Open access

Synergistic Interaction Between Aflatoxin and Hepatitis B Virus in Hepatocarcinogenesis

Written By

Michael C. Kew

Submitted: 13 March 2012 Published: 23 January 2013

DOI: 10.5772/51396

From the Edited Volume

Aflatoxins - Recent Advances and Future Prospects

Edited by Mehdi Razzaghi-Abyaneh

Chapter metrics overview

2,302 Chapter Downloads

View Full Metrics

1. Introduction

Based on the number of new cases of cancer in humans each year, hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide [20], the fifth in males and the seventh in females [37]. In the most recently reported year, 748,000 new cases of the tumour were recorded, constituting 9.2% of all new cancers [20]. Furthermore, the number of new cases of the tumour continues to increase year by year. Not only is HCC common, it also carries an especially grave prognosis, ranking third in annual cancer mortality rates. In the year mentioned, the total death rate from the tumour was 695,900. Of the patients who died, 93% did so within 12 months of the onset of symptoms. This 12 month fatality ratio in HCC (0.93 - 0.96) is the highest of any human tumour.

HCC does not have a uniform geographical distribution. Rather, of all the new cases of the cancer recorded during recent years, approximately 84% occurred in resource-constrained (developing) countries [20], particularly in sub-Saharan Africa and the Asia Pacific region. In these regions the dominant cause of HCC is chronic hepatitis B virus (HBV) infection. This infection is almost invariably acquired very early in life, either as a result of perinatal transmission of the virus or of horizontal transmission in infancy or early childhood, times at which the infection very often becomes chronic [42]. The tumour resulting from the HBV infection frequently occurs at a young or relatively young age, and it carries a particularly grave prognosis.

In addition to chronic HBV infection, the other major cause of HCC in these high-risk regions is dietary exposure to aflatoxins, the toxic secondary metabolites of the fungi, Aspergillus flavus and Aspergillus parasiticus. These viral and fungal risk factors are largely responsible for the striking geographical variation in incidence of HCC. Both aflatoxin exposure and chronic HBV infection are more common in rural than in urban dwellers in resource-constrained regions [42, 63]. In these regions, the association between aflatoxin exposure and the development of HCC is closest in sub-Saharan Africa [51].

Aflatoxins are structurally-related difuranocoumarin derivatives, some of which are mutagenic and carcinogenic in humans and animals [89, 87]. These toxins are widely distributed in nature. Because atmospheric humidity and moisture content of plants are important factors in determining growth of, and toxin production by, these moulds, contamination of crops occurs mainly in tropical and sub-tropical climates with high humidity and temperature. These conditions exist in sub-Saharan Africa, the Asia Pacific region, and parts of South America. Contamination is particularly likely to occur in subsistence farming communities in regions with these climates and where regulations to control exposure to the fungi are either non-existent or unenforceable in practice.

In these regions, the moulds contaminate a variety of staple foods, especially maize and groundnuts [89, 34, 87]. Because most rural dwellers can afford only limited food variation, these staples make up a significant portion of their diets. Contamination of crops with aflatoxins occurs either during their growth or as a result of their storage under conditions that promote fungal growth and toxin production [31, 32, 33]. Exposure begins in utero as a result of trans-placental transmission of the toxins [86] and in the postnatal period as a result of breast-feeding [85], and continues throughout life. Exposure increases with increasing age - for example, in Malaysia evidence of exposure was more common in the population aged 31 to 50 years than that aged 18 to 30 years [46].

Approximately 4.5 billion of the world’s population are believed to be exposed to aflatoxins [88]. Between 25,200 new cases of HCC each year (or 4.6% of all cases of the tumour worldwide) and 155,000 new cases each year (or 28.2% of all cases of the tumour world wide) may be attributed to this exposure [51]. It has been estimated that aflatoxins play a causative role in at least 4.6% and at most 28.2% of all cases of HCC worldwide [51]. These large ranges stem from the considerable uncertainty and variability in data on cancer potency factors, HBV prevalence, aflatoxin exposure, and other risk factors [51].

Although the parent aflatoxin molecule is harmless, it is converted by members of the cytochrome 450 superfamily into electrophilic intermediates that are mutagenic and carcinogenic [90, 87, 40, 71]. Of the four naturally occurring aflatoxins, aflatoxin B1 (AFB1), B2, G1 and G2, toxigenic strains of A. flavus typically produce only aflatoxins B1 and B2, whereas most strains of A. parasiticus produce all of the aflatoxins [18]. AFB1 is the most potent experimental hepatocarcinogen known to man -- no animal model exposed to the toxin thus far has failed to develop HCC. AFM1, the hydroxylation product of AFB1, is found in milk and milk products when animals intended for dairy production consume aflatoxin-contaminated feed [70]. In rodents exposed to AFB1 in equivalent doses to those occurring in humans, levels of aflatoxin adduct in the serum have correlated with levels of hepatic DNA damage and with development of HCC [83].

AFB1 is the aflatoxin most often found in contaminated human foodstuffs [78], and exposure to AFB1 is causally related to the development of HCC in humans [34]. The correlation between the degree of exposure to AFB1 and the incidence of HCC is direct. It has been estimated that by reducing dietary AFB1 levels to below detectable limits in Asia and sub-Saharan Africa, between 72,800 and 98,800 new cases of HCC could be prevented each year [49].

The major human cytochrome P450 (CYP) enzymes involved in aflatoxin metabolism are CYP3A4, 3A5, 3A7 and 1A2, and the predominant site of metabolism is the liver [87, 39]. AFB1 is metabolized to an AFB1-8,9-exo-epoxide and, to a lesser extent, an AFB1-8,9-endo-epoxide. The exo-epoxide binds to DNA to form the predominant promutagenic 8,9-dihydro-8-(N7-guanyl)-9-hydroxy AFB1 (AFB1-N7-Gua) adduct. AFB1-N7-Gua can result in two secondary forms, an apurinic site and a more stable ring-opened AFB1-formamidopyrimidine (AFB1-FABY) adduct, which is far more persistent in vivo. This adduct causes G to T transversion mutations [28, 4, 87], the most prevalent of which are targeted to the site of the original adduct. AFB1-FABY exists as a mixture of two rotameric forms. In Escherichia coli AFB1-FABY induced a six-fold higher G to T mutation frequency than AFB1-N7-Gua, with mutations also occurring adjacent to the site of adduct formation [82]. AFB1-FABY also resulted in blocked replication. Subsequent studies showed that the form of AFB1-FABY normally present in double-stranded DNA is mutagenic, whereas the dominant species in single-stranded DNA blocks replication [5].

Chronic liver injury and regenerative hyperplasia are critical to the development of HCC [30]. AFB1-induced DNA adducts may therefore be fixed as mutations consequent to an HBV-related increase in cell proliferation and hyperplasia. Inflammation and oxidative stress associated with chronic active hepatitis and aflatoxin exposure may also result directly in DNA damage and mutations [52].

The ‘DNA damage checkpoint response’ acts as an anti-tumour mechanism against genotoxic agents. By playing a central role in co-ordinating DNA repair and cell cycle progression, ‘DNA damage checkpoint response’ proteins play a key role in preventing mutations [66]. Genotoxic doses of AFB1 induce an incomplete and inefficient ‘DNA damage checkpoint response’, which may contribute to the carcinogenic properties of the toxin [27].

AFB1 has a geographical distribution similar to that of chronic HBV infection, colonizing a variety of foodstuffs in the same Far Eastern and sub-Saharan African countries. Accordingly, a synergistic interaction between the hepatocarcinogenic effects of HBV and AFB1 would offer a plausible explanation for the very high incidence of HCC, and perhaps also the young age of the patients, in these regions.

Advertisement

2. Evidence for a synergistic hepatocarcinogenic interaction between aflatoxins and hepatitis B virus

Although a study in Guanxi, China published in the mid-1980s showed that HCC occurring in individuals infected with HBV who lived in villages with a “high” consumption of aflatoxins had a mortality rate that was 10 times higher than that in individuals living in villages with a “low” consumption [93], other early studies of the consequences of exposure to aflatoxins did not include data on the HBV status of the populations studied. All of these studies showed a statistically significant increase in incidence of HCC in those individuals who were exposed to the fungal toxin [31, 53, 32]. But, given the high frequency of chronic HBV infection in this region at that time, the probability exists that at least some, and possibly a significant number, of the subjects included in these early studies were also chronically infected with HBV and that the virus, rather than the exposure to aflatoxins, could have caused the malignant transformation or, at least, contributed to it. In two studies, one earlier and the other later, the roles of AFB1 and HBV in explaining the varying frequencies of HCC in different areas of Swaziland in southern Africa [61] and in Guanxi Province in China [93] were assessed. Both analyses concluded that with simultaneous exposure to the two potential carcinogenic agents, AFB1 exposure was the more important determinant of geographical variation in the incidence of HCC than was HBV infection, at least in those regions. However, no attempt was made in either study to evaluate a possible interactive hepatocarcinogenic effect between the two risk factors.

The first published evidence consistent with synergism between AFB1 and HBV in the genesis of HCC was provided by experiments in which transgenic mice over-expressing the large envelope polypeptide of HBV were fed AFB1. These mice produced more rapid and extensive hepatocyte dysplasia than did their unexposed littermates, and HCCs developed [67]. Shortly thereafter, further experimental evidence for a positive interaction between AFB1 and another member of the Hepadnaviridae family, the woodchuck hepatitis virus, in the development of HCC was presented [3]. Woodchucks infected with woodchuck hepatitis virus and exposed to AFB1 developed, after six to 26 months of exposure, a high incidence of preneoplastic foci of altered hepatocytes followed by hepatocellular adenomas and HCCs. Moreover, woodchucks infected with woodchuck hepatitis virus had earlier been shown to have enhanced activation of the biologically inactive AFB1 to AFB1-8,9-epoxide [17]. The development of liver tumours was also reported in ducks infected with duck hepatitis virus and exposed to AFB1 [14] and in tree shrews (Tupaia belangeri chinensis) infected with HBV and exposed to AFB1 [48].

Following the introduction of methods to measure aflatoxin metabolites and aflatoxin-DNA adducts in urine and aflatoxin-albumin adducts in serum, biomarkers that were a far more accurate and reliable indicator of AFB1 exposure than the hitherto used food sampling and dietary questionnaires, five large cohort studies were undertaken in Shanghai and Qidong county, China and in Taiwan to assess the carcinogenic effects of AFB1 and HBV alone and in combination. In four of the studies an hepatocarcinogenic effect of AFB1 alone was shown, with increased odds ratios ranging from 1.9 to 32.0 with a mean ratio of 13.7 [65, 64, 54, 59] (Table 1). These studies proved that exposure to AFB1 alone could cause malignant transformation of hepatocytes in humans. The fifth study failed to show an increased odds ratio of AFB1 exposure alone [79].

As expected, these studies (including the one that did not show an increased odds ratio for the development of HCC for AFB1 alone [79]) confirmed an hepatocarcinogenic effect of HBV alone - odds ratios ranged from 3.3 to 17.4, with a mean ratio of 10.0 (Table 1).

HBV alone AFB1 alone HBV and AFB1
RR (95% CL)* RR (95% CL) RR (5% CL)
[65] 4.8 (1.2 -19.7) 1.9 (0.5 - 7.5) 60.1 (6.4 - 561.8)
[64] 7.3 (2.2 - 24.4) 3.4 (1.1 - 10.0) 59.4 (15.6 - 212)
[80] 17.4 (3.6 – 143.4)
425.4)
0.3 (0 - 3.6)
70.0 (11.5 –
[54] 17.0 (2.8 - 103.9) 17.4 (3.4 - 90.3) 67.6 (12.2 - 373.2)
[59] 3.3 (1.3 - 8.3) 32.0 (4.0 - 255.8) 40.7 (12.7- 130.9)

Table 1.

Findings in five studies comparing the risk of HBV infection alone, dietary exposure to AFB1 alone, and the two risk factors together in the genesis of HCC. * Relative risk (95% confidence limits).

A synergistic interaction between AFB1 exposure and chronic HBV infection in causing HCC was evident in each of the five studies - odds ratios ranged from 40.7 to 70.0 with a mean of 59.6 (Table 1). In three of these studies there was a striking multiplicative effect, and in the other two a sub-multiplicative effect between exposure to AFB1 alone and exposure to AFB1 in the presence of chronic HBV infection in inducing HCC, in comparison with each carcinogen alone. The study which did not show an increased odds ratio with AFB1 alone had the highest odds ratios for both HBV infection alone and for co-existing AFB1 exposure and HBV infection [79]. The finding in this study that exposure to AFB1 alone did not increase the risk of HCC development [79] could conceivably be the source of the erroneous view held by some hepatologists and oncologists that AFB1 alone does not cause HCC and is important only as a co-carcinogen with HBV.

In other investigations, also in countries with high rates of contamination of foodstuffs by AFB1, only individuals chronically infected with HBV were studied and the influence of AFB1 exposure in further increasing their risk of HCC development was analysed. In Qidong county, China, over a 10-year prospective follow-up period, the risk of HCC in male carriers of the virus was shown to be increased 3-fold (95% confidence limits 1.2, 8.7) in those with detectable urinary levels of AFB1 metabolites in comparison with those without these metabolites [72]. This result was later confirmed in a longer follow-up of the same cohort of HBV carriers, when the risk of HCC was increased 3.5-fold (95% confidence limits 1.5, 8.1) [57]. A dose-response relationship between urinary AFB1 metabolites and the risk of HCC was shown in HBV carriers in Taiwan [94]. Comparing high and low urinary levels of the aflatoxin metabolite, AFM1, a multivariate-adjusted odds ratio of 6.0 (95% confidence limits1.2, 29) was calculated. The risk was greater (odds ratio 10.0: 95% confidence limits 1.6 ; 60.9) when both AFM1 and AFB1-N7- gua metabolites were tested for and detected in the urine. In another study performed in chronic carriers of HBV in the same country, a statistically significant relationship was noted between detectable levels of AFB1 adducts in serum and the risk of HCC, with an age-adjusted odds ratio of 2.0 (95% confidence limits 1.1, 3.7) [73]. A recent meta-analysis has shown that the population attributable risk of developing HCC in individuals exposed to dietary aflatoxins is 17%, with the risk being 21% in those individuals also chronically infected with HBV [49]. Individuals infected with HBV alone have a population attributable risk of 8.8% [49]. If the one study in the meta-analysis that contributed most to heterogeneity in the analysis was excluded, the summarised odds ratio of HCC (with 95% confidence limits) was 73 (36 to 148.3) for the combined effects of AFB1 and HBV, 11.3 (6.75 to 18.9) for HBV alone, and 6.37 (3.74 to 10.86) for AFB1 alone [49]. The effect of a synergistic interaction between AFB1 and HBV on the age of onset of HCC was specifically addressed in a study of Taiwanese patients. HBV-infected patients in whom tumour tissue was shown by histochemical staining to be positive for AFB1-N7-gua adducts were on average 10 years younger than those with adduct-negative tumours [10].

Although they have had limitations, various animal models with natural hepatitis viral infections have been used to examine the interaction between hepadnaviruses and AFB1 [84]. In woodchucks and tree shrews, animal species with hepadnaviral-induced liver pathology similar to that observed in HBV-infected humans, the administration of AFB1 resulted in a higher incidence of liver tumours than in infected animals not receiving AFB1 [92, 3]. Moreover, HBsAg transgenic mice over-expressing the large envelope protein of HBsAg in the liver developed HCC when exposed to aflatoxin, whereas their littermates not exposed to carcinogens did not [67, 47].

In those human populations in which an interaction between the fungal toxin and HBV has been described, the infection is predominantly acquired in infancy or early childhood. During the early years of HBV infection, a state of immune tolerance towards the virus exists and little if any cellular damage occurs. With loss of this tolerance, the ongoing infection results in recurring cell damage. Exposure to AFB1 in contaminated foodstuffs also occurs in young children [81].

Nevertheless, it is likely, certainly in China and Taiwan, where perinatal transmission of HBV is the predominant mode of infection, and also probably in Africa, where slightly later horizontal infection is the major route of infection, that the HBV carrier state is established, not before exposure to, but before heavy exposure to the toxin.

Advertisement

3. Possible mechanisms of interaction between AFB1 and HBV in hepatocarcinogenesis

A number of possible mechanisms for the interaction between HBV and AFB1 in causing HCC have been suggested. One is that HBV infection directly or indirectly sensitizes hepatocytes to the carcinogenic effects of AFB1. One way in which this may be accomplished is that the specific cytochrome P450s that metabolize AFB1 to AFB1-8,9-epoxide may be induced either by chronic hepatitis caused by HBV infection or by the presence of the virus itself. Induction of these phase I enzymes has been described in HBV transgenic mice [21, 6], where this effect appeared to result from hepatocyte injury induced by the virus rather than the presence of the virus per se [6]. The observation that Gambian and Taiwanese children and adolescents chronically infected with HBV have higher concentrations of AFB1 adducts than uninfected individuals [2, 76, 11] is consistent with this mechanism. But studies in adults in China, Taiwan and The Gambia have either failed to show a significant difference in serum AFB1 adduct levels between HBsAg-positive and -negative subjects [23, 79, 12] or showed only a marginally significant difference [74]. Moreover, a study in woodchucks with chronic woodchuck hepatitis virus infection did not show enhanced activation of AFB1 [75, 44].

The generated aflatoxin-8,9-epoxide has been shown to bind to proteins, causing acute toxicity, or to DNA inducing changes that over time increase the risk of malignant transformation [26]. DNA damage can also increase the chance of integration of the viral DNA into the host genome [16]. This effect could be exerted directly by AFB1 or indirectly by oxidative stress induced by chronic viral hepatitis.

A guanine to thymine transversion at the third base of codon 249 of the p53 tumour suppressor gene (arginine to serine substitution; 249ser, R249S) is present in between 40 and 66% of HCC patients in regions with heavy dietary exposure to AFB1 [28, 4, 45, 30]. The mutation is also detectable in circulating cell-free DNA from the plasma of HCC patients and healthy subjects from these regions [77]. The exact timing of the development of the 249ser mutation remains uncertain, although it has been shown to be an early event. The mutation abrogates the normal functions of p53, including those in cell cycle control, DNA repair, and apoptosis, thereby contributing to the multistep process of hepatocarcinogenesis. This mutation is extremely uncommon in tumors other than HCC [58].

A specific and close association between this inactivating mutation, the presence of AFB1 biomarkers, and the development of HCC was recognised in epidemiological studies in regions with high or low AFB1 exposure rates [4, 60, 15, 19, 45, 30, 62, 22], and evidence that the mutation induced chromosomal instability was found [62]. Arising from the observation of the co-existence of the p53 mutation and AFB1 exposure, the presence of the 249ser mutation was believed to be a primary genetic event in hepatocarcinogenesis. It occurs early in the series of events leading to AFB1-associated HCC, and may thus provide an early biomarker of exposure to the fungal toxin and AFB1-induced hepatocarcinogenesis [36].

But the findings have been inconsistent with support for an aetiological association being provided by some but not all studies. In an investigation of Taiwanese patients with HCC the mutation was present in 36.3% of HBV-infected patients with HCC, compared with 11.7% of those without HBV markers [79]. In a second analysis in Taiwan, all of the 249ser mutations occurred in patients positive for HBsAg, giving an odds ratio of 10.0 (95% confidence limits 1.6; 17.5) [54]. In a study in The Gambia patients positive for HBsAg alone had an increased relative risk of 10, those with 249ser mutation alone of13, and those with both an estimated risk of 399 [45]. Other studies, however, showed a similar, but not a statistically significant trend [68, 19], and in yet other analyses from a variety of countries no association could be found [listed in reference: 69]. Furthermore, in a meta-analysis of 49 published studies using a method that takes into account both within-study and study-to-study variability, little evidence for HBV-AFB1 interaction in modulating the 249ser mutation was found [69]. In addition, the absence of the 249ser mutation from the serum of patients from countries with a low incidence of HBV-induced HCC to date suggests that chronic HBV infection alone is insufficient to result in the development of the 249ser mutation [82].

Another suggested possibility is that the activity of phase II detoxification enzymes (glutathione S transferase (GST) and epoxide hydrolase(EPHX)) may play a role in the genesis of HCC induced jointly by AFB1 and HBV [94; 73; 56]. A multiplicative interaction in the genesis of HCC in West African and Chinese patients was demonstrated between HBV infection and mutations of EPHX [56]: patients with chronic HBV infection but with normal EPHX alleles were at a 15-fold increase in risk, and those with both HBV infection and at least one EPHX mutant were at a 77-fold increased risk. In further studies in these patients a positive interaction between HBV and AFB1 seemed to depend on the presence of a polymorphism of the GST M1, GST T1, and EPHX genes that are normally responsible for converting the carcinogenic AFB1 -8,9-epoxide to non-reactive metabolites [56, 8, 94, 73]. But again no consistent pattern has emerged. In one analysis in Taiwan the risk of HCC formation was greater in HBV carriers who had the GST M1 null genotype compared with the non-null genotype [94], in a second study the risk appeared to depend on the presence of a GST T1 null genotype [73], and in a third the risk was considerably greater in those with null genotypes of both GST M1 and GST T1 [8].

Another possible mechanism for a carcinogenic interaction between AFB1 and HBV is that increased hepatocyte necrosis and proliferation cause by chronic HBV infection increases the likelihood of both AFB1 mutations, including 249ser, and the subsequent clonal expansion of cells containing these mutations [13]. Chronic necroinflammatory hepatic disease, including that resulting from HBV infection, results in the generation of oxygen and nitrogen reactive species [50, 35]. Both of the latter are mutagenic, but, in addition, increased oxidative stress has been shown to induce 249ser mutations [29].

The HBV x gene is frequently included in sequences of the virus that are integrated into cellular DNA [43]. AFB1-DNA adducts are normally repaired by the nucleotide excision repair pathway. The HBV X protein interferes with the nuclear excision repair pathway [38; 43] and might, by this means, favour persistence of existing mutations or impaired DNA. DNA repair is also compromised by the rapid cell turnover rate in chronic hepatitis. In the presence of dietary exposure to AFB1, the HB X protein may contribute to the uncontrolled cell proliferation in other ways. The transcription of p21 waf1/ cip1, which induces cell cycle arrest at the G1-S checkpoint, is activated by HB X protein in a dose-dependent manner in the presence of functional p53. This transcription is, however, repressed by HB X protein when p53 is not functional or is functional at a low level [1]. The expression of HB X protein also correlates with an increase in the overall frequency of DNA mutations in transgenic mice and a 2-fold increase the incidence of the 249ser mutation in transgenic mice exposed to AFB1 [55].

Altered methylation of genes may play a role in hepatocarcinogenesis [43]. For example, the methylation status of the human ras association domain gene (RASSF1A) and the P16 gene has been incriminated in the pathogenesis of HCC [95]. No association was found between methylation status and P53 status {95]. A statistically significant association was, however, found between RASSF1A methylation status and the level of AFB1-DNA adducts in HCC tissues [95].

An understanding of the mechanisms responsible for the heightened risk of malignant transformation in patients chronically infected with HBV and exposed to AFB1 is far from complete, and there is clearly a need for further research to be undertaken into the pathogenetic mechanisms involved in this interaction between the two common hepatocarcinogens in resource-constrained geographical regions.

References

  1. 1. Ahn J. I. Jung E. Y. Kwun H. J. et al. 2002 Dual effects of hepatitis B virus X protein on the regulation of cell cycle depending on the status of cellular p53 J Gen Virol 83 2765 2772
  2. 2. Allen S. J. Wild C. P. Wheeler J. G. et al. 1992 Aflatoxin exposure and hepatitis B infection in rural Gambian children. Trans Roy Soc Trop Med Hyg 86 426 430
  3. 3. Bannasch P. Khoshkhou N. I. Hacker H. J. et al. 1995 Synergistic hepatocarcinogenic effect of hepadnaviral infection and dietary aflatoxin B1 in woodchucks. Cancer Res 55 3318 3330
  4. 4. Bressac B. Kew M. C. Wands J. R. et al. 1991 Selective G to T mutations of p53 gene in hepatocellular carcinoma from southern Africa. Nature Lond 350 429 430
  5. 5. Brown K. L. Deng J. Z. Iver R. S. et al. 2006 Unravelling the aflatoxin-FABY conundrum: structural basis for differential replicative processing of isomeric forms of the formaminopyrimidine DNA adduct of aflatoxin B1. J Am Chem Soc 128 15188 15199
  6. 6. Chemin I. Ohgaki H. Chisari F. V. et al. 1999 Altered expression of hepatic carcinogen metabolizing enzymes with liver injury in HBV transgenic mice lineages expressing various amounts of hepatitis B surface antigen. Liver 19 81 87
  7. 7. Chen C. J. Liang K. Y. Chang A. S. et al. 1991 Effect of hepatitis B virus, alcohol drinking, cigarette smoking and familial tendency on hepatocellular carcinoma. Hepatology 13 398 406
  8. 8. Chen C. J. Wang L. Y. Lu S. N. et al. 1996 Elevated aflatoxin exposure and increased risk of hepatocellular carcinoma. Hepatology 24 38 42
  9. 9. Chen C. J. Yu M. W. Liaw Y. F. et al. 1996 Chronic hepatitis B carriers with null genotypes of glutathione S-tranferase M1 and T1 polymorphisms who are exposed to aflatoxin are at increased risk of hepatocellular carcinoma. Am J Hum Genet 59 128 134
  10. 10. Chen C. J. Zhang Y. J. Lu S. N. et al. 1992 Aflatoxin B1 adducts in smeared liver tissue of hepatocellular carcinoma patients. Hepatology 16 1150 1155
  11. 11. Chen S. Y. Chen C. J. Chou S. R. et al. 2001 Association of afltoxin B1-albumin adduct levels with hepatitis B surface antigen status among adolescents in Taiwan. Cancer Epidemiol Biomarkers Prev 10 1223 1226
  12. 12. Chen S. Y. Chen C. J. Tsai W. Y. et al. 2000 Associations of plasma aflatoxin B1-albumin adduct level and genetic polymorphisms of glutathione-S- transferase MI and T1. Nutr Cancer 38 179 185
  13. 13. Chisari F. V. Klopchin K. Moriyama T. et al. 1989 Molecular pathogenesis of hepatocellular carcinoma in hepatitis B transgenic mice. Cell 59 1145 1156
  14. 14. Cova L. Wild C. P. Mehrotta V. et al. 1990 Contribution of aflatoxin B1 and hepatitis B virus infection in the induction of liver tumors in ducks. Cancer Res 50 2156 2163
  15. 15. Coursaget P. Depril N. Chabaud M. et al. 1993 High prevalence of mutations at codon 249 of the p53 gene in hepatocellular carcinoma in Senegal. Br J Cancer 67 1395 1397
  16. 16. Dandri M. Burda M. R. Burkle A. et al. 2002 Increase in de novo HBV DNA integrations in response to oxidative DNA damage or inhibition of poly(ADP-ribosyl)ation. Hepatology 35 217 223
  17. 17. De Flora S. Hietane E. Bartsch H. et al. 1989 Enhanced metabolic activation of chemical hepatocarcinogenesis in woodchucks infected with woodchuck hepatitis virus. Carcinogenesis 10 1099 1106
  18. 18. Dorner J. W. 2004 Biological control of crops. J Toxicol Toxin Rev 23 445 450
  19. 19. Eaton D. L. Gallagher E. P. 1994 Mechanisms of aflatoxin carcinogenesis. Annu Rev Pharmacol Toxicol 34 275 301
  20. 20. Ferlay J Shin H. R. Bray F et al. 2010 Establishment of world-wide burden of cancer in 2008. GLOBOCAN 2008. Int J Cancer 127 2893 2917
  21. 21. Gemechu-Hatewu M. Platt K. L. Oesch F. et al. 1997 Metabolic activation of aflatoxin B1 to aflatoxin B1-8,9-epoxide in woodchucks undergoing chronic active hepatitis. Int J Cancer 73 587 591
  22. 22. Gouas D. Shi H. Hainaup P. 2009 The aflatoxin-induced TP53 muation at codon 249(R249S): biomarker of exposure, early detection and target for therapy. Cancer Lett 286 29 37
  23. 23. Groopman J. D. Hall A. J. Whittle W. H. et al. 1992 Molecular dosimetry of aflatoxin-N7 guanine in human urine obtained in The Gambia. Cancer Epidemiol Biomarkers Prev 1 221 227
  24. 24. Groopman J. D. Kensler T. W. Wild C. P. 2008 Protective interventions to prevent aflatoxin-induced carcinogenesis in developing countries. Annu Rev Public Health 28 187 203
  25. 25. Groopman J. D. Wang B. J. Scholl P. 1996 Molecular biomarkers of aflatoxins: from adducts to gene mutations to human liver cancer. Can J Physiol Pharmacol 74 203 209
  26. 26. Groopman J. D. Scholl P. Wang J. S. et al. 1996 Epidemiology of human aflatoxin exposures and their relationship to liver cancer. Progr Clin Biol Res 395 211 222
  27. 27. Gursoy-Yuzugullu O. Yuzullu H. Yilmaz M. et al. 2011 Aflatoxin genotoxicity is associated with a defective DNA damage response by bypassing p53 activation. Liv Internat 31 561 571
  28. 28. Hsu I. C. Metcalf R. A. Sun T. et al. 1991 Mutational hotspot in the p53 gene in human hepatocellular carinomas. Nature (Lond) 350 427 428
  29. 29. Hussain S. P. Aquilar F Amstad P et al. Oncogene Oxyradical-induced mutagenesis of hotspot codons 248 and 249 of the human p53 gene. 1994 9 2277 2281
  30. 30. Hussain S. P. Schwank S. J. Staib F et al. 2007 TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene 26 2166 2176
  31. 31. IARC. 1987 Overall evaluations of Carcinogenicity: An Updating of IARC Monographs 1-42. IARC Press, Lyon.
  32. 32. IARC. 1993 Some naturally occurring substances: Food Items and Constituents. Heterocyclic Aromatic Amines and Mycotoxins. IARC Press, Lyon.
  33. 33. IARC 2002 Monographs on the evaluation of Carcinogenic Risks to Humans. International Agency for Research on Cancer: Lyon France 82 59
  34. 34. IARC. 2002 Some Traditional Herbal Medicines, Some Mycotoxins, Naphthalene and Styrene. IARC Press Lyon
  35. 35. Ishima H. Bartsch H. 1994 Chronic infection and inflammatory process as cancer risk factors: possible role for nitric oxide in carcinogenesis. Mutation Res 305 253 264
  36. 36. Jackson P. E. Kuang S. Y. Wang G. B. et al. 2003 Prospective detection of codon 249 mutations in plasma of hepatocellular carcinoma patients Carcinogenesis 24 1657 1663
  37. 37. Jemal A. Bray F. Center MM et al. 2011 Global cancer statistics. CA Cancer J Clin 61 69 90
  38. 38. Jia L. Wang X. W. Harris C. C. 1999 Hepatitis B virus x protein inhibits nucleotide excision repair. Int J Cancer 80 875 879
  39. 39. Kamdem L. K. Meineke I. Godtel-Armbrust U. et al. 2006 Dominant contribution of P450 3A4 to the hepatic carcinogenic activation of aflatoxin B1. Chem Res Toxicol 19 577 586
  40. 40. Kensler T. W. Qian G. S. Chen J. G. et al. 2003 Translational strategies for cancer prevention in liver. Nat Rev Cancer 3 321 329
  41. 41. Kew M. C. 2007 Hepatocellular carcinoma in Africa. In: About Cancer in Africa. Paris: National Cancer Institute of France (INCa) 221 241
  42. 42. Kew M. C. 2010 Epidemiology of hepatitis B virus infection, hepatocellular carcinoma and hepatitis B virus-induced hepatocellular carcinoma. Pathologie Biologie (Paris) 58 273 277
  43. 43. Kew M. C. 2011 Hepatitis B virus x gene in the pathogenesis of hepatitis B virus-induced hepatocellular carcinoma. J Gastroenterol Hepatol 1 144 152
  44. 44. Kirby G. Chemin I. Montesana R. et al. 1994 Induction of specific cytochrome p450 sinvolved in aflatoxin metabolism in hepatitis B transgenic mice. Mol Carcinogenesis 11 74 80
  45. 45. Kirk G. D. Lesi O. A. Mendy M. et al. 2005 249 (Ser) TP53 mutation in plasma DNA, hepatitis B virus infection, and risk of hepatocellular carcinoma. Oncogene 24 5858 5867
  46. 46. Leong Y. H. Rosma M. A. Latiff A. A. et al. 2012 Association of serum aflatoxin B1 adduct level with socio-demographic factors in a study in Malaysia and aflatoxins intake from nuts and nut products in Malaysia. Int J Hyg Environ Health 215 368 372
  47. 47. Lian M. Liu Y. Yu S. Z. et al. 2006 Hepatitis B virus x gene and cyanobacterial toxins promote hepato-tumorigenesis in mice. World J Gastroenterol 12 3065 3072
  48. 48. Li Y. Su J. Qin C. et al. 1999 Synergistic effect of hepatitis B virus and aflatoxin B1 in hepatocarcinogenesis in tree shrews. Ann Acad Med Singapore 28 68 71
  49. 49. Liu Y. Chang C. C. Marsh G. M. et al. 2012 Population attributable risk of aflatoxin-related liver cancer: a systematic review and meta-analysis. Eur J Cancer Mar 8th. Epub ahead of print.
  50. 50. Liu R. H. Jacob J. R. Hotchkiss J. H. et al. 1994 Woodchuck hepatitis virus surface antigen induces nitric oxide synthesis in hepatocytes: possible role in hepatocarcinogenesis. Carcinogenesis 15 2875 2877
  51. 51. Liu Y. Wu F. 2010 Global burden of aflatoxin-induced hepatocellular carcinoma: a risk assessment Environ Health Perspect 118 818 824
  52. 52. Liu Z. M. Li L. Q. Peng M. H. et al. 2008 Hepatitis B virus infection contributes to oxidative stress in a population exposed to aflatoxin B1 and high-risk for hepatocellular carcinoma. Cancer Lett 263 745 749
  53. 53. Liu S. N. Lin T. M. Chen C. J. et al. 1988 A case-control study of primary hepatocellular carcinoma. Cancer 62 2051 2055
  54. 54. Lunn R. M. Zhang Y. J. Wang L. Y. et al. 1997 p53 mutations, chronic hepatitis B virus infection, and aflatoxin exposure in hepatocellular carcinoma in Taiwan. Cancer Res 57 3471 3477
  55. 55. Madden C. R. Finegold M. J. Slagel B. L. 2002 Altered DNA mutation spectrum in aflatoxin treated transgenic mice that express the hepatitis B virus X protein. J Virol 76 11770 11774
  56. 56. McGlynn K. A. Rosvold E. A. Lustbader E. D. et al. 1995 Susceptibility to hepatocellular carcinoma is associated with genetic variation in the enzymatic detoxification of aflatoxin B1. Proc Natl Acad Sci 92 2384 2387
  57. 57. Ming L. Thorgeirsson S. Gail M. H. et al. 2002 Dominant role of hepatitis B virus and co-factor role of aflatoxin in hepatocarcinogenesis in Qidong, China. Hepatology 36 1214 1220
  58. 58. Montesano R Hainaut P Wild C. P. 1997 Hepatocellular carcinoma; from gene to public health. J Natl Cancer Inst 89 1844 1851
  59. 59. Omer R. E. Kuijsten A. Kadaru M. Y. K. et al. 2004 Population attributable risk of dietary aflatoxins and hepatitis B virus infection with respect to hepatocellular carcinoma. Nutr Cancer 48 15 21
  60. 60. Ozturk M. Bressac B. Pusieux A. et al. 1991 A p53 mutational hotspot in primary liver cancer is geographically localised to high aflatoxin areas of the world. Lancet 338 260 265
  61. 61. Peers F. G. Gilman G. A. Linsell C. A. 1976 Dietary aflatoxins and human liver cancer. A study in Swaziland. Int J Cancer 1976 17 167
  62. 62. Pineau P. Marchio A. Battiston C. et al. 2008 Chromosome instability in human hepatocellular carcinoma depends upon p53 status and aflatoxin exposure. Mutat Res 653 6 13
  63. 63. Plymoth A. Vivani S. Hainaut P. 2008 Control of hepatocellular carcinoma through hepatitis B vaccination in areas of high endomicity: perspectives for global liver cancer prevention. Cancer Lett 286 15 21
  64. 64. Qian G. S. Ross R. K. Yu M. C. et al. 1994 A follow-up study of urinary markers of aflatoxin exposure and liver cancer risk in Shanghai, People’s republic of China. Epidemiol Biomarkers Prev 3 3 10
  65. 65. Ross R. K. Yuan J. M. Yu M. C. et al. 1992 Urinary aflatoxin biomarkers and risk of hepatocellular carcinoma. Lancet 339 943 946
  66. 66. Sancar A. Lindsey-Boltz L. A. Unsal-Kacmaz K. et al. 2004 Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu Rev Biochem 73 39 85
  67. 67. Sell S. Hunt J. M. Dunsford H. A. et al. 1991 Synergy between hepatitis B virus expression and chemical hepatocarcinogenesis in transgenic mice. Cancer Res 511 1278 1285
  68. 68. Sheu J. C. Huang G. T. Lee P. H. et al. 1992 Mutations of p53 in hepatocellular carcinoma in Taiwan. Cancer Res 52 6089 6100
  69. 69. Stern M. C. Umbach D. M. Yu M. C. et al. 2001 Hepatitis B, aflatoxin B1 p53 codon 249 mutation in hepatocellular carcinoma from Guanxi, People’s Republic of China, and a meta-analysis of existing studies. Cancer Epidemiol Biomarkers Prev 10 617 625
  70. 70. Strosnider H Baumgartner-Azziz F Banzinger M et al. 2006 Workgroup report: Public health strategies for reducing aflatoxin exposure in developing countries. Environ Health Perspect 114 1898 1903
  71. 71. Sudaikin D. I. 2003 Dietary aflatoxin exposure and chemoprevention of cancer: a clinical review. J Toxicol Clin Toxicol 41 195 204
  72. 72. Sun Z. Lu P. Gail M. H. et al. 1999 Increased risk of hepatocellular carcinoma in male hepatitis B surface antigen carriers with chronic hepatitis who have detectable uruinary aflatoxin metabolite M1. Hepatology 30 379 383
  73. 73. Sun CA Wang LY Chen CJ et al. 2001 Genetic polymorphisms of glutathione S-transferases M1 and T1 associated with susceptibility to aflatoxin-related hepatocarcinogenesis among chronic hepatitis B carriers: a nested case-control study in Taiwan. Carcinogenesis 22 1289 1294
  74. 74. Sun C. A. Wu D. M. Wang L. Y. et al. 2002 Determinants of formation of aflatoxin-albumin : a seven township study in Taiwan. Br J Cancer 87 966 970
  75. 75. Tennant B. Horbuckle W. E. Yeager A. E. et al. 1990 Effects of aflatoxin B1 on experimental woodchuck hepatitis virus infection and hepatocellular carcinomas. In: Hollinger FB, Lemon SM, Margolis H, editors. Viral Hepatitis and Liver Disease. Baltimore Williams and Wilkens 599 600
  76. 76. Turner P. C. Mendy M. Whittle H. et al. 2000 Hepatitis B infection and aflatoxin levels in Gambian children. Trop Med Int Health 5 837 841
  77. 77. Villar S. Le Roux-Goglin E. Gouas D. A. et al. 2011 Seasonal variation in TP53 R249S-mutated serum DNA with aflatoxina exposure and hepatitis B virus infection. Environ Health Perspect 119 1635 1640
  78. 78. Wang J. S. Groopman J. D. 1999 DNA damage by mycotoxins. Mutat Res 424 167 181
  79. 79. Wang L. Y. Hatch M. Chen C. J. et al. 1996 Aflatoxin exposure and risk of hepatocellular carcinoma. Int J Cancer 67 620 630
  80. 80. Wang J. S. Qian G. S. Zarba A. et al. 1996 Temporal patterns of aflatoxin-albumin adducts in hepatitis B surface antigen positive and negative residents of Davin, Qidong County, People’s Republic of China. Cancer Epidemiol Biomarkers Prev 5 253 261
  81. 81. Wild C. P. Fortuin M. Donato F. et al. 1993 Aflatoxin, liver enzymes and hepatitis B virus infection in Gambian children. Cancer Epidemiol Biomarkers Prev 2 555 561
  82. 82. Wild C. P. Gong Y. Y. 2010 Mycotoxins and human disease: a largely ignored global health issue Carcinogenesis 31 71 82
  83. 83. Wild C. P. Hasegawa R. Barraud L. et al. 1996 Aflatoxin-albumin adducts: a basis for comparative carcinogenesis between animals and humans. Cancer Epidemiol Biomarkers Prev 5 179 189
  84. 84. Wild C. P. Montesano R. 2009 A model of interaction: aflatoxin B1 and hepatitis viruses in liver cancer aetiology. Cancer Lett 286 22 28
  85. 85. Wild C. P. Pionneau F. A. Montesano R. et al. 1987 Aflatoxin detected in human breast milk by radioimmunoassay. Int J Cancer 40 328 333
  86. 86. Wild C. P. Shrestha S. M. Anwar W. A. et al. 1992 Field studies of aflatoxin exposure, metabolism and induction of genetic alterations in relation to HBV infection and hepatocellular carcinomas in The Gambia and Thailand Toxicol Lett 455 461
  87. 87. Wild C. P. Turner P. C. 2002 The toxicity of aflatoxins as a basis for public health decisions. Mutagenesis 17 471 481
  88. 88. Williams J. H. Phillips T. D. Jolly P. E. et al. 2004 Human aflatoxicosis in developing countries: review of toxicology, exposure, potential health consequences, and interventions. Am J Clin Clin Nutr 80 10106 1022
  89. 89. Wogan G. N. 1997 Aflatoxin exposure as a risk factor in the etiology of hepatocellular carcinoma. In:Liver Cancer. Okuda K, Tabor E (eds). New York Churchill Livingstone S 1 S8
  90. 90. Wogan G. N. 2000 Impact of chemicals on liver cancer risk. Semin Cancer Biol 10 201 210
  91. 91. Wu H. C. Wang Q. Yang H. I. et al. 2009 Aflatoxin B1 exposure, hepatitis B virus infection and hepatocellular carcinoma in Taiwan. Cancer Epidemiol Biomarkers Prev. 18 846 853
  92. 92. Yan R. Q. 1989 A study of primary liver cancer in tree shrews induced by human hepatitis B virus and aflatoxin B1. Zhongua Bing Li Xue Za Zhi 18 19 22
  93. 93. Yeh F. S. Mo C. C. Yen R. C. et al. 1989 Risk factors for hepatocellular carcinoma in Guanxi, People’s republic of China. Nati Cancer Inst Monog 69 47 48
  94. 94. Yu M. W. Lien J. P. Chiu Y. H. et al. 1997 Effect of aflatoxin metabolism and DNA adduct formation on hepatocellular carcinoma among chronic hepatitis B carriers in Taiwan. J Hepatol 27 320 330
  95. 95. Zhang Y. J. Ahsan H. Chen Y. et al. 2002 High frequency of promoter hypermethylation of RASSFIA and p16 and its relationship to aflatoxin B1-DNA adducts in human hepatocellular carcinoma. Mol Carcinog 35 85 92

Written By

Michael C. Kew

Submitted: 13 March 2012 Published: 23 January 2013