has been noted by several investigators that tobacco use has been falling in many jurisdictions worldwide whilst cannabis use has been rising [70,71,72] constituting a major trend difference which can be exploited by regression studies and other epidemiological techniques. Moreover, this particular source of confounding has been clarified by the many laboratory studies and the numerous studies in preclinical animal models referenced above.
It is important that cannabinoid genotoxicity has been repeatedly shown to have an exponential dose–response relationship in both laboratory-based metabolic and mutagenic assays [41,42,43,44,73,74,75,76] and in epidemiological field studies [19,27,28,29,66,68,77,78,79].
The findings relating to acute myeloid leukaemia (AML) are intriguing and clearly invite further investigation. Data showed that 6.5 times as many AML-related genes were triggered by cannabis withdrawal compared to cannabis dependence and the median p-value fell 2.7-fold. Since some AML cases occur in early childhood (prior to ten years of age) this may imply that the cannabis withdrawal involved in birth transgenerationally triggers early life leukaemogenesis. This hypothesis would need to be tested further experimentally. It could also be tested in other cannabis-related heritable pediatric malignancies including neuroblastoma, rhabdomyosarcoma and acute lymphoid leukaemia [32,80] but is not immediately apparent from the Schrott dataset as these results have been presented.
As study findings are broadly consistent with earlier results from this research group [65] it may be that the present results are broadly generalizable. The clear concordance between the present epigenomic and epidemiological findings provides external validation to these findings and lends further credence to their reliability. It is however important that these results be replicated by other researchers and other laboratories.
It should be emphasized however that whilst the present results are important and intruiging they do not formally demonstrate causality. The present results are strongly hypothesis generating. They do however powerfully call for further research in the laboratory and with animal models to further investigate the intruiging findings reported.
Recently a very powerful single cell RNA sequencing technique which allows the sequential transcriptomic analysis of the same cells across time has been described called “Live-Seq” [81]. Future studies could therefore be envisioned of whole animals and organoids which combine experimentally modelled cannabis dependence and withdrawal with studies of DNA methylation, histone modifications and transcriptomic output with a focus on specific organs and organoids of interest including brain, heart, testis [82] and ovary and the various major cancerogenic tissues. It could be imagined that important mechanistic insights may emerge from such studies including the identification of key genomic vulnerabilities in cannabinoid carcinogenesis and the identification of potential therapeutic targets.

4.1. Concise Oncogenic Mechanistic Considerations

Limitations of space necessarily constrain any detailed discussion of the numerous oncogenic mechanisms of cannabis which have been described in detail by previous researchers [19,26,27,28,29,30,32,33,83,84,85,86] however a tightly truncated selection of described effects might be considered as follows.
THC is known to suppress the synthesis of DNA, RNA, proteins and histones [39,40,87,88,89] thereby disrupting the key elements of genomic architecture. Cannabis induces nuclear blebs and chromosomal bridges in sperm, lymphocytes and oocytes changes which are themselves signs of nuclear aging and of major genomic—chromosomal errors [90,91,92]. Cannabis is known to disrupt the replacement of histones by protamines during condensation of the sperm nucleus which necessarily disrupts gene function globally [93,94,95]. Cannabis and several cannabinoids (including cannabidiol) have been shown to oxidize DNA bases which is known to be a potent oncogenic mechanism [42].
Cannabis (including cannabidiol) is well described as causing chromosomal and DNA breaks [62] and tests positively in the dramatic comet assay for DNA breaks (where the broken DNA forms a comet-like tail behind the main mass of DNA moving in a gel under an electrophoretic gradient) [42]. This will lead to breakage-fusion-bridge cycles which is highly oncogenic [96]. This process has been implicated in the rapidly accelerated carcinogenesis occurring in cannabis induced-testicular cancer [26,86]. Cannabis has been shown to induce ring and chain chromosome formation [62] and the formation of microchromsomes and chromosomal circles which form micronuclei which are now known to constitute the central engine of chromothripsis which is a known powerful engine for aggressive cancer formation [64,97,98,99]. Cannabis is known to induce tripolar, quadripolar and higher order mitotic spindles which are associated with disrupted polyvalent cell division [91,92]. Additionally, where a tumour suppressor is silenced or excised or a tumour promoter is aberrantly or constitutively activated by such major megabase scale genomic rearrangements, these changes are a well established pathway to oncogenesis [20,21]. Epigenomic effects can bring a gene enhancer (or superenhancer) into functional contact with a gene promoter and lead to either constitutive activation of a gene promoter or disruption of a tumour suppressor in a manner functionally analogous to that induced by gene rearrangements [20,21].
The vital contribution of RNA exosomes to sperm in the tail of the epididymis (so-called “epididymosomes”) is under endocannabinoid control and is disrupted by exogenous cannabinoids [100]. These exosomes have a critical function in gene expression in the early zygote and impact at least the initial cell divisions. In general global disruption of gene expression is frequently pro-carcinogenic [20,21].
The well described mitochondrial inhibition induced by many cannabinoids (including cannabidiol) [53,54,55,56,59,101,102] will disrupt the supply of energy to the genomic and epigenomic machinery as most reactions involved in genome maintenance are energy consuming. This will directly induce genomic instability which is a well described precursor to malignant change [20,21]. Mitochondrial metabolism also provides many of the intermediate substrates for the epigenomic machinery and signals stress to the nuclear genome via mitonuclear balance [61]. Hence, perturbation of mitochondrial respiration necessarily disrupts normal epigenomic regulation and is pro-carcinogenic. Dozens of other carcinogenic pathways have been described elsewhere [27,28,29,30,103,104].

4.2. Strengths and Limitations

There are many strengths of the present study. It is known that the investigative power of the longitudinal design is impressive for epigenomic studies. The epigenomic studies have been performed by a leading laboratory for such work in the world. They are also strongly consistent with major recent epidemiological from USA [27,28,29]. Study limitations relate mainly to the need for these results to be replicated in other laboratories.

5. Conclusions

Meanwhile the prior contribution of the Schrott group remains an intriguing and tantalizing data resource to be mined for years to come. The presently reviewed results are strongly hypothesis generating and together suggest much further labotary and preclinical model studies to further investigate the suggested links in an organ-specific manner. Clearly this is a subject which requires extensive future research. Pending such further investigations it is nevertheless prudent from the results described that the strong epigenotoxic findings relating cannabis to malignant disease be carefully considered to understand the strong epidemiological signals which have been reported in relation to cannabis, that cannabinoid genotoxicity be seriously considered as a major matter of public health importance, that the exponential dose response effects of cannabinoids be carefully taken into account, that the food chain be protected, and that appropriate attention be give to the substantial genotoxic and epigenotoxic effects of numerous cannabinoids for the present and subsequent generations. Clearly data imply the need to carefully protect populations from exposure to genotoxic and epigenotoxic cannabinoid compounds of various types. Meanwhile much further research is indicated on these intriguing and important results.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14010032/s1. Table S1: Input data extracted from source data in the Schrott et al. technical appendix (reference [43]). Input Data for Analysis (as text file.txt named “CaEpi.txt”) which is easily convertible into a.csv file in MS Excel. R code for analysis (supplied as an MS Word file but easily copied into an R Script). Named “R_Code for Cancer Epigenomics Paper”. Figure S1: (A) Median Numbers of genes annotated and (B) median p-values for each cancer type for cannabis dependence (from Table 2).

Author Contributions

A.S.R. assembled the data, designed and conducted the analyses, and wrote the first manuscript draft. G.K.H. provided technical and logistic support, co-wrote the paper, assisted with gaining ethical approval, provided advice on manuscript preparation and general guidance to study conduct. A.S.R. had the idea for the article, performed the literature search, wrote the first draft and is the guarantor for the article. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The Human Research Ethics Committee of the University of Western Australia provided ethical approval for the study to be undertaken 24 September 2021 (No. RA/4/20/4724).

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analysed during this study along with the relevant computational code in R are included in this published article and its supplementary information files.

Acknowledgments

All authors had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Conflicts of Interest

The authors declare that they have no competing interest.

References

  1. Aldington, S.; Harwood, M.; Cox, B.; Weatherall, M.; Beckert, L.; Hansell, A.; Pritchard, A.; Robinson, G.; Beasley, R. Cannabis use and risk of lung cancer: A case-control study. Eur. Respir. J. 2008, 31, 280–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Voirin, N.; Berthiller, J.; Benhaim-Luzon, V.; Boniol, M.; Straif, K.; Ayoub, W.B.; Ayed, F.B.; Sasco, A.J. Risk of lung cancer and past use of cannabis in Tunisia. J. Thorac. Oncol. 2006, 1, 577–579. [Google Scholar] [CrossRef] [PubMed]
  3. Berthiller, J.; Straif, K.; Boniol, M.; Voirin, N.; Benhaim-Luzon, V.; Ayoub, W.B.; Dari, I.; Laouamri, S.; Hamdi-Cherif, M.; Bartal, M.; et al. Cannabis smoking and risk of lung cancer in men: A pooled analysis of three studies in Maghreb. J. Thorac. Oncol. 2008, 3, 1398–1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Zhang, Z.F.; Morgenstern, H.; Spitz, M.R.; Tashkin, D.P.; Yu, G.P.; Marshall, J.R.; Hsu, T.C.; Schantz, S.P. Marijuana use and increased risk of squamous cell carcinoma of the head and neck. Cancer Epidemiol. Biomark. Prev. 1999, 8, 1071–1078. [Google Scholar]
  5. Hashibe, M.; Ford, D.E.; Zhang, Z.F. Marijuana smoking and head and neck cancer. J. Clin. Pharmacol. 2002, 42 (Suppl. S11), 103S–107S. [Google Scholar] [CrossRef] [PubMed]
  6. Sidney, S.; Quesenberry, C.P., Jr.; Friedman, G.D.; Tekawa, I.S. Marijuana use and cancer incidence (California, United States). Cancer Causes Control. 1997, 8, 722–728. [Google Scholar] [CrossRef]
  7. Daling, J.R.; Doody, D.R.; Sun, X.; Trabert, B.L.; Weiss, N.S.; Chen, C.; Biggs, M.L.; Starr, J.R.; Dey, S.K.; Schwartz, S.M. Association of marijuana use and the incidence of testicular germ cell tumors. Cancer 2009, 115, 1215–1223. [Google Scholar] [CrossRef] [Green Version]
  8. Efird, J.T.; Friedman, G.D.; Sidney, S.; Klatsky, A.; Habel, L.A.; Udaltsova, N.V.; Van den Eeden, S.; Nelson, L.M. The risk for malignant primary adult-onset glioma in a large, multiethnic, managed-care cohort: Cigarette smoking and other lifestyle behaviors. J. Neuro.-Oncol. 2004, 68, 57–69. [Google Scholar] [CrossRef]
  9. Moiche Bokobo, P.; Atxa de la Presa, M.A.; Cuesta Angulo, J. Transitional cell carcinoma in a young heavy marihuana smoker. Arch. Esp. De Urol. 2001, 54, 165–167. [Google Scholar]
  10. Chacko, J.A.; Heiner, J.G.; Siu, W.; Macy, M.; Terris, M.K. Association between marijuana use and transitional cell carcinoma. Urology 2006, 67, 100–104. [Google Scholar] [CrossRef]
  11. Nieder, A.M.; Lipke, M.C.; Madjar, S. Transitional cell carcinoma associated with marijuana: Case report and review of the literature. Urology 2006, 67, 200. [Google Scholar] [CrossRef] [PubMed]
  12. Hashibe, M.; Straif, K.; Tashkin, D.P.; Morgenstern, H.; Greenland, S.; Zhang, Z.F. Epidemiologic review of marijuana use and cancer risk. Alcohol 2005, 35, 265–275. [Google Scholar] [CrossRef] [PubMed]
  13. Hashibe, M.; Morgenstern, H.; Cui, Y.; Tashkin, D.P.; Zhang, Z.F.; Cozen, W.; Mack, T.M.; Greenland, S. Marijuana use and the risk of lung and upper aerodigestive tract cancers: Results of a population-based case-control study. Cancer Epidemiol. Biomark. Prev. 2006, 15, 1829–1834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Volkow, N.D.; Compton, W.M.; Weiss, S.R. Adverse health effects of marijuana use. N. Engl. J. Med. 2014, 371, 878–879. [Google Scholar] [CrossRef] [Green Version]
  15. Bluhm, E.C.; Daniels, J.; Pollock, B.H.; Olshan, A.F. Maternal use of recreational drugs and neuroblastoma in offspring: A report from the Children’s Oncology Group (United States). Cancer Causes Control. 2006, 17, 663–669. [Google Scholar] [CrossRef]
  16. Robison, L.L.; Buckley, J.D.; Daigle, A.E.; Wells, R.; Benjamin, D.; Arthur, D.C.; Hammond, G.D. Maternal drug use and risk of childhood nonlymphoblastic leukemia among offspring. An epidemiologic investigation implicating marijuana (a report from the Childrens Cancer Study Group). Cancer 1989, 63, 1904–1911. [Google Scholar] [CrossRef]
  17. Wen, W.Q.; Shu, X.O.; Steinbuch, M.; Severson, R.K.; Reaman, G.H.; Buckley, J.D.; Robison, L.L. Paternal military service and risk for childhood leukemia in offspring. Am. J. Epidemiol. 2000, 151, 231–240. [Google Scholar] [CrossRef] [Green Version]
  18. Trivers, K.F.; Mertens, A.C.; Ross, J.A.; Steinbuch, M.; Olshan, A.F.; Robison, L.L.; Children’s Cancer Group. Parental marijuana use and risk of childhood acute myeloid leukaemia: A report from the Children’s Cancer Group (United States and Canada). Paediatr. Perinat. Epidemiol. 2006, 20, 110–118. [Google Scholar] [CrossRef]
  19. Reece, A.S.; Hulse, G.K. Epidemiological overview of multidimensional chromosomal and genome toxicity of cannabis exposure in congenital anomalies and cancer development. Sci. Rep. 2021, 11, 13892–13912. [Google Scholar] [CrossRef]
  20. Grobner, S.N.; Worst, B.C.; Weischenfeldt, J.; Buchhalter, I.; Kleinheinz, K.; Rudneva, V.A.; Johann, P.D.; Balasubramanian, G.P.; Segura-Wang, M.; Brabetz, S.; et al. The landscape of genomic alterations across childhood cancers. Nature 2018, 555, 321–327. [Google Scholar] [CrossRef] [Green Version]
  21. Ma, X.; Liu, Y.; Liu, Y.; Alexandrov, L.B.; Edmonson, M.N.; Gawad, C.; Zhou, X.; Li, Y.; Rusch, M.C.; Easton, J.; et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature 2018, 555, 371–376. [Google Scholar] [CrossRef] [PubMed]
  22. Tomar, R.S.; Beaumont, J.; Hsieh, J.C.Y.; Reproductive and Cancer Hazard Assessment Branch, Office of Environmental Health Hazard Assessment, Californian Environmental Protection Agency. Evidence on the Carcinogenicity of Marijuana Smoke. Calif. Health Dep. 2009, 1, 1–150. [Google Scholar]
  23. Trabert, B.; Sigurdson, A.J.; Sweeney, A.M.; Strom, S.S.; McGlynn, K.A. Marijuana use and testicular germ cell tumors. Cancer 2011, 117, 848–853. [Google Scholar] [CrossRef] [PubMed]
  24. Lacson, J.C.; Carroll, J.D.; Tuazon, E.; Castelao, E.J.; Bernstein, L.; Cortessis, V.K. Population-based case-control study of recreational drug use and testis cancer risk confirms an association between marijuana use and nonseminoma risk. Cancer 2012, 118, 5374–5383. [Google Scholar] [CrossRef] [Green Version]
  25. Callaghan, R.C.; Allebeck, P.; Akre, O.; McGlynn, K.A.; Sidorchuk, A. Cannabis Use and Incidence of Testicular Cancer: A 42-Year Follow-up of Swedish Men between 1970 and 2011. Cancer Epidemiol. Biomark. Prev. 2017, 26, 1644–1652. [Google Scholar] [CrossRef] [Green Version]
  26. Reece, A.S.; Hulse, G.K. Causal inference multiple imputation investigation of the impact of cannabinoids and other substances on ethnic differentials in US testicular cancer incidence. BMC Pharm. Toxicol. 2021, 22, 40–71. [Google Scholar] [CrossRef]
  27. Reece, A.S.; Hulse, G.K. Geotemporospatial and Causal Inferential Epidemiological Overview and Survey of USA Cannabis, Cannabidiol and Cannabinoid Genotoxicity Expressed in Cancer Incidence 2003–2017: Part 1–Continuous Bivariate Analysis. Arch. Public Health 2022, 80, 99–133. [Google Scholar] [CrossRef]
  28. Reece, A.S.; Hulse, G.K. Geotemporospatial and Causal Inferential Epidemiological Overview and Survey of USA Cannabis, Cannabidiol and Cannabinoid Genotoxicity Expressed in Cancer Incidence 2003–2017: Part 2–Categorical Bivariate Analysis and Attributable Fractions. Arch. Public Health 2022, 80, 100–135. [Google Scholar] [CrossRef]
  29. Reece, A.S.; Hulse, G.K. Geotemporospatial and Causal Inferential Epidemiological Overview and Survey of USA Cannabis, Cannabidiol and Cannabinoid Genotoxicity Expressed in Cancer Incidence 2003–2017: Part 3–Spatiotemporal, Multivariable and Causal Inferential Pathfinding and Exploratory Analyses of Prostate and Ovarian Cancers. Arch. Public Health 2022, 80, 100–136. [Google Scholar]
  30. Reece, A.S.; Hulse, G.K. Cannabis Genotoxicity and Cancer Incidence: A Highly Concordant Synthesis of European and USA Datasets. In Cannabis, Cannabinoids and Endocannabinoids; Preedy., V., Patel, V., Eds.; Elsevier: London, UK, 2022; Volume 1, in press. [Google Scholar]
  31. Reece, A.S.; Hulse, G.K. Epidemiological Overview of Cannabis- and Substance-Carcinogenesis in Europe: A Lagged Causal Inferential Panel Regression Modelling and Marginal Effects Study. 2022; submitted. [Google Scholar]
  32. Reece, A.S.; Hulse, G.K. Cannabinoid exposure as a major driver of pediatric acute lymphoid Leukaemia rates across the USA: Combined geospatial, multiple imputation and causal inference study. BMC Cancer 2021, 21, 984–1017. [Google Scholar] [CrossRef]
  33. Reece, A.S.; Hulse, G.K. A geospatiotemporal and causal inference epidemiological exploration of substance and cannabinoid exposure as drivers of rising US pediatric cancer rates. BMC Cancer 2021, 21, 197–230. [Google Scholar] [CrossRef] [PubMed]
  34. Nahas, G.G. Cannabis Physiopathology Epidemiology Detection; CRC Press Revivals: Boca Raton, FL, USA, 1990; Volume 1. [Google Scholar]
  35. Nahas, G.G. Keep Off the Grass; Elsevier: Middlebury, VT, USA, 1990; Volume 1. [Google Scholar]
  36. Nahas, G.G.; Morishima, A.; Desoize, B. Effects of cannabinoids on macromolecular synthesis and replication of cultured lymphocytes. Fed. Proc. 1977, 36, 1748–1752. [Google Scholar] [PubMed]
  37. Price, P.J.; Suk, W.A.; Spahn, G.J.; Freeman, A.E. Transformation of Fischer rat embryo cells by the combined action of murine leukemia virus and (-)-trans- 9 -tetrahydrocannabinol. Proc. Soc. Exp. Biol. Med. 1972, 140, 454–456. [Google Scholar] [CrossRef] [PubMed]
  38. Busch, F.W.; Seid, D.A.; Wei, E.T. Mutagenic activity of marihuana smoke condensates. Cancer Lett. 1979, 6, 319–324. [Google Scholar] [CrossRef]
  39. Zimmerman, A.M.; Raj, A.Y. Influence of cannabinoids on somatic cells in vivo. Pharmacology 1980, 21, 277–287. [Google Scholar] [CrossRef]
  40. Tahir, S.K.; Trogadis, J.E.; Stevens, J.K.; Zimmerman, A.M. Cytoskeletal organization following cannabinoid treatment in undifferentiated and differentiated PC12 cells. Biochem. Cell Biol. 1992, 70, 1159–1173. [Google Scholar] [CrossRef]
  41. Koller, V.J.; Ferk, F.; Al-Serori, H.; Misik, M.; Nersesyan, A.; Auwarter, V.; Grummt, T.; Knasmuller, S. Genotoxic properties of representatives of alkylindazoles and aminoalkyl-indoles which are consumed as synthetic cannabinoids. Food Chem. Toxicol. 2015, 80, 130–136. [Google Scholar] [CrossRef]
  42. Russo, C.; Ferk, F.; Misik, M.; Ropek, N.; Nersesyan, A.; Mejri, D.; Holzmann, K.; Lavorgna, M.; Isidori, M.; Knasmuller, S. Low doses of widely consumed cannabinoids (cannabidiol and cannabidivarin) cause DNA damage and chromosomal aberrations in human-derived cells. Arch. Toxicol. 2018, 93, 179–188. [Google Scholar] [CrossRef] [Green Version]
  43. Fish, E.W.; Murdaugh, L.B.; Zhang, C.; Boschen, K.E.; Boa-Amponsem, O.; Mendoza-Romero, H.N.; Tarpley, M.; Chdid, L.; Mukhopadhyay, S.; Cole, G.J.; et al. Cannabinoids Exacerbate Alcohol Teratogenesis by a CB1-Hedgehog Interaction. Sci. Rep. 2019, 9, 16057–16075. [Google Scholar] [CrossRef] [Green Version]
  44. Hölzel, B.N.; Pfannkuche, K.; Allner, B.; Allner, H.T.; Hescheler, J.; Derichsweiler, D.; Hollert, H.; Schiwy, A.; Brendt, J.; Schaffeld, M.; et al. Following the adverse outcome pathway from micronucleus to cancer using H2B-eGFP transgenic healthy stem cells. Arch. Toxicol. 2020, 94, 3265–3280. [Google Scholar] [CrossRef]
  45. Schrott, R.; Murphy, S.K.; Modliszewski, J.L.; King, D.E.; Hill, B.; Itchon-Ramos, N.; Raburn, D.; Price, T.; Levin, E.D.; Vandrey, R.; et al. Refraining from use diminishes cannabis-associated epigenetic changes in human sperm. Environ. Epigenetics 2021, 7, 1–10. [Google Scholar] [CrossRef]
  46. SEER Explorer. Available online: https://seer.cancer.gov/explorer/application.html (accessed on 1 February 2022).
  47. Leuchtenberger, C.; Leuchtenberger, R. Morphological and cytochemical effects of marijuana cigarette smoke on epithelioid cells of lung explants from mice. Nature 1971, 234, 227–229. [Google Scholar] [CrossRef]
  48. Leuchtenberger, C.; Leuchtenberger, R.; Schneider, A. Effects of marijuana and tobacco smoke on human lung physiology. Nature 1973, 241, 137–139. [Google Scholar] [CrossRef] [PubMed]
  49. Stenchever, M.A.; Kunysz, T.J.; Allen, M.A. Chromosome breakage in users of marihuana. Am. J. Obs. Gynecol. 1974, 118, 106–113. [Google Scholar] [CrossRef] [PubMed]
  50. Zimmerman, A.M.; Zimmerman, S. Cytogenetic Studies of Cannabinoid Effects. In Genetic and Perinatal Effects of Abused Substances; Braude, M.C., Zimmerman, A.M., Eds.; Academic Press Inc.: New York, NY, USA, 1987; Harcourt, Brace Jovanovich; Volume 1, pp. 95–112. [Google Scholar]
  51. Zimmerman, A.M.; Zimmerman, S.; Raj, A.Y. Effects of Cannabinoids on Spermatogensis in Mice. In Marijuana and Medicine, 1st ed.; Nahas, G.G., Sutin, K.M., Harvey, D.J., Agurell, S., Eds.; Humana Press: Totowa, NJ, USA, 1999; Volume 1, pp. 347–358. [Google Scholar]
  52. Zimmerman, S.; Zimmerman, A.M. Genetic effects of marijuana. Int. J. Addict. 1990, 25, 19–33. [Google Scholar] [CrossRef] [PubMed]
  53. Mahoney, J.M.; Harris, R.A. Effect of 9-tetrahydrocannabinol on mitochondrial processes. Biochem. Pharmacol. 1972, 21, 1217–1226. [Google Scholar] [CrossRef]
  54. Bartova, A.; Birmingham, M.K. Effect of delta9-tetrahydrocannabinol on mitochondrial NADH-oxidase activity. J. Biol. Chem. 1976, 251, 5002–5006. [Google Scholar] [CrossRef]
  55. Benard, G.; Massa, F.; Puente, N.; Lourenco, J.; Bellocchio, L.; Soria-Gomez, E.; Matias, I.; Delamarre, A.; Metna-Laurent, M.; Cannich, A.; et al. Mitochondrial CB(1) receptors regulate neuronal energy metabolism. Nat. Neurosci. 2012, 15, 558–564. [Google Scholar] [CrossRef]
  56. Hebert-Chatelain, E.; Reguero, L.; Puente, N.; Lutz, B.; Chaouloff, F.; Rossignol, R.; Piazza, P.V.; Benard, G.; Grandes, P.; Marsicano, G. Cannabinoid control of brain bioenergetics: Exploring the subcellular localization of the CB1 receptor. Mol. Metab. 2014, 3, 495–504. [Google Scholar] [CrossRef]
  57. Koch, M.; Varela, L.; Kim, J.G.; Kim, J.D.; Hernandez-Nuno, F.; Simonds, S.E.; Castorena, C.M.; Vianna, C.R.; Elmquist, J.K.; Morozov, Y.M.; et al. Hypothalamic POMC neurons promote cannabinoid-induced feeding. Nature 2015, 519, 45–50. [Google Scholar] [CrossRef] [Green Version]
  58. Wolff, V.; Schlagowski, A.I.; Rouyer, O.; Charles, A.L.; Singh, F.; Auger, C.; Schini-Kerth, V.; Marescaux, C.; Raul, J.S.; Zoll, J.; et al. Tetrahydrocannabinol induces brain mitochondrial respiratory chain dysfunction and increases oxidative stress: A potential mechanism involved in cannabis-related stroke. Biomed. Res. Int. 2015, 2015, 323706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Hebert-Chatelain, E.; Desprez, T.; Serrat, R.; Bellocchio, L.; Soria-Gomez, E.; Busquets-Garcia, A.; Pagano Zottola, A.C.; Delamarre, A.; Cannich, A.; Vincent, P.; et al. A cannabinoid link between mitochondria and memory. Nature 2016, 539, 555–559. [Google Scholar] [CrossRef] [PubMed]
  60. Jimenez-Blasco, D.; Busquets-Garcia, A.; Hebert-Chatelain, E.; Serrat, R.; Vicente-Gutierrez, C.; Ioannidou, C.; Gómez-Sotres, P.; Lopez-Fabuel, I.; Resch-Beusher, M.; Resel, E.; et al. Glucose metabolism links astroglial mitochondria to cannabinoid effects. Nature 2020, 583, 603–608. [Google Scholar] [CrossRef] [PubMed]
  61. Canto, C.; Menzies, K.J.; Auwerx, J. NAD(+) Metabolism and the Control of Energy Homeostasis: A Balancing Act between Mitochondria and the Nucleus. Cell Metab. 2015, 22, 31–53. [Google Scholar] [CrossRef]
  62. Huang, H.F.S.; Nahas, G.G.; Hembree, W.C. Effects of Marijuana Inhalation on Spermatogenesis of the Rat. In Marijuana in Medicine; Nahas, G.G., Sutin, K.M., Harvey, D.J., Agurell, S., Eds.; Humana Press: Totowa, NJ, USA, 1999; Volume 1, pp. 359–366. [Google Scholar]
  63. Hall, W.; Degenhardt, L. Adverse health effects of non-medical cannabis use. Lancet 2009, 374, 1383–1391. [Google Scholar] [CrossRef]
  64. Reece, A.S.; Hulse, G.K. Chromothripsis and epigenomics complete causality criteria for cannabis- and addiction-connected carcinogenicity, congenital toxicity and heritable genotoxicity. Mutat. Res. 2016, 789, 15–25. [Google Scholar] [CrossRef]
  65. Murphy, S.K.; Itchon-Ramos, N.; Visco, Z.; Huang, Z.; Grenier, C.; Schrott, R.; Acharya, K.; Boudreau, M.H.; Price, T.M.; Raburn, D.J.; et al. Cannabinoid exposure and altered DNA methylation in rat and human sperm. Epigenetics 2018, 13, 1208–1221. [Google Scholar] [CrossRef] [Green Version]
  66. Reece, A.S.; Hulse, G.K. Geotemporospatial and Causal Inference Epidemiological Analysis of US Survey and Overview of Cannabis, Cannabidiol and Cannabinoid Genotoxicity in Relation to Congenital Anomalies 2001–2015. BMC Pediatr. 2022, 22, 47–124. [Google Scholar] [CrossRef]
  67. Reece, A.S.; Hulse, G.K. Canadian Cannabis Consumption and Patterns of Congenital Anomalies: An Ecological Geospatial Analysis. J. Addict. Med. 2020, 14, e195–e210. [Google Scholar] [CrossRef]
  68. Reece, A.S.; Hulse, G.K. Epidemiological Association of Cannabinoid- and Drug- Exposures and Sociodemographic Factors with Limb Reduction Defects Across USA 1989-2016: A Geotemporospatial Study. Spat. Spatiotemporal Epidemiol. 2022, 41, 100480–100490. [Google Scholar] [CrossRef]
  69. Schauer, G.L.; Rosenberry, Z.R.; Peters, E.N. Marijuana and tobacco co-administration in blunts, spliffs, and mulled cigarettes: A systematic literature review. Addict. Behav. 2017, 64, 200–211. [Google Scholar] [CrossRef] [PubMed]
  70. Key Substance Use and Mental Health Indicators in the United States: Results from the 2020 National Survey on Drug Use and Health (NSDUH). Available online: https://www.samhsa.gov/data/sites/default/files/reports/rpt35325/NSDUHFFRPDFWHTMLFiles2020/2020NSDUHFFR1PDFW102121.pdf (accessed on 1 February 2022).
  71. Reece, A.S.; Hulse, G.K. Cannabis Teratology Explains Current Patterns of Coloradan Congenital Defects: The Contribution of Increased Cannabinoid Exposure to Rising Teratological Trends. Clin. Pediatr. 2019, 58, 1085–1123. [Google Scholar] [CrossRef] [PubMed]
  72. Reece, A.S.; Hulse, G.K. Broad Spectrum epidemiological contribution of cannabis and other substances to the teratological profile of northern New South Wales: Geospatial and causal inference analysis. BMC Pharm. Toxicol. 2020, 21, 75–103. [Google Scholar] [CrossRef] [PubMed]
  73. Shoyama, Y.; Sugawa, C.; Tanaka, H.; Morimoto, S. Cannabinoids act as necrosis-inducing factors in Cannabis sativa. Plant Signal. Behav. 2008, 3, 1111–1112. [Google Scholar] [CrossRef] [PubMed]
  74. Fisar, Z.; Singh, N.; Hroudova, J. Cannabinoid-induced changes in respiration of brain mitochondria. Toxicol. Lett. 2014, 231, 62–71. [Google Scholar] [CrossRef] [PubMed]
  75. Koller, V.J.; Auwarter, V.; Grummt, T.; Moosmann, B.; Misik, M.; Knasmuller, S. Investigation of the in vitro toxicological properties of the synthetic cannabimimetic drug CP-47,497-C8. Toxicol. Appl. Pharmacol. 2014, 277, 164–171. [Google Scholar] [CrossRef] [PubMed]
  76. Singh, N.; Hroudova, J.; Fisar, Z. Cannabinoid-Induced Changes in the Activity of Electron Transport Chain Complexes of Brain Mitochondria. J. Mol. Neurosci. 2015, 56, 926–931. [Google Scholar] [CrossRef]
  77. Reece, A.S.; Hulse, G.K. Cannabis in Pregnancy–Rejoinder, Exposition and Cautionary Tales. Psychiatr. Times 2020, 37. Available online: https://www.psychiatrictimes.com/view/cannabis-pregnancy-rejoinder-exposition-cautionary-tales (accessed on 20 December 2022).
  78. Reece, A.S.; Hulse, G.K. Contemporary epidemiology of rising atrial septal defect trends across USA 1991-2016: A combined ecological geospatiotemporal and causal inferential study. BMC Pediatr. 2020, 20, 539–550. [Google Scholar] [CrossRef]
  79. Reece, A.S.; Hulse, G.K. Cannabinoid- and Substance-Relationships of European Congenital Anomaly Patterns: A Space-Time Panel Regression and Causal Inferential Study. Environ. Epigenet. 2022, 8, 1–40. [Google Scholar] [CrossRef]
  80. Reece, A.S. Chronic toxicology of cannabis. Clin. Toxicol. 2009, 47, 517–524. [Google Scholar] [CrossRef] [PubMed]
  81. Chen, W.; Guillaume-Gentil, O.; Rainer, P.Y.; Gäbelein, C.G.; Saelens, W.; Gardeux, V.; Klaeger, A.; Dainese, R.; Zachara, M.; Zambelli, T.; et al. Live-seq enables temporal transcriptomic recording of single cells. Nature 2022, 608, 733–740. [Google Scholar] [CrossRef] [PubMed]
  82. Schrott, R.; Modliszewski, J.L.; Hawkey, A.B.; Grenier, C.; Holloway, Z.; Evans, J.; Pippen, E.; Corcoran, D.L.; Levin, E.D.; Murphy, S.K. Sperm DNA methylation alterations from cannabis extract exposure are evident in offspring. Epigenetics Chromatin 2022, 15, 33. [Google Scholar] [CrossRef] [PubMed]
  83. Reece, A.S. Rapid Response: Cannabinoid Genotoxic Trifecta-Cancerogenesis, Clinical Teratogenesis and Cellular Ageing. Br. Med. J. 2022, 376, n3114. [Google Scholar]
  84. Reece, A.S.; Hulse, G.K. Geospatiotemporal and Causal Inference Study of Cannabis and Other Drugs as Risk Factors for Female Breast Cancer USA 2003-2017. Environ. Epigenetics 2022, 2022, 1–22. [Google Scholar] [CrossRef]
  85. Reece, A.S.; Hulse, G.K. Epigenomic Basis and Transgenerational Impacts of Cannabinoid Genotoxicity Manifesting as Cannabinoid -Teratogenesis, -Cancerogenesis and -Aging Acceleration. Int. J. Environ. Res. Public Health, 2022; submitted. [Google Scholar]
  86. Reece, A.S.; Hulse, G.K. State Trends of Cannabis Liberalization as a Causal Driver of Increasing Testicular Cancer Rates across the USA. Int. J. Environ. Res. Public Health 2022, 19, 12759–12796. [Google Scholar] [CrossRef]
  87. McClean, D.K.; Zimmerman, A.M. Action of delta 9-tetrahydrocannabinol on cell division and macromolecular synthesis in division-synchronized protozoa. Pharmacology 1976, 14, 307–321. [Google Scholar] [CrossRef]
  88. Tahir, S.K.; Zimmerman, A.M. Influence of marihuana on cellular structures and biochemical activities. Pharmacol. Biochem. Behav. 1991, 40, 617–623. [Google Scholar] [CrossRef]
  89. Parker, S.J.; Zuckerman, B.S.; Zimmermann, A.M. The Effects of Maternal Marijuana Use During Pregnancy on Fetal Growth. In Marijuana in Medicine; Nahas, G.G., Sutin, K.M., Harvey, D.J., Agurell, S., Eds.; Humana Press: Totowa, NJ, USA, 1999; Volume 1, pp. 461–468. [Google Scholar]
  90. Morishima, A.; Henrich, R.T.; Jayaraman, J.; Nahas, G.G. Hypoploid metaphases in cultured lymphocytes of marihuana smokers. Adv. Biosci. 1978, 22–23, 371–376. [Google Scholar]
  91. Henrich, R.T.; Nogawa, T.; Morishima, A. In vitro induction of segregational errors of chromosomes by natural cannabinoids in normal human lymphocytes. Environ. Mutagen. 1980, 2, 139–147. [Google Scholar] [CrossRef]
  92. Morishima, A. Effects of cannabis and natural cannabinoids on chromosomes and ova. NIDA Res. Monogr. 1984, 44, 25–45. [Google Scholar] [PubMed]
  93. Chioccarelli, T.; Cacciola, G.; Altucci, L.; Lewis, S.E.; Simon, L.; Ricci, G.; Ledent, C.; Meccariello, R.; Fasano, S.; Pierantoni, R.; et al. Cannabinoid receptor 1 influences chromatin remodeling in mouse spermatids by affecting content of transition protein 2 mRNA and histone displacement. Endocrinology 2010, 151, 5017–5029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Rossato, M.; Pagano, C.; Vettor, R. The cannabinoid system and male reproductive functions. J. Neuroendocrinol. 2008, 20 (Suppl. S1), 90–93. [Google Scholar] [CrossRef] [PubMed]
  95. Rossato, M.; Ion Popa, F.; Ferigo, M.; Clari, G.; Foresta, C. Human sperm express cannabinoid receptor Cb1, the activation of which inhibits motility, acrosome reaction, and mitochondrial function. J. Clin. Endocrinol. Metab. 2005, 90, 984–991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. McClintock, B. The Stability of Broken Ends of Chromosomes in Zea Mays. Genetics 1941, 26, 234–282. [Google Scholar] [CrossRef]
  97. Zhang, C.Z.; Spektor, A.; Cornils, H.; Francis, J.M.; Jackson, E.K.; Liu, S.; Meyerson, M.; Pellman, D. Chromothripsis from DNA damage in micronuclei. Nature 2015, 522, 179–184. [Google Scholar] [CrossRef] [Green Version]
  98. Kuznetsova, A.Y.; Seget, K.; Moeller, G.K.; de Pagter, M.S.; de Roos, J.A.; Durrbaum, M.; Kuffer, C.; Muller, S.; Zaman, G.J.; Kloosterman, W.P.; et al. Chromosomal instability, tolerance of mitotic errors and multidrug resistance are promoted by tetraploidization in human cells. Cell Cycle 2015, 14, 2810–2820. [Google Scholar] [CrossRef]
  99. Kloosterman, W.P.; Hoogstraat, M.; Paling, O.; Tavakoli-Yaraki, M.; Renkens, I.; Vermaat, J.S.; van Roosmalen, M.J.; van Lieshout, S.; Nijman, I.J.; Roessingh, W.; et al. Chromothripsis is a common mechanism driving genomic rearrangements in primary and metastatic colorectal cancer. Genome Biol. 2011, 12, R103. [Google Scholar] [CrossRef] [Green Version]
  100. Mobisson, S.K.; Ikpi, D.E.; Wopara, I.; Obembe, A.O. Cannabis sativa exacerbate testicular function by increased oxidative stress, altered male reproductive hormones, sperm quality/quantity and cellular architecture of the testis. Andrologia 2022, 54, e14492. [Google Scholar] [CrossRef]
  101. Drummond-Main, C.D.; Ahn, Y.; Kesler, M.; Gavrilovici, C.; Kim, D.Y.; Kiroski, I.; Baglot, S.L.; Chen, A.; Sharkey, K.A.; Hill, M.N.; et al. Cannabidiol Impairs Brain Mitochondrial Metabolism and Neuronal Integrity. Cannabis Cannabinoid Res. 2022. [Google Scholar] [CrossRef]
  102. Rupprecht, A.; Theisen, U.; Wendt, F.; Frank, M.; Hinz, B. The Combination of Δ(9)-Tetrahydrocannabinol and Cannabidiol Suppresses Mitochondrial Respiration of Human Glioblastoma Cells via Downregulation of Specific Respiratory Chain Proteins. Cancers 2022, 14, 3129. [Google Scholar] [CrossRef] [PubMed]
  103. Reece, A.S.; Hulse, G.K. Geospatiotemporal and Causal Inferential Epidemiological Survey and Exploration of Cannabinoid- and Substance- Related Carcinogenesis in USA 2003–2017. In Epidemiology of Cannabis: Genotoxicity and Neurotoxicity, Epigenomics and Aging; Elsevier: New York, NY, USA, 2023; Volume 1, p. 2500, in press. [Google Scholar]
  104. Reece, A.S.; Hulse, G.K. Chapter 5: Multivalent Cannabinoid Epigenotoxicities and Multigenerational Aging. In Epidemiology of Cannabis: Genotoxicity and Neurotoxicity, Epigenomics and Aging; Elsevier: New York, NY, USA, 2023; Volume 1, p. 2500, in press. [Google Scholar]
Figure 1. (A) Median Numbers of genes annotated and (B) median p-values for each cancer type for all results overall (from Table 1). Data relate to differential DNA methylation data from sperm for genes previously linked with cancer. The experimental condition considered in this Figure is the overall data (cannabis dependence and cannabis withdrawal considered together). All data from the Schrott data appendix [45].
Figure 1. (A) Median Numbers of genes annotated and (B) median p-values for each cancer type for all results overall (from Table 1). Data relate to differential DNA methylation data from sperm for genes previously linked with cancer. The experimental condition considered in this Figure is the overall data (cannabis dependence and cannabis withdrawal considered together). All data from the Schrott data appendix [45].
Genes 14 00032 g001
Figure 2. Ratio of (A) median numbers of genes annotated and (B) median p-values in cannabis dependence to withdrawal (from Table 2). Data relate to differential DNA methylation data from sperm for genes previously linked with cancer. The experimental condition considered in this Figure is the ratio of the data in cannabis dependence to that in cannabis withdrawal. All data from the Schrott data appendix [45].
Figure 2. Ratio of (A) median numbers of genes annotated and (B) median p-values in cannabis dependence to withdrawal (from Table 2). Data relate to differential DNA methylation data from sperm for genes previously linked with cancer. The experimental condition considered in this Figure is the ratio of the data in cannabis dependence to that in cannabis withdrawal. All data from the Schrott data appendix [45].
Genes 14 00032 g002
Table 1. Overall Significance Levels and Gene Numbers Grouped by Cancer Type.
Table 1. Overall Significance Levels and Gene Numbers Grouped by Cancer Type.
No.CancerNumber AnnotationsMedian p-ValueMedian Gene NumberCumulative Gene NumberCumulative p-ValueMean p-Value
1Thyroid31.26 × 10−173187520.16222.16 × 10−4
2Testis31.60 × 10−81513640.04051.11 × 10−4
3Stomach59.77 × 10−81035450.20393.74 × 10−4
4Liver52.52 × 10−71798900.00202.24 × 10−6
5Oropharynx31.82 × 10−6972398.94E-043.74 × 10−6
6Melanoma43.86 × 10−61988040.00809.94 × 10−6
7Pancreas124.65 × 10−583.58810.70678.02 × 10−4
8Brain284.74 × 10−59837260.27777.45 × 10−5
9Esophagus76.80 × 10−5645820.27554.73 × 10−4
10Urinary102.69 × 10−4185.518700.44572.38 × 10−4
11Prostate75.33 × 10−4595570.38776.96 × 10−4
12Colorectal185.34 × 10−47621860.82143.76 × 10−4
13Ovary75.62 × 10−4715300.26695.04 × 10−4
14NHL157.77 × 10−4203650.30478.35 × 10−4
15AML38.96 × 10−49470.04750.0010
16Breast100.0011818510.77659.12 × 10−4
17Kidney20.001944.5890.17730.0020
18CML80.00262220.05850.0027
19ALL140.00284.51410.25070.0018
20Myeloma30.00624100.03980.0040
Table key: The experimental condition considered in this Table is the overall results (cannabis dependence and withdrawal combined). Cancers were grouped by cancer type. The table lists the various classes of gene numbers and p-values as described in the column headings. All data is taken from results reported from the epigenomic data appendix in the study of Schrott and colleagues as referenced [45].
Table 2. Significance Levels and Gene Numbers Grouped by Cancer Type and Cannabis Dependence or Withdrawal Status.
Table 2. Significance Levels and Gene Numbers Grouped by Cancer Type and Cannabis Dependence or Withdrawal Status.
Cancerp-ValuesGene Numbers
Median p-Value DependenceMedian p-Value Withdrawalp-Value RatioMedian Gene Number DependenceMedian Gene Number WithdrawalGene No. Ratio Dependence/Withdrawal
Thyroid1.24 × 10−170.00141.14 × 1014318.5115.02.77
Melanoma1.36 × 10−143.49 × 10−52.56 × 109289.5112.52.57
Stomach1.53 × 10−120.00271.74 × 109169.051.03.31
Colorectal7.38 × 10−118.67 × 10−41.17 × 107197.072.02.74
Urinary1.12 × 10−103.94 × 10−43.53 × 106268.0111.52.40
Testis1.37 × 10−86.75 × 10−44.93 × 104152.060.02.53
Esophagus4.89 × 10−80.00204.14 × 104136.038.03.58
Liver2.52 × 10−7-179.0--
Prostate8.39 × 10−70.00364.24 × 103128.049.02.61
Oropharynx9.73 × 10−71.60 × 10−516.4597.544.02.22
Brain5.82 × 10−61.60 × 10−427.38179.082.02.18
Pancreas1.65 × 10−50.0061368.3992.519.54.74
NHL2.08 × 10−40.003617.3335.04.57.78
ALL2.23 × 10−40.003415.0611.54.52.56
Breast3.45 × 10−40.004011.51127.042.52.99
Ovary4.10 × 10−40.007017.1272.51.072.50
AML0.00176.26 × 10−40.365.536.00.15
CML0.00180.00311.744.02.02.00
Kidney0.0019--44.5--
Myeloma-0.0062--4.0-
Table key: This Table considers the experimental conditions of cannabis dependence and cannabis withdrawal separately. Cancers were grouped by cancer type. The table lists the various classes of gene numbers and p-values and their ratios as described in the column headings. All data is taken from results reported from the epigenomic data appendix in the study of Schrott and colleagues as referenced [45].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Reece, A.S.; Hulse, G.K. Novel Insights into Potential Cannabis-Related Cancerogenesis from Recent Key Whole Epigenome Screen of Cannabis Dependence and Withdrawal: Epidemiological Commentary and Explication of Schrott et al. Genes 2023, 14, 32. https://doi.org/10.3390/genes14010032

AMA Style

Reece AS, Hulse GK. Novel Insights into Potential Cannabis-Related Cancerogenesis from Recent Key Whole Epigenome Screen of Cannabis Dependence and Withdrawal: Epidemiological Commentary and Explication of Schrott et al. Genes. 2023; 14(1):32. https://doi.org/10.3390/genes14010032

Chicago/Turabian Style

Reece, Albert Stuart, and Gary Kenneth Hulse. 2023. "Novel Insights into Potential Cannabis-Related Cancerogenesis from Recent Key Whole Epigenome Screen of Cannabis Dependence and Withdrawal: Epidemiological Commentary and Explication of Schrott et al." Genes 14, no. 1: 32. https://doi.org/10.3390/genes14010032

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop